Next Article in Journal
Gold Nanoparticles Modification with Liquid Crystalline Polybenzylic Dendrons via 1,3-Dipolar Cycloaddition
Next Article in Special Issue
Micro and Nano Interdigitated Electrode Array (IDEA)-Based MEMS/NEMS as Electrochemical Transducers: A Review
Previous Article in Journal
Highly Bright Gold Nanowires Arrays for Sensitive Detection of Urea and Urease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Nanomaterials in the Fabrication of bioNEMS/MEMS for Biomedical Applications and towards Pioneering Food Waste Utilisation

1
Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur 50603, Malaysia
2
Centre for Innovation in Medical Engineering (CIME), Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur 50603, Malaysia
3
Centre for Printable Electronics, Universiti Malaya, Kuala Lumpur 50603, Malaysia
4
Faculty of Technical Sciences, University of Novi Sad, T. Dositeja Obradovića 6, 21000 Novi Sad, Serbia
5
Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia
*
Authors to whom correspondence should be addressed.
Nanomaterials 2022, 12(22), 4025; https://doi.org/10.3390/nano12224025
Submission received: 19 October 2022 / Revised: 11 November 2022 / Accepted: 13 November 2022 / Published: 16 November 2022
(This article belongs to the Special Issue Nanocomposites for Bio-MEMS/NEMS Applications)

Abstract

:
bioNEMS/MEMS has emerged as an innovative technology for the miniaturisation of biomedical devices with high precision and rapid processing since its first R&D breakthrough in the 1980s. To date, several organic including food waste derived nanomaterials and inorganic nanomaterials (e.g., carbon nanotubes, graphene, silica, gold, and magnetic nanoparticles) have steered the development of high-throughput and sensitive bioNEMS/MEMS-based biosensors, actuator systems, drug delivery systems and implantable/wearable sensors with desirable biomedical properties. Turning food waste into valuable nanomaterials is potential groundbreaking research in this growing field of bioMEMS/NEMS. This review aspires to communicate recent progress in organic and inorganic nanomaterials based bioNEMS/MEMS for biomedical applications, comprehensively discussing nanomaterials criteria and their prospects as ideal tools for biomedical devices. We discuss clinical applications for diagnostic, monitoring, and therapeutic applications as well as the technological potential for cell manipulation (i.e., sorting, separation, and patterning technology). In addition, current in vitro and in vivo assessments of promising nanomaterials-based biomedical devices will be discussed in this review. Finally, this review also looked at the most recent state-of-the-art knowledge on Internet of Things (IoT) applications such as nanosensors, nanoantennas, nanoprocessors, and nanobattery.

1. Introduction

Biological (or biomedical) applications of nano-/micro-electro-mechanical systems (bioNEMS/MEMS) are emerging advanced devices for rapid, sensitive, and real-time analysis of biological samples. Micro and nanofabrication started with bulk and surface micromachining for silicon in the electronic industry. The rapid development of single-crystal silicon using MEMS technology has led to an interest in employing glass and polymer-based materials to fabricate MEMS devices. As a result, this technology branched out to cater to other industries such as biomedical industries, which later formed the foundation for bioNEMS/MEMS technology [1,2]. bioNEMS/MEMS devices are expected to perform sample screening and analysis cycles in the millisecond to picosecond range, necessitating careful consideration of factors such as device design, sensor sensitivity, and material selection. Nanomaterials-based bioNEMS/MEMS have found potential in a myriad of biomedical applications, from drug delivery systems, cell manipulation (e.g., sorting–separation technology and cell patterning) to point-of-care testing (e.g., surgical sensors, implant sensors for tissue engineering applications, and rapid home monitoring devices) [3,4,5,6]. Hence, the development of high-throughput nanomaterials-based bioNEMS/MEMS devices is needed.
The primary goal of this review is to explore the progress of nanotechnology and bioNEMS/MEMS by focusing on the fabrication strategies and clinical applications that have rapidly evolved in the past years, as summarised in Figure 1. In this framework, we discuss the developmental trend of nanomaterials-based bioNEMS/MEMS and the prospects for future biomedical devices. The amalgamation of multidisciplinary fields between engineering, materials science, chemistry, biology, and computer science has contributed to the rapid advancement of this technology. In this instance, the integration of bioNEMS/MEMS to the Internet of Things (IoT) technology could be realised in the near future. Therefore, we also discuss the current state-of-art of IoT platforms and their potential to integrate information into bioNEMS/MEMS nanodevices. Lastly, challenges and future opportunities for further development of nanomaterials-based bioNEMS/MEMS will be thoroughly highlighted in this review.

