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 -- 1565 2023-10-24 16:09:48 |
2 only format change Meta information modification 1565 2023-10-25 03:26:02 |

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.
Yu, X.; Zhao, J.; Fan, D. The Application of Dissolving Microneedles in Biomedicine. Encyclopedia. Available online: https://encyclopedia.pub/entry/50747 (accessed on 21 July 2024).
Yu X, Zhao J, Fan D. The Application of Dissolving Microneedles in Biomedicine. Encyclopedia. Available at: https://encyclopedia.pub/entry/50747. Accessed July 21, 2024.
Yu, Xueqing, Jing Zhao, Daidi Fan. "The Application of Dissolving Microneedles in Biomedicine" Encyclopedia, https://encyclopedia.pub/entry/50747 (accessed July 21, 2024).
Yu, X., Zhao, J., & Fan, D. (2023, October 24). The Application of Dissolving Microneedles in Biomedicine. In Encyclopedia. https://encyclopedia.pub/entry/50747
Yu, Xueqing, et al. "The Application of Dissolving Microneedles in Biomedicine." Encyclopedia. Web. 24 October, 2023.
The Application of Dissolving Microneedles in Biomedicine
Edit

Microneedle technology has been widely used for the transdermal delivery of substances, showing improvements in drug delivery effects with the advantages of minimally invasive, painless, and convenient operation. With the development of nano- and electrochemical technology, different types of microneedles are increasingly being used in other biomedical fields. Dissolving microneedles have achieved remarkable results in the fields of dermatological treatment, disease diagnosis and monitoring, and vaccine delivery, and they have a wide range of application prospects in various biomedical fields, showing their great potential as a form of clinical treatment. 

dissolving microneedles drug delivery cancer therapy wound healing cutaneous disease

1. Introduction

Subcutaneous injection is one of the common drug administration methods in clinics. However, it can be painful and invasive, can produce sharp and biodangerous waste, and needs to be performed [1][2] by trained healthcare personnel [3]. In recent years, the strategy of transdermal drug delivery has become an important therapeutic option to complement oral, subcutaneous, and intravenous injections [4][5]. Transdermal administration avoids first-pass liver metabolism compared to subcutaneous administration; it is painless compared to injection; and it is a needle-free device that avoids the risk of disease transmission associated with the re-use of needles, thus reducing the unsafe factors commonly caused by medical waste, especially in developing countries [6][7]. However, the stratum corneum (SC), a layer of skin formed by dead corner cells and located in the outermost layer of the skin, with a thickness of 10–15 μm, is the main barrier to transdermal drug delivery, and can severely reduce the efficiency of drug delivery and limit the types of drugs that can be delivered transdermally [8][9][10].
In 1976, a new technology called microneedles was first introduced to overcome the limitations of traditional transdermal drug delivery [11]. MNs consist of micron-scale needle arrays with heights of approximately 50–900 μm. MNs can be manufactured with microfabrication techniques using a variety of materials and geometrics, because they disrupt the SC and epidermal layer and form microscale drug delivery channels without touching the nerve fibers or blood vessels located in the dermis of the epidermis and epidermal layer. Microneedle technology can significantly improve the efficiency of drug delivery [12][13]. It is also possible to increase the types of drugs transported in a painless and minimally invasive manner [14]. As a result, the device is easy to use and painless compared to traditional invasive injection and/or oral strategies with functional advantages [15][16]. MNs can not only help drug molecules bypass first-pass metabolism and gastrointestinal degradation, but also broaden the application range of their drug types, regardless of their molecular weight or hydrophilicity [17][18]. There are currently no confirmed reports that MNs cause or increase the chance of skin infections [19], nor that they affect normal skin function [20]. Numerous preclinical studies and a limited number of clinical trials have now shown that MNs can be used to deliver DNA, vaccines, insulin, and human growth hormone. In addition, MNs have been extensively studied for blood sampling, signal monitoring, and biosensors. This means that MNs have a broad market for transdermal drug delivery, vaccine preparation and biologics. Large amounts of MN have entered clinical trials for the treatment of various diseases, showing its universal effectiveness [12][21][22].
Over the past few decades, dissolving microneedles have been used in various fields, including anti-cancer, dermatology treatment, vaccine delivery, insulin delivery, and biomarker detection. Furthermore, in addition to single drug delivery, the design of microneedles allows for customizable special structures and intelligent response systems, such as controlled drug release; multi-therapy; and targeted delivery to specific sites, such as the heart [23], blood vessels [24], brain [25], etc.

