Complimenting CRISPR: Improving Genetic Delivery Techniques with Nanoparticles

Christina Cai and Erin Parlow

Authors

            Christina Cai is a rising high school junior. She will be attending the Texas Academy of Math and Science in the fall where she will continue to expand upon her passion for biology and other STEM courses. Although college applications are still a year away she wishes to enter a university majoring in biochemical engineering. In her free time she enjoys dancing, reading and traveling with friends and family.

Erin Parlow is a rising high school senior. She attends Cardinal Mooney Catholic High School in Marine City, Michigan. She is very excited about biology, particularly nanobiomaterials and genetics. She will soon begin the college application process and hopes to secure a spot at an accredited university where she will study biomedical engineering and material science. In her free time, Erin enjoys playing soccer, doing community service, and working with her robotics team.

 

Introduction

The recent sequencing of the human genome, and the consequential knowledge about the genetic code that defines humanity imparts novel capabilities in medicine and genetic engineering. Using biological cues, scientists can engineer an organism’s genome and constitute significant changes to cell function. However, despite the major strides that have been made in genetic understanding, the ability to feasibly make genetic changes still remains out of reach. This is in part due to the lack of efficient delivery systems to accompany genetic technology.8,31,32,37 As systems for genetic editing have been perfected, an increasing amount of attention is being delegated to developing delivery systems to accompany them. No matter how precise editing technology becomes, worth can only be measured in the accuracy, specificity, and lessened toxicity with which a system is introduced to target cells and their nuclei. In order for the potential of editing technology to be maximized, a delivery mechanism that effectively addresses these concerns is essential to further progress.

 

CRISPR Cas-9

New understanding of the CRISPR Cas-9 immunity system has led to the discovery of a more sophisticated technique for genetic editing. The CRISPR system is a major component of the immune systems of several species of bacteria and archaea.4,15,20,25By incorporating pieces of foreign RNA into its own loci, CRISPR is able to recognize and attack identical sequences of pathogenic DNA that invade bacteria.20 CRISPR can include RNA from virtually any organism.25 Upon introduction to a cell of the same organism, CRISPR is able to use RNA to target the matching DNA sequence within the cell’s genome (Figure 1).  CRISPR splices host DNA and introduces new genes or silences the expression of genes that are already present.25 One of the earliest demonstrations of CRISPR altering the genetic material of organisms was demonstrated with cell exposure to the bacterium Streptococcus thermophilus (S. thermophilus).9 In this study, RNA spacer molecules that code for immunity to different phages were derived from various mutant versions of S. thermophilus. These were incorporated into CRISPR systems and inserted into the genome of cells, which resulted in added immunity to each of the phages.9  CRISPR machinery can transpose genes from multiple sources, inducing minimal toxicity and mutagenic activity.15 As impressive as the CRISPR vision is, it still suffers from the fundamental limitation of genetic editing. This is the absence of a proper delivery technique to transfer prepared editing systems to the targeted gene.15,25

Traditionally, cellular delivery of CRISPR has been facilitated by viral or retroviral vectors. These systems make use of biological viruses that have been genetically altered to be non-pathogenic.12 Despite these changes, there are still significant instances of viral immune response, inflammation, tissue degradation, and overall toxicity to the targeted cell.21,38 These hindrances are key motivations for the development of new delivery systems that do not rely on the infectious properties of viruses and instead make use of other nonviral particles, particularly those that are defined on the nanoscale.

 

Nanoparticles and LNPs

Nanoparticles are synthetic or naturally occurring molecules that measure 1-100 nanometers in at least one dimension.32,34,27 Studies have shown that with pertinence to genetic editing, the size of the particle for delivery is relevant, with nanoscale design allowing for the development of many different particles with a varied range of specifications.26  Within the nanoscale range, there are nuances in optimal particle size. Larger particles are unable to efficiently diffuse through the membrane of the cell and deliver their product.10 Conversely, particles that are too small are unable to encapsulate required machinery.30

Among nanoparticles, the most commonly studied for potential gene editing transportation are lipid nanoparticles (LNPs). Occupying an optimal size range of 10-100 nm, LNPs are formulated by synthesis of a phospholipid bilayer with positively charged lipid molecules.18,29 These capsules mimic the physical and chemical makeup of the cell membrane, allowing LNPs to more readily be taken into the cell interior.29 Once inside the cell, LNPs protect genetic machinery until successful delivery into the nucleus. One study reported that a certain LNP-mediated delivery resulted in 90% success rate of genetic recombination within the inner mouse ear.33 Success of this magnitude could suggest that nonviral nanoparticle delivery of CRISPR might be able to overtake that of traditional viral methods.

