Nanomaterials for Viral Diseases Diagnosis, Prevention, and Treatment: Comparison
Please note this is a comparison between Version 2 by Peter Tang and Version 1 by Hyun-Ouk Kim.

Nanomaterials can be tailored for specific uses by modulating physical and chemical properties, including size, morphology, surface charge, and solubility. Due to these controllable properties, nanomaterials have been used in biosensors to potentiate target-specific reactions that respond to biochemical environments, such as temperature, pH, and the presence of enzymes.

  • nanomaterials
  • viral diseases
  • vaccines
  • diagnosis
  • therapeutics

1. Introduction

Since 2009, numerous infectious diseases have been occurring periodically, including those caused due to infection by influenza A virus (IAV; H1N1), West African Ebola virus, Middle East respiratory syndrome coronavirus (MERS-CoV), and the recent severe acute respiratory syndrome coronavirus 2 [SARS-CoV-2; known as coronavirus disease (COVID-19)]. SARS-CoV-2 has infected numerous individuals, which has resulted in millions of deaths. Owing to the rapid global spread of SARS-CoV-2 infections and the consequent damage, the World Health Organization (WHO) declared a COVID pandemic [1].
Strict measures, such as quarantining infected individuals, and technology-based approaches have been employed to prevent the further spread of COVID. Various types of healthcare technologies, including on-site rapid testing kits, vaccines, and antiviral therapeutics for COVID, have been developed and applied in a short period of time via Emergency Use Authorization (EUA) approvals. Most governments have carried out extensive testing to identify potential infections and have rolled out mass vaccination programs to prevent infection and reduce economic loss and human damage [2]. FDA-approved antiviral agents, such as remdesivir, have been used to treat SAR-CoV-2-infected patients, to reduce the number of severe cases that pose a serious burden on public health systems. It is now being recognized that the peak of the pandemic has passed, although the morbidity and mortality rates require further reduction. However, infection rates are fluctuating due to sporadic, rapidly rising infections with viral variants. The current pandemic situation is a potent warning indicating that the emergence or re-emergence of viral diseases is unpredictable but inevitable. Therefore, it is imperative to develop technology-based countermeasures for diagnosis, prevention, and treatment to cope with potential viral diseases in the future.
The introduction of nanomaterials in the development of technological countermeasures has advantages in many respects, such as facile functionalization, control of surface chemistry, and availability as a delivery carrier [3]. Nanomaterials can be tailored for specific uses by modulating physical and chemical properties, including size, morphology, surface charge, and solubility. Due to these controllable properties, nanomaterials have been used in biosensors to potentiate target-specific reactions that respond to biochemical environments, such as temperature, pH, and the presence of enzymes (Table 1) [4]. Furthermore, drug delivery carriers constructed with biocompatible nanomaterials have been actively researched to improve the efficacy of therapeutics, especially in cancer therapy. Nanomaterials can be applied in diagnosis, prophylaxis, and treatment systems to combat infectious diseases [5] (Figure 1). 
Table 1. Nanomaterial-based diagnostics for emerging and re-emerging viral diseases.

Nanomaterials

Diagnostic Techniques

Target

LOD

Time

Ref.

Carbon nanotubes

RDT

DENV

8.4 × 102 TCID50/mL

>10 min

[6]

 

SARS-CoV-2

0.55 fg/mL

>5 min

[7]

Immunological

Influenza A Virus (H1N1)

1 PFU/ml

30 min

[8]

Graphene

Immunological

JEV/AIV

1 fM/10 fM

1 h

[9]

 

AIV

1.6 pg/mL

30 min

[10]

 

HIV-1

2.3 × 10−14 M

1 h

[11]

 

Influenza A Virus (H5N1)

25 PFU/mL

15 min

[12]

 

Zika Virus

450 pmol/L

5 min

[13]

AuNPs

Optical

SARS-CoV-2

0.18 ng/μL

>10 min

[14]

   

50 RNA copies per reaction

30 min

[15]

   

4 copies/μL

40 min

[16]

 

Hepatitis B virus

100 fg/mL

10–15 min

[17]

Immunological

SARS-CoV-2

370 vp/mL

15 min

[18]

   

0.08 ng/mL

30 min

[19]

