JenKemBeijingRnDJenKem Technology provides high quality activated homobifunctional polyethylene glycol derivatives (PEGs) with high purity and low polydispersity.

JenKem Technology’s homobifunctional PEG derivatives have numerous applications as cross-linkers, including PEGylation of proteins and peptides, or nanoparticle and surface modifications [1, 2]. Homobifunctional PEG derivatives have the general structure: X―PEG―X, where X is a functional reactive group.

Homobifunctional PEG products with molecular weights and functional groups not listed in our online catalog may be available by custom synthesis. Please inquire at tech@jenkemusa.com about pricing and availability of custom homofunctional PEGs.

JenKem Technology provides GMP grade PEG derivatives and bulk orders via custom synthesis, offering the opportunity to match customers’ special quality requirements. JenKem Technology is capable of development and synthesis of a wide range of GMP PEG derivatives starting at 200g up to 40 kg or greater batches, under ISO 9001 and ISO 13485 certified quality management system, following ICH Q7A guidelines. For inquiries on cGMP production of PEG derivatives and PEGylation services please contact us at tech@jenkemusa.com.

For global distribution, please visit link. Please click the buttons below to order directly from JenKem Technology:

PEG DERIVATIVE SUBSTITUTION REACTIVITY DETAILS
≥ 95% PEG (Acetic Acid)2 (PEG dicarboxyl, PEG diacetic acid, CM-PEG-CM, COOH-PEG-COOH, Carboxyl PEG Carboxyl) Amino reactive PEG crosslinker. [1-4]

≥ 95% Discrete PEG8 (Acetic Acid)2 and PEG12 (Acetic Acid)2  (Discrete PEG dicarboxyl, PEG diacetic acid, CM-PEG-CM, COOH-PEG-COOH)  Monodisperse discrete amino reactive PEG crosslinker [5].

≥ 95% Discrete PEG8 (Propionic Acid)2 and PEG12 (Propionic acid)2 (Monodisperse discrete PEG8 and PEG 12 dipropionic acid, PA-PEG PA, PEG dipropanoic acid). Monodisperse discrete amino reactive PEG crosslinker.
≥ 95% SCM PEG SCM (PEG diNHS, PEG diSuccinimidyl Carboxymethyl Ester, SCM-PEG-SCM, NHS-PEG-NHS, or PEG diSCM).  Amino reactive PEG diNHS ester crosslinker for lysine(s) or N-terminal amine. [6-7]
> 90% SS PEG SS (PEG diSuccinimidyl Succinate Ester, SS-PEG-SS, or PEG diSS).  Amino reactive PEG diNHS ester crosslinker for lysine(s) or N-terminal amine, with cleavable linker.
≥ 95% Amine PEG Amine (PEG diamine, PEG (Amine)2). Crosslinker PEG, attaches via stable linkages, such as amide, urethane, urea, secondary amine. [8-19]

≥ 95% Discrete PEG8 (Amine)2 and PEG12 (Amine)2 (Monodisperse PEG diamine, PEG (Amine)2). Crosslinker PEG, attaches via stable linkages, such as amide, urethane, urea, secondary amine.
> 90% Maleimide PEG Maleimide (PEG dimaleimide, PEG(Maleimide)2). Selective crosslinker for thiol groups on cystein side chains. [20-24]
≥ 95% Acrylate-PEG-Acrylate (PEG-DA, PEG diacrylate, PEG (Acrylate)2). PEG crosslinker used in vinyl polymerization or co-polymerization. [25-30]
≥ 95% Thiol PEG Thiol (PEG dithiol, PEG(Thiol)2). Thiol reactive PEG crosslinker, reacts with HS groups on cysteine side chains under mild reaction conditions. [31-33]
> 90% Vinylsulfone PEG Vinylsulfone (PEG-divinylsulfone, PEG (Vinylsulfone)2). Sulfhydryl reactive PEG crosslinker.[34-37]
≥ 90% Tosylate PEG Tosylate (diTosylate PEG, PEG diToluenesulfonyl , or PEG (Tosylate)2). Reactive homobifunctional PEG crosslinker employed for amine and thiol PEGylation
> 90% Alkyne-PEG-Alkyne (diAcetylene PEG, di Propargyl PEG, PEG)Alkyne)2, or dialkyne PEG). Click PEG crosslinking reagent for reaction with azide groups

> ≥95% Discrete PEG8 Dihydroxyl, PEG9 Dihydroxyl and PEG12-Dihydroxyl (HO-PEG8-OH, HO-PEG9-OH and HO-PEG12-OH)Monodisperse dihydroxyl PEG, dialcohol PEG). Monodisperse polyethylene glycol raw material.