2. Dissolving Microneedle for Cancer Therapy

As one of the main diseases threatening human health, the treatment of cancer has been the focus of attention all over the world. In addition to commonly used chemotherapy drugs, with the development of nanotechnology, more and more treatment methods are constantly improving the survival rates of cancer patients and reducing the recurrence of cancer. In addition, the on-demand delivery and responsive release of photothermal and photodynamic, gene, and immunotherapy drugs can also be achieved through microneedles. In the past decade, MN has been widely used for the transdermal delivery of anticancer drugs, showing improved drug utilization, reduced toxic side effects, enhanced tumor targeting ability, and low off-target toxicity [26]. Initially, microneedles were used as delivery tools for some first-line anticancer drugs to improve drug availability and target cancer cells [27][28]. Photothermal therapy is also often combined with microneedles for the treatment of cancer [29].
In addition to photothermal therapy, in recent years, combining the microneedle system with photodynamic therapy to allow for greater uniformity and depth in photosensitizer delivery to tumor sites, thus providing better treatment outcomes and an easier pathway, offers a promising strategy for the clinical application of PDT [30][31]. Tham et al. [32] developed a mesoporous nanocarrier with dual loading of photosensitizers and clinically relevant drugs for combination therapy while utilizing microneedle technology to promote its penetration into deep skin tissues. Skin fluorescence imaging shows that microneedles can encourage the nanocarrier to penetrate the skin epidermis and reach deep melanoma sites. The combination of PDT and targeted therapy with nanocarriers has been proven to have a superior therapeutic effect in the xenotransplantation of melanoma mice.
The use of MN broadens the delivery mode of cancer therapy, with various hydrophilic and hydrophobic drugs being able to penetrate the skin and reach the lesion area through microneedles, greatly improving drug utilization. In addition, MNs improve drug stability, making synergies of multiple therapies possible and reducing the need for trained operators, thus making them promising tools.

3. Dissolving Microneedles in Wound Healing

As the largest organ of the human body, the skin is easily damaged by the surrounding environment [33]. Bacterial infections, diabetic wounds, bedsores, burns, and other skin lesions pose a serious threat to people’s lives and health. Wound healing goes through a series of roughly continuous, but overlapping, stages: hemostasis, inflammation, proliferation, and remodeling [34]. Each stage is crucial to the final healing of the wound. Microneedle arrays can be used to improve delivery efficiency, thereby improving healing. MN improves the efficiency of transport by bypassing various physical and chemical barriers to deliver treatment to the target area with an improved spatial distribution [35]. In chronic wounds, the presence of eschar, exudate discharge, and harsh chemical microenvironments rich in various enzymes can undermine the effectiveness of local drug delivery therapies [36][37]. The MN system can increase the availability of various therapeutic agents by controlling the drug content of a single needle in a controlled spatial distribution [38][39][40]. During other stages of wound healing, growth factors, immunomodulators, etc., can also be delivered to the wound through microneedles to promote rapid wound healing [41][42][43].
As a new type of transdermal drug delivery method, microneedles can break down barriers at the wound site; improve drug delivery efficiency; and achieve various antibacterial, proliferative, and angiogenesis-related effects to improve wound healing.

4. Dissolving Microneedles for Diagnostics and Monitoring

Although MNs were originally designed to be developed for the transdermal delivery of drugs and vaccines, their suitable size is large enough to penetrate the cuticle of the skin and enter the dermal ISF without triggering pain-sensing neurons deep in the skin. Thus, microneedles are also a technology whose properties are well-suited for direct access to the dermal ISF in a minimally invasive manner. They provide an excellent platform for transdermal diagnosis and monitoring [44]. In addition to their ability to cross the stratum corneum, MNs’ ability to come into direct contact with the dermal ISF provides an opportunity to sample fluids for external analysis or for directly measuring physiological parameters within the skin. MN-based medical sensing technologies can be divided into two categories. The first is electrochemical biosensors; generally, solid and hollow MNs are used as electrode substrates for further modification [45][46][47][48]. The other is the direct extraction of interstitial fluid (ISF) by means of polymer MNs or hydrogel MNs. In an MN-based electrochemical biosensor, the concentration of the target sample (glucose, lactic acid, alcohol, urea, amino acids, therapeutic drugs, etc.) reflecting the human condition is converted into an electrical signal through the MN electrode [49]. In MN-based direct ISF extraction, because ISF exists in the skin in large quantities, when hydrogel MNs with three-dimensional cross-linked network structures penetrate the skin, ISF is absorbed to form a swollen state [50]. By extracting ISF or by real-time detection, researchers can analyze these therapeutic drugs, proteins, and ions that reflect the physiological conditions of the body.
In addition to detecting blood sugar and providing insulin as needed, microneedles are also used to detect DNA and RNA [51][52][53][54], cytokines [55][56][57][58], exosomes [59], small molecules [60][61][62], etc. By combining them with electrochemistry, the monitoring of movement status and wounds can also be achieved [63][64][65].
Therefore, with MN-assisted monitoring of technology or extraction of ISF, healthy people or patients without professional training can self-diagnose, simplify the monitoring process, and avoid the potential dangers and problems caused by delayed medical guidance. Combined with MN assistive technology, more information regarding physical conditions can be obtained in a minimally invasive, fast, and convenient way.