2. Overview of Nanomaterials for bioNEMS/MEMS Applications

2.1. Classification of Nanomaterials

Nanomaterials in biomedical applications enable the miniaturisation of bioNEMS/MEMS with multifunctional capabilities, which facilitates control and regulation of the biological environment [7,8]. The miniaturisation of devices is important to allow their operation in a specific area for a selected duration of time. Materials at the nanoscale range exhibit outstanding electrical, mechanical, chemical, and optical properties and an increase in the surface area of a material also enhances its catalytic ability [9,10]. However, the biocompatibility of nanomaterials-based bioNEMS/MEMS such as biosensors, actuation systems, drug delivery systems and smart stents is an important concern to prevent the toxicity and loss of functionality of the device in both in vitro and in vivo conditions [11]. Some of the important criteria of nanomaterials that are promising for the fabrication of bioNEMS/MEMS are their fascinating tuneable electronic, mechanical, and optical properties [12,13]. Furthermore, nanomaterials also promote the adhesion between the substrate and biological molecular layer and the substrate, which enables the tunnelling-based charge transport of the nanocomposites [14]. In addition to that, other properties such as chemical stability, thermal stability, heat conductance, porosity, and surface area are also important features of the nanomaterials-based bioNEMS/MEMS [15,16].
Metal nanoparticles, carbon nanomaterials including polymer nanocomposites, are the most commonly adopted nanomaterials for the fabrication of bioNEMS/MEMS (Figure 2) [17,18]. The heterogeneous integration of bioNEMS/MEMS devices with nanomaterials has created a breakthrough in the biomedical field for applications such as sensing, diagnostics, imaging, and therapeutics. The nanomaterial-bioNEMS/MEMS developed for drug delivery applications include micropumps, microneedles, and micro-reservoirs to achieve targeted drug delivery. The nanomaterials-based micro/nanoneedles are small-scale bioNEMS/MEMS devices that possess unique properties whereby they can be modified for selective transport of drug molecules, and the superior electronic properties of nanomaterials enable the control of the delivery by applying an electric field [11]. For example, a novel approach of integrating carbon nanotube (CNT) nanofilters with SU-8 microneedle array devices was fabricated for transdermal drug delivery through the lithography process [19]. The fabricated device is capable of penetrating the stratum corneum layer and with the help of CNT, the drug molecules are selectively delivered to specific tissues with an applied electric field. Similarly, for drug delivery into the tissue, a nanosized drug delivery vehicle bioNEMS/MEMS that employs the micro-nano reservoirs concept was used [20]. Paclitaxel drug delivery into human glioblastoma cells and human umbilical vein endothelial cells tissues was achieved with the help of a nanomaterial coated biocompatible co-polymer block of poly(lactide)-poly(ethylene glycol) (PLA-PEG). The co-polymer was found to reduce the in-vitro toxicity and the surface properties of nanomaterials prevent tissue aggregation and improve the drug loading to increase therapeutic efficacy.
In the field of biosensing, the nanomaterial-coated bioNEMS/MEMS plays a significant role in the biomedical field due to some unique features such as portability, fast response, and high sensitivity. The unique biological and physical properties of nanomaterials enable target biomolecule identification and cause–effect transduction of an electronic signal in bioNEMS/MEMS biosensors [11]. The integration of nanomaterials in the system allows the miniaturisation of the system into a single chip to facilitate the fabrication of point-of-care devices. A study by micro-nanocantilevers increased immobilisation of the cholesterol oxidase enzyme for sensitive detection of cholesterol levels up to 100 femto molar within a short period of response time. Similarly, Sharma, et al. [21] explored the effect of gold nanoparticles on the electrical conductivity, electroactive surface area and electrochemical activity of nano-sized carbon interdigitated electrodes. This amperometric MEMS biosensor exhibited a wide sensing range (0.005–10 mM), high selectivity and high sensitivity with a limit of detection (LOD) of ~1.28 µM for cholesterol detection.

2.2. Nanomaterials Synthesised from Food Waste

In past years, a plethora of nanoparticles have been produced through various techniques and adopted to advance technologies for biomedical applications [22]. With the advancement of knowledge in the field of materials science and engineering, researchers are keener on ensuring the sustainability of the nanomaterials production processes. Through these initiatives, many alternative inputs for nanomaterials production as well as green synthesis techniques were introduced widely. Rapid urbanisation led to a massive increase in food waste or food-derived waste which includes agricultural waste [23]. Around 1.3 billion tons per year of edible food produced for human consumption were wasted globally. The wastage of food starts from the initial stage of agricultural production down to the final household consumption [24]. For medium- and high-income countries, food wastage is high at the final consumer stage, whereas in low-income countries the wastage occurs initial or middle stage of the food supply [25]. Moreover, in Europe and North America, food wastage accounts for 280–300 kg/year, which is equivalent to 95–115 kg/year per capita. However, in Sub-Saharan Africa and South/Southeast Asia, food wastage accounts for 120–170 kg/year, which is equivalent to only 6–11 kg/year per capita [26]. According to the recent food waste index published in 2021 as summarised in Figure 3, European countries produce approximately 3400 kg/capita/year, whereas Asia is one of the highest contributors of food waste, producing more than 4300 kg/capita/year of household food waste [27].
Aside from the startling statistics, the negative repercussions of wasted food on society take many other forms, ranging from immediate financial losses to environmental pollution including air and land contamination. Products with low and negative values (waste) can be converted into useful resources. Its utilization as a resource for the production of high-value materials is a profitable route to improving sustainability and lowering financial expenditure of many industries and scientific disciplines including biomedicine and nanomaterials [28]. The wastes can be utilized to produce valuable nanomaterials such as carbon-based nanomaterials, silver, zinc, gold and chitosan that can be used for biomedical applications as they add integrated functionalities to the bioNEMS/MEMS devices [23,29,30]. Interestingly, food wastes are a valuable source for the green synthesis of nanomaterials due to their low toxicity, cost-effectiveness, nano-size, and high stability properties. These features of nanomaterials are important for integration with bioNEMS/MEMS especially, for therapeutic and diagnostic purposes as the system must be biocompatible as their use should not cause cell toxicity [31]. Due to the organic nature of most of the food waste, the nanomaterials can be synthesized using green ways to complement the traditional synthesis method such as chemical, hydrothermal, and high-pressure treatments, which can be toxic due to the use of adverse chemicals and that limits its biomedical applications. Nanomaterials from food waste are often synthesized using the pyrolysis method, carbonization, thermal decomposition and other chemical-free green synthesis methods [32,33,34,35].
Up to now, very limited studies reported on utilizing food waste such as eggshells, peanut shells, coconut shells and coconut noir to fabricate MEMS devices [36,37,38]. Despite that, with the aid of two different kinds of agricultural waste, especially coconut coir and coconut shell, the method for converting waste into high-value capacitor-reduced graphene oxide electrodes has been proposed [37]. It has also been shown that negative value peanut shells, a common agricultural waste, can be converted into graphene-like nanosheets consisting of several layers [38]. Because of the low cost and enormous amount, which can help in addressing global problems such as water and energy crises, this food waste reuse strategy has enormous significance for the upcoming management of food waste and material science. It also holds truly excellent for a sustainable society. In this view, food waste-based bioNEMS/MEMS could open a new research niche following explosive interests in food waste-based nanomaterials. Repurposing food waste into innovative products is one of the promising circular economy approaches in an attempt to tackle the food waste problem. Therefore, the exploration of new research niches could be a blueprint to achieve sustainable food waste management in future. Furthermore, this new research area is in alignment with the 2030 Sustainable Development Goals set by the United Nations General Assembly in its effort to reduce 50% of food waste by 2030 [31,39].