3ARM, 4ARM, 6ARM, and 8ARM Homofunctional PEGs

Linear, 3ARM, 4ARM, 6ARM, and 8ARM PEG Raw Materials (Hydroxyl PEGs)

References:

  1. Wang, C., et al., Synthesis and formation mechanism of bone mineral, whitlockite nanocrystals in tri-solvent system, Journal of Colloid and Interface Science, 2020, V. 569, P. 1-11.
  2. Ji, F., et al., A Dual pH/Magnetic Responsive Nanocarrier Based on PEGylated Fe3O4 Nanoparticles for Doxorubicin Delivery, Journal of Nanoscience and Nanotechnology, 2018, 18.7: 4464-4470.
  3. Xiao, S., et al., Aptamer-mediated gene therapy enhanced antitumor activity against human hepatocellular carcinoma in vitro and in vivo, Journal of Controlled Release, 2017.
  4. Kwon, J., et al., Excitation-dependent emissive FeSe nanoparticles induced by chiral interlayer expansion and their multi-color bio-imaging. Nano Today, 2022, 43, p.101424.
  5. Gajbhiye, V., et al., Drug-loaded nanoparticles induce gene expression in human pluripotent stem cell derivatives. Nanoscale, 2014, 6(1): p. 521-31.
  6. Inostroza-Brito, K.E., et al., Cross-linking of a Biopolymer-Peptide Co-Assembling System, Acta Biomaterialia, 2017.
  7. Lu, C., et al., Poly(ethylene glycol) crosslinked multi-armed poly(ε-benzyloxycarbonyl-L-lysine)s as super-amphiphiles: Synthesis, self-assembly, and evaluation as efficient delivery systems for poorly water-soluble drugs, Colloids and Surfaces B: Biointerfaces, 2019, 182.
  8. Yang, S.-B., et al., The colossal role of H-MnO2-PEG in ischemic stroke, Nanomedicine: Nanotechnology, Biology and Medicine, 2021, 33, 102362.
  9. Zhang, T., et al., Magnetic/pH dual-responsive nanocomposites loading doxorubicin hydrochloride for cancer therapy, Micro & Nano Letters, 2019.
  10. Goh, S.C., et al., Polydopamine-polyethylene glycol-albumin antifouling coatings on multiple substrates: variations, Journal of Materials Chemistry B, 2018.
  11. Kawanami, T., et al., A novel diclofenac-hydrogel conjugate system for intraarticular sustained release: Development of 2-pyridylamino-substituted 1-phenylethanol (PAPE) and its derivatives as tunable traceless linkers, International Journal of Pharmaceutics, 2020, 585, 119519.
  12. Goor, O., et al., Introduction of anti-fouling coatings at the surface of supramolecular elastomeric materials via post-modification of reactive supramolecular additives, Polymer Chemistry, 2017.
  13. Goh, S.C.M, Universal Aqueous-Based Antifouling Coatings For Multi-Material Devices, McMaster University, 2017.
  14. Kim, H.C., et al., Highly stable and reduction responsive micelles from a novel polymeric surfactant with a repeating disulfide-based gemini structure for efficient drug delivery, Polymer, 2017.
  15. Bai, J., et al., Triple-Modal Imaging of Magnetically-Targeted Nanocapsules in Solid Tumours In Vivo, Theranostics, 2016, 6(3):342-356.
  16. Jain, S., et al., Estradiol functionalized multi-walled carbon nanotubes as renovated strategy for efficient gene delivery, RSC Advances, 2016, 6(13):10792-801.
  17. Yu, H., et al., Polyvinylpyrrolidone functionalization induces deformable structure of graphene oxide nanosheets for lung-targeting delivery, Nano Today, 2021, V. 38.
  18. Liao, H. T., et al, A bioactive multi-functional heparin-grafted aligned poly(lactide-co-glycolide)/curcumin nanofiber membrane to accelerate diabetic wound healing, Materials Science and Engineering: C, 2021, V. 120.
  19. Mou, J., et al., A New Green Titania with Enhanced NIR Absorption for Mitochondria-Targeted Cancer Therapy, Theranostics, 2017, 7(6):1531-1542.
  20. Day, J.R., et al., The impact of functional groups of poly(ethylene glycol) macromers on the physical properties of photo-polymerized hydrogels and the local inflammatory response in the host, Acta Biomaterialia, 2018, V. 67, P. 42-52.