5. Dissolving Microneedle for Cutaneous Disease

As a global public health problem, skin diseases seriously affect the quality of life of patients [66]. The presence of the stratum corneum (SC) of the skin also severely impedes the transdermal penetration of drugs, making it extremely difficult for them to cross the skin [67]. Microneedles can increase skin permeability, increase drug concentration in local skin lesions, and reduce systemic toxicity. Microneedles have been used to treat a variety of common skin conditions, such as acne, hair loss [68][69][70], atopic dermatitis (AD) [71][72][73][74][75], psoriasis [76][77][78][79], and scarring [80][81][82][83][84][85][86].

References

  1. Allen, E.A.; O'Mahony, C.; Cronin, M.; O'Mahony, T.; Moore, A.C.; Crean, A.M. Dissolvable microneedle fabrication using piezoelectric dispensing technology. Int. J. Pharm. 2016, 500, 1–10.
  2. Badran, M.M.; Kuntsche, J.; Fahr, A. Skin penetration enhancement by a microneedle device (Dermaroller) in vitro: Dependency on needle size and applied formulation. Eur. J. Pharm. Sci. 2009, 36, 511–523.
  3. Engelke, L.; Winter, G.; Hook, S.; Engert, J. Recent insights into cutaneous immunization: How to vaccinate via the skin. Vaccine 2015, 33, 4663–4674.
  4. Li, D.; Hu, D.; Xu, H.; Patra, H.K.; Liu, X.; Zhou, Z.; Tang, J.; Slater, N.; Shen, Y. Progress and perspective of microneedle system for anti-cancer drug delivery. Biomaterials 2021, 264, 120410.
  5. DeMuth, P.C.; Su, X.; Samuel, R.E.; Hammond, P.T.; Irvine, D.J. Nano-layered microneedles for transcutaneous delivery of polymer nanoparticles and plasmid DNA. Adv. Mater. 2010, 22, 4851–4856.
  6. Miller, M.A.; Pisani, E. The cost of unsafe injections. Bull. World Health Organ. 1999, 77, 808–811.
  7. Lee, J.W.; Prausnitz, M.R. Drug delivery using microneedle patches: Not just for skin. Expert Opin. Drug Deliv. 2018, 15, 541–543.
  8. Donnelly, R.; Douroumis, D. Microneedles for drug and vaccine delivery and patient monitoring. Drug Deliv. Transl. Res. 2015, 5, 311–312.
  9. Kim, Y.-C.; Park, J.-H.; Prausnitz, M.R. Microneedles for drug and vaccine delivery. Adv. Drug Deliv. Rev. 2012, 64, 1547–1568.
  10. Jepps, O.G.; Dancik, Y.; Anissimov, Y.G.; Roberts, M.S. Modeling the human skin barrier--towards a better understanding of dermal absorption. Adv. Drug Deliv. Rev. 2013, 65, 152–168.
  11. Prausnitz, M.R.; Langer, R. Transdermal drug delivery. Nat. Biotechnol. 2008, 26, 1261–1268.
  12. Lee, B.-M.; Lee, C.; Lahiji, S.F.; Jung, U.-W.; Chung, G.; Jung, H. Dissolving Microneedles for Rapid and Painless Local Anesthesia. Pharmaceutics 2020, 12, 366.
  13. Henry, S.; McAllister, D.V.; Allen, M.G.; Prausnitz, M.R. Microfabricated microneedles: A novel approach to transdermal drug delivery. J. Pharm. Sci. 1998, 87, 922–925.
  14. Hao, Y.; Li, W.; Zhou, X.; Yang, F.; Qian, Z. Microneedles-Based Transdermal Drug Delivery Systems: A Review. J. Biomed. Nanotechnol. 2017, 13, 1581–1597.
  15. Sabbagh, F.; Kim, B.-S. Ex Vivo Transdermal Delivery of Nicotinamide Mononucleotide Using Polyvinyl Alcohol Microneedles. Polymers 2023, 15, 31.
  16. Sabbagh, F.; Kim, B.S. Microneedles for transdermal drug delivery using clay-based composites. Expert Opin. Drug Deliv. 2022, 19, 1099–1113.
