yingweiwo

Linrodostat mesylate

Alias: Linrodostat mesylate; BMS-986205; BMS 986205; BMS986205 mesylate; ONO-7701; ONO 7701 mesylate; ONO7701; F-001287; F001287; F001287; 2221034-29-1; OS7OBU191R; Linrodostat mesylate [USAN]; Linrodostat [USAN]; Linrodostat
Cat No.:V24189 Purity: ≥98%
Linrodostat mesylate (formerly known as BMS-986205 mesylate, ONO-7701mesylate and F001287 mesylate) is a novel, potent, orally bioactive and selective inhibitor of IDO (indoleamine 2,3-dioxygenase 1) with potential anticancer activities.
Linrodostat mesylate
Linrodostat mesylate Chemical Structure CAS No.: 2221034-29-1
Product category: New1
This product is for research use only, not for human use. We do not sell to patients.
Size Price
500mg
1g
Other Sizes

Other Forms of Linrodostat mesylate:

  • Linrodostat (BMS-986205)
Official Supplier of:
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Top Publications Citing lnvivochem Products
Product Description

Linrodostat mesylate (formerly known as BMS-986205 mesylate, ONO-7701 mesylate and F001287 mesylate) is a novel, potent, orally bioactive and selective inhibitor of IDO (indoleamine 2,3-dioxygenase 1) with potential anticancer activities. It is licensed by Flexus Inc. to Bristol-Myers Squibb for clinical development. This compound appears to act as an irreversible inhibitor of high potency (~2 nmol/L) with superior pharmacokinetics relative to epacadostat and navoximod. In 2015, BMS-986205 entered a phase I/II study in melanoma patients as monotherapy and in combination with nivolumab (NCT02658890). An interim report noted the compound was well tolerated in patients. A second phase I study of the same combination is being conducted in various other types of advanced cancer patients (NCT03192943).

Biological Activity I Assay Protocols (From Reference)
Targets
Indoleamine 2,3-dioxygenase 1 (IDO1) (IC50 = 1.1 nM)
ln Vitro
When SKOV-3 and Jurkat clone E6-1 cells are treated with linrodostat (0.01-100 μM) for 72 hours, the proportion of viable cells is lower than when the control group is left untreated. With an IC50 of 6.3 μM, linrodostat likewise causes cell death at substantially lower concentrations[1].
In support of clinical testing, BMS-986205 was evaluated preclinically. In vitro characteristics included potent inhibition of kynurenine (kyn) production in IDO1-HEK293 cells (IC50 = 1.1 nM) but not in TDO-HEK293 cells, sustained inhibition in IDO1 cell-based assays after washout, and single-digit nM potency in human tolerogenic MLR assays. [2]
Linrodostat exhibited potent cellular activity, suppressing kynurenine production in HEK293 cells overexpressing human IDO1 and HeLa cells stimulated with IFNγ, with no activity against tryptophan 2,3-dioxygenase or murine indoleamine 2,3-dioxygenase 2 detected. Linrodostat restored T-cell proliferation in a mixed-lymphocyte reaction of T cells and allogeneic IDO1-expressing dendritic cells [3].

In vitro activity: BMS-986205 is a novel, potent, orally bioactive and selective inhibitor of IDO (indoleamine 2,3-dioxygenase 1) with potential anticancer activities. It is licensed by Flexus Inc. to Bristol-Myers Squibb for clinical development. This compound appears to act as an irreversible inhibitor of high potency (~2 nmol/L) with superior pharmacokinetics relative to epacadostat and navoximod. In 2015, BMS-986205 entered a phase I/II study in melanoma patients as monotherapy and in combination with nivolumab (NCT02658890). An interim report noted the compound was well tolerated in patients. A second phase I study of the same combination is being conducted in various other types of advanced cancer patients (NCT03192943).