  21. Yu, W., et al., PEGylated recombinant human interferon-ω as a long-acting antiviral agent: Structure, antiviral activity and pharmacokinetics. Antiviral Research, 2014, 108, p. 142-147.
  22. Tang, L., et al., Separation and detection of bis-maleimide-polyethylene glycol and mono-maleimide-polyethylene glycol by reversed-phase high pressure liquid chromatography, Journal of Chromatography A, 2012, 1246, p: 117–122.
  23. Ozdemir, T., et al., Bottom-up assembly of salivary gland microtissues for assessing myoepithelial cell function, Biomaterials, 2017, V. 142, P. 124-135.
  24. He, X., et al., Intelligent lesion blood brain barrier targeting nano-missiles for Alzheimer’s disease treatment by anti-neuroinflammation and neuroprotection. Acta Pharmaceutica Sinica B. 2022.
  25. Gottipati, A., et al., Gelatin Based Polymer Cell Coating Improves Bone Marrow-Derived Cell Retention in the Heart after Myocardial Infarction, Stem Cell Reviews and Reports, 2019.
  26. Day, J.R., et al., The impact of functional groups of poly(ethylene glycol) macromers on the physical properties of photo-polymerized hydrogels and the local inflammatory response in the host, Acta Biomaterialia, 2018, V. 67, P. 42-52.
  27. Tan, J.J., et al., Impact of substrate stiffness on dermal papilla aggregates in microgels, Biomaterials science, 2018.
  28. Marozas, I. A., et al., Adaptable boronate ester hydrogels with tunable viscoelastic spectra to probe timescale dependent mechanotransduction, Biomaterials, 2019, 223.
  29. Yu, T., et al., An injectable and self-healing hydrogel with dual physical crosslinking for in-situ bone formation, Materials Today Bio, 19, 2023.
  30. Huang, Y., et al., Structural aspects controlling the mechanical and biological properties of tough, double network hydrogels. Acta Biomaterialia. 2022.
  31. Liu, S., et al., Effects of the proportion of two different cross-linkers on the material and biological properties of enzymatically degradable PEG hydrogels, Polymer Degradation and Stability, 2020, V. 172.
  32. Thai, V. L., et al., Hydrogel degradation promotes angiogenic and regenerative potential of cell spheroids for wound healing, Materials Today Bio, 22, 2023.
  33. Bae, S.W., et al., Dynamic, Bioresponsive Hydrogels via Changes in DNA Aptamer Conformation, Macromolecular bioscience, 2019, 19(2):1800353.
  34. Stoichevska, V., et al., Engineering specific chemical modification sites into a collagen‐like protein from Streptococcus pyogenes, Journal of Biomedical Materials Research Part A., 2017.
  35. Li, H., et al., Bioengineered three-dimensional scaffolds to elucidate the effects of material biodegradability on cell behavior using POSS-PEG hybrid hydrogels, Polymer Degradation and Stability, 2019, 164:118-26.
  36. Day, J.R., et al., The impact of functional groups of poly(ethylene glycol) macromers on the physical properties of photo-polymerized hydrogels and the local inflammatory response in the host, Acta Biomaterialia, 2018, 67, P. 42-52.
  37. Rutz, A. L., et al., Employing PEG crosslinkers to optimize cell viability in gel phase bioinks and tailor post printing mechanical properties, Acta Biomaterialia, 2019.

Founded in 2001 by experts in PEG synthesis and PEGylation, JenKem Technology specializes exclusively in the development and manufacturing of high quality polyethylene glycol (PEG) products and derivatives, and related custom synthesis and PEGylation services. JenKem Technology is ISO 9001 and ISO 13485 certified, and adheres to ICH Q7A guidelines for GMP manufacture. The production of JenKem® PEGs is back-integrated to in-house polymerization from ethylene oxide, enabling facile traceability for regulated customers. JenKem Technology caters to the PEGylation needs of the pharmaceutical, biotechnology, medical device and diagnostics, and emerging chemical specialty markets, from laboratory scale through large commercial scale.