3. Perspective on Nanotechnology as an Enabling Tool for the Design of New Functional Materials and Devices for bioNEMS/MEMS in Medical Applications

The integration of functional nanomaterials in the fabrication of bioNEMS/MEMS created a breakthrough in the biomedical field. Nanomaterials introduce new features and increase the overall performance in terms of sensitivity, portability, specificity, and throughput without affecting the economic feasibility of the system. Nanomaterials with bioNEMS/MEMS improve the overall mechanical properties of the system and allow the researchers to better understand the cellular interactions at the molecular level [40,41]. Moreover, nanomaterials (e.g., carbon-based nanoparticles, graphene nanosheets, silver, zinc and gold nanoparticles) in biomedical applications provide good control and regulation of the biological environment because the micro-sized devices can be tuned to acquire controlled multifunctional capabilities.
Metal nanoparticles are used in the fabrication of bioNEMS/MEMS for numerous applications due to their ease of scalability, high surface-to-volume ratio and good optical properties [42,43]. In particular, gold nanoparticles are widely used in biomedical applications mainly due to their low toxicity, good biocompatibility, tuneable surface plasmon resonance (SPR), easy biofunctionalization and detection [44]. MEMS are usually coated with gold nanoparticles which are photoresponsive nanomaterials that create laser-based actuators for applications such as studying the effect of mechanical forces on cells, and remotely triggering cardiac muscle stimulators [45]. The metal nanoparticles-MEMS hybrid acquires good biocompatibility which helps create infrared light-driven cellular scale mechanical actuators that can be distributed throughout various types of tissue samples [46]. For instance, Schlicke, et al. [47] fabricated micro- and nanoelectromechanical systems with freestanding organically cross-linked gold nanoparticle (AuNPs) membranes with a thickness of 29–45 nm deposited through lithographic patterning in a SU-8 resist. This finding can be used for the fabrication of electrostatic actuators due to the conductive and flexible nature of the hybrid system and can be tailored for different applications by tuning the particle size or structure of the cross-linker for mechanical, electrical, and optical properties based on the different requirements.
Currently, bioNEMS/MEMS devices rely on the usage of silicon, but it has poor mechanical and thermal stability, which creates a demand for more promising materials to be integrated into the fabrication of the system for biomedical applications [48]. Carbon nanomaterials such as graphene, nanodiamonds, and carbon nanotubes are some of the promising alternatives due to some remarkable properties such as high Young’s modulus, hydrophobic surface, low mass, tailorable electronic configuration and high thermal conductivity [49]. Traditional bioNEMS/MEMS face major hurdles such as contamination diffusion barrier properties, high variability, low yields, low physical and chemical stability, tribology and wear rates and low reproducibility of surface functionalization which limits their commercial applications [50]. Thus, with integration with carbon nanomaterials, commercial feasibility and scalability are made possible.
Graphene is a two-dimensional (2D) material with high strength and stiffness that can be used in bioNEMS and MEMS applications [51]. Graphene has high mechanical strength, high carrier mobility, low density, and remarkable mechanical and electrical properties which makes it a suitable transducer candidate for beams and membranes in bioNEMS/MEMS applications [52]. Moreover, it also provides substantial device scalability without compromising the performance of the device. Its excellent electrical conductivity enables integrated electrical transduction and its planar geometry allows itself easy compatibility with patterning using a standard lithographic process [53]. For biomedical applications, graphene is mostly used for sensing purposes. For instance, Rahman, et al. [54] researched the feasibility of applying graphene membrane as a pressure sensor diaphragm in the MEMS piezoresistive pressure sensor for sensitive intracranial pressure monitoring. The study reported that the proposed graphene-based MEMS device structure is suitable to detect pressure-induced strain even at low-pressure applications. Moreover, Khan and Song [55] fabricated graphene field-effect transistor (GFETs)-based bioMEMS devices for the detection of interleukin-6 (IL-6) protein biomarkers. In this study, pyrene-tagged deoxyribonucleic acid (DNA) aptamers were immobilised on the sensing platform easily due to the presence of graphene which aids the π–π stacking bonding with the negatively charged DNA aptamer in the presence of negative electric field. Graphene increased the immobilization rate and the surface coverage for a more sensitive IL-6 protein detection up to the picomolar range. Graphene has explored significant potential, especially as the negative piezoconductive effect of mono-layer graphene which can be used for flexible sensors for electronic skin and wearable devices [56]. Chen, et al. [57] developed a nanowires/graphene heterostructure-based pressure sensor for static pressure measurement. The synergistic effect between the strain-induced polarisation charges of piezoelectric nanowire and the high carrier mobility of graphene achieved sensitivity for static pressure measurement up to 1.06 × 102 kPa of within 5–7 ms response time. The nanowires/graphene heterostructure-based device design developed in this study is suitable for wearable human health inspection.
Carbon nanotubes offer great potential for electronic and biomedical applications owing to their outstanding mechanical, electrical, optoelectrical, thermal, and physical solid-state surface properties [58,59]. With a controlled synthesis condition, structural properties (chirality, semi-conducting/metallic properties, single or multi-walled), and size (diameter/length), CNT extend their applicability to bioNEMS/MEMS [50]. With these advances in the area of biomolecule–CNT hybrid systems, many potential applications within the biomedical field can be explored. The CNT can also be integrated into a carbon microstructure using various MEMS fabrication techniques. The large active surface area of CNT-modified carbon microstructures shows great potential and performance in biosensing. Xi, et al. [60] studied the efficiency and performance of a multi-walled CNT-modified C-MEMS electrode for the detection of glucose. In this study, the glucose oxidase was immobilised in polymerised polypyrrole film onto the CNT carbon microstructures in which the presence of CNT leads to increased electroactive sites for enzyme loading and facilitates rapid electron transfer rate. This CNT-based MEMS amperometric glucose biosensor results in the sensitive detection of glucose with a linear detection range of 5 to 80 mM and a limit of detection of 97.3 mA M−1 cm−2 within 10 s of response time. Moreover, MEMS technology also paves a way for the fabrication of microelectrode arrays (MEA) with uniform electrochemical characteristics for various biomedical applications. Xu, et al. [61] developed an MEA-based bioMEMS device for electrochemical detection of 5-hydroxytryptamine and dopamine for neural function analysis at both the single cell and network levels. The microelectrodes were coated with chitosan multi-walled carbon nanotubes (MWCNT) hybrid through electrochemical deposition. This nanomaterial film allows high loading of electrochemically active (bio-) molecules and improves the electrochemical currents of diffusing electroactive species, which leads to high signals. The high catalytic activity of the hybrid film bioMEMS resulted in oxidation peak separation of the electroactive neurotransmitters along with improved current response with a detection range of 5 × 10−6 M to 2 × 10−4 M and 1 × 10−5 M to 3 × 10−4 M for dopamine and 5-hydroxytryptamine, respectively. Carbon nanotubes are also often coupled with polydimethylsiloxane as polymer nanocomposites for biomedical applications. For instance, these nanocomposites have been used for the fabrication of flexible pressure sensors to detect endovascular aneurysm repair problems [62]. The feasibility of CNT-based NEMS/MEMS has been proved by many researchers. This integration will result in the next generation of nanotransducers.