 

Influenza A Virus

7.8 HAU

30 min

[20]

 

Zika Virus

0.82 pmol/L

50 min

[21]

Quantum dots

ELISA

Influenza A Virus

22 pfu/mL

>35 min

[22]

 

Influenza A Virus (H5N1)

0.016 HAU

>15 min

[23]

 

SARS-CoV-2

5 pg/mL

>15 min

[24]

Immunological

HEV3

1.23 fM

20 min

[25]

 

SARS-CoV

0.1 pg/mL

1 h

[26]

Synthetic polymer

Immunological

Influenza A Virus (H1N1)

5 × 103~104 TCID50

9 min

[27]

Nanofibers

Immunological

SARS-CoV-2

0.8 pg/mL

20 min

[28]

Figure 1. Application of nanomaterials for diagnosis with advanced sensitivity and selectivity. (a) Schematic of using carbon nanotubes for H5N2 isolation and concentration, directly from in situ samples. Transmission electron microscopy (TEM) and scanning electron microscopy (SEM) images of H5N2 separated by carbon nanotubes are shown. Reproduced with permission from [30][29]. Copyright (2016) American Association for Advancement of Science. (b) Schematic representation of graphene as a sensing material for detecting SARS-CoV-2. The SARS-CoV-2 spike antibody binds to graphene and the reaction with the target is converted into an electrochemical signal. Reproduced with permission from [31][30]. Copyright (2020) American Chemical Society. (c) AuNP-based colorimetric diagnosis of coronavirus disease (COVID-19). The surface-modified AuNPs with antibodies bind to the virus, which shifts the absorption wavelength of AuNPs. The shift in absorption wavelength changes the color of AuNPs from red to purple. Reproduced with permission from [32][31]. Copyright (2020) American Chemical Society. (d) Synthesis schematic of CdSe/CdS/ZnS quantum dots (QDs) and mechanism of application in rapid diagnostic strips. TEM images and fluorescence spectrum of synthesized QDs. Influenza A virus was detected using the fluorescence emission spectrum of the QDs on the rapid diagnostic strip. Reproduced with permission from [33][32]. Copyright (2020) Elsevier.

2. Diagnosis

Diagnostic tests are a key component of any successful strategy aimed at suppressing emerging and re-emerging viral diseases and play an important role at all stages from early detection to final resolution [34][33]. Diagnostic tests appropriate for epidemic prevention and suppression are technically difficult to develop, validate, and implement and in-volve complex and time-consuming processes.
The accuracy of nucleic acid amplification tests is highly dependent on the time of obtaining the sample, as well as the type, storage, and handling of the sample. These tests can only diagnose active infections. False-negative results can occur if the sample is not obtained appropriately or if the subject is tested too soon or too late after exposure to the virus. In addition, the tests are complex and require specialized laboratory equipment and reagents, as well as specialists to perform the tests, which can be problematic due to the prolonged time required for obtaining the results.
Many researchers have attempted to overcome the limitations of reverse transcription polymerase chain reaction (RT-PCR) assays [35,36,37,38][34][35][36][37]. Antibody detection assays present several advantages over RT-PCR. Antibodies are more stable than RNA and are less degradable during transport and storage, thus reducing the risk of false-negative results. Although the research on antibody-based tests is ongoing, there are limitations to overcome. The main reason for this is the lack of specificity. Additionally, there is a lag phase from the initial virus exposure to the antibody response against infection. According to the accumulated immunological data, the antibody response peaks at approximately 11 days, indicating an insufficient time period for preventing the rapid spread of infectious diseases at early stages of infection. Therefore, antibody detection assays are less effective in diagnosing emerging and re-emerging viral diseases.
There is a need to develop new diagnostic platforms that are accurate, specific, fast, and easy to use, to facilitate rapid screening. Currently, research dynamics have shifted towards rapid diagnostics based on nanomaterials [39,40,41,42][38][39][40][41]. In this regard, nanotechnology-based applications can greatly improve the sensitivity of previously developed detection techniques, such as RT-PCR and immunoassays. Nanoparticles (NPs) have the characteristics of high adsorption capacity, the quantum size effect, and high reactivity. The large surface area of NPs can enhance detection effectiveness, as it allows efficient interaction with target analytes. Therefore, through physical or chemical bonding, nanomaterial-based diagnosis can be developed to increase selectivity and specificity and reduce detection time. Appropriately using advanced nanomaterials is the key to achieving improvements in nanotechnology. Nanomaterials are the basis for the design of a wide range of virus diagnostic tools. The unique characteristics of nanomaterials make them suitable for application in state-of-the-art virus detection technologies.