Kinase Assay: In vitrocharacteristics included potent inhibition of kynurenine (kyn) production in IDO1-HEK293 cells (IC50= 1.1 nM) but not in TDO-HEK293 cells, sustained inhibition in IDO1 cell-based assays after washout, and single-digit nM potency in human tolerogenic MLR assays.

ln Vivo
Based on preclinical data, a 150 mg QD human dose was projected to maximally inhibit IDO1. In the ongoing clinical study, 42 pts have been treated. All treatment-related adverse events were grade 1/2 except three grade 3 toxicities (autoimmune hepatitis [dose limiting; BMS-986205 100 mg/nivo 240 mg], rash, and asymptomatic hypophosphatemia). Day 14 individual trough concentrations began exceeding the human whole blood IC50 starting with 25 mg QD, and the IC90 starting with 50 mg QD; all pts treated at 200 mg QD exceeded the IC90. Serum kyn was substantially reduced at all doses (> 45% mean reduction at each dose), with > 60% mean reduction at the 100 and 200 mg QD doses. Importantly, intratumoral kyn was reduced up to 90% in evaluable paired pre- and on-treatment samples.
Linrodostat PK/PD profile in human SKOV3 xenografts [3]
Next, we assessed PK/PD properties of linrodostat using SKOV3 cells in a mouse xenograft model. Dose-dependent PD activity of linrodostat was assessed by measuring serum and intratumoral kynurenine levels with dosing once a day at 5, 25, or 125 mg/kg (Supplementary Table S1; PK reported as AUC0–24, μmol/L × h). AUEC measurements for the percent kynurenine reduction in linrodostat-treated tumors were 60%, 63%, and 76% at 5, 25, and 125 mg/kg, respectively, demonstrating dose-dependent PD activity in tumors. Of the time points assessed, maximal PD effects (96% kynurenine reduction) were observed at 6 hours after treatment (Fig. 4A). Reduced kynurenine levels (≥ 30%) were maintained 24 hours after the last dose of 10 mg/kg linrodostat and 100 mg/kg epacadostat. Derived from these PK/PD analyses, the in vivo median IC50 of linrodostat was 3.4 nmol/L in serum from SKOV3 tumors, which was similar to the in vitro median IC50 of 9.4 nmol/L in human whole blood (Fig. 4B). We also examined the potency of another IDO1 inhibitor, epacadostat, and detected a median IC50 of 227- and 163 nmol/L potency in vivo and in vitro, respectively (Fig. 4C). These data demonstrate that linrodostat shows single-digit nanomolar potency.
Enzyme Assay
IDO1 inhibition [3]
SKOV3 human ovarian cancer cells (ATCCs) were treated with media containing 1 mmol/L tryptophan, 50 μmol/L succinylacetone, and 50 ng/mL human IFNγ to induce IDO1 synthesis, and either vehicle (DMSO), Linrodostat , or epacadostat at 10, 50, or 100 nmol/L for 72 hours. Kynurenine levels were measured with the Ehrlich colorimetric assay. Colorimetric changes were detected with a plate reader (490 nm). Kynurenine concentrations were determined from kynurenine standard curves.
Reversal of IDO1 inhibition [3]
SKOV3 cells were treated with 5 μg/mL cycloheximide to prevent further IDO1 synthesis and incubated with 50 μmol/L succinylacetone. Media were replaced with media containing 1 mmol/L tryptophan, 50 μmol/L succinylacetone, and 5 μg/mL cycloheximide, and duplicates were treated with vehicle (DMSO) or 10 μmol/L heme. Kynurenine levels were measured as described above at 0, 2, 4, 6, 8, and 10 hours.
Aryl hydrocarbon receptor assay [3]
The Human AhR Reporter Assay System (96-well format) was used according to the manufacturer's instructions. Reporter cells dispensed into a 96-well plate were immediately dosed with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD; 1 or 10 nmol/L), kynurenine (1 or 10 μmol/L), or Linrodostat (1 or 10 μmol/L). After 22 to 24 hours' incubation, media were removed, and luciferase detection reagent (100 μL) was added. After approximately 5 minutes, luciferase activity was detected per well using a plate-reading luminometer.
SKOV-3 IDO1 assay (cell plating, IDO1 induction, kynurenine determination) [1]
The cells were plated at 3 × 104 cells/well and allowed to attach overnight. The next day IFNγ was added to the cell culture at 100 ng/mL final concentration to induce IDO1 expression followed by 24 h incubation at 37°C and 5% CO2. A kynurenine standard curve was used to interpolate kynurenine concentrations in the test samples.
Caspase 3/7 activation [1]
Caspase 3/7 activity was measured using Caspase 3/7 Glo reagent form Promega according to manufacturer's instructions.
Cell Assay
Half maximal inhibitory concentration in cell-based assays [3]
All cell lines used in the study were routinely tested for mycoplasma, and passages were tracked per standard protocols. HEK293 cells overexpressing human IDO1, human TDO, murine Ido1, or murine Ido2 were seeded using RPMI phenol red–free media containing 10% fetal bovine serum (FBS). Linrodostat was prepared as previously described in Patent Cooperation Treaty Int Appl WO 2016073774 (example 19; refs. 32, 33). Linrodostat (100 nL; 10 μmol/L–10 pmol/L) was added, and cells were incubated for 20 hours at 37°C in 5% CO2. HeLa or M109 cells were seeded using RPMI phenol red–free media containing 10% FBS. Linrodostat (50 μL; 10 μmol/L–10 pmol/L) was added, and cells were incubated at 37°C for 2 hours. Recombinant human IFNγ (final concentration, 10 ng/mL) or recombinant murine IFNγ (final concentration, 5 ng/mL) was added to induce IDO1. Cells were incubated for 18 hours at 37°C in 5% CO2. Treatment was stopped, and IC50 values were calculated.
