Need Assistance?
  • US & Canada:
    +
  • UK: +

RGD-4C

* Please kindly note that our products are not to be used for therapeutic purposes and cannot be sold to patients.

RGD-4C preferentially binds to integrins αvβ3 and αvβ5, both of which are overexpressed in many cancers, such as non-small cell lung cancer, melanoma, and growing vessels.

Category
Peptide Inhibitors
Catalog number
BAT-014782
CAS number
332179-76-7
Molecular Formula
C42H60N14O16S4
Molecular Weight
1145.29
IUPAC Name
2-[(1R,4S,7R,12R,15S,18R,21S,27S)-7-[[(2S)-2-aminopropanoyl]amino]-15-benzyl-4-(carboxymethyl)-12-(carboxymethylcarbamoyl)-27-[3-(diaminomethylideneamino)propyl]-3,6,14,17,20,23,26,29-octaoxo-9,10,31,32-tetrathia-2,5,13,16,19,22,25,28-octazabicyclo[16.11.4]tritriacontan-21-yl]acetic acid
Synonyms
H-Ala-Cys-Asp-Cys-Arg-Gly-Asp-Cys-Phe-Cys-Gly-OH (Disulfide bridge: Cys2-Cys10, Cys4-Cys8); L-alanyl-L-cysteinyl-L-alpha-aspartyl-L-cysteinyl-L-arginyl-glycyl-L-alpha-aspartyl-L-cysteinyl-L-phenylalanyl-L-cysteinyl-glycine (2->10),(4->8)-bis(disulfide); N-{[(1R,4S,7R,12R,15S,18R,21S,27S)-7-(L-Alanylamino)-15-benzyl-27-(3-carbamimidamidopropyl)-4,21-bis(carboxymethyl)-3,6,14,17,20,23,26,29-octaoxo-9,10,31,32-tetrathia-2,5,13,16,19,22,25,28-octaazabicyclo[16.11.4]tritriacont-12-yl]carbonyl}glycine
Appearance
White Powder
Purity
≥95%
Sequence
ACDCRGDCFCG (Disulfide bridge: Cys2-Cys10, Cys4-Cys8)
Storage
Store at -20°C
Solubility
Soluble in Acetonitrile, DMSO, Water
InChI
InChI=1S/C42H60N14O16S4/c1-19(43)33(64)53-26-16-74-73-15-25(35(66)48-14-32(62)63)54-36(67)22(10-20-6-3-2-4-7-20)51-41(72)28-18-76-75-17-27(56-38(69)24(12-31(60)61)52-40(26)71)39(70)50-21(8-5-9-46-42(44)45)34(65)47-13-29(57)49-23(11-30(58)59)37(68)55-28/h2-4,6-7,19,21-28H,5,8-18,43H2,1H3,(H,47,65)(H,48,66)(H,49,57)(H,50,70)(H,51,72)(H,52,71)(H,53,64)(H,54,67)(H,55,68)(H,56,69)(H,58,59)(H,60,61)(H,62,63)(H4,44,45,46)/t19-,21-,22-,23-,24-,25-,26-,27-,28-/m0/s1
InChI Key
DXIKEERYADAVLF-CFFDDJBSSA-N
Canonical SMILES
CC(C(=O)NC1CSSCC(NC(=O)C(NC(=O)C2CSSCC(C(=O)NC(C(=O)NCC(=O)NC(C(=O)N2)CC(=O)O)CCCN=C(N)N)NC(=O)C(NC1=O)CC(=O)O)CC3=CC=CC=C3)C(=O)NCC(=O)O)N
1. Effect of RGD-4C position is more important than disulfide bonds on antiangiogenic activity of RGD-4C modified endostatin derived synthetic polypeptide
Runting Yin, Heng Zheng, Tao Xi, Han-Mei Xu Bioconjug Chem. 2010 Jul 21;21(7):1142-7. doi: 10.1021/bc900292y.
ES-2 (IVRRADRAAVP), an endostatin-derived synthetic polypeptide, contains the amino acids 50-60 of endostatin from its N terminus, and it had no inhibitory effects on tumor growth in vivo. In order to increase the targeted delivery of ES-2 to tumors and further enhance the activity, the polypeptide RGD-4C (ACDCRGDCFC) was introduced into ES-2, and the effects of RGD-4C position and RGD-4C disulfide bonds on polypeptides activity were investigated. When RGD-4C polypeptides (with or without disulfide bonds) were introduced to the N-terminals of synthesized ES-2, the modified ES-2 showed significant antitumor activity in vivo. Cell proliferation and chicken chorioallantoic membrane (CAM) assay results showed that disulfide bonds had no significant effects on RGD-4C-modified ES-2 antiangiogenic activity. Furthermore, the target of modified peptides was integrin alpha5beta1, rather than integrin alphavbeta3 as previous studies mentioned.
2. Targeted delivery of neural progenitor cell-derived extracellular vesicles for anti-inflammation after cerebral ischemia