3. NP Vaccines for Emerging Viruses

Vaccines are the most effective and cost-efficient means for preventing infectious diseases. Despite the significant successes of various vaccines, the ongoing development of new, safer, and more potent vaccines is required because of the emergence of new pathogens, recurrence of old pathogens, and mutations in existing pathogens. Typically, vaccines incorporate adjuvants, which are supplementary substances that compensate for the poor immunogenicity of antigens and enhance the cellular and humoral immunity. Several nanoplatforms that improve vaccine immunogenicity by enhancing the delivery of antigens to the immune system or via a depot effect have been developed. The WHO Emergency Use Listing for combating the COVID-19 pandemic includes vaccines that incorporate a delivery system based on NPs, such as lipid NPs (LNPs) (Moderna, MA, US and Pfizer-BioNTech, NY, US) and adenovirus viral vectors (Janssen, NJ, US and AstraZeneca, UK) (Figure 2).
Figure 2. Overview of nanoplatform-based vaccines approved by the World Health Organization (WHO) for the prevention of emerging infectious diseases. Viral vectors and lipid NPs (LNPs) elicit potent immune responses by stably delivering DNA and mRNA encoding antigens, respectively, to antigen-presenting cells.
Delivery systems formulated with NPs are being used in next-generation vaccines for effectively delivering antigens and/or intrinsic immunostimulants [93,94,95,96,97][42][43][44][45][46]. These vaccines are being engineered not only for the prevention of emerging infectious diseases, but also for treating chronic diseases, such as diseases caused by hepatitis C infections [98[47][48],99], human immunodeficiency virus (HIV) infections [100[49][50],101], herpes [102][51], and cancer. Next-generation vaccines aim to induce both humoral and cellular immune responses, while being applied prophylactically and therapeutically [103,104][52][53]. Various nanoplatforms have been incorporated into delivery systems and immunogenicity-enhancing strategies, such as LNPs, polymeric NPs, nano-complexes, virus-like particles, and inorganic NPs. The main principle of delivery systems is to deliver the vaccine antigens or immunopotentiators to the antigen-presenting cells (APCs; including macrophages and dendritic cells) responsible for the induction of innate immune responses. A delivery system that is similar in size to nano-sized pathogens is expected to be advantageous for APC phagocytosis and for presenting antigens to naïve T cells in lymphoid tissues. Further, nanoparticulate delivery agents can protect the payload, deliver it in an intact native conformation to a target site within the immune system, and create a sustained release of antigens over time. In the future, the usage of such next-generation nano-delivery systems in vaccine technology is expected continue with sufficient safety and stability. Herein, we discuss the use of several representative nanoplatforms (LNPs and polymer particles) in delivery systems that enhance vaccine efficacy. We also review nanotechnology-based adjuvants that enhance the intensity and quality of cellular and humoral immune responses for the development of vaccines.

4. Treatment

The development of effective antiviral agents is essential for treating or alleviating severe symptoms and preventing death in infected patients. Timely antiviral therapy is an important measure to reduce the burden on the health care system. A variety of synthetic and natural antiviral agents have been developed, including chemical compounds, peptides, and essential oils. These agents exhibit antiviral activity against various types of viruses [139][54]. Other FDA-approved therapeutics, such as oseltamivir, zanamivir, and abacavir, are being utilized in antiviral therapy for influenza and HIV infection [140][55]. Remdesivir is now being used for the treatment of COVID-19, and its usage is associated with a significant reduction in the mortality of infected patients [141][56]. Despite the contributions of antiviral agents, there are several challenges associated with their usage, such as limited efficacy because of poor solubility, low biostability, and toxicity. Additionally, improvements can be made regarding the therapeutic effects of antiviral agents, which mostly block viral proteins and cellular receptors involved in viral infection pathways, such as site-specific delivery of antiviral agents to enhance efficacy and reduce off-target effects. Thus, there is a need for efficient delivery of antiviral agents [142][57].

