Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1621 2022-08-26 11:22:54 |
2 format correction + 1 word(s) 1622 2022-08-29 02:52:32 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Mirón-Barroso, S.;  Correia, J.S.;  Frampton, A.E.;  Lythgoe, M.P.;  Clark, J.;  Tookman, L.;  Ottaviani, S.;  Castellano, L.;  Porter, A.E.;  Georgiou, T.K.; et al. Polymeric Carriers. Encyclopedia. Available online: https://encyclopedia.pub/entry/26540 (accessed on 03 July 2024).
Mirón-Barroso S,  Correia JS,  Frampton AE,  Lythgoe MP,  Clark J,  Tookman L, et al. Polymeric Carriers. Encyclopedia. Available at: https://encyclopedia.pub/entry/26540. Accessed July 03, 2024.
Mirón-Barroso, Sofía, Joana S. Correia, Adam E. Frampton, Mark P. Lythgoe, James Clark, Laura Tookman, Silvia Ottaviani, Leandro Castellano, Alexandra E. Porter, Theoni K. Georgiou, et al. "Polymeric Carriers" Encyclopedia, https://encyclopedia.pub/entry/26540 (accessed July 03, 2024).
Mirón-Barroso, S.,  Correia, J.S.,  Frampton, A.E.,  Lythgoe, M.P.,  Clark, J.,  Tookman, L.,  Ottaviani, S.,  Castellano, L.,  Porter, A.E.,  Georgiou, T.K., & Krell, J. (2022, August 26). Polymeric Carriers. In Encyclopedia. https://encyclopedia.pub/entry/26540
Mirón-Barroso, Sofía, et al. "Polymeric Carriers." Encyclopedia. Web. 26 August, 2022.
Polymeric Carriers
Edit

RNA therapeutics have gained popularity due to their ability to affect targets that small molecules cannot. Additionally, they can be manufacture more rapidly and cost-effectively than small molecules or recombinant proteins. RNA therapeutics can be synthesised chemically and altered quickly, which can enable a more personalised approach to treatment. Even though a wide range of RNA therapeutics are being developed for various indications, none has reached the clinic to date. One of the main reasons for this is attributed to the lack of safe and effective delivery systems for this type of therapeutic. Polymeric carriers have been widely studied for the delivery of RNA therapeutics because of their versatility, potential multi-functionality and relative low cost.

RNA cancer therapeutics polymeric carriers

1. Introduction

Polymeric carriers have been widely studied for the delivery of RNA therapeutics because of their versatility, potential multi-functionality and relative low cost. Polymers are macromolecules that can be defined by different characteristics such as their composition, architecture, molecular mass or charge [1].

