Sirolimus (Rapamycin)
new
featured

    WARNING: This product is for research use only, not for human or veterinary use.

MedKoo CAT#: 100766

CAS#: 53123-88-9

Description: Sirolimus, also known as rapamycin, is a natural macrocyclic lactone produced by the bacterium Streptomyces hygroscopicus, with immunosuppressant properties. In cells, sirolimus binds to the immunophilin FK Binding Protein-12 (FKBP-12) to generate an immunosuppressive complex that binds to and inhibits the activation of the mammalian Target Of Rapamycin (mTOR), a key regulatory kinase. This results in inhibition of T lymphocyte activation and proliferation that occurs in response to antigenic and cytokine (IL-2, IL-4, and IL-15) stimulation and inhibition of antibody production.


Chemical Structure

img
Sirolimus (Rapamycin)
CAS# 53123-88-9

Theoretical Analysis

MedKoo Cat#: 100766
Name: Sirolimus (Rapamycin)
CAS#: 53123-88-9
Chemical Formula: C51H79NO13
Exact Mass: 913.56
Molecular Weight: 914.170
Elemental Analysis: C, 67.01; H, 8.71; N, 1.53; O, 22.75

Price and Availability

Size Price Availability Quantity
50mg USD 110 Ready to ship
100mg USD 180 Ready to ship
200mg USD 300 Ready to ship
500mg USD 625 Ready to ship
1g USD 1050 Ready to ship
2g USD 1750 Ready to ship
5g USD 3750 Ready to ship
Bulk inquiry

Synonym: RAPA; RAP; RPM; SLM; AY 22989; AY22989; AY-22989; SILA 9268A; WY090217; WY-090217; WY 090217; C07909; D00753; sirolimus; rapamycin; Rapamune.

IUPAC/Chemical Name: (3S,6R,7E,9R,10R,12R,14S,15E,17E,19E,21S,23S,26R,27R,34aS)-9,10,12,13,14,21,22,23,24,25,26,27,32,33,34, 34a-hexadecahydro-9,27-dihydroxy-3-[(1R)-2-[(1S,3R,4R)-4-hydroxy-3-methoxycyclohexyl]-1-methylethyl]-10,21-dimethoxy-6,8,12,14,20,26-hexamethyl-23,27-epoxy-3H-pyrido[2,1-c][1,4] oxaazacyclohentriacontine-1,5,11,28,29 (4H,6H,31H)-pentone

InChi Key: QFJCIRLUMZQUOT-PYYJPVDBSA-N

InChi Code: InChI=1S/C51H79NO13/c1-30-16-12-11-13-17-31(2)42(61-8)28-38-21-19-36(7)51(60,65-38)48(57)49(58)52-23-15-14-18-39(52)50(59)64-43(33(4)26-37-20-22-40(53)44(27-37)62-9)29-41(54)32(3)25-35(6)46(56)47(63-10)45(55)34(5)24-30/h11-13,16-17,25,30,32-34,36-40,42-44,46-47,53,56,60H,14-15,18-24,26-29H2,1-10H3/b13-11+,16-12+,31-17+,35-25+/t30-,32-,33-,34-,36-,37+,38+,39+,40-,42+,43+,44?,46-,47+,51-/m1/s1

SMILES Code: C[C@@H](C([C@H](OC)[C@H](O)/C(C)=C/[C@@H](C)C(C[C@@H]([C@H](C)C[C@@H]1CC[C@@H](O)C(OC)C1)OC2=O)=O)=O)C[C@H](C)/C=C/C=C/C=C(C)/[C@@H](OC)C[C@H]3O[C@](C(C(N4[C@H]2CCCC4)=O)=O)(O)[C@H](C)CC3

Appearance: White to off-white solid powder

Purity: >98% (or refer to the Certificate of Analysis)

Shipping Condition: Shipped under ambient temperature as non-hazardous chemical. This product is stable enough for a few weeks during ordinary shipping and time spent in Customs.

Storage Condition: Dry, dark and at 0 - 4 C for short term (days to weeks) or -20 C for long term (months to years).

Solubility: Soluble in DMSO, not in water

Shelf Life: >2 years if stored properly

Drug Formulation: This drug may be formulated in DMSO

Stock Solution Storage: 0 - 4 C for short term (days to weeks), or -20 C for long term (months).