References

  1. Park, S.E. Epidemiology, virology, and clinical features of severe acute respiratory syndrome -coronavirus-2 (SARS-CoV-2; coronavirus disease-19). Clin. Exp. Pediatr. 2020, 63, 119–124.
  2. Chintagunta, A.D.; Sai Krishna, M.; Nalluru, S.; Sampath Kumar, N.S. Nanotechnology: An emerging approach to combat COVID-19. Emergent Mater. 2021, 1–12.
  3. Díez-Pascual, A.M. Recent progress in antimicrobial nanomaterials. Nanomaterials 2020, 10, 2315.
  4. Huang, H.; Lovell, J.F. Advanced functional nanomaterials for theranostics. Adv. Funct. Mater. 2017, 27, 1603524.
  5. Look, M.; Bandyopadhyay, A.; Blum, J.S.; Fahmy, T.M. Application of nanotechnologies for improved immune response against infectious diseases in the developing world. Adv. Drug. Deliv. Rev. 2010, 62, 378–393.
  6. Wasik, D.; Mulchandani, A.; Yates, M.V. A heparin-functionalized carbon nanotube-based affinity biosensor for dengue virus. Biosens. Bioelectron. 2017, 91, 811–816.
  7. Shao, W.; Shurin, M.R.; Wheeler, S.E.; He, X.; Star, A. Rapid detection of SARS-CoV-2 antigens using high-purity semiconducting single-walled carbon nanotube-based field-effect transistors. ACS Appl. Mater. Interfaces 2021, 13, 10321–10327.
  8. Singh, R.; Sharma, A.; Hong, S.; Jang, J. Electrical immunosensor based on dielectrophoretically-deposited carbon nanotubes for detection of influenza virus h1n1. Analyst 2014, 139, 5415–5421.
  9. Roberts, A.; Chauhan, N.; Islam, S.; Mahari, S.; Ghawri, B.; Gandham, R.K.; Majumdar, S.; Ghosh, A.; Gandhi, S. Graphene functionalized field-effect transistors for ultrasensitive detection of japanese encephalitis and avian influenza virus. Sci. Rep. 2020, 10, 1–12.
  10. Huang, J.; Xie, Z.; Xie, Z.; Luo, S.; Xie, L.; Huang, L.; Fan, Q.; Zhang, Y.; Wang, S.; Zeng, T. Silver nanoparticles coated graphene electrochemical sensor for the ultrasensitive analysis of avian influenza virus h7. Anal. Chim. Acta 2016, 913, 121–127.
  11. Gong, Q.; Wang, Y.; Yang, H. A sensitive impedimetric DNA biosensor for the determination of the hiv gene based on graphene-nafion composite film. Biosens. Bioelectron. 2017, 89, 565–569.
  12. Li, C.; Zou, Z.; Liu, H.; Jin, Y.; Li, G.; Yuan, C.; Xiao, Z.; Jin, M. Synthesis of polystyrene-based fluorescent quantum dots nanolabel and its performance in h5n1 virus and SARS-CoV-2 antibody sensing. Talanta 2021, 225, 122064.
  13. Afsahi, S.; Lerner, M.B.; Goldstein, J.M.; Lee, J.; Tang, X.; Bagarozzi, D.A., Jr.; Pan, D.; Locascio, L.; Walker, A.; Barron, F.; et al. Novel graphene-based biosensor for early detection of zika virus infection. Biosens. Bioelectron. 2018, 100, 85–88.
  14. Moitra, P.; Alafeef, M.; Dighe, K.; Frieman, M.B.; Pan, D. Selective naked-eye detection of SARS-CoV-2 mediated by n gene targeted antisense oligonucleotide capped plasmonic nanoparticles. ACS Nano 2020, 14, 7617–7627.
  15. Jiang, Y.; Hu, M.; Liu, A.A.; Lin, Y.; Liu, L.; Yu, B.; Zhou, X.; Pang, D.W. Detection of SARS-CoV-2 by crispr/cas12a-enhanced colorimetry. ACS Sens. 2021, 6, 1086–1093.