2. Polymer Composition

A variety of polymers are being developed for the delivery of RNA therapeutics (Figure 1a, Table 1. They can be classified in homopolymers, composed of only one type of monomer, or co-polymers if they include several types of monomers (Figure 1b).
Figure 1. (a) Chemical structures of commonly used polymers in RNA therapeutics. (b) Schematical illustrations of different polymer architectures and topologies.
Table 1. Polymers for RNA delivery.
Polymer Advantages Limitations Ref.
PEI High transfection efficiency High toxicity and immunogenicity [2][3]
Chitosan Biocompatibility, biodegradability, low toxicity and immunogenicity Premature release and low transfection efficiency [4][5][6]
PLL Biodegradability, high transfection efficiency Toxicity [7][8]
PLGA FDA approved, biodegradability and biocompatibility Low efficiency [9][10][11]
PAMAM Dendrimers highly efficiency Toxicity [12][13]
PBAE Biodegradability and biocompatibility Limited ability to sustain delivery over long timespans, toxicity [14][15]
PDMAEMA High transfection efficiency Non-biodegradable [16][17]
The most widely studied cationic polymer for RNA delivery is polyethyleneimine (PEI) due to its high transfection efficiency. Its primary, secondary and tertiary amines are protonated at physiological pH and enable nucleic acid complexation, cellular internalisation and endosomal escape. However, PEI presents high toxicity and immunogenicity that has hindered its translation into the clinic. Combination of PEI with poly(ethylene glycol) [2] or hydrophobic moieties such as cholesterol [3] is being studied to decrease its toxicity and enable a safe and effective delivery of RNA therapeutics.
Chitosan is a naturally sourced polysaccharide widely studied for RNA delivery due to its biocompatibility, biodegradability, low toxicity and immunogenicity. Furthermore, the ability to fine-tune several of its parameters such as the degrees of deacetylation (DDA) or its charge by altering the fractions of protonatable amine has made it appealing for the development of gene delivery systems [4]. This cationic co-polymer is composed of β-linked N-acetyl glucosamine and D-glucosamine, and its amino groups are protonated at physiological pH which allows it to interact with negatively charged nucleic acids [5]. However, these interactions with nucleic acids are not very strong and can cause premature release and low efficiency; several strategies are being developed to overcome these issues [6].
Poly(L-Lysine) (PLL) is a biodegradable homopolymer which contains primary amines that can be protonated to interact with RNA but can cause toxicity in vivo. Novel architectures, such as PLL dendrigrafts, are being developed to deliver RNA therapeutics [7]. Approaches to reduce PLL toxicity such as complexation with anionic compounds are being studied [8].
Poly(lactic-co-glycolic acid) (PLGA) is a copolymer composed of lactic and glycolic acid, widely used for drug delivery. It is FDA approved, biodegradable and biocompatible. Its tuneable properties, such as the ratio of lactic acid to glycolic acid, enable the controlled release of encapsulated therapeutics. Systems based on PLGA are being developed for the delivery of RNA therapeutics [9][10]. The combination of PLGA with cationic polymers such as PEI are being studied to improve RNA condensation [11].
Polyamidoamine (PAMAM) dendrimers have also been developed for delivery of RNA [12]. Strategies, such as grafting targeting moieties, are being studied to increase their selectivity towards diseased cells [13][18]. Higher dendrimer generations lead to higher efficacy, but also increased toxicity; the balance between these parameters is key in the design of PAMAM gene delivery systems [19].
Poly(β-amino esters) (PBAE) are biodegradable and biocompatible polymers that can be easily modified. The application of PBAE for RNA delivery is being studied. However, there is a need to optimise the balance between their toxicity and efficiency in vivo [14] as well as their stability in order to accomplish their translation into the clinic [15].
Poly[2-(dimethylamino)ethyl methacrylate] (PDMAEMA) is a promising polymer for delivery of RNA therapeutics. It contains tertiary amines that interact with RNA and allow endosomal escape and cellular internalisation [16][17][20].
A common co-monomer that is often introduced to cationic polymer chains is poly(ethylene glycol) (PEG) because of its biocompatibility. It is present in the formulation of many FDA approved products, such as the COVID-19 vaccines. Thus, many studies reported that by introducing PEG or PEG based monomers like oligo(ethylene glycol) methyl ether methacrylate (OEGMA) resulted in decreased toxicity and prolonged circulation time [16][17][20][21].

2.1. Polymer Architectures

In copolymers, monomers can be arranged in different manners which can results in statistical, alternating, gradient and block copolymers. The effect of the different arrangement of monomers on gene delivery efficiency is being studied [22]. Statistical copolymers that include cationic and non-ionic or anionic monomers have reported higher efficacy and toxicity than block copolymers with the same composition. This might be due to the lack of a hydrophilic block that hinders interaction with cellular membranes. However, block copolymers were observed to have increased colloidal stability probably due to the steric hindrance of the hydrophilic blocks [23][24][25].
Polymers can also present different spatial architectures (Figure 1B). In linear polymers monomers are only bond to one or two other monomers. Incorporation of crosslinkers that bind more than two monomers can result in different architectures such as stars, grafts, branched polymers or dendrimers [22].
Branched architectures have been shown to increase efficiency over linear polymers [26]. They include branched copolymers in which secondary polymer chains are linked to a primary backbone and dendrimers [1].
Dendrimers consist of a central core and highly branched arms. They are synthesised in a controlled manner and are characterised by their generation which refers to the number of branches additions. With each generation the volume and surface increase as well as the number of terminal groups. Generally, dendrimers are characterised by a very narrow size distribution. The most commonly used dendrimers for gene delivery are poly(amidoamine) (PAMAM) [12][13][18][19] and poly(propylenimine) (PPI) [27][28] dendrimers.
Another architecture emerging for promising delivery systems for nucleic acids are star copolymers. They consist of several linear homo- or co-polymers bond to a core forming a star shaped structure [29][30][31][32]. Star shaped polymers have reported higher transfection efficiencies than their linear counterparts which can be due to a higher condensation of the nucleic acids [33].