  17. van der Maaden, K.; Jiskoot, W.; Bouwstra, J. Microneedle technologies for (trans)dermal drug and vaccine delivery. J. Control. Release 2012, 161, 645–655.
  18. Donnelly, R.F.; Raj Singh, T.R.; Woolfson, A.D. Microneedle-based drug delivery systems: Microfabrication, drug delivery, and safety. Drug Deliv. 2010, 17, 187–207.
  19. Donnelly, R.F.; Moffatt, K.; Alkilani, A.Z.; Vicente-Pérez, E.M.; Barry, J.; McCrudden, M.T.C.; Woolfson, A.D. Hydrogel-forming microneedle arrays can be effectively inserted in skin by self-application: A pilot study centred on pharmacist intervention and a patient information leaflet. Pharm. Res. 2014, 31, 1989–1999.
  20. Vicente-Perez, E.M.; Larrañeta, E.; McCrudden, M.T.C.; Kissenpfennig, A.; Hegarty, S.; McCarthy, H.O.; Donnelly, R.F. Repeat application of microneedles does not alter skin appearance or barrier function and causes no measurable disturbance of serum biomarkers of infection, inflammation or immunity in mice in vivo. Eur. J. Pharm. Biopharm. 2017, 117, 400–407.
  21. Howells, O.; Blayney, G.J.; Gualeni, B.; Birchall, J.C.; Eng, P.F.; Ashraf, H.; Sharma, S.; Guy, O.J. Design, fabrication, and characterisation of a silicon microneedle array for transdermal therapeutic delivery using a single step wet etch process. Eur. J. Pharm. Biopharm. 2022, 171, 19–28.
  22. Ventrelli, L.; Marsilio Strambini, L.; Barillaro, G. Microneedles for Transdermal Biosensing: Current Picture and Future Direction. Adv. Healthc. Mater. 2015, 4, 2606–2640.
  23. Chen, H.; Fan, L.; Peng, N.; Yin, Y.; Mu, D.; Wang, J.; Meng, R.; Xie, J. Galunisertib-Loaded Gelatin Methacryloyl Hydrogel Microneedle Patch for Cardiac Repair after Myocardial Infarction. ACS Appl. Mater. Interfaces 2022, 14, 40491–40500.
  24. Zhang, X.; Cheng, Y.; Liu, R.; Zhao, Y. Globefish-Inspired Balloon Catheter with Intelligent Microneedle Coating for Endovascular Drug Delivery. Adv. Sci. 2022, 9, e2204497.
  25. Zhou, J.-X.; Ding, F.; Tang, L.-N.; Li, T.; Li, Y.-H.; Zhang, Y.-J.; Gong, H.-Y.; Li, Y.-T.; Zhang, G.-J. Monitoring of pH changes in a live rat brain with MoS2/PAN functionalized microneedles. Analyst 2018, 143, 4469–4475.
  26. Banerjee, D.; Cieslar-Pobuda, A.; Zhu, G.H.; Wiechec, E.; Patra, H.K. Adding Nanotechnology to the Metastasis Treatment Arsenal. Trends Pharmacol. Sci. 2019, 40, 403–418.
  27. Lan, X.; She, J.; Lin, D.-A.; Xu, Y.; Li, X.; Yang, W.-F.; Lui, V.W.Y.; Jin, L.; Xie, X.; Su, Y.-X. Microneedle-Mediated Delivery of Lipid-Coated Cisplatin Nanoparticles for Efficient and Safe Cancer Therapy. ACS Appl. Mater. Interfaces 2018, 10, 33060–33069.
  28. Wang, C.; Zeng, Y.; Chen, K.-F.; Lin, J.; Yuan, Q.; Jiang, X.; Wu, G.; Wang, F.; Jia, Y.-G.; Li, W. A self-monitoring microneedle patch for light-controlled synergistic treatment of melanoma. Bioact. Mater. 2023, 27, 58–71.
  29. Hao, Y.; Chen, Y.; He, X.; Yang, F.; Han, R.; Yang, C.; Li, W.; Qian, Z. Near-infrared responsive 5-fluorouracil and indocyanine green loaded MPEG-PCL nanoparticle integrated with dissolvable microneedle for skin cancer therapy. Bioact. Mater. 2020, 5, 542–552.
  30. Dai, J.; Wei, S.; Xu, J.; Xue, H.; Chen, Z.; Wu, M.; Chen, W.; Lou, X.; Xia, F.; Wang, S. Microneedle Device Delivering Aggregation-Induced Emission Photosensitizers for Enhanced Metronomic Photodynamic Therapy of Cancer. ACS Appl. Mater. Interfaces 2023, 15, 16526–16538.