Relative viability determination [1]
Cell viability was assessed using Cell-Titer Fluro (Promega) according to manufacturer's instructions. Relative cell viability was calculated based on the reading of the non-treated control. % Relative Viability = TestValue/Non-treatedControlValue × 100.
Cell Viability Assay[1]
Cell Types: SKOV-3 and Jurkat clone E6-1 cells
Tested Concentrations: 0.01-100 μM
Incubation Duration: 72 hrs (hours)
Experimental Results: decreased the number of viable cells compared with the non-treated control and induced cell death at much lower concentrations.
Animal Protocol
During dose escalation, patients (pts) with advanced cancers were treated in escalating cohorts with BMS-986205 (25-200 mg as of Jan 5, 2017) orally once daily (QD) for 2 wk, followed by BMS-986205 + nivo 240 mg IV every 2 wk. Objectives included safety (primary), PK, and PD. [2]
Human SKOV3 xenograft tumor model [3]
Female nu/nu mice (7–12 weeks old; Envigo) received food and water ad libitum and were maintained in a controlled environment according to Association for Assessment and Accreditation of Laboratory Animal Care International regulations. SKOV3 cells were maintained in RPMI 1640 with 10% FBS and were harvested in the log growth phase (6 × 107 cells/mL in Hank's balanced salt solution), mixed 1:1 with Matrigel, and s.c. implanted into the mouse flank (0.1 mL or 3 × 106 cells/mouse). After implantation (15 days), tumor volumes were measured, and tumor-bearing mice were randomized (five animals/group). Linrodostat was formulated as a solution in the vehicle ethanol/polyethylene glycol (PEG) 400/propylene glycol/d-α-tocopheryl PEG 1000 succinate (volume ratio, 5:55:20:20). Each group was orally administered a vehicle, Linrodostat (5, 25, or 125 mg/kg once a day), or epacadostat (30 or 100 mg/kg twice daily), for 5 days. Tumor volumes were measured before tumors were snap frozen. Sera were harvested (day 5) at designated times after dosing to quantify kynurenine and compound levels.
PK analyses [3]
PK parameters were determined by a noncompartmental model of analysis of plasma concentration versus time data using Phoenix 6.3.0.395. AUCs from time zero to last sampling time (AUC0-T) and time zero to infinity (AUCINF) were calculated using the linear-log trapezoidal method. The total plasma clearance (CLTp), steady-state volume of distribution (Vss), and apparent elimination half-life (t1/2) were estimated after IV administration. t1/2 estimations were calculated at ≥ 3 time points. The total blood clearance (CLTb) was calculated as the CLTp divided by the blood-to-plasma concentration ratio. The absolute oral bioavailability (F) was estimated as the ratio of dose-normalized AUCs following oral and IV doses.
ADME/Pharmacokinetics
Linrodostat PK profile in vivo [3]
To extend these SKOV3 xenograft studies, we assessed PK parameters of IV and oral linrodostat in higher species including rat, dog, and monkey (Table 1). After IV administration, the CLTp of linrodostat was comparable across species, with levels of 27, 25, and 19 mL/min/kg in rats, dogs, and monkeys, respectively. The CLTp were ≤ 48% of the respective reported liver blood flows, indicating that linrodostat has low-to-moderate systemic clearance. After IV administration, Vss values in rats, dogs, and monkeys were 3.8, 5.7, and 4.1 L/kg, respectively, indicating extravascular distribution. The t1/2 of linrodostat was 3.9, 4.7, and 6.6 hours in rats, dogs, and monkeys, respectively. After oral administration, the Tmax ranged from 0.5 to 1.7 hours in rats, dogs, and monkeys. The absolute oral bioavailability of linrodostat given as a solution was 64%, 39%, and 10% in rats, dogs, and monkeys, respectively. The rate of linrodostat metabolism by hepatocytes was higher in rats than dogs, monkeys, and humans. Based on the in vitro–in vivo correlation of hepatocyte clearance in animal species, clearance in humans is projected to be moderate. These data lead to a projected human t1/2 of 23 hours, supporting dosing once a day in a clinical setting.
To extend these preclinical, in vivo PD/PK studies, we assessed kynurenine PD with linrodostat + nivolumab treatment in patients with advanced cancers. Substantial decreases in mean serum kynurenine levels were detected at all linrodostat doses evaluated, with an approximately 60% reduction observed with 100 and 200 mg once a day (Fig. 5A and B). Furthermore, pre- and on-treatment tumor samples from 13 patients demonstrated that intratumoral kynurenine levels were reduced across all doses following treatment, even in the presence of relatively high pretreatment kynurenine levels (Fig. 5C). These results demonstrate that linrodostat + nivolumab leads to substantial reductions in serum and intratumoral kynurenine levels.
References