2.2. Molecular Mass

Molecular mass distribution of polymers is one of the most studied characteristics. Increasing molecular mass have generally shown to increase efficiency and cytotoxicity [34]. This can be due to the increase of the probability of interaction with cellular membranes. Molecular mass distribution can also impact the ability of polymers to escape the endosome. Higher molecular mass polymers reported increased endosomal escape [35]. Optimizing the molecular mass to balance efficiency and toxicity is a key consideration in the design of polymeric delivery systems [26].

2.3. Polyplexes Formulation

The formation of polyplexes is mostly driven by electrostatic interactions. A key parameter in polyplex formulation is the N/P ratio (the ratio of nitrogen groups of the polymer to the phosphate groups of the nucleic acid). Higher N/P ratios lead to higher transfection efficiency and colloidal stability due to the electrostatic repulsion of the positive charges in the surface of the polyplexes. However, high N/P ratios can also cause toxicity as a result of the interactions of the polymer’s positive charges with negatively charged proteins and cellular membranes [36].
Other preparation methods, such as the buffer used or the mixing of reagents, can have an influence on the physicochemical characteristics of the polyplexes and ultimately their transfection efficiency. Mixing the reagents by pipetting instead of dropwise addition leads to lower hydrodynamic diameters and narrower size distributions, as well as lower transfection efficiency [37].

2.4. Characterisation Techniques

In order to reach the clinical setting, polyplexes need to be thoroughly characterised. Size is one of the key parameters that has a great impact on the pharmacokinetic profile of polyplexes. Several techniques have been developed to evaluate the size distribution of nanosised systems.
Dynamic light scattering (DLS) determines the hydrodynamic diameter of the polyplexes by relating it to their Brownian motion using the Stokes–Einstein equation. DLS is ideal to determine the hydrodynamic diameter distribution of mono-population, nanosised particles. Fluorescent correlation spectroscopy (FCS) is also used measure the size and diffusion coefficient of fluorescently labelled polyplexes [38].
Atomic force microscopy (AFM) allows the visualisation particles’ surface and morphology at high resolutions scanning the sample with a cantilever tip. Scanning electron microscopy (SEM) is used to determine the surface, morphology and composition by creating images from the scattered electrons. Transmission electron microscopy (TEM) provides information on the inner structure, size and morphology as well as on the cellular internalisation of the polyplexes. It creates images from the electrons transmitted through the sample [39].
The charge at the surface of the polyplexes can be determined by their zeta potential. The zeta potential can be measured by electrophoretic mobility, observing how the particles move when an electric field is applied. This parameter is crucial for the polyplexes’ stability as well as its’ safety and efficiency [40].
The molecular mass and composition are also key parameters for polymer characterisation. Gel permeation chromatography (GPC) is the standard method for determining the molecular mass. Nuclear magnetic resonance (NMR) spectroscopy can also be used to determine the polymer’s molecular mass as well as to accurately determine monomer composition for copolymers [41]. Fourier transform infrared spectrometry (FTIR) can also be used to characterise polymers and determine their composition [42].