4. NEMS/MEMS Fabrication Technologies with Nanomaterials

In this section, NEMS and MEMS fabrication technologies, which pertain to nanomaterial manipulation, will be described, as well as the ones which are most commonly used, such as bulk micromachining techniques. The aforementioned techniques are micromachining techniques, and the following ones will be described: photolithography, electron beam lithography, scanning probe lithography, soft lithography, nanoimprint lithography, bulk micromachining, surface micromachining, electro-discharged micromachining, laser micromachining and suspended carbon nanowire technique (Figure 4). In the process of fabrication these techniques can be used sequentially.

4.1. Photolithography

Photolithography, like other lithography processes, uses a substrate, most popularly silicon wafers, onto which a layer of coating is applied. Photoresists are coated on a layer of nitride, oxide, or any metal film that has been either deposited or grown on the aforementioned silicon wafer during the photolithography process. Depending on the type of photoresist, exposing it to light either removes or leaves the sensitive layer behind (positive or negative, respectively). As a result, the pattern is transferred from the overlaying mask to the photoresist [63]. Following that, other techniques such as etching are used. Photolithography can be used multiple times, layer by layer [64].
Photolithography, also known as optical lithography or ultraviolet (UV) lithography, is highly resistant to chemical agents, bases, and acids. It is also distinguished by its high photosensitivity to radiation. It has adequate adhesion to the surface of the substrate. Working resolution is limited by the diffraction of the used light and also the wavelength of the used radiation beam [65]. Photolithography’s rapid and selective UV-curing allows for faster fabrication than soft lithography. In light of this, there has been an increase in the preparation of preceramic photoresists [63].
As it is a relatively common method in practice, it has been beneficial in developing novel approaches to different types of sensors. Micro-machined Al shadow masks have been reported to be used in photolithography to create resonant sensors [66]. Furthermore, a three-step photolithographic process for releasing a resonator structure also has been proposed [66]. Sui, et al. [67], used photolithography to define the chromium (Cr) hard mask after the completion of chromium deposition with the goal of fabricating GaN/AIN. A different photolithography technique known as two-photon lithography has been used for manufacturing complex 3D structures on the micro- and nanoscale [68]. Three-dimensional direct laser lithography based on multiphoton absorption has also been used for nanoscale structure fabrication [69]. However, both photolithography and conventional lithography have been shown to be time-consuming processes; as a result, laser machining techniques are becoming more popular, overcoming this limitation [68].
Aside from directly fabricating NEMS/MEMS, photolithography is used as a fabrication step for cantilevers. UV lithography was used as the first step in the fabrication of cantilevers by Udara, et al. [70]. X-ray photolithography is used to create patterns in relatively thick photoresist layers [71].

4.2. Electron Beam Lithography

Aside from the variations discussed in the following paragraph, electron beam lithography (EBL) is a serial write patterning process that produces patterns as a direct result of a small spot of electrons being deflected across a substrate [72]. The previously mentioned process is mostly used in nanoscale pattern creation [73].
The advantages of EBL over other methods include the absence of a mask for pattern transfer [73]. Moreover, it gives precise control over the energy and dose of the electron beam, as well as accurate registration over small areas on a wafer and lower defect densities [64]. However, the application of EBL is constrained by its slow exposure speed, high cost, and resolution limitations caused by electron beam diffraction in solids [64,74].

4.3. Scanning Probe Lithography

Scanning probe lithography (SPL) is another type of atomic-scale write patterning process based on scanning tunnelling microscopy, which generates images by passing a conducting tip over a surface with a constant tunnelling current [64]. The advantage of SPL over EBL is that it has higher resolution and lower energy electrons with lateral scattering [64,75]. Aside from that, it has demonstrated other advantages over other lithographic processes. These include low costs, 3D patterning, high-quality patterning, and so on [76].
One significant disadvantage is that SPL, particularly mechanical SPL (m-SPL), which uses force to pattern, is relatively slow [76]. This can be overcome by using different types of SPL, such as thermal SPL [77]. Thermal SPL (t-SPL) uses concentrated heat to activate endothermic decomposition and evaporation of thermally sensitive resist [76].
SPL maximises friction-induced selective etching, which uses a diamond tip to scratch the surface of a resist. It has higher resolution and flexibility, making it a promising method for fabricating micro/nanostructures [78].

4.4. Soft Lithography

Soft lithography, which is primarily used for the fabrication of nano/microstructures, is a method that uses a master stamp to transfer the pattern onto a substrate using a special ink [64]. Because of its biocompatibility and low cost, polydimethylsiloxane (PDMS) has been most commonly used as a stamping material. Soft lithography is most commonly used for the development of microfluidic devices [79]. It is used for developing lab-on-chip and flexible electronics [80].
Soft lithography is typically comprised of six stages [81].
  • Soft mould creation;
  • Preparation of metal and ceramic slurries;
  • Filling of the soft mould with the prepared slurries;
  • Drying or curing and demoulding;
  • De-binding;
  • Sintering.
As previously stated, the soft lithography process is relatively slow due to the extensive steps and processes involved. As a result, photolithography is preferred for ease of fabrication [63]. There are five types of soft lithography, which include micro-moulding in capillaries, micro-transfer moulding, micro-contact printing, replica moulding and solvent-assisted micro-moulding [81].

4.5. Nanoimprint Lithography

Nanoimprint lithography (NIL) is a high resolution (more than 10 nm) and high efficiency lithography technique that employs a patterning technology that involves the deposition and exposure of a low viscosity resist onto a substrate [82]. This process utilises capillary action in which the fluid quickly flows into the lowered pattern mask and the resist will be exposed under UV light for curing. Finally, the pattern mask is removed, leaving the resist on the substrate [83]. NIL is used for nano structuring Si wafers [78], as well as crystalline silicon pillars [84].
Depending on the polymer characteristics, there are three families of NIL [85]:
  • Electrical NIL;
  • Thermal NIL;
  • UV NIL.
NIL has drawbacks despite being a high-efficiency, high-resolution method that is more widely used than other lithography techniques. The pattern stamp’s positioning is critical, and if there is a mechanical shift, the imprinting will be misaligned [75]. Aside from that, controlling defectivity and template life is difficult [74].