 

Material Composition

Although LNPs form the basis of nanoparticle delivery systems, other materials have been tested to determine whether other alternative nanoparticles exist. Tests have shown that increased efficiency can be achieved through the use of gold nanoparticles rather than LNPs in certain situations.17 Gold nanoparticles with less than a 10 nm diameter are able to easily and efficiently enter the nucleus of a breast cancer cell to delivery the gene editing system.17 Virus-Like Particles (VLPs) are nanoparticle protein structures derived from bacteriophages.27 These structures behave like viruses with lessened possibility of initiating an immune response.27 It has been shown that the use of VLPs can increase the efficiency of CRISPR targeting due to ability to resist degradation in tissue fluid.27 The fact that different compositions of nanoparticles yield varying delivery results suggests that success is dependant on the material that is being used for delivery.

 

Toxicity and Limitations

Despite the positive implications that result from nanoparticle delivery systems, there are still detrimental effects that can occur. Toxicity, the measure of a material’s ability to cause cell death,  is still a concern with nanoparticle systems. LNPs are able to cause lethal toxicity in laboratory mice at certain dosages.24 Toxicity has also been shown to result from nanoparticle delivery systems that are based off the polymer polyethlylenimine (PEI).28 This polymer alone has fatal effects on cell activity, although in vitro tests have shown that when combined with DNA, these toxic effects are reduced.28 After DNA is released from PEI, toxicity returns and has adverse effects on the cell. Several other polymers and synthetic lipid carriers follow this model of toxicity.28 Other instances of nanoparticle toxicity result from their interference with enzymes that promote critical metabolic processes.24 The interference of foreign material with enzymes (enzymatic inhibition) is also a form of toxicity.24 The incidence of cell toxicity caused by nanoparticle delivery systems is a point of further research to improve efficiency and biocompatibility. This alone stands as the final barrier between the successful implementation of nanoparticles as the ideal genetic editing delivery mechanism.

 

Conclusion

Perfecting genetic editing could lead to clinic incorporation of genetic editing techniques, but is currently limited by the absence of an efficient delivery system. Nanoparticles provide promising evidence for a solution to the lack of non-toxic, consistent, and efficient delivery system. Nanoparticles of various sizes and material specifications are demonstrating an increased ability to deliver their products to ideal locations. As suitable nanoparticles are for gene delivery systems, there are still challenges that need to be addressed in order to maximize the potential of this technology. Increasing the efficiency of vector transport and abolishing any toxicity that might be related are such examples. Recent proof of concept for genetic editing with CRISPR opens the doors to the vast possibilities of genetic engineering. All that remains is the discovery of a partner to accompany and facilitate delivery. Recent trends indicate that this position might soon be occupied by a nanoparticle

 

The Future

In the future, there will be an ideal (size, material, charge) genetic delivery system that evades the foreign body reaction, prevents inflammation, and allows for targeted delivery to cells. This will give humans the ability to consistently make changes to organisms with techniques that operate in vivo. Genetic engineering has the possibility to eradicate previously terminal genetic diseases, differentiate stem cells, or correct genetic mutations in cancerous tumors. There is an unlimited possibility that begins only with us.          

 

Acknowledgments

            We would like to express the most sincere gratitude to those that have allowed this blog article to come to fruition. Special thanks to our Teacher’s Assistants, Danielle Nikal and Chia-Ying Lee, for their dedication and guidance throughout the entire process of blog writing. Finally, thanks to our Stanford Pre-Collegiate Summer Institutes Professor Dr. Jagannath Padmanabhan for inspiring us to think creatively and critically about science, and for challenging us to learn and accurately communicate the field of science.