References

  1. Kumar, R.; Chalarca, C.F.S.; Bockman, M.R.; Van Bruggen, C.; Grimme, C.J.; Dalal, R.J.; Hanson, M.G.; Hexum, J.K.; Reineke, T.M. Polymeric Delivery of Therapeutic Nucleic Acids. Chem. Rev. 2021, 121, 11527–11652.
  2. Safari, F.; Tamaddon, A.M.; Zarghami, N.; Abolmali, S.; Akbarzadeh, A. Polyelectrolyte complexes of hTERT siRNA and polyethyleneimine: Effect of degree of PEG grafting on biological and cellular activity. Artif. Cells Nanomed. Biotechnol. 2016, 44, 1561–1568.
  3. Wu, P.K.; Luo, X.P.; Wu, H.; Zhang, Q.Y.; Wang, K.K.; Sun, M.J.; Oupicky, D. Combined Hydrophobization of Polyethylenimine with Cholesterol and Perfluorobutyrate Improves siRNA Delivery. Bioconjug. Chem. 2020, 31, 698–707.
  4. Alameh, M.; Lavertu, M.; Tran-Khanh, N.; Chang, C.-Y.; Lesage, F.; Bail, M.; Darras, V.; Chevrier, A.; Buschmann, M.D. siRNA Delivery with Chitosan: Influence of Chitosan Molecular Weight, Degree of Deacetylation, and Amine to Phosphate Ratio on in Vitro Silencing Efficiency, Hemocompatibility, Biodistribution, and in Vivo Efficacy. Biomacromolecules 2018, 19, 112–131.
  5. Rudzinski, W.E.; Palacios, A.; Ahmed, A.; Lane, M.A.; Aminabhavi, T.M. Targeted delivery of small interfering RNA to colon cancer cells using chitosan and PEGylated chitosan nanoparticles. Carbohydr. Polym. 2016, 147, 323–332.
  6. De Souza, R.; Dalla Picola, I.P.; Shi, Q.; Petronio, M.S.; Benderdour, M.; Fernandes, J.C.; Lima, A.M.F.; Martins, G.O.; Martinez, A.M.; Tiera, V.A.D.; et al. Diethylaminoethyl-chitosan as an efficient carrier for siRNA delivery: Improving the condensation process and the nanoparticles properties. Int. J. Biol. Macromol. 2018, 119, 186–197.
  7. Ye, L.; Liu, H.; Fei, X.; Ma, D.; He, X.; Tang, Q.; Zhao, X.; Zou, H.; Chen, X.; Kong, X.; et al. Enhanced endosomal escape of dendrigraft poly-L-lysine polymers for the efficient gene therapy of breast cancer. Nano Res. 2022, 15, 1135–1144.
  8. Kodama, Y.; Kuramoto, H.; Mieda, Y.; Muro, T.; Nakagawa, H.; Kurosaki, T.; Sakaguchi, M.; Nakamura, T.; Kitahara, T.; Sasaki, H. Application of biodegradable dendrigraft poly-L-lysine to a small interfering RNA delivery system. J. Drug Target. 2017, 25, 49–57.
  9. Hazekawa, M.; Nishinakagawa, T.; Mori, T.; Yoshida, M.; Uchida, T.; Ishibashi, D. Preparation of siRNA–PLGA/Fabʹ–PLGA mixed micellar system with target cell-specific recognition. Sci. Rep. 2021, 11, 16789.
  10. Devulapally, R.; Sekar, N.M.; Sekar, T.V.; Foygel, K.; Massoud, T.F.; Willmann, J.K.; Paulmurugan, R. Polymer Nanoparticles Mediated Codelivery of AntimiR-10b and AntimiR-21 for Achieving Triple Negative Breast Cancer Therapy. ACS Nano 2015, 9, 2290–2302.
  11. Bilecen, D.S.; Rodriguez-Cabello, J.C.; Uludag, H.; Hasirci, V. Construction of a PLGA based, targeted siRNA delivery system for treatment of osteoporosis. J. Biomater. Sci. Polym. Ed. 2017, 28, 1859–1873.
  12. Ghaffari, M.; Dehghan, G.; Baradaran, B.; Zarebkohan, A.; Mansoori, B.; Soleymani, J.; Dolatabadi, J.E.N.; Hamblin, M.R. Co-delivery of curcumin and Bcl-2 siRNA by PAMAM dendrimers for enhancement of the therapeutic efficacy in HeLa cancer cells. Colloids Surf. B Biointerfaces 2020, 188, 110762.
  13. Li, T.J.; Chen, Q.W.; Zheng, Y.; Zhang, P.F.; Chen, X.X.; Lu, J.N.; Lv, Y.H.; Sun, S.G.; Zeng, W.X. PAMAM-cRGD mediating efficient siRNA delivery to spermatogonial stem cells. Stem Cell Res. Ther. 2019, 10, 399.