  16. Zhang, Y.; Chen, M.; Liu, C.; Chen, J.; Luo, X.; Xue, Y.; Liang, Q.; Zhou, L.; Tao, Y.; Li, M.; et al. Sensitive and rapid on-site detection of SARS-CoV-2 using a gold nanoparticle-based high-throughput platform coupled with crispr/cas12-assisted rt-lamp. Sens. Actuators B Chem. 2021, 345, 130411.
  17. Kim, J.; Oh, S.Y.; Shukla, S.; Hong, S.B.; Heo, N.S.; Bajpai, V.K.; Chun, H.S.; Jo, C.H.; Choi, B.G.; Huh, Y.S.; et al. Heteroassembled gold nanoparticles with sandwich-immunoassay lspr chip format for rapid and sensitive detection of hepatitis b virus surface antigen (hbsag). Biosens. Bioelectron. 2018, 107, 118–122.
  18. Huang, L.; Ding, L.; Zhou, J.; Chen, S.; Chen, F.; Zhao, C.; Xu, J.; Hu, W.; Ji, J.; Xu, H.; et al. One-step rapid quantification of SARS-CoV-2 virus particles via low-cost nanoplasmonic sensors in generic microplate reader and point-of-care device. Biosens. Bioelectron. 2021, 171, 112685.
  19. Funari, R.; Chu, K.Y.; Shen, A.Q. Detection of antibodies against SARS-CoV-2 spike protein by gold nanospikes in an opto-microfluidic chip. Biosens. Bioelectron. 2020, 169, 112578.
  20. Liu, Y.J.; Zhang, L.Q.; Wei, W.; Zhao, H.Y.; Zhou, Z.X.; Zhang, Y.J.; Liu, S.Q. Colorimetric detection of influenza a virus using antibody-functionalized gold nanoparticles. Analyst 2015, 140, 3989–3995.
  21. Steinmetz, M.; Lima, D.; Viana, A.G.; Fujiwara, S.T.; Pessoa, C.A.; Etto, R.M.; Wohnrath, K. A sensitive label-free impedimetric DNA biosensor based on silsesquioxane-functionalized gold nanoparticles for zika virus detection. Biosens. Bioelectron. 2019, 141, 111351.
  22. Bai, Z.K.; Wei, H.J.; Yang, X.S.; Zhu, Y.H.; Peng, Y.J.; Yang, J.; Wang, C.W.; Rong, Z.; Wang, S.Q. Rapid enrichment and ultrasensitive detection of influenza a virus in human specimen using magnetic quantum dot nanobeads based test strips. Sens. Actuators B Chem. 2020, 325, 128780.
  23. Wu, F.; Yuan, H.; Zhou, C.H.; Mao, M.; Liu, Q.; Shen, H.B.; Cen, Y.; Qin, Z.F.; Ma, L.; Li, L.S. Multiplexed detection of influenza a virus subtype h5 and h9 via quantum dot-based immunoassay. Biosens. Bioelectron. 2016, 77, 464–470.
  24. Wang, C.; Yang, X.; Zheng, S.; Cheng, X.; Xiao, R.; Li, Q.; Wang, W.; Liu, X.; Wang, S. Development of an ultrasensitive fluorescent immunochromatographic assay based on multilayer quantum dot nanobead for simultaneous detection of SARS-CoV-2 antigen and influenza a virus. Sens. Actuators B Chem. 2021, 345, 130372.
  25. Ngo, D.B.; Chaibun, T.; Yin, L.S.; Lertanantawong, B.; Surareungchai, W. Electrochemical DNA detection of hepatitis e virus genotype 3 using pbs quantum dot labelling. Anal. Bioanal. Chem. 2021, 413, 1027–1037.
  26. Roh, C.; Jo, S.K. Quantitative and sensitive detection of sars coronavirus nucleocapsid protein using quantum dots-conjugated rna aptamer on chip. J. Chem. Technol. Biot. 2011, 86, 1475–1479.