 

References

  1. Bahal, R., Ali, N., Quijano, E., Liu, Y., Sulkowski, P., Turchick, A., . . . Glazer, P. M. (2016, October 26). In vivo correction of anaemia in β-thalassemic mice by γPNA-mediated gene editing with nanoparticle delivery. Retrieved July 03, 2017, from https://www.ncbi.nlm.nih.gov/pubmed/27782131
  2. Bowie, A. G., & Unterholzner, L. (2008). Viral evasion and subversion of pattern-recognition receptor signalling. Nature Reviews Immunology,8(12), 911-922. doi:10.1038/nri2436
  3. Chen, S., Tam, Y. Y., Lin, P. J., Sung, M. M., Tam, Y. K., & Cullis, P. R. (2016, August 10). Influence of particle size on the in vivo potency of lipid nanoparticle formulations of siRNA. Retrieved July 11, 2017, from https://www.ncbi.nlm.nih.gov/pubmed/27238441
  4. Chira, S., Gulei, D., Hajitou, A., Zimta, A. A., Cordelier, P., & Berindan-Neagoe, I. (2017, June 16). CRISPR/Cas9: Transcending the Reality of Genome Editing. Retrieved July 03, 2017, from https://www.ncbi.nlm.nih.gov/pubmed/28624197
  5. Coelho, T., Adams, D., Silva, A., Lozeron, P., Hawkins, P. N., Mant, T., . . . Suhr, O. B. (2013, August 29). Safety and Efficacy of RNAi Therapy for Transthyretin Amyloidosis — NEJM. Retrieved July 12, 2017, from http://www.nejm.org/doi/10.1056/NEJMoa1208760
  6. Cong, L., Ran, F. A., Cox, D., Lin, S., Barretto, R., Habib, N., . . . Zhang, F. (2013, February 15). Multiplex Genome Engineering Using CRISPR/Cas Systems. Retrieved July 03, 2017, from http://science.sciencemag.org/content/339/6121/819
  7. File:CRISPR-Cas9 mode of action.png. (n.d.). Retrieved July 13, 2017, from https://commons.wikimedia.org/wiki/File:CRISPR-Cas9_mode_of_action.png
  8. Cullis, P. R., & Hope, M. J. (2017, July 05). Lipid Nanoparticle Systems for Enabling Gene Therapies. Retrieved July 03, 2017, from https://www.ncbi.nlm.nih.gov/pubmed/28412170
  9. Deveau, H., Barrangou, R., Garneau, J. E., Labonté, J., Fremaux, C., Boyaval, P., . . . Moineau, S. (2008, February). Phage Response to CRISPR-Encoded Resistance in Streptococcus thermophilus. Retrieved July 03, 2017, from https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2238228/#fn1
  10. Florence, A. T., Sakthivel, T., & Toth, I. (2000, February 25). Oral uptake and translocation of a polylysine dendrimer with a lipid surface. Retrieved July 12, 2017, from http://www.sciencedirect.com/science/article/pii/S0168365999002370
  11. Fu, Y., Foden, J. A., Khayter, C., Maeder, M. L., Reyon, D., Joung, J. K., & Sander, J. D. (2013). High-frequency off-target mutagenesis induced by CRISPR-Cas nucleases in human cells. Nature Biotechnology,31(9), 822-826. doi:10.1038/nbt.2623
  12. Fujita, A., Sakagami, K., Kanegae, Y., Saito, I., & Kobayashi, I. (1995, October). Gene targeting with a replication-defective adenovirus vector. Retrieved July 11, 2017, from https://www.ncbi.nlm.nih.gov/pmc/articles/PMC189515/
  13. Gilleron, J., Querbes, W., Zeigerer, A., Borodovsky, A., Marsico, G., Schubert, U., . . . Zerial, M. (2013). Image-based analysis of lipid nanoparticle–mediated siRNA delivery, intracellular trafficking and endosomal escape. Nature Biotechnology,31(7), 638-646. doi:10.1038/nbt.2612
  14. Godbey, W., Wu, K. K., & Mikos, A. G. (2001, January 4). Poly(ethylenimine)-mediated gene delivery affects endothelial cell function and viability. Retrieved July 03, 2017, from http://www.sciencedirect.com/science/article/pii/S0142961200002039