HS Tariff Code: 2934.99.9001

More Info: According to http://en.wikipedia.org/wiki/Sirolimus, Sirolimus (also known as rapamycin) is a chemical that was discovered by Suren Sehgal,[1] as a product of bacteria discovered on Easter Island (the island is also known as Rapa Nui).[2] It was approved by the US Food and Drug Administration in September 1999 and is marketed under the trade name Rapamune by Pfizer (formerly by Wyeth). Sirolimus was originally developed as an antifungal agent. However, this use was abandoned when it was discovered to have potent immunosuppressive and antiproliferative properties. It has since been shown to prolong the life of mice and might also be useful in the treatment of certain cancers.   Mechanism of Action Mechanism of Action Sirolimus inhibits T-lymphocyte activation and proliferation that occurs in response to antigenic and cytokine (Interleukin [IL]-2, IL-4, and IL-15) stimulation by a mechanism that is distinct from that of other immunosuppressants. Sirolimus also inhibits antibody production. In cells, sirolimus binds to the immunophilin, FK Binding Protein-12 (FKBP-12), to generate an immunosuppressive complex. The sirolimus:FKBP-12 complex has no effect on calcineurin activity. This complex binds to and inhibits the activation of the mammalian Target Of Rapamycin (mTOR), a key regulatory kinase. This inhibition suppresses cytokine-driven T-cell proliferation, inhibiting the progression from the G1 to the S phase of the cell cycle. Studies in experimental models show that sirolimus prolongs allograft (kidney, heart, skin, islet, small bowel, pancreatico-duodenal, and bone marrow) survival in mice, rats, pigs, and/or primates. Sirolimus reverses acute rejection of heart and kidney allografts in rats and prolongs the graft survival in presensitized rats. In some studies, the immunosuppressive effect of sirolimus lasts up to 6 months after discontinuation of therapy. This tolerization effect is alloantigen-specific. In rodent models of autoimmune disease, sirolimus suppresses immune-mediated events associated with systemic lupus erythematosus, collagen-induced arthritis, autoimmune type I diabetes, autoimmune myocarditis, experimental allergic encephalomyelitis, graft-versus-host disease, and autoimmune uveoretinitis. Sirolimus inhibits T-lymphocyte activation and proliferation that occurs in response to antigenic and cytokine (Interleukin [IL]-2, IL-4, and IL-15) stimulation by a mechanism that is distinct from that of other immunosuppressants. Sirolimus also inhibits antibody production. In cells, sirolimus binds to the immunophilin, FK Binding Protein-12 (FKBP-12), to generate an immunosuppressive complex. The sirolimus:FKBP-12 complex has no effect on calcineurin activity. This complex binds to and inhibits the activation of the mammalian Target Of Rapamycin (mTOR), a key regulatory kinase. This inhibition suppresses cytokine-driven T-cell proliferation, inhibiting the progression from the G1 to the S phase of the cell cycle. Studies in experimental models show that sirolimus prolongs allograft (kidney, heart, skin, islet, small bowel, pancreatico-duodenal, and bone marrow) survival in mice, rats, pigs, and/or primates. Sirolimus reverses acute rejection of heart and kidney allografts in rats and prolongs the graft survival in presensitized rats. In some studies, the immunosuppressive effect of sirolimus lasts up to 6 months after discontinuation of therapy. This tolerization effect is alloantigen-specific. In rodent models of autoimmune disease, sirolimus suppresses immune-mediated events associated with systemic lupus erythematosus, collagen-induced arthritis, autoimmune type I diabetes, autoimmune myocarditis, experimental allergic encephalomyelitis, graft-versus-host disease, and autoimmune uveoretinitis.

Biological target: Rapamycin (Sirolimus; AY 22989) is a potent and specific mTOR inhibitor with an IC50 of 0.1 nM in HEK293 cells.
In vitro activity: To further investigate the role of autophagy in the regulation of PTSC senescence and the potential mechanisms involved, this study examined the protein expression levels of autophagy markers and SASP markers with bleomycin and rapamycin treatments. First, the expression of p62, which was inversely correlated with autophagy, was increased in the bleomycin-treated PTSCs (Figure 3(a)). The addition of rapamycin reversed the p62 expression to the basal level as in the control group (Figures 3(a) and 3(b)). Meanwhile, as an indicator of autophagy activation, the LC3 II/LC3 I expression ratio was significantly reduced by bleomycin treatment in the PTSCs, but rapamycin antagonized this decrease (Figures 3(a) and 3(c)). Moreover, rapamycin at the dose of 25 nM completely inhibited the downstream responder S6 phosphorylation, indicating an inhibition of the mTOR signaling pathway (Figure 3(a)). Reference: Stem Cells Int. 2021; 2021: 6638249. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7870298/
In vivo activity: As expected, rapamycin treatment effectively prevented the AA-induced increase in mTOR and S6K1 phosphorylation, which led to marked elevations in AA-induced renal expression of the autophagy markers (Fig. 2). This suggested that inhibiting mTOR activity by rapamycin further activated renal autophagy. Autophagy and apoptosis are two connected pathological processes involved in the development of AAN. To determine the effects of rapamycin on renal apoptosis in AAN mice, the protein expression of Bcl-2 and Bax, common markers of apoptosis, was assessed in kidney tissues using western blotting. The results indicated that the kidney tissue of AA-treated mice presented with decreased expression of Bcl-2 and increased expression of Bax, which were reversed by rapamycin treatment (Fig. 2A, G and H), suggesting that rapamycin inhibited apoptosis in the kidneys of AA-treated renal injury mice. Taken together, these observations indicate that rapamycin supplementation inhibits the renal activity of mTOR, which promotes renal autophagy, thereby probably suppressing the apoptosis of kidney tissues in mice with AA-induced renal injury. Reference: Mol Med Rep. 2021 Jul; 24(1): 495. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8127069/