4.6. Bulk Micromachining

Bulk micromachining is a MEMS fabrication process that involves removing specific amounts of the silicon substrate to create the desired structure [71,80]. Etching is a significant subtype of bulk micromachining that will be discussed in detail below [71].
This method is most often performed by depositing a masking material layer and then patterning it by exposing the areas on the substrate which will be etched [86]. It has been used in a wide range of different MEMS applications (MEMS silicon accelerometers) [87], as well as cantilever fabrication [88,89]. Bulk micromachining techniques can be used along with other methods such as wafer bonding [90]. Aside from etching techniques, some bulk micromachining techniques are silicon wet bulk micromachining techniques.
  • Silicon wet bulk micromachining
Used mainly for the fabrication of cantilevers and cavities, silicon wet bulk micromachining is used in laboratories and the industry for MEMS applications [91]. It is used for surface texturing with the goal of minimising the reflectance of light [92].
2.
Etching
Etching is the process of removing materials from a specified area [79]. It is divided into two groups: dry and wet etching. During the process of dry etching, microfeatures are achieved by milling through the usage of an ion beam or using reactive ion etching, whereas wet etching uses etchant solutions [81].
(a)
Wet etching
The amount of etching of a substrate is controlled by parameters such as etching solution concentration, etching temperature, rate, and roughness [79], making wet etching highly modulable. The ability of batch fabrication is an important aspect of wet etching, resulting in it being widely used in the industry [91]. When it comes to NEMS/MEMS fabrication, wet etching can be used in creating anisotropic directional etches on crystalline materials [66] for the production of cantilevers [70], as well as in the fabrication of microfluidic devices [93].
(b)
Dry etching
As the name implies, dry etching does not use any etchant solution. There are two types of dry etching, namely ion beam etching and reactive ion etching [79]. The ion beam etching produces smooth edges on curves, whereas reactive ion etching uses inductively coupled plasma (ICP-RIE) to remove the chromium oxide sacrificial layer at a cryogenic temperature [84,94]. Dry etching is a common method used in NEMS/MEMS applications. For example, this method was adapted in the process of making 3D silicon nanowires for FET biosensors [95]. With the goal of making PIN diodes, Schottky diodes, junction field-effect transistors (JFETs), and metal oxide semiconductor field-effect transistors (MOSFETs), the previously mentioned ICP-RIE is used [96]. ICP-RIE is also used in the fabrication of resonators for high-frequency bands [97]. Moreover, dry etching is used as a step-in fabrication of silicon-on-glass MEMS [98].

4.7. Surface Micromachining

As the opposite of bulk micromachining, surface micromachining techniques implore additive processes that utilise thin films. A sacrificial layer is usually used for creating a beam or membrane for moving parts. The most commonly used structural materials are polysilicon and metal [64]. It has various uses, mainly for NEMS/MEMS applications, capacitive MEMS accelerometer [87], and bulk acoustic resonators [66].
The drawback of using surface micromachining in comparison to bulk micromachining is the fact that there is a need for a post-process with the goal of stress release [64].

4.8. Combined Micromachining

Since both surface and bulk micromachining techniques possess disadvantages, a technique based on aspects of both of these micromachining techniques was developed [64]. A combination of both of these methods utilises etching techniques followed by deposition techniques which in turn create more complex structures [79].

4.9. Electron-Discharge Micromachining

Electro-discharge machining (EDM) is the process of machining any conductive material regardless of its chemical and mechanical properties, whilst getting thermo-electric energy as a side product [99]. Similar to EDM, micro-electro-discharge machining or electro-discharge micromachining were utilised in the fabrication of microscale features on any conductive material that gives a crucial advantage [100]. Therefore, it is frequently used in NEMS/MEMS applications [73]. It is used for the development of micro-electrodes and micro-gears [101], as well as accelerometers [102].

4.10. Laser Micromachining

Laser micromachining is a NEMS/MEMS fabrication technique which does not require a mask, nor any post fabrication steps [79]. Aside from not needing a mask during fabrication, laser micromachining is precise, fast and has fewer heat-affected zones [68]. In addition to its frequent use in the fabrication of biomedical technologies, laser micromachining is widely used in optoelectronics, photonics and applications utilising glass [93]. It is suitable for rapid prototyping and is commonly used in fabrication of microfluidic devices [79]. However, there is still on-going investigation into its parameter optimization for better performance [103].

4.11. Electrospinning

The production of nanofibers from polymer with the aid of a high electrostatic force is called electrospinning. During this process, the jet moves towards the receptor and when it is within a distance of a few nanometres with the help of Casimir force, the jet and the receptor attract which in turn attaches nanofibers to the surface of the receptor. These nanofibers are in nano-range diameters and can be used in many NEMS/MEMS-based devices [104,105].
The advantages and disadvantages of the discussed fabrication strategies are summarised in Table 1.

5. Clinical Applications of Nanomaterials-Based bioNEMS/MEMS

Emerging as one of the most advanced miniaturised electronics, bioNEMS/MEMS offer the indispensable potential for various analytical biology characterisation in biomedical fields. In the current research, increasing interests are directed at nanomaterials (e.g., nanocrystal, organic/inorganic nanoparticles, carbon nanoparticles and composite-nanoparticles) based nano/microsystems to sense or actuate biological components targeting diagnostics (e.g., detection early sign of diseases—cancers, cardiovascular diseases, oral diseases), monitoring (e.g., disease monitoring—diabetes, glucose and blood coagulation monitoring) therapeutics (e.g., drug delivery actuators) and cell manipulation applications [3,111,112,113]. This section highlights the clinical applications of nanomaterials-based bioNEMS/MEMS classified into diagnostic, monitoring, therapeutic and cell manipulation applications in great detail.