[1]. Cell based functional assays for IDO1 inhibitor screening and characterization. Oncotarget. 2018 Jul 20;9(56):30814-30820.

[2]. Abstract CT116: BMS-986205, an optimized indoleamine 2,3-dioxygenase 1 (IDO1) inhibitor, is well tolerated with potent pharmacodynamic (PD) activity, alone and in combination with nivolumab (nivo) in advanced cancers in a phase 1/2a trial. AACR; Cancer Res. 2017; 77 (13 Suppl): Abstract nr CT116.

[3]. Preclinical Characterization of Linrodostat Mesylate, a Novel, Potent, and Selective Oral Indoleamine 2,3-Dioxygenase 1 Inhibitor. Mol Cancer Ther. 2021 Mar;20(3):467-476.

Additional Infomation
Linrodostat is under investigation in clinical trial NCT03247283 (Pharmacokinetics and Metabolism Study in Healthy Male Participants).
Linrodostat is an orally available inhibitor of indoleamine 2,3-dioxygenase 1 (IDO1), with potential immunomodulating and antineoplastic activities. Upon administration, linrodostat specifically targets and binds to IDO1, a cytosolic enzyme responsible for the oxidation of the amino acid tryptophan into the immunosuppressive metabolite kynurenine. By inhibiting IDO1 and decreasing kynurenine in tumor cells, BMS-986205 restores and promotes the proliferation and activation of various immune cells, including dendritic cells (DCs), natural killer (NK) cells, and T-lymphocytes, and causes a reduction in tumor-associated regulatory T-cells (Tregs). Activation of the immune system, which is suppressed in many cancers, may induce a cytotoxic T-lymphocyte (CTL) response against the IDO1-expressing tumor cells, thereby inhibiting the growth of IDO1-expressing tumor cells. IDO1, overexpressed by multiple tumor cell types, plays an important role in immunosuppression. Tryptophan depletion inhibits T-lymphocyte proliferation and activation, and subsequently suppresses the immune system.
Indoleamine 2,3-dioxygenase 1 (IDO1) is a new immune-oncology target and its inhibitors have shown promise in the clinic especially in combination with other immune-stimulating agents. Here we describe two robust cell-based assays for screening IDO1 inhibitors. Both assays can be easily adopted by most laboratories and utilized for screening of IDO1 inhibitors. Endogenous IDO1 expression is induced in a cancer cell line with interferon gamma and its activity is assessed by measuring kynurenine secreted into the media. The effect of cancer cell IDO1 induction and inhibition on T cell activation is evaluated in a co-culture assay using Jurkat T cell line. Additional readouts assessing cell viability are employed for early detection of false positive IDO1 inhibitors and toxic compounds. Clinical candidates epacadostat and BMS-986205 were evaluated in the assays as control compounds, the former can completely inhibit IDO1 activity while the maximum effect of the later is limited (to about 80% in our system) consistent with the differences in their interaction with IDO1. Nanomolar concentrations of both compounds rescued IDO1 mediated inhibition of T cell activation. However, treatment with micromolar concentrations of BMS-986205 blocked Jurkat T cell activation and after prolonged incubation induced cell death.[1]