Tian Tian, Lei Cao, Chuan He, Qing Ye, Ruyu Liang, Weiyan You, Huixin Zhang, Jiahuan Wu, Jinhai Ye, Bakhos A Tannous, Jun Gao Theranostics. 2021 Apr 19;11(13):6507-6521. doi: 10.7150/thno.56367. eCollection 2021.
Ischemic stroke remains a major cause of death, and anti-inflammatory strategies hold great promise for preventing major brain injury during reperfusion. In the past decade, stem cell-derived extracellular vesicles (EVs) have emerged as novel therapeutic effectors in immune modulation. However, the intravenous delivery of EVs into the ischemic brain remains a challenge due to poor targeting of unmodified EVs, and the costs of large-scale production of stem cell-derived EVs hinder their clinical application. Methods: EVs were isolated from a human neural progenitor cell line, and their anti-inflammatory effects were verified in vitro. To attach targeting ligands onto EVs, we generated a recombinant fusion protein containing the arginine-glycine-aspartic acid (RGD)-4C peptide (ACDCRGDCFC) fused to the phosphatidylserine (PS)-binding domains of lactadherin (C1C2), which readily self-associates onto the EV membrane. Subsequently, in a middle cerebral artery occlusion (MCAO) mouse model, the RGD-C1C2-bound EVs (RGD-EV) were intravenously injected through the tail vein, followed by fluorescence imaging and assessment of proinflammatory cytokines expression and microglia activation. Results: The neural progenitor cell-derived EVs showed intrinsic anti-inflammatory activity. The RGD-EV targeted the lesion region of the ischemic brain after intravenous administration, and resulted in a strong suppression of the inflammatory response. Furthermore, RNA sequencing revealed a set of 7 miRNAs packaged in the EVs inhibited MAPK, an inflammation related pathway. Conclusion: These results point to a rapid and easy strategy to produce targeting EVs and suggest a potential therapeutic agent for ischemic stroke.
3. A Novel RGD-4C-Saporin Conjugate Inhibits Tumor Growth in Mouse Models of Bladder Cancer
Stefania Zuppone, et al. Front Oncol. 2022 Apr 11;12:846958. doi: 10.3389/fonc.2022.846958. eCollection 2022.
Although toxin may have some advantages compared to chemotherapeutic drugs in cancer therapy, e.g. a potent cytotoxic activity and a reduced risk of resistance, their successful application in the treatments to solid tumors still remains to be fully demonstrated. In this study, we genetically modified the structure of the plant-derived single-chain ribosome inactivating protein saporin (SAP) by fusing its N-terminus to the ACDCRGDCFCG peptide (RGD-4C), an αv-integrin ligand, and explored the anti-tumor activity of the resulting protein (called RGD-SAP) in vitro and in vivo, using a model of muscle invasive bladder cancer. We found that the RGD-4C targeting domain enhances the cytotoxic activity of SAP against various tumor cell lines, in a manner dependent on αv-integrin expression levels. In a subcutaneous syngeneic model of bladder cancer, RGD-SAP significantly reduced tumor growth in a dose-dependent manner. Furthermore, systemic administration of RGD-SAP in combination with mitomycin C, a chemotherapeutic drug currently used to treat patients with bladder cancer, increased the survival of mice bearing orthotopic bladder cancer with no evidence of systemic toxicity. Overall, the results suggest that RGD-SAP represents an efficient drug that could be exploited, either alone or in combination with the state-of-the-art therapies, for the treatment of bladder cancer and, potentially, of other solid tumors.
Online Inquiry
Verification code
Inquiry Basket