5.1. Diagnostic Applications

The ability to manufacture bioNEMS and MEMS in nano and sub-micron scales has facilitated the development of user-friendly diagnostic devices. Advancement of NEMS/MEMS in various industries such as lab-on-a-chip (LoC), miniaturised biosensors and point-of-care testing enables rapid diagnosis and monitoring of chronic diseases such as asthma, diabetes, Alzheimer’s disease, cardiovascular diseases, and cancers. Most notably, early detection of cancer biomarkers is feasible with the help of these biosensors. Table 2 summarises recent research on diagnostic and monitoring applications using bioNEMS and MEMS technology. The transition of NEMS and MEMS technology to carbon-based NEMS/MEMS (C-NEMS/C-MEMS) for cancer biomarkers detection is an attempt to increase the sensitivity of the sensing devices. Carbon-NEMS/C-MEMS involves the patterning of carbon-based materials followed by pyrolysis in an oxygen-free environment. According to Forouzanfar, et al. [114], C-NEMS/MEMS approaches offer facile fabrication of electrochemical-based sensors equipped with tailored glassy-like carbon structures (e.g., three-dimensional micropillar, microchannel, microarray) and active surfaces for functionalization due to the high presence of carboxyl groups (-COOH). This has been observed in several studies that developed label-free C-MEMS aptasensors through the introduction of carbon-rich materials (e.g., reduced graphene oxide, SU-8 photoresist) for early detection of cancer biomarkers. In both studies, electrochemical characterisation revealed a comparable linear range of detection [114,115].
The pressure sensor is another class of sophisticated biosensors with the ability to transduce external pressure into electrical signals. Using NEMS and MEMS technology, diagnosis and prevention of progressive vision impairment disorders such as glaucoma can be achieved by employing pressure detection. Glaucoma is known to be caused by elevated intraocular pressure (IOP) and without proper treatment, can lead to permanent blindness due to chronic optic nerve damage [116]. In most cases, glaucoma is non-detectable until a later chronic stage. A research group led by Liu, et al. [117] developed a contact lens sensor using self-assembly graphene (SAG) film and MEMS technology. The advanced sensor expressed ultra-high sensitivity towards simulated IOP fluctuation as shown in Figure 5. Furthermore, the sensor showed reliable IOP detection in vitro tests on silicone (Figure 5a) and porcine eyes. The contact lens biosensor showed enhanced sensitivity owing to the excellent strain sensitivity and desirable graphene properties with flexible overlapping morphology as illustrated in Figure 5c. To date, limited studies were invested in pressure-based biosensors using NEMS and MEMS technology despite promising findings in recent research that show the applicability of this technology for early diagnosis of glaucoma. Despite promising findings in recent research that show the applicability of this technology for early diagnosis of glaucoma, limited studies have been invested in pressure-based biosensors using NEMS and MEMS technology to date. With the estimated projection of glaucoma to reach 112 million cases by 2040, immediate interventions and research investment are required [116]. Examples of recent bioNEMS/MEMS with nanomaterials in diagnostic, monitoring and therapeutic applications are summarised in Table 2.
Figure 5. (a) Photomicrograph of the test platform configuration using silicon cornea, (b) schematic depiction of activities that could induce acute intraocular hypertension and (c) schematic illustration of the sensing mechanism of contact lens integrated with SAG film. Reprinted with permission from ref [117]. Copyright 2022 John Wiley and Sons.
Figure 5. (a) Photomicrograph of the test platform configuration using silicon cornea, (b) schematic depiction of activities that could induce acute intraocular hypertension and (c) schematic illustration of the sensing mechanism of contact lens integrated with SAG film. Reprinted with permission from ref [117]. Copyright 2022 John Wiley and Sons.
Nanomaterials 12 04025 g005
Table 2. Example studies on nanomaterials-based bioNEMS/MEMS for diagnostic, monitoring and therapeutic applications (selected studies between 2017–2022).
Table 2. Example studies on nanomaterials-based bioNEMS/MEMS for diagnostic, monitoring and therapeutic applications (selected studies between 2017–2022).
TechnologyType of NanomaterialsBiomedical Application(s)Tested
Biological
Samples
Outcome(s)Application(s)Ref
NEMS1D photonic
crystal
Cancers-
Improved optical sensitivity (13 452 nm/RIU) figure of merit (FOM) (7 912 RIU−1) compared to existing biosensors
Label-free diagnostic biosensor
(diagnostic)
[112]
Mesoporous silica nanoparticlesCancers-
Fast delivery of drug loaded nanocarriers within 30 min without the need for external stimulation
Controlled release of payloads
Targeted drug delivery for cancer therapy
(therapeutic)
[3]
MEMS Titanium dioxide/tungsten trioxide (TiO2/WO3) nanocomposites Cardiovascular diseases-
Highly selective towards isoprene (breath biomarker for cholesterol abnormalities) ranging between 20 ppb (abnormal cholesterol) to 80 ppb (normal cholesterol)
Cholesterol breath analyser
(diagnostic)
[118]
Monolayer grapheneCoronavirus disease (e.g., COVID-19)Nasopharyngeal liquids
Highly sensitive and label-free detection of unamplified SARS-COV-2 as low as 0.02 copies/μL in nasopharyngeal swap samples
Point-of-care testing
(diagnostic)
[4]
Biofunctionalized reduced graphene oxideDiabetes Human blood serum
Ultra-high selectivity towards glucose compared to uric acid, cholesterol and ascorbic acid
4.3 times higher glucose sensitivity for 3D carbon electrodes compare to 2D carbon electrodes
Electrochemical glucose biosensor
(monitoring)
[113]
Reduced graphene oxide, gold and platinum alloy nanoparticlesDiabetes Human sweat
High sensitivity with favourable glucose detection limits
Comparable efficiency to the commercial sensor with glucose recovery between 93 to 96%
Wearable glucose sensor
(monitoring)
[119]
Cerium oxide-polyethylene glycol-glucose oxidase
nanoparticles
Diabetes Artificial tear
In vivo: Detectable changes in reflection spectrum corresponded to glucose concentration in diabetic mouse group compared to non-diabetic mouse group
Contact lens
biosensor
(monitoring)
[120]
Magnetic nanoparticlesCardiovascular diseases-
-
Negligible increase in simulated body temperature (~2 °C), thus reducing risk of induced hyperthermia
-
In vivo catheter implantation showed good biocompatibility
Implantable catheter
(therapeutic)
[118]
C-MEMS Gold (Au) nanoparticles Cholesterol-associated diseases-
Au nanoparticles deposition facilitated selective immobilisation of cholesterol oxidase
High sensitivity and reliability to detect the low concentration of cholesterol
Cholesterol rapid detection
(diagnostic)
[21]
Interdigitated array nanoelectrodeCardiovascular disease (myocardial infarction)Human serumHighly sensitive for cardiac myoglobin detection as low as 0.43 pg/mLImmunosensor for cardiac Biomarker
(diagnostic)
[121]
Gold particles with dendritic nanostructures (AuNs)Diabetes-
AuNs/nanoporous sponge-like networks electrodes showed 3.8-fold higher sensitivity and 27.7-fold increase in detection limit compared to AuNs/bare carbon electrodes
Non-enzymatic glucose sensor
(monitoring)
[122]