Background: IDO1 is highly expressed in multiple cancers and may be an immunosuppressive mechanism for tumor escape via its production of metabolites that inhibit T-cell function. Nivo, a mAb that targets PD-1, causes IDO1 upregulation, supporting a rationale for combining it with an IDO1 inhibitor. Our preclinical program aimed to identify a best in class IDO1 inhibitor with favorable pharmacokinetic (PK) characteristics (Hunt J, et al. AACR 2017 [abst 6774]). Here we present BMS-986205, a selective IDO1 inhibitor validated in a novel phase 1/2a trial alone and in combination with nivo. Methods: During dose escalation, patients (pts) with advanced cancers were treated in escalating cohorts with BMS-986205 (25-200 mg as of Jan 5, 2017) orally once daily (QD) for 2 wk, followed by BMS-986205 + nivo 240 mg IV every 2 wk. Objectives included safety (primary), PK, and PD. Preclinical analyses included measurement of enzyme inhibition in HEK293 cells overexpressing human IDO1 or tryptophan 2,3-dioxygenase (TDO) and IFNγ-stimulated HeLa cells. Results: In support of clinical testing, BMS-986205 was evaluated preclinically. In vitro characteristics included potent inhibition of kynurenine (kyn) production in IDO1-HEK293 cells (IC50 = 1.1 nM) but not in TDO-HEK293 cells, sustained inhibition in IDO1 cell-based assays after washout, and single-digit nM potency in human tolerogenic MLR assays. Based on preclinical data, a 150 mg QD human dose was projected to maximally inhibit IDO1. In the ongoing clinical study, 42 pts have been treated. All treatment-related adverse events were grade 1/2 except three grade 3 toxicities (autoimmune hepatitis [dose limiting; BMS-986205 100 mg/nivo 240 mg], rash, and asymptomatic hypophosphatemia). Day 14 individual trough concentrations began exceeding the human whole blood IC50 starting with 25 mg QD, and the IC90 starting with 50 mg QD; all pts treated at 200 mg QD exceeded the IC90. Serum kyn was substantially reduced at all doses (> 45% mean reduction at each dose), with > 60% mean reduction at the 100 and 200 mg QD doses. Importantly, intratumoral kyn was reduced up to 90% in evaluable paired pre- and on-treatment samples. Conclusions: BMS-986205 is an optimized, once-daily, selective and potent oral IDO1 inhibitor at clinically relevant concentrations. It is well tolerated up to at least 200 mg in combination with nivo in this novel trial. Evidence of substantial serum kyn reduction was observed at doses as low as 25 mg QD; inhibition at 100 and 200 mg QD appears greater than that reported for other in-class compounds. In addition, we have presented the first evidence of intratumoral kyn reduction by an IDO1 inhibitor. These data suggest the potential of BMS-986205 as an IDO1 inhibitor with superior PD properties and support further evaluation in combination with nivo.[2]
Tumors can exploit the indoleamine 2,3-dioxygenase 1 (IDO1) pathway to create an immunosuppressive microenvironment. Activated IDO1 metabolizes tryptophan into immunosuppressive kynurenine, leading to suppressed effector T-cell (Teff) proliferation, allowing for tumor escape from host immune surveillance. IDO1 inhibition counteracts this immunosuppressive tumor microenvironment and may improve cancer outcomes, particularly when combined with other immunotherapies. Linrodostat mesylate (linrodostat) is a potent, selective oral IDO1 inhibitor that occupies the heme cofactor-binding site to prevent further IDO1 activation and is currently in multiple clinical trials for treatment of patients with advanced cancers. Here, we assess the in vitro potency, in vivo pharmacodynamic (PD) activity, and preclinical pharmacokinetics (PKs) of linrodostat. Linrodostat exhibited potent cellular