Solubility Data

Solvent Max Conc. mg/mL Max Conc. mM
Solubility
DMSO 63.3 69.27
DMF 10.0 10.94
Ethanol 80.0 87.51

Preparing Stock Solutions

The following data is based on the product molecular weight 914.17 Batch specific molecular weights may vary from batch to batch due to the degree of hydration, which will affect the solvent volumes required to prepare stock solutions.

Recalculate based on batch purity %
Concentration / Solvent Volume / Mass 1 mg 5 mg 10 mg
1 mM 1.15 mL 5.76 mL 11.51 mL
5 mM 0.23 mL 1.15 mL 2.3 mL
10 mM 0.12 mL 0.58 mL 1.15 mL
50 mM 0.02 mL 0.12 mL 0.23 mL
Formulation protocol: 1. Nie D, Zhang J, Zhou Y, Sun J, Wang W, Wang JH. Rapamycin Treatment of Tendon Stem/Progenitor Cells Reduces Cellular Senescence by Upregulating Autophagy. Stem Cells Int. 2021 Feb 1;2021:6638249. doi: 10.1155/2021/6638249. PMID: 33603790; PMCID: PMC7870298. 2. Sahni A, Narra HP, Sahni SK. Activation of Mechanistic Target of Rapamycin (mTOR) in Human Endothelial Cells Infected with Pathogenic Spotted Fever Group Rickettsiae. Int J Mol Sci. 2020 Sep 29;21(19):7179. doi: 10.3390/ijms21197179. PMID: 33003310; PMCID: PMC7582468. 3. Lin F, Liu Y, Tang L, Xu X, Zhang X, Song Y, Chen B, Ren Y, Yang X. Rapamycin protects against aristolochic acid nephropathy in mice by potentiating mammalian target of rapamycin‑mediated autophagy. Mol Med Rep. 2021 Jul;24(1):495. doi: 10.3892/mmr.2021.12134. Epub 2021 May 6. PMID: 33955513; PMCID: PMC8127069. 4. Wu J, Zhong W, Zhang H, Yin Y. Mammalian Target of Rapamycin Signaling Enhances Ovalbumin-Induced Neutrophilic Airway Inflammation by Promoting Th17 Cell Polarization in Murine Noneosinophilic Asthma Model. Pediatr Allergy Immunol Pulmonol. 2020 Mar;33(1):25-32. doi: 10.1089/ped.2019.1088. PMID: 33406024; PMCID: PMC7875112.
In vitro protocol: 1. Nie D, Zhang J, Zhou Y, Sun J, Wang W, Wang JH. Rapamycin Treatment of Tendon Stem/Progenitor Cells Reduces Cellular Senescence by Upregulating Autophagy. Stem Cells Int. 2021 Feb 1;2021:6638249. doi: 10.1155/2021/6638249. PMID: 33603790; PMCID: PMC7870298. 2. Sahni A, Narra HP, Sahni SK. Activation of Mechanistic Target of Rapamycin (mTOR) in Human Endothelial Cells Infected with Pathogenic Spotted Fever Group Rickettsiae. Int J Mol Sci. 2020 Sep 29;21(19):7179. doi: 10.3390/ijms21197179. PMID: 33003310; PMCID: PMC7582468.
In vivo protocol: 1. Lin F, Liu Y, Tang L, Xu X, Zhang X, Song Y, Chen B, Ren Y, Yang X. Rapamycin protects against aristolochic acid nephropathy in mice by potentiating mammalian target of rapamycin‑mediated autophagy. Mol Med Rep. 2021 Jul;24(1):495. doi: 10.3892/mmr.2021.12134. Epub 2021 May 6. PMID: 33955513; PMCID: PMC8127069. 2. Wu J, Zhong W, Zhang H, Yin Y. Mammalian Target of Rapamycin Signaling Enhances Ovalbumin-Induced Neutrophilic Airway Inflammation by Promoting Th17 Cell Polarization in Murine Noneosinophilic Asthma Model. Pediatr Allergy Immunol Pulmonol. 2020 Mar;33(1):25-32. doi: 10.1089/ped.2019.1088. PMID: 33406024; PMCID: PMC7875112.