  14. Liu, Y.; Chen, J.; Tang, Y.; Li, S.H.; Dou, Y.S.; Zheng, J.W. Synthesis and Characterization of Quaternized Poly(beta-amino ester) for Highly Efficient Delivery of Small Interfering RNA. Mol. Pharm. 2018, 15, 4558–4567.
  15. Dosta, P.; Ramos, V.; Borros, S. Stable and efficient generation of poly(beta-amino ester)s for RNAi delivery. Mol. Syst. Des. Eng. 2018, 3, 677–689.
  16. Üzgün, S.; Akdemir, Ö.; Hasenpusch, G.; Maucksch, C.; Golas, M.M.; Sander, B.; Stark, H.; Imker, R.; Lutz, J.-F.; Rudolph, C. Characterization of Tailor-Made Copolymers of Oligo(ethylene glycol) Methyl Ether Methacrylate and N,N-Dimethylaminoethyl Methacrylate as Nonviral Gene Transfer Agents: Influence of Macromolecular Structure on Gene Vector Particle Properties and Transfection Efficiency. Biomacromolecules 2010, 11, 39–50.
  17. Georgiou, T.K.; Vamvakaki, M.; Phylactou, L.A.; Patrickios, C.S. Synthesis, Characterization, and Evaluation as Transfection Reagents of Double-Hydrophilic Star Copolymers: Effect of Star Architecture. Biomacromolecules 2005, 6, 2990–2997.
  18. Ayatollahi, S.; Salmasi, Z.; Hashemi, M.; Askarian, S.; Oskuee, R.K.; Abnous, K.; Ramezani, M. Aptamer-targeted delivery of Bcl-xL shRNA using alkyl modified PAMAM dendrimers into lung cancer cells. Int. J. Biochem. Cell Biol. 2017, 92, 210–217.
  19. Li, J.; Liang, H.; Liu, J.; Wang, Z. Poly (amidoamine) (PAMAM) dendrimer mediated delivery of drug and pDNA/siRNA for cancer therapy. Int. J. Pharm. 2018, 546, 215–225.
  20. Tan, J.-K.Y.; Choi, J.L.; Wei, H.; Schellinger, J.G.; Pun, S.H. Reducible, dibromomaleimide-linked polymers for gene delivery. Biomater. Sci. 2015, 3, 112–120.
  21. Porfiryeva, N.; Moustafine, R.I.; Khutoryanskiy, V.V. PEGylated Systems in Pharmaceutics. Polym. Sci. Ser. C 2020, 62, 62–74.
  22. Rinkenauer, A.C.; Schubert, S.; Traeger, A.; Schubert, U.S. The influence of polymer architecture on in vitro pDNA transfection. J. Mater. Chem. B 2015, 3, 7477–7493.
  23. Tan, E.; Lv, J.; Hu, J.; Shen, W.; Wang, H.; Cheng, Y. Statistical versus block fluoropolymers in gene delivery. J. Mater. Chem. B 2018, 6, 7230–7238.
  24. Sprouse, D.; Reineke, T.M. Investigating the Effects of Block versus Statistical Glycopolycations Containing Primary and Tertiary Amines for Plasmid DNA Delivery. Biomacromolecules 2014, 15, 2616–2628.
  25. Deshpande, M.C.; Garnett, M.; Vamvakaki, M.; Bailey, L.; Armes, S.; Stolnik, S. Influence of polymer architecture on the structure of complexes formed by PEG–tertiary amine methacrylate copolymers and phosphorothioate oligonucleotide. J. Control. Release 2002, 81, 185–199.
  26. Synatschke, C.V.; Schallon, A.; Jérôme, V.; Freitag, R.; Müller, A.H.E. Influence of Polymer Architecture and Molecular Weight of Poly(2-(dimethylamino)ethyl methacrylate) Polycations on Transfection Efficiency and Cell Viability in Gene Delivery. Biomacromolecules 2011, 12, 4247–4255.
  27. Lee, G.J.; Ryu, K.; Kim, K.; Choi, J.-Y.; Kim, T.-I. Crosslinked Polypropylenimine Dendrimers With Bioreducible Linkages for Gene Delivery Systems and Their Reductive Degradation Behaviors. Macromol. Biosci. 2015, 15, 1595–1604.
  28. Somani, S.; Laskar, P.; Altwaijry, N.; Kewcharoenvong, P.; Irving, C.; Robb, G.; Pickard, B.; Dufès, C. PEGylation of polypropylenimine dendrimers: Effects on cytotoxicity, DNA condensation, gene delivery and expression in cancer cells. Sci. Rep. 2018, 8, 9410.
  29. Georgiou, T.K. Star polymers for gene delivery. Polym. Int. 2014, 63, 1130–1133.