  31. Song, C.; Wu, X.; Wang, J.; Liu, R.; Zhao, Y. Photosensitizer-immunotherapy integrated microneedles for preventing tumor recurrence and metastasis. Nano Today 2023, 51, 101913.
  32. Tham, H.P.; Xu, K.; Lim, W.Q.; Chen, H.; Zheng, M.; Thng, T.G.S.; Venkatraman, S.S.; Xu, C.; Zhao, Y. Microneedle-Assisted Topical Delivery of Photodynamically Active Mesoporous Formulation for Combination Therapy of Deep-Seated Melanoma. ACS Nano 2018, 12, 11936–11948.
  33. Roger, M.; Fullard, N.; Costello, L.; Bradbury, S.; Markiewicz, E.; O'Reilly, S.; Darling, N.; Ritchie, P.; Määttä, A.; Karakesisoglou, I.; et al. Bioengineering the microanatomy of human skin. J. Anat. 2019, 234, 438–455.
  34. Han, G.; Ceilley, R. Chronic Wound Healing: A Review of Current Management and Treatments. Adv. Ther. 2017, 34, 599–610.
  35. Park, S.Y.; Lee, H.U.; Lee, Y.-C.; Kim, G.H.; Park, E.C.; Han, S.H.; Lee, J.G.; Choi, S.; Heo, N.S.; Kim, D.L.; et al. Wound healing potential of antibacterial microneedles loaded with green tea extracts. Mater. Sci. Eng. C Mater. Biol. Appl. 2014, 42, 757–762.
  36. Caffarel-Salvador, E.; Kearney, M.-C.; Mairs, R.; Gallo, L.; Stewart, S.A.; Brady, A.J.; Donnelly, R.F. Methylene Blue-Loaded Dissolving Microneedles: Potential Use in Photodynamic Antimicrobial Chemotherapy of Infected Wounds. Pharmaceutics 2015, 7, 397–412.
  37. Jamaledin, R.; Yiu, C.K.Y.; Zare, E.N.; Niu, L.-N.; Vecchione, R.; Chen, G.; Gu, Z.; Tay, F.R.; Makvandi, P. Advances in Antimicrobial Microneedle Patches for Combating Infections. Adv. Mater. 2020, 32, e2002129.
  38. Guo, Z.; Liu, H.; Shi, Z.; Lin, L.; Li, Y.; Wang, M.; Pan, G.; Lei, Y.; Xue, L. Responsive hydrogel-based microneedle dressing for diabetic wound healing. J. Mater. Chem. B 2022, 10, 3501–3511.
  39. Lei, Q.; He, D.; Ding, L.; Kong, F.; He, P.; Huang, J.; Guo, J.; Brinker, C.J.; Luo, G.; Zhu, W.; et al. Microneedle Patches Integrated with Biomineralized Melanin Nanoparticles for Simultaneous Skin Tumor Photothermal Therapy and Wound Healing. Adv. Funct. Mater. 2022, 32, 2113269.
  40. Ning, T.; Yang, F.; Chen, D.; Jia, Z.; Yuan, R.; Du, Z.; Liu, S.; Yu, Y.; Dai, X.; Niu, X.; et al. Synergistically Detachable Microneedle Dressing for Programmed Treatment of Chronic Wounds. Adv. Healthc. Mater. 2022, 11, e2102180.
  41. Li, S.; Wang, X.; Yan, Z.; Wang, T.; Chen, Z.; Song, H.; Zheng, Y. Microneedle Patches with Antimicrobial and Immunomodulating Properties for Infected Wound Healing. Adv. Sci. 2023, 10, e2300576.
  42. Liu, W.; Zhai, X.; Zhao, X.; Cai, Y.; Zhang, X.; Xu, K.; Weng, J.; Li, J.; Chen, X. Multifunctional Double-Layer and Dual Drug-Loaded Microneedle Patch Promotes Diabetic Wound Healing. Adv. Healthc. Mater. 2023, 12, e2300297.
  43. Gao, S.; Zhang, W.; Zhai, X.; Zhao, X.; Wang, J.; Weng, J.; Li, J.; Chen, X. An antibacterial and proangiogenic double-layer drug-loaded microneedle patch for accelerating diabetic wound healing. Biomater. Sci. 2023, 11, 533–541.
  44. Kaushik, S.; Hord, A.H.; Denson, D.D.; McAllister, D.V.; Smitra, S.; Allen, M.G.; Prausnitz, M.R. Lack of pain associated with microfabricated microneedles. Anesth. Analg. 2001, 92, 502–504.