5.2. Monitoring Applications

Apart from diagnostic applications, these nano/microsystems have shown promising potential in various disease monitoring applications. Undoubtedly, research on monitoring glucose levels for diabetic patients receives much attention from the mainstream scientific community as diabetes is one of the top four leading non-communicable diseases behind cardiovascular diseases, cancers, and respiratory diseases. It is estimated that 693 million patients will be diagnosed with diabetes and at least half of the world’s population will remain undiagnosed by 2045 [123]. As a result, numerous studies are being directed towards early diagnosis of diabetes, monitoring of blood glucose levels and delivery of therapeutic agents (e.g., insulin). In recent years, the use of biological fluids such as whole blood, sweat and tears has shown promising outcomes for monitoring glucose levels [113,119,120] (Table 2). Monitoring glucose with enzymatic-based sensors (e.g., electrochemical and optical sensors) that rely on the detection of the glucose oxidase enzyme can be difficult. This is because of the fact that the glucose oxidase enzyme is extremely sensitive to changes in pH, body temperature, and ionic strength [124]. Despite challenges in regulating sweat behaviour in situ, current proof-of-concept studies have demonstrated the ability of their biosensors to detect and monitor glucose levels from biological sweat [119,125]. In this case, future research could look into how different real sweat conditions affect enzyme-based biosensor sensitivity and reliability.
Surgical technology has recently followed the trend of incorporating nano/micro-systems to monitor surgical performance and post-operative treatments. Because of advancements in sensor technology, real-time monitoring of various post-operative care and recovery such as fracture healing, early signs of infection determination that could potentially lead to sepsis if not properly assessed and treated, and wound recovery monitoring is now possible [126]. Implantable biosensors are also useful for many invasive implant surgeries (e.g., orthopaedic implants—total knee and hip replacements, spine implants) because they provide real-time implant monitoring or post-implantation data for improved clinical outcomes [5,126]. Anderson, Wilson and Holdsworth [5] demonstrated technical modifications to a MEMS pressure sensor that allowed them to measure changes in deformation up to 350 μm.

5.3. Therapeutic Applications

Recent MEMS technology advancements enable the delivery of multiple therapeutics at a predetermined time within a miniaturised implantable “pharmacy-on-a-chip” design. In 2012, Farra, et al. [127] pioneered the first human clinical trial by subcutaneously implanting a “pharmacy-on-a-chip” device to deliver an anti-osteoporosis drug an alternative anabolic osteoporosis treatment. The trial confirmed systematic delivery of the anti-osteoporosis drug (up to 20 doses per device) equivalent to the conventional subcutaneous injections. Furthermore, eight osteoporotic postmenopausal women who volunteered for the trial experienced no cytotoxicity side effects from the implanted devices and experienced minimal pain before and after implantation. Their promising findings served as a stepping stone for further development of future devices that can deliver precise dosage by opening up to 100 different reservoirs within microseconds [128]. A recent study by Li, et al. [129] developed hybridised gefitinib-loaded nanocarriers for lung tumour-targeted delivery. The hybridised nanocarrier systems embedded within the NEMS platform demonstrated a pH-responsive with prolonged in vitro drug release. Their study showed that the NEMS system was reliable for improving the delivery of gefitinib to the targeted lung tumour cells. Overall, the system has a lot of potential as a multifunctional and high-performance biomaterial for targeted lung tumour drug delivery with enhanced antitumor effect and sustained release behaviour. The latest research has demonstrated effective transmembrane delivery of drug nanoparticles through the adaptation of NEMS to the resonator technology. Specifically, Lu, Palanikumar, Choi, Huskens, Ryu, Wang, Pang and Duan [3] established an acoustic method for direct delivery of doxorubicin (DOX) across biological barriers of the cell membrane and endosomes. In this research, the delivery of DOX-loaded mesoporous silica nanoparticles to the targeted cytoplasm was navigated via controlled hypersound from the NEMS resonator. The hypersound-influenced drug delivery facilitated cellular uptake of drug nanocarriers with a size ranging between 100 to 200 nm showing the versatility to deliver a wide range of therapeutic drug nanoparticles for cancer therapy. Another interesting and impactful research niche to improve the lives of Alzheimer’s patients correlates to the in vitro screening and monitoring of Alzheimer’s therapeutic drugs containing acetylcholinesterase (AChE) inhibitors. In this instance, Chae, et al. [130] developed a low-cost graphene-based enzyme biosensor for screening and monitoring enzymatic reactions between AChE and AChE inhibitor-therapeutic drugs. As compared to conventional in vivo screening using transgenic animals or heavy dependence on high-tech research instruments to analyse physiological fluids, the low-cost graphene-based enzyme biosensor successfully screened two different AChE inhibitor-therapeutic drugs (i.e., donepezil and rivastigmine). They also observed a significant enzymatic reaction between AChE and AChE inhibitors with the ability of the biosensor to detect low up to micromolar concentrations of AChE.
Nanochannels have been employed in recent studies using NEMS technology for drug delivery systems (DDS). DDS can be divided into passive and active targeting strategies. Nanochannels are part of the passive targeting strategy by having a supersaturated drug reservoir for the passive release of drugs at the targeted sites through concentration-driven transport [131]. By carefully adjusting the size and surface chemistry of microchannels, it is possible to achieve sustained drug release. In vitro and in vivo assessments using rodents, dogs, pigs and non-human primates have validated the applicability of nanochannel NEMS technology as drug nanocarriers [132]. Since the discovery of nanochannels, several promising nanochannel innovations have been commercialised and available in the market with controlled drug release properties. Recently, NanoMedical Systems launched the nStrada platform, a nanochannel chip drug delivery technology platform. At present, the company is leading the innovation of nanochannel chips by having a refillable system with high dosing properties compared to their competitors [133].

5.4. Cell Manipulation

The future of bioNEMS/MEMS is still blooming with limitless design possibilities considering its multifunctionalities in a myriad of biomedical applications. The technology has become an ideal tool for cell manipulation in the field of cell patterning and cell-sorting applications as illustrated in Figure 6 [111,134]. Sorting and patterning of cells or biological molecules using microchannels or nanochannels can be very useful for various clinical diagnoses, stem cell research, cell-based drug screening, and tissue engineering [135]. The guidance of cellular movement within the micro/nanochannels can be achieved via passive (e.g., channel/surface geometry, hydrodynamic forces) or active mechanisms. Active mechanisms rely on external stimulation such as dielectrophoresis (DEP), optical, magnetophoresis and acoustophoresis [111,135,136]. Integrating nano/microfluidic and LoC with bioNEMS and MEMS technology allows the fabrication of low-cost, high-throughput devices for cell sorting and separation of biological samples. The current state-of-the-art technology demonstrated promising sorting and separation of rare blood cell types (e.g., sickle-cell red blood cells), circulating cancer, or parasites from biological blood samples [136,137]. The design of microfluidic systems using nanomaterials-based bioMEMS technology can be traced back to a conceptual microfluidic bioparticle sorter conducted by Nieuwenhuis and co-workers in 2005 [138]. In this study, gold (300 nm) and chromium made up the different configurations of sorter electrodes (i.e., triangular and tetrahedral) for potential cell alignment and DEP separation. The shift towards sorting and manipulation of other biological components shows the potential of this technology for other biomedical applications. For example, Fujiwara, Morikawa, Endo, Hisamoto and Sueyoshi [6] developed a micro-nanofluidic system with nanochannels for size-sorting of exosomes, a subtype of extracellular vesicles. Classification of exosomes depending on sizes may yield more information on the exosomes’ biogenesis and a better understanding of extracellular vesicle mechanisms as potential biomarkers for liquid biopsy of cancers. In this instance, the micro-nanofluidic design developed by Fujiwara and colleagues demonstrated the ability to control the size-sorting of exosomes by manipulating the electric double layers of the integrated nanochannels.