  30. Boyer, C.; Teo, J.; Phillips, P.; Erlich, R.B.; Sagnella, S.; Sharbeen, G.; Dwarte, T.; Duong, H.T.T.; Goldstein, D.; Davis, T.P.; et al. Effective Delivery of siRNA into Cancer Cells and Tumors Using Well-Defined Biodegradable Cationic Star Polymers. Mol. Pharm. 2013, 10, 2435–2444.
  31. Teo, J.; McCarroll, J.A.; Boyer, C.; Youkhana, J.; Sagnella, S.M.; Duong, H.T.; Liu, J.; Sharbeen, G.; Goldstein, D.; Davis, T.P.; et al. A Rationally Optimized Nanoparticle System for the Delivery of RNA Interference Therapeutics into Pancreatic Tumors in Vivo. Biomacromolecules 2016, 17, 2337–2351.
  32. Cho, H.Y.; Srinivasan, A.; Hong, J.; Hsu, E.; Liu, S.; Shrivats, A.; Kwak, D.; Bohaty, A.K.; Paik, H.-J.; Hollinger, J.O.; et al. Synthesis of Biocompatible PEG-Based Star Polymers with Cationic and Degradable Core for siRNA Delivery. Biomacromolecules 2011, 12, 3478–3486.
  33. Liao, X.; Walden, G.; Falcon, N.D.; Donell, S.; Raxworthy, M.J.; Wormstone, M.; Riley, G.P.; Saeed, A. A direct comparison of linear and star-shaped poly(dimethylaminoethyl acrylate) polymers for polyplexation with DNA and cytotoxicity in cultured cell lines. Eur. Polym. J. 2017, 87, 458–467.
  34. Monnery, B.D.; Wright, M.; Cavill, R.; Hoogenboom, R.; Shaunak, S.; Steinke, J.H.; Thanou, M. Cytotoxicity of polycations: Relationship of molecular weight and the hydrolytic theory of the mechanism of toxicity. Int. J. Pharm. 2017, 521, 249–258.
  35. Kongkatigumjorn, N.; Cortez-Jugo, C.; Czuba, E.; Wong, A.S.M.; Hodgetts, R.Y.; Johnston, A.P.R.; Such, G.K. Probing Endosomal Escape Using pHlexi Nanoparticles. Macromol. Biosci. 2017, 17, 1600248.
  36. Mahajan, S.; Tang, T. Polyethylenimine–DNA Ratio Strongly Affects Their Nanoparticle Formation: A Large-Scale Coarse-Grained Molecular Dynamics Study. J. Phys. Chem. B 2019, 123, 9629–9640.
  37. Pezzoli, D.; Giupponi, E.; Mantovani, D.; Candiani, G. Size matters for in vitro gene delivery: Investigating the relationships among complexation protocol, transfection medium, size and sedimentation. Sci. Rep. 2017, 7, 44134.
  38. Troiber, C.; Kasper, J.C.; Milani, S.; Scheible, M.; Martin, I.; Schaubhut, F.; Küchler, S.; Rädler, J.; Simmel, F.C.; Friess, W.; et al. Comparison of four different particle sizing methods for siRNA polyplex characterization. Eur. J. Pharm. Biopharm. 2013, 84, 255–264.
  39. Rumschöttel, J.; Kosmella, S.; Prietzel, C.; Appelhans, D.; Koetz, J. Change in size, morphology and stability of DNA polyplexes with hyperbranched poly(ethyleneimines) containing bulky maltose units. Colloids Surf. B Biointerfaces 2016, 138, 78–85.
  40. Varenne, F.; Coty, J.-B.; Botton, J.; Legrand, F.-X.; Hillaireau, H.; Barratt, G.; Vauthier, C. Evaluation of zeta potential of nanomaterials by electrophoretic light scattering: Fast field reversal versus Slow field reversal modes. Talanta 2019, 205, 120062.
  41. Izunobi, J.U.; Higginbotham, C.L. Polymer Molecular Weight Analysis by 1H NMR Spectroscopy. J. Chem. Educ. 2011, 88, 1098–1104.
  42. Dwyer, J.L.; Zhou, M. Polymer Characterization by Combined Chromatography-Infrared Spectroscopy. Int. J. Spectrosc. 2011, 2011, 694645.
More
Information
Subjects: Polymer Science
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , , , , , ,
View Times: 459
Entry Collection: Biopharmaceuticals Technology
Revisions: 2 times (View History)
Update Date: 29 Aug 2022
1000/1000
Video Production Service