activity, suppressing kynurenine production in HEK293 cells overexpressing human IDO1 and HeLa cells stimulated with IFNγ, with no activity against tryptophan 2,3-dioxygenase or murine indoleamine 2,3-dioxygenase 2 detected. Linrodostat restored T-cell proliferation in a mixed-lymphocyte reaction of T cells and allogeneic IDO1-expressing dendritic cells. In vivo, linrodostat reduced kynurenine levels in human tumor xenograft models, exhibiting significant PD activity. Linrodostat demonstrated a PK/PD relationship in the xenograft model, preclinical species, and samples from patients with advanced cancers, with high oral bioavailability in preclinical species and low to moderate systemic clearance. Our data demonstrate that linrodostat potently and specifically inhibits IDO1 to block an immunosuppressive mechanism that could be responsible for tumor escape from host immune surveillance with favorable PK/PD characteristics that support clinical development.[3]
These protocols are for reference only. InvivoChem does not independently validate these methods.
Physicochemical Properties
Exact Mass
506.14423
Elemental Analysis
C, 59.22; H, 5.57; Cl, 6.99; F, 3.75; N, 5.53; O, 12.62; S, 6.32
CAS #
2221034-29-1
Related CAS #
1923833-60-6;2221034-29-1 (mesylate)
PubChem CID
134687067
Appearance
Typically exists as solid at room temperature
Hydrogen Bond Donor Count
2
Hydrogen Bond Acceptor Count
6
Rotatable Bond Count
4
Heavy Atom Count
34
Complexity
638
Defined Atom Stereocenter Count
1
InChi Key
LZNFBNWGSDOVIO-QEFLZESTSA-N
InChi Code
InChI=1S/C24H24ClFN2O.CH4O3S/c1-15(24(29)28-20-9-6-18(25)7-10-20)16-2-4-17(5-3-16)21-12-13-27-23-11-8-19(26)14-22(21)231-5(2,3)4/h6-17H,2-5H2,1H3,(H,28,29)1H3,(H,2,3,4)/t15-,16-,17+/m1./s1
Chemical Name
(R)-N-(4-chlorophenyl)-2-((1s,4S)-4-(6-fluoroquinolin-4-yl)cyclohexyl)propanamide methanesulfonic acid
Synonyms
Linrodostat mesylate; BMS-986205; BMS 986205; BMS986205 mesylate; ONO-7701; ONO 7701 mesylate; ONO7701; F-001287; F001287; F001287; 2221034-29-1; OS7OBU191R; Linrodostat mesylate [USAN]; Linrodostat [USAN]; Linrodostat
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 Data
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
(e.g. IP/IV/IM/SC)
Injection Formulation 1: DMSO : Tween 80: Saline = 10 : 5 : 85 (i.e. 100 μL DMSO stock solution 50 μL Tween 80 850 μL Saline)
*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 : PEG300Tween 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)]
*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.
Injection Formulation 5: 2-Hydroxypropyl-β-cyclodextrin : Saline = 50 : 50 (i.e. 500 μL 2-Hydroxypropyl-β-cyclodextrin 500 μL Saline)
Injection Formulation 6: DMSO : PEG300 : castor oil : Saline = 5 : 10 : 20 : 65 (i.e. 50 μL DMSO 100 μLPEG300 200 μL castor oil 650 μL Saline)
Injection Formulation 7: Ethanol : Cremophor : Saline = 10: 10 : 80 (i.e. 100 μL Ethanol 100 μL Cremophor 800 μL Saline)
Injection Formulation 8: Dissolve in Cremophor/Ethanol (50 : 50), then diluted by Saline
Injection Formulation 9: EtOH : Corn oil = 10 : 90 (i.e. 100 μL EtOH 900 μL Corn oil)
Injection Formulation 10: EtOH : PEG300Tween 80 : Saline = 10 : 40 : 5 : 45 (i.e. 100 μL EtOH 400 μLPEG300 50 μL Tween 80 450 μL 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
Oral Formulation 4: Suspend in 0.2% Carboxymethyl cellulose
Oral Formulation 5: Dissolve in 0.25% Tween 80 and 0.5% Carboxymethyl cellulose
Oral Formulation 6: Mixing with food powders