  15. Gupta, R. M., & Musunuru, K. (2014, October). Expanding the genetic editing tool kit: ZFNs, TALENs, and CRISPR-Cas9. Retrieved July 09, 2017, from https://www.ncbi.nlm.nih.gov/pubmed/25271723
  16. Hartono, S. B., Phuoc, N. T., Yu, M., Jia, Z., Monteiro, M. J., Qiao, S., & Yu, C. (2013, November 12). Functionalized large pore mesoporous silica nanoparticles for gene delivery featuring controlled release and co-delivery. Retrieved July 03, 2017, from http://pubs.rsc.org/-/content/articlehtml/2014/tb/c3tb21015d
  17. Huo, S., Jin, S., Ma, X., Xue, X., Yang, K., Kumar, A., . . . Liang, X. (2014, May 13). Ultrasmall Gold Nanoparticles as Carriers for Nucleus-Based Gene Therapy Due to Size-Dependent Nuclear Entry. Retrieved July 03, 2017, from http://pubs.acs.org/doi/abs/10.1021/nn5008572
  18. Jackson, D. A., Juranek, S., & Lipps, H. J. (2016, December 14). Designing Nonviral Vectors for Efficient Gene Transfer and Long-Term Gene Expression. Retrieved July 11, 2017, from http://www.sciencedirect.com/science/article/pii/S152500160600181X
  19. Jayaraman, M., Ansell, S. M., Mui, B. L., Tam, Y. K., Chen, J., Du, X., . . . Hope, M. J. (2012, August 20). Maximizing the potency of siRNA lipid nanoparticles for hepatic gene silencing in vivo. Retrieved July 03, 2017, from https://www.ncbi.nlm.nih.gov/pubmed/22782619
  20. Jinek, M., Chylinski, K., Fonfara, I., Hauer, M., Doudna, J. A., & Charpentier, E. (2012, August 17). A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Retrieved July 10, 2017, from https://www.ncbi.nlm.nih.gov/pubmed/22745249
  21. Knowles, M. R., Hohneker, K. W., Zhou, Z., Olsen, J. C., Noah, T. L., Hu, P. C., . . . Jones, K. R. (1995, September 28). A controlled study of adenoviral-vector-mediated gene transfer in the nasal epithelium of patients with cystic fibrosis. Retrieved July 11, 2017, from https://www.ncbi.nlm.nih.gov/pubmed/7544439
  22. Li, L., Song, L., Yang, X., Li, X., Wu, Y., He, T., . . . Gong, C. (2016, September 29). Multifunctional “core-shell” nanoparticles-based gene delivery for treatment of aggressive melanoma. Retrieved July 03, 2017, from http://www.sciencedirect.com/science/article/pii/S0142961216305208
  23. Liu, C., & Zhang, N. (n.d.). Nanoparticles in gene therapy principles, prospects, and challenges. Retrieved July 03, 2017, from https://www.ncbi.nlm.nih.gov/pubmed/22093228
  24. Lv, H., Zhang, S., Wang, B., Cui, S., & Yan, J. (2006, May 13). Toxicity of cationic lipids and cationic polymers in gene delivery. Retrieved July 03, 2017, from http://www.sciencedirect.com/science/article/pii/S0168365906002045
  25. Musunuru, K. (2017, June 14). The Hope and Hype of CRISPR-Cas9 Genome Editing: A Review. Retrieved July 03, 2017, from https://www.ncbi.nlm.nih.gov/pubmed/28614576
  26. Putnam, D. (2006). Polymers for gene delivery across length scales. Nature Materials,5(6), 439-451. doi:10.1038/nmat1645
  27. Qazi, S., Miettinen, H. M., Wilkinson, R. A., McCoy, K., Douglas, T., & Wiedenheft, B. (2016, March 07). Programmed Self-Assembly of an Active P22-Cas9 Nanocarrier System. Retrieved July 03, 2017, from https://www.ncbi.nlm.nih.gov/pubmed/26894836
  28. Ramamoorth, M., & Narvekar, A. (2015, January). Non Viral Vectors in Gene Therapy- An Overview. Retrieved July 09, 2017, from https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4347098/