  45. Cheng, J.; Huang, J.; Xiang, Q.; Dong, H. Hollow microneedle microfluidic paper-based chip for biomolecules rapid sampling and detection in interstitial fluid. Anal. Chim. Acta 2023, 1255, 341101.
  46. Downs, A.M.; Bolotsky, A.; Weaver, B.M.; Bennett, H.; Wolff, N.; Polsky, R.; Miller, P.R. Microneedle electrochemical aptamer-based sensing: Real-time small molecule measurements using sensor-embedded, commercially-available stainless steel microneedles. Biosens. Bioelectron. 2023, 236, 115408.
  47. Huang, X.; Zheng, S.; Liang, B.; He, M.; Wu, F.; Yang, J.; Chen, H.-J.; Xie, X. 3D-assembled microneedle ion sensor-based wearable system for the transdermal monitoring of physiological ion fluctuations. Microsyst. Nanoeng. 2023, 9, 25.
  48. Li, Z.; Kadian, S.; Mishra, R.K.; Huang, T.; Zhou, C.; Liu, S.; Wang, Z.; Narayan, R.; Zhu, Z. Electrochemical detection of cholesterol in human biofluid using microneedle sensor. J. Mater. Chem. B 2023, 11, 6075–6081.
  49. Moon, J.-M.; Thapliyal, N.; Hussain, K.K.; Goyal, R.N.; Shim, Y.-B. Conducting polymer-based electrochemical biosensors for neurotransmitters: A review. Biosens. Bioelectron. 2018, 102, 540–552.
  50. Kim, M.; Jung, B.; Park, J.-H. Hydrogel swelling as a trigger to release biodegradable polymer microneedles in skin. Biomaterials 2012, 33, 668–678.
  51. Yang, B.; Kong, J.; Fang, X. Programmable CRISPR-Cas9 microneedle patch for long-term capture and real-time monitoring of universal cell-free DNA. Nat. Commun. 2022, 13, 3999.
  52. Yang, B.; Fang, X.; Kong, J. In Situ Sampling and Monitoring Cell-Free DNA of the Epstein-Barr Virus from Dermal Interstitial Fluid Using Wearable Microneedle Patches. ACS Appl. Mater. Interfaces 2019, 11, 38448–38458.
  53. Yang, B.; Fang, X.; Kong, J. Engineered Microneedles for Interstitial Fluid Cell-Free DNA Capture and Sensing Using Iontophoretic Dual-Extraction Wearable Patch. Adv. Funct. Mater. 2020, 30, 2000591.
  54. Yang, Q.; Wang, Y.; Liu, T.; Wu, C.; Li, J.; Cheng, J.; Wei, W.; Yang, F.; Zhou, L.; Zhang, Y.; et al. Microneedle Array Encapsulated with Programmed DNA Hydrogels for Rapidly Sampling and Sensitively Sensing of Specific MicroRNA in Dermal Interstitial Fluid. ACS Nano 2022, 16, 18366–18375.
  55. Song, N.; Xie, P.; Shen, W.; Oh, H.; Zhang, Y.; Vitale, F.; Javanmard, M.; Allen, M.G. A microwell-based impedance sensor on an insertable microneedle for real-time in vivo cytokine detection. Microsyst. Nanoeng. 2021, 7, 96.
  56. Xu, J.; Yang, B.; Kong, J.; Zhang, Y.; Fang, X. Real-Time Monitoring and Early Warning of a Cytokine Storm In Vivo Using a Wearable Noninvasive Skin Microneedle Patch. Adv. Healthc. Mater. 2023, e2203133.
  57. Cai, Y.; Huang, S.; Zhang, Z.; Zhang, J.; Zhu, X.; Chen, X.; Ding, X. Bioinspired Rotation Microneedles for Accurate Transdermal Positioning and Ultraminimal-Invasive Biomarker Detection with Mechanical Robustness. Research 2022, 2022, 9869734.
  58. Lee, K.H.; Kim, J.D.; Jeong, D.H.; Kim, S.M.; Park, C.O.; Lee, K.H. Development of a novel microneedle platform for biomarker assessment of atopic dermatitis patients. Skin Res. Technol. 2023, 29, e13413.
  59. Park, W.; Maeng, S.-W.; Mok, J.W.; Choi, M.; Cha, H.J.; Joo, C.-K.; Hahn, S.K. Hydrogel Microneedles Extracting Exosomes for Early Detection of Colorectal Cancer. Biomacromolecules 2023, 24, 1445–1452.