  27. Son, S.U.; Seo, S.B.; Jane, S.; Choi, J.; Lim, J.W.; Lee, D.K.; Kim, H.; Seo, S.; Kang, T.; Jung, J.; et al. Naked-eye detection of pandemic influenza a (ph1n1) virus by polydiacetylene (pda)-based paper sensor as a point-of-care diagnostic platform. Sens. Actuators B Chem. 2019, 291, 257–265.
  28. Eissa, S.; Zourob, M. Development of a low-cost cotton-tipped electrochemical immunosensor for the detection of SARS-CoV-2. Anal. Chem. 2021, 93, 1826–1833.
  29. Yeh, Y.T.; Tang, Y.; Sebastian, A.; Dasgupta, A.; Perea-Lopez, N.; Albert, I.; Lu, H.G.; Terrones, M.; Zheng, S.Y. Tunable and label-free virus enrichment for ultrasensitive virus detection using carbon nanotube arrays. Sci. Adv. 2016, 2, e1601026.
  30. Seo, G.; Lee, G.; Kim, M.J.; Baek, S.H.; Choi, M.; Ku, K.B.; Lee, C.S.; Jun, S.; Park, D.; Kim, H.G.; et al. Rapid detection of COVID-19 causative virus (SARS-CoV-2) in human nasopharyngeal swab specimens using field-effect transistor-based biosensor (vol 14, pg 5135, 2020). ACS Nano 2020, 14, 12257–12258.
  31. Della Ventura, B.; Cennamo, M.; Minopoli, A.; Campanile, R.; Censi, S.B.; Terracciano, D.; Portella, G.; Velotta, R. Colorimetric test for fast detection of SARS-CoV-2 in nasal and throat swabs. ACS Sens. 2020, 5, 3043–3048.
  32. Yezhelyev, M.V.; Gao, X.; Xing, Y.; Al-Hajj, A.; Nie, S.M.; O’Regan, R.M. Emerging use of nanoparticles in diagnosis and treatment of breast cancer. Lancet Oncol. 2006, 7, 657–667.
  33. Kevadiya, B.D.; Machhi, J.; Herskovitz, J.; Oleynikov, M.D.; Blomberg, W.R.; Bajwa, N.; Soni, D.; Das, S.; Hasan, M.; Patel, M.; et al. Diagnostics for SARS-CoV-2 infections. Nat. Mater. 2021, 20, 593–605.
  34. Hou, H.Y.; Wang, T.; Zhang, B.; Luo, Y.; Mao, L.; Wang, F.; Wu, S.J.; Sun, Z.Y. Detection of igm and igg antibodies in patients with coronavirus disease 2019. Clin. Transl. Immunol. 2020, 9, e1136.
  35. Long, Q.X.; Liu, B.Z.; Deng, H.J.; Wu, G.C.; Deng, K.; Chen, Y.K.; Liao, P.; Qiu, J.F.; Lin, Y.; Cai, X.F.; et al. Antibody responses to SARS-CoV-2 in patients with COVID-19. Nat. Med. 2020, 26, 845.
  36. Padoan, A.; Sciacovelli, L.; Basso, D.; Negrini, D.; Zuin, S.; Cosma, C.; Faggian, D.; Matricardi, P.; Plebani, M. Iga-ab response to spike glycoprotein of SARS-CoV-2 in patients with COVID-19: A longitudinal study. Clin. Chim. Acta. 2020, 507, 164–166.
  37. Petherick, A. Developing antibody tests for SARS-CoV-2. Lancet 2020, 395, 1101–1102.
  38. Hildebrandt, N.; Spillmann, C.M.; Algar, W.R.; Pons, T.; Stewart, M.H.; Oh, E.; Susumu, K.; Diaz, S.A.; Delehanty, J.B.; Medintz, I.L. Energy transfer with semiconductor quantum dot bioconjugates: A versatile platform for biosensing, energy harvesting, and other developing applications. Chem. Rev. 2017, 117, 536–711.
  39. Kim, H.O.; Na, W.; Yeom, M.; Choi, J.; Kim, J.; Lim, J.W.; Yun, D.; Chun, H.; Park, G.; Park, C.; et al. Host cell mimic polymersomes for rapid detection of highly pathogenic influenza virus via a viral fusion and cell entry mechanism. Adv. Funct. Mater. 2018, 28, 1800960.