Note: Please be aware that the above formulations are for reference only. InvivoChem strongly recommends customers to read literature methods/protocols carefully before determining which formulation you should use for in vivo studies, as different compounds have different solubility properties and have to be formulated differently.

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

Molarity Calculator allows you to calculate the mass, volume, and/or concentration required for a solution, as detailed below:

  • Calculate the Mass of a compound required to prepare a solution of known volume and concentration
  • Calculate the Volume of solution required to dissolve a compound of known mass to a desired concentration
  • Calculate the Concentration of a solution resulting from a known mass of compound in a specific volume
An example of molarity calculation using the molarity calculator is shown below:
What is the mass of compound required to make a 10 mM stock solution in 5 ml of DMSO given that the molecular weight of the compound is 350.26 g/mol?
  • Enter 350.26 in the Molecular Weight (MW) box
  • Enter 10 in the Concentration box and choose the correct unit (mM)
  • Enter 5 in the Volume box and choose the correct unit (mL)
  • Click the “Calculate” button
  • The answer of 17.513 mg appears in the Mass box. In a similar way, you may calculate the volume and concentration.

Dilution Calculator allows you to calculate how to dilute a stock solution of known concentrations. For example, you may Enter C1, C2 & V2 to calculate V1, as detailed below:

What volume of a given 10 mM stock solution is required to make 25 ml of a 25 μM solution?
Using the equation C1V1 = C2V2, where C1=10 mM, C2=25 μM, V2=25 ml and V1 is the unknown:
  • Enter 10 into the Concentration (Start) box and choose the correct unit (mM)
  • Enter 25 into the Concentration (End) box and select the correct unit (mM)
  • Enter 25 into the Volume (End) box and choose the correct unit (mL)
  • Click the “Calculate” button
  • The answer of 62.5 μL (0.1 ml) appears in the Volume (Start) box
g/mol

Molecular Weight Calculator allows you to calculate the molar mass and elemental composition of a compound, as detailed below:

Note: Chemical formula is case sensitive: C12H18N3O4  c12h18n3o4
Instructions to calculate molar mass (molecular weight) of a chemical compound:
  • To calculate molar mass of a chemical compound, please enter the chemical/molecular formula and click the “Calculate’ button.
Definitions of molecular mass, molecular weight, molar mass and molar weight:
  • Molecular mass (or molecular weight) is the mass of one molecule of a substance and is expressed in the unified atomic mass units (u). (1 u is equal to 1/12 the mass of one atom of carbon-12)
  • Molar mass (molar weight) is the mass of one mole of a substance and is expressed in g/mol.
/

Reconstitution Calculator allows you to calculate the volume of solvent required to reconstitute your vial.

  • Enter the mass of the reagent and the desired reconstitution concentration as well as the correct units
  • Click the “Calculate” button
  • The answer appears in the Volume (to add to vial) box
In vivo Formulation Calculator (Clear solution)
Step 1: Enter information below (Recommended: An additional animal to make allowance for loss during the experiment)
Step 2: Enter in vivo formulation (This is only a calculator, not the exact formulation for a specific product. Please contact us first if there is no in vivo formulation in the solubility section.)
+
+
+

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.

Clinical Trial Information
Nivolumab and BMS986205 in Treating Patients With Stage II-IV Squamous Cell Cancer of the Head and Neck
CTID: NCT03854032
Phase: Phase 2
Status: Active, not recruiting
Date: 2025-02-04
An Investigational Study of Immunotherapy Combinations in Participants With Solid Cancers That Are Advanced or Have Spread
CTID: NCT03459222
Phase: Phase 1/Phase 2
Status: Active, not recruiting
Date: 2024-08-20
Nivolumab, BMS-986205, and Radiation Therapy With or Without Temozolomide in Treating Patients With Newly Diagnosed Glioblastoma
CTID: NCT04047706
Phase: Phase 1
Status: Active, not recruiting
Date: 2024-08-15
Study of BMS-986205 and Nivolumab in Endometrial Cancer or Endometrial Carcinosarcoma That Has Not Responded to Treatment
CTID: NCT04106414
Phase: Phase 2
Status: Active, not recruiting
Date: 2024-07-03
An Investigational Immuno-therapy Study of BMS-986205 Given in Combination With Nivolumab and in Combination With Both Nivolumab and Ipilimumab in Cancers That Are Advanced or Have Spread
CTID: NCT02658890
Phase: Phase 1/Phase 2
Status: Completed
Date: 2023-08-28
Contact Us