  60. Keyvani, F.; Zheng, H.; Kaysir, M.R.; Mantaila, D.F.; Ghavami Nejad, P.; Rahman, F.A.; Quadrilatero, J.; Ban, D.; Poudineh, M. A Hydrogel Microneedle Assay Combined with Nucleic Acid Probes for On-Site Detection of Small Molecules and Proteins. Angew. Chem. Int. Ed Engl. 2023, 62, e202301624.
  61. Lu, M.; Zhang, X.; Xu, D.; Li, N.; Zhao, Y. Encoded Structural Color Microneedle Patches for Multiple Screening of Wound Small Molecules. Adv. Mater. 2023, 35, e2211330.
  62. Zhu, J.; Zhou, X.; Kim, H.-J.; Qu, M.; Jiang, X.; Lee, K.; Ren, L.; Wu, Q.; Wang, C.; Zhu, X.; et al. Gelatin Methacryloyl Microneedle Patches for Minimally Invasive Extraction of Skin Interstitial Fluid. Small 2020, 16, e1905910.
  63. Gao, B.; Guo, M.; Lyu, K.; Chu, T.; He, B. Intelligent Silk Fibroin Based Microneedle Dressing (i-SMD). Adv. Funct. Mater. 2021, 31, 2006839.
  64. Guo, M.; Wang, Y.; Gao, B.; He, B. Shark Tooth-Inspired Microneedle Dressing for Intelligent Wound Management. ACS Nano 2021, 15, 15316–15327.
  65. Huang, Y.; Li, H.; Hu, T.; Li, J.; Yiu, C.K.; Zhou, J.; Li, J.; Huang, X.; Yao, K.; Qiu, X.; et al. Implantable Electronic Medicine Enabled by Bioresorbable Microneedles for Wireless Electrotherapy and Drug Delivery. Nano Lett. 2022, 22, 5944–5953.
  66. Qu, F.; Geng, R.; Liu, Y.; Zhu, J. Advanced nanocarrier- and microneedle-based transdermal drug delivery strategies for skin diseases treatment. Theranostics 2022, 12, 3372–3406.
  67. Pham, Q.D.; Björklund, S.; Engblom, J.; Topgaard, D.; Sparr, E. Chemical penetration enhancers in stratum corneum-Relation between molecular effects and barrier function. J. Control. Release 2016, 232, 175–187.
  68. Hong, C.; Zhang, G.; Zhang, W.; Liu, J.; Zhang, J.; Chen, Y.; Peng, H.; Cheng, Y.; Ding, X.; Xin, H.; et al. Hair grows hair: Dual-effective hair regrowth through a hair enhanced dissolvable microneedle patch cooperated with the pure yellow light irradiation. Appl. Mater. Today 2021, 25, 101188.
  69. Shi, Y.; Zhao, J.; Li, H.; Yu, M.; Zhang, W.; Qin, D.; Qiu, K.; Chen, X.; Kong, M. A Drug-Free, Hair Follicle Cycling Regulatable, Separable, Antibacterial Microneedle Patch for Hair Regeneration Therapy. Adv. Healthc. Mater. 2022, 11, e2200908.
  70. Yang, G.; Chen, Q.; Wen, D.; Chen, Z.; Wang, J.; Chen, G.; Wang, Z.; Zhang, X.; Zhang, Y.; Hu, Q.; et al. A Therapeutic Microneedle Patch Made from Hair-Derived Keratin for Promoting Hair Regrowth. ACS Nano 2019, 13, 4354–4360.
  71. Ben David, N.; Richtman, Y.; Gross, A.; Ibrahim, R.; Nyska, A.; Ramot, Y.; Mizrahi, B. Design and Evaluation of Dissolvable Microneedles for Treating Atopic Dermatitis. Pharmaceutics 2023, 15, 1109.
  72. Chen, M.-C.; Chen, C.-S.; Wu, Y.-W.; Yang, Y.-Y. Poly-γ-Glutamate microneedles as transdermal immunomodulators for ameliorating atopic dermatitis-like skin lesions in Nc/Nga mice. Acta Biomater. 2020, 114, 183–192.
  73. Chen, Y.-L.; Chang, C.-C.; Lin, Y.-C.; Chen, M.-C. Double-layered PLGA/HA microneedle systems as a long-acting formulation of polyphenols for effective and long-term management of atopic dermatitis. Biomater. Sci. 2023, 11, 4995–5011.