  40. Farzin, L.; Shamsipur, M.; Samandari, L.; Sheibani, S. Hiv biosensors for early diagnosis of infection: The intertwine of nanotechnology with sensing strategies. Talanta 2020, 206, 120201.
  41. Tymm, C.; Zhou, J.H.; Tadimety, A.; Burklund, A.; Zhang, J.X.J. Scalable COVID-19 detection enabled by lab-on-chip biosensors. Cell Mol. Bioeng. 2020, 13, 313–329.
  42. Reed, S.G.; Orr, M.T.; Fox, C.B. Key roles of adjuvants in modern vaccines. Nat. Med. 2013, 19, 1597–1608.
  43. Smith, D.M.; Simon, J.K.; Baker Jr, J.R. Applications of nanotechnology for immunology. Nat. Rev. Immunol. 2013, 13, 592–605.
  44. Kim, M.-G.; Park, J.Y.; Shon, Y.; Kim, G.; Shim, G.; Oh, Y.-K. Nanotechnology and vaccine development. Asian J. Pharm. Sci. 2014, 9, 227–235.
  45. van Riet, E.; Ainai, A.; Suzuki, T.; Kersten, G.; Hasegawa, H. Combatting infectious diseases; nanotechnology as a platform for rational vaccine design. Adv. Drug Deliv. 2014, 74, 28–34.
  46. Kim, E.; Lim, E.K.; Park, G.; Park, C.; Lim, J.W.; Lee, H.; Na, W.; Yeom, M.; Kim, J.; Song, D. Advanced nanomaterials for preparedness against (re-) emerging viral diseases. Adv. Mater. 2021, 2005927.
  47. Elberry, M.H.; Darwish, N.H.; Mousa, S.A. Hepatitis c virus management: Potential impact of nanotechnology. Virol. J. 2017, 14, 1–10.
  48. Yan, Y.; Wang, X.; Lou, P.; Hu, Z.; Qu, P.; Li, D.; Li, Q.; Xu, Y.; Niu, J.; He, Y. A nanoparticle-based hepatitis c virus vaccine with enhanced potency. J. Infect. Dis. 2020, 221, 1304–1314.
  49. Tian, Y.; Wang, H.; Liu, Y.; Mao, L.; Chen, W.; Zhu, Z.; Liu, W.; Zheng, W.; Zhao, Y.; Kong, D.; et al. A peptide-based nanofibrous hydrogel as a promising DNA nanovector for optimizing the efficacy of hiv vaccine. Nano Lett. 2014, 14, 1439–1445.
  50. Liu, Y.; Chen, C. Role of nanotechnology in hiv/aids vaccine development. Adv. Drug Deliv. Rev. 2016, 103, 76–89.
  51. Shin, H.N.; Iwasaki, A. A vaccine strategy that protects against genital herpes by establishing local memory t cells. Nature 2012, 491, 463.
  52. Luo, M.; Wang, H.; Wang, Z.; Cai, H.; Lu, Z.; Li, Y.; Du, M.; Huang, G.; Wang, C.; Chen, X. A sting-activating nanovaccine for cancer immunotherapy. Nat. Nanotechnol. 2017, 12, 648–654.
  53. Zhu, G.; Zhang, F.; Ni, Q.; Niu, G.; Chen, X. Efficient nanovaccine delivery in cancer immunotherapy. ACS Nano 2017, 11, 2387–2392.
  54. Lou, Z.Y.; Sun, Y.N.; Rao, Z.H. Current progress in antiviral strategies. Trends Pharmacol. Sci. 2014, 35, 86–102.
  55. De Clercq, E. Antiviral agents active against influenza a viruses. Nat. Rev. Drug Discov. 2006, 5, 1015–1025.
  56. Riva, L.; Yuan, S.F.; Yin, X.; Martin-Sancho, L.; Matsunaga, N.; Pache, L.; Burgstaller-Muehlbacher, S.; De Jesus, P.D.; Teriete, P.; Hull, M.V.; et al. Discovery of SARS-CoV-2 antiviral drugs through large-scale compound repurposing. Nature 2020, 586, 113.
  57. Medhi, R.; Srinoi, P.; Ngo, N.; Tran, H.V.; Lee, T.R. Nanoparticle-based strategies to combat COVID-19. ACS Appl. Nano Mater. 2020, 3, 8557–8580.
More