  29. Semple, S. C., Akinc, A., Chen, J., Sandhu, A. P., Mui, B. L., Cho, C. K., . . . Hope, M. J. (2010, January 17). Rational design of cationic lipids for siRNA delivery. Retrieved July 03, 2017, from https://www.ncbi.nlm.nih.gov/pubmed/20081866
  30. Semple, S. C., Klimuk, S. K., Harasym, T. O., Dos, N., Ansell, S. M., Wong, K. F., . . . Scherrer, P. (2001, February 09). Efficient encapsulation of antisense oligonucleotides in lipid vesicles using ionizable aminolipids: formation of novel small multilamellar vesicle structures. Retrieved July 12, 2017, from https://www.ncbi.nlm.nih.gov/pubmed/11342155
  31. Sun, N., Liu, Z., Huang, W., Tian, A., & Hu, S. (2013, October 12). The research of nanoparticles as gene vector for tumor gene therapy. Retrieved July 03, 2017, from http://www.sciencedirect.com/science/article/pii/S1040842813002163
  32. Wang, K., Kievit, F. M., & Zhang, M. (2016, October 19). Nanoparticles for cancer gene therapy: Recent advances, challenges, and strategies. Retrieved July 03, 2017, from http://www.sciencedirect.com/science/article/pii/S1043661816310751
  33. Wang, M., Zuris, J. A., Meng, F., Rees, H., Sun, S., Deng, P., . . . Xu, Q. (2016, March 15). Efficient delivery of genome-editing proteins using bioreducible lipid nanoparticles. Retrieved July 03, 2017, from https://www.ncbi.nlm.nih.gov/pubmed/26929348
  34. Yan, M., Wen, J., Liang, M., Lu, Y., Kamata, M., & Chen, I. S. (2015, June 02). Modulation of Gene Expression by Polymer Nanocapsule Delivery of DNA Cassettes Encoding Small RNAs. Retrieved July 09, 2017, from https://www.ncbi.nlm.nih.gov/pubmed/26035832
  35. Yang, Y., Wang, L., Bell, P., McMenamin, D., He, Z., White, J., . . . Wilson, J. M. (2016, February 1). A dual AAV system enables the Cas9-mediated correction of a metabolic liver disease in newborn mice. Retrieved July 09, 2017, from https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4786489/
  36. Yin, H., Song, C. Q., Dorkin, J. R., Zhu, L. J., Li, Y., Wu, Q., . . . Anderson, D. G. (2016, March). Therapeutic genome editing by combined viral and non-viral delivery of CRISPR system components in vivo. Retrieved July 03, 2017, from https://www.ncbi.nlm.nih.gov/pubmed/26829318
  37. Yin, H., Kauffman, K. J., & Anderson, D. G. (2017). Delivery technologies for genome editing. Nature Reviews Drug Discovery,16(6), 387-399. doi:10.1038/nrd.2016.280
  38. Zhu, B., Cao, Y., Xin, K. Q., Wang, X. T., Summerhayes, I. C., Liu, P. Y., & Tang, J. B. (2006, December). Tissue reactions of adenoviral, adeno-associated viral, and liposome-plasmid vectors in tendons and comparison with early-stage healing responses of injured flexor tendons. Retrieved July 11, 2017, from https://www.ncbi.nlm.nih.gov/pubmed/17145387
  39. Zimmermann, T. S., Lee, A. C., Akinc, A., Bramlage, B., Bumcrot, D., Fedoruk, M. N., . . . MacLachlan, I. (2006, March 26). RNAi-mediated gene silencing in non-human primates. Retrieved July 12, 2017, from https://www.nature.com/nature/journal/v441/n7089/abs/nature04688.html
  40. Zuris, J. A., Thompson, D. B., Shu, Y., Guilinger, J. P., Bessen, J. L., Hu, J. H., . . . Liu, D. R. (2014). Cationic lipid-mediated delivery of proteins enables efficient protein-based genome editing in vitro and in vivo. Nature Biotechnology,33(1), 73-80. doi:10.1038/nbt.3081