  74. Jang, M.; Kang, B.M.; Yang, H.; Ohn, J.; Kwon, O.; Jung, H. High-Dose Steroid Dissolving Microneedle for Relieving Atopic Dermatitis. Adv. Healthc. Mater. 2021, 10, e2001691.
  75. Yang, Y.; Chen, B.Z.; Zhang, X.P.; Zheng, H.; Li, Z.; Zhang, C.Y.; Guo, X.D. Conductive Microneedle Patch with Electricity-Triggered Drug Release Performance for Atopic Dermatitis Treatment. ACS Appl. Mater. Interfaces 2022, 14, 31645–31654.
  76. Korkmaz, E.; Friedrich, E.E.; Ramadan, M.H.; Erdos, G.; Mathers, A.R.; Burak Ozdoganlar, O.; Washburn, N.R.; Falo, L.D. Therapeutic intradermal delivery of tumor necrosis factor-alpha antibodies using tip-loaded dissolvable microneedle arrays. Acta Biomater. 2015, 24, 96–105.
  77. Lin, Y.; Chen, Y.; Deng, R.; Qin, H.; Li, N.; Qin, Y.; Chen, H.; Wei, Y.; Wang, Z.; Sun, Q.; et al. Delivery of neutrophil membrane encapsulated non-steroidal anti-inflammatory drugs by degradable biopolymer microneedle patch for rheumatoid arthritis therapy. Nano Today 2023, 49, 101791.
  78. Wang, Y.; Zhang, X.; Chen, G.; Lu, M.; Zhao, Y. Multifunctional structural color triboelectric microneedle patches for psoriasis treatment. Matter 2023, 6, 1555–1568.
  79. Zhang, W.; Chen, Y.; Zhao, Z.; Zheng, H.; Wang, S.; Liao, Z.; Sheng, T.; Zhao, S.; Hou, W.; Yu, X.; et al. Adoptive Treg therapy with metabolic intervention via perforated microneedles ameliorates psoriasis syndrome. Sci. Adv. 2023, 9, eadg6007.
  80. Chien, P.N.; Jeong, J.H.; Nam, S.Y.; Lim, S.Y.; van Long, N.; Zhang, X.R.; Jeong, J.H.; Heo, C.Y. Nanomicelle-generating Microneedles Loaded with Tranilast for Treatment of Hypertrophic Scars in a Rabbit Model. In Vivo 2022, 36, 1734–1744.
  81. Chun, Y.Y.; Tan, W.W.R.; Vos, M.I.G.; Chan, W.K.; Tey, H.L.; Tan, N.S.; Tan, T.T.Y. Scar prevention through topical delivery of gelatin-tyramine-siSPARC nanoplex loaded in dissolvable hyaluronic acid microneedle patch across skin barrier. Biomater. Sci. 2022, 10, 3963–3971.
  82. Lin, S.; Quan, G.; Hou, A.; Yang, P.; Peng, T.; Gu, Y.; Qin, W.; Liu, R.; Ma, X.; Pan, X.; et al. Strategy for hypertrophic scar therapy: Improved delivery of triamcinolone acetonide using mechanically robust tip-concentrated dissolving microneedle array. J. Control. Release 2019, 306, 69–82.
  83. Liu, F.; Luo, Y.; Chen, H.; Xu, S.; Zhang, D.; Sang, H.; Xu, C.; Zhang, M. Comparison of the efficacy of seven types of microneedles for treating a rabbit hypertrophic scar model. Nanoscale Adv. 2023, 5, 927–933.
  84. Ning, X.; Wiraja, C.; Chew, W.T.S.; Fan, C.; Xu, C. Transdermal delivery of Chinese herbal medicine extract using dissolvable microneedles for hypertrophic scar treatment. Acta Pharm. Sin. B 2021, 11, 2937–2944.
  85. Zhang, Q.; Shi, L.; He, H.; Liu, X.; Huang, Y.; Xu, D.; Yao, M.; Zhang, N.; Guo, Y.; Lu, Y.; et al. Down-Regulating Scar Formation by Microneedles Directly via a Mechanical Communication Pathway. ACS Nano 2022, 16, 10163–10178.
  86. Zhao, B.; Guo, W.; Zhou, X.; Xue, Y.; Wang, T.; Li, Q.; Tan, L.-L.; Shang, L. Ferroptosis-Mediated Synergistic Therapy of Hypertrophic Scarring Based on Metal–Organic Framework Microneedle Patch. Adv. Funct. Mater. 2023, 33, 2300575.
More
Information
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: 303
Revisions: 2 times (View History)
Update Date: 25 Oct 2023
1000/1000
Video Production Service