Molarity Calculator

Calculate the mass, volume, or concentration required for a solution.
=
x
x
g/mol

*When preparing stock solutions always use the batch-specific molecular weight of the product found on the vial label and SDS / CoA (available online).

Reconstitution Calculator

The reconstitution calculator allows you to quickly calculate the volume of a reagent to reconstitute your vial. Simply enter the mass of reagent and the target concentration and the calculator will determine the rest.

=
÷

Dilution Calculator

Calculate the dilution required to prepare a stock solution.
x
=
x

1: Knoll GA, Kokolo MB, Mallick R, Beck A, Buenaventura CD, Ducharme R, Barsoum R, Bernasconi C, Blydt-Hansen TD, Ekberg H, Felipe CR, Firth J, Gallon L, Gelens M, Glotz D, Gossmann J, Guba M, Morsy AA, Salgo R, Scheuermann EH, Tedesco-Silva H, Vitko S, Watson C, Fergusson DA. Effect of sirolimus on malignancy and survival after kidney transplantation: systematic review and meta-analysis of individual patient data. BMJ. 2014 Nov 24;349:g6679. doi: 10.1136/bmj.g6679. Review. PubMed PMID: 25422259; PubMed Central PMCID: PMC4241732.

2: Peng ZF, Yang L, Wang TT, Han P, Liu ZH, Wei Q. Efficacy and safety of sirolimus for renal angiomyolipoma in patients with tuberous sclerosis complex or sporadic lymphangioleiomyomatosis: a systematic review. J Urol. 2014 Nov;192(5):1424-30. doi: 10.1016/j.juro.2014.04.096. Epub 2014 May 9. Review. PubMed PMID: 24813310.

3: Woudstra P, de Winter RJ, Beijk MA. Next-generation DES: the COMBO dual therapy stent with Genous endothelial progenitor capturing technology and an abluminal sirolimus matrix. Expert Rev Med Devices. 2014 Mar;11(2):121-35. doi: 10.1586/17434440.2014.882046. Epub 2014 Feb 3. Review. PubMed PMID: 24484431.

4: Choe H, Aldoss I, Chaudhary P, Donovan J, Petrovic L, Pullarkat V. Nodular regenerative hyperplasia causing portal hypertension in a patient with chronic graft versus host disease: response to sirolimus. Acta Haematol. 2014;132(1):49-52. doi: 10.1159/000356736. Epub 2014 Jan 15. Review. PubMed PMID: 24434665.

5: Qiao Y, Bian Y, Yan X, Liu Z, Chen Y. Efficacy and safety of sirolimus-eluting stents versus bare-metal stents in coronary artery disease patients with diabetes: a meta-analysis. Cardiovasc J Afr. 2013 Aug;24(7):274-9. doi: 10.5830/CVJA-2013-062. Review. PubMed PMID: 24217305; PubMed Central PMCID: PMC3807685.

6: Dasari TW, Patel B, Saucedo JF. Systematic review of effectiveness of oral sirolimus after bare-metal stenting of coronary arteries for prevention of in-stent restenosis. Am J Cardiol. 2013 Nov 1;112(9):1322-7. doi: 10.1016/j.amjcard.2013.05.074. Epub 2013 Aug 6. Review. PubMed PMID: 23932188.

7: Lemos PA, Bienert I. The Supralimus sirolimus-eluting stent. Expert Rev Med Devices. 2013 May;10(3):295-300. doi: 10.1586/erd.12.91. Epub 2013 Apr 18. Review. PubMed PMID: 23597097.

8: Tartarin P, Froment P. [mTORC1 and sirolimus: a link with fertility]. Med Sci (Paris). 2013 Feb;29(2):200-5. doi: 10.1051/medsci/2013292019. Epub 2013 Feb 28. Review. French. PubMed PMID: 23452608.

9: Park KW, Kang SH, Velders MA, Shin DH, Hahn S, Lim WH, Yang HM, Lee HY, Van Boven AJ, Hofma SH, Kang HJ, Koo BK, Oh BH, Park YB, Kandzari DE, Kim HS. Safety and efficacy of everolimus- versus sirolimus-eluting stents: a systematic review and meta-analysis of 11 randomized trials. Am Heart J. 2013 Feb;165(2):241-50.e4. doi: 10.1016/j.ahj.2012.08.007. Epub 2012 Dec 27. Review. PubMed PMID: 23351828.

10: Halleck F, Duerr M, Waiser J, Huber L, Matz M, Brakemeier S, Liefeldt L, Neumayer HH, Budde K. An evaluation of sirolimus in renal transplantation. Expert Opin Drug Metab Toxicol. 2012 Oct;8(10):1337-56. doi: 10.1517/17425255.2012.719874. Epub 2012 Aug 28. Review. PubMed PMID: 22928953.