6. Integrating NEMS/MEMS with IoT Applications with the Help of Nanomaterials for Biomedical Applications

Nanomaterials not only play an important role in sensing technology but also contribute to the boosting of wireless body networks and the emergence of the sixth generation (6G) mobile networks. The progress of nanotechnology to some extent, has had an impact on the size of wireless devices and led to the rise of nanodevices [139]. The integration of information from nanodevices with communication technologies, the Internet of Things (IoT) as well as the Internet of Bodies (IoB) has an enormous impact on our ability to improve human health and longevity [140]. Highly functional nanodevices for real-time monitoring and response against threats before they yield catastrophic consequences are highly dependent on modern telemedicine. However, processing power and memory will be the limitations of nanodevices due to their tiny form factors [139]. To overcome these limitations, nanodevices are incorporated into either whole or parts of nanocircuits namely nanosensor, nanoprocessor, nanomemory, nanoantenna, and nanobattery for robust technologies [140]. Adopting nanomaterials within nanocircuits not only ensures the functionality, energy efficiency, and accuracy of nanodevices while reducing their size, but also ameliorates the device properties and function itself compared to normal device size.
This section will review the role of nanomaterial in the development of nanodevice and IoT for healthcare applications.

6.1. Nanosensor

Nanosensors are one of the important parts of a nanodevice that are capable of sensing physical, chemical, and biological monitoring [141]. It comes in a nanoscale size, nanosensor is a miniaturised traditional bioanalytical method, such as immunoassay, chromatography, and spectroscopy, by integration of microfluidics, and microelectronics in lab-on-a-chip [142]. Nanomaterials and nanotechnology are widely used during the fabrication process of nanosensors to enhance accuracy, precision, and sensing detection [143,144]. The integration of nanosensor with IoT and body area network (BAN) technology in wearable and implantable sensors offer many advantages in providing real-time point-of-care diagnostics and therapeutics. Furthermore, the IoT and BAN platforms are considerate of patients’ health and information privacy, as well as security concerns [145]. A comprehensive table highlighting the nanomaterials based nanosensors in wearable devices and implants for various biomedical applications is provided in Table 3.

6.2. Nanoantenna

Antennas are an important part of wireless communication data transfer, including body area networks (BAN) and IoT. In BAN, signals from nanosensors or biosensors are transmitted to the IoT gateway or bridge by using Bluetooth (IEEE 802.15.1) and Zigbee/Xbee (IEEE 802.15.4) connectivity modules within the permitted frequency bands for medical devices according to the Industrial, Scientific, and Medical (ISM) radio band, 2.45 GHz. The IoT gateway or bridge then transmits the received signals from the nanosensors or biosensors to the cloud storage via an internet connection (Wi-fi, 4G/5G, ethernet, etc.).
The nanoantenna is a nanoscale version of radio-frequency (RF) or microwave antennas used for signal transmission and reception [152]. Desirable properties such as radiation, bandwidth, and transmission efficiency of the nanoantenna are not only maintained but enhanced with the use of nanomaterials (e.g., quantum dots, metallic nanostructures/nanoparticles, graphene, indium-doped tin oxide (ITO)-based CNT and silver-nanowires) compared to the conventional metal antenna [153,154,155]. Recently, 2D titanium carbide (MXene) coating is explored in wireless communication and offers an opportunity to produce transparent antennas [156]. To date, the nanomaterials-based nanoantenna not only improves the transmission performance of existing wireless modules and communication technologies such as Bluetooth, Zigbee/Xbee RF modules, and 5G, but also contributes to the 6G communication technology. The evolution of the 6G network has become an important point in the healthcare sector toward smart healthcare treatments that eliminate time and space barriers through patient remote monitoring and telesurgery, for instance [157].
Aside from high-speed communication, nanoantenna radiation and electromagnetic wave properties have the potential to be used for biosensing and imaging [154,158], energy harvesting [159], optical circuits [160], and in vitro and in vivo applications [161]. Table 4 summarises the benefits of nanomaterials-based nanoantennas for wireless sensing technology and healthcare applications.

6.3. Nanoprocessor

The processor is the heart and brain of every electronic device. It is primarily made from silicon and consists of transistors. The nanoprocessor and its core (nanocore) are constructed from intrinsically nanometre-scale building blocks and designed to be implemented with minimal logic resources to control memories for small tasks [165,166]. In IoT applications, the main feature of a nanoprocessor is the transfer of the data flows from far-end devices through the communication channels in real time. Small energy consumption and low cost yet high speed are among the important features for nanoprocessors to function as IoT nanodevices [165].
The development of the nanoprocessor is yet to surface and is still in its state-of-the-art stage [167]. However, the use of nanomaterials in processor and core fabrication demonstrates their performance ability compared to silicon-semiconductor-based materials [168]. Table 5 summarises the role of nanomaterials in reducing energy consumption, boosting processor speed and moving towards flexible read-access memory for IoT and wearable smart data storage.

6.4. Nanobattery

Nanobattery is not only defined based on its nanosize but uses nanotechnology in a macro-sized battery to enhance its performance and lifetime [172]. Nanobattery performance metrics include higher power density, faster charging times, and longer shelf life. Nanomaterials with different morphologies such as confined nanoparticles, nanosphere, nanopyramid, nanowires, nanostraws, nanotubes, and nanosheets with different compositions can function as an anode, cathode, separator, and electrolytes in nanobatteries [173,174]. The nanomaterials play important roles in rechargeable lithium (Li) batteries in enhancing Li storage hosting of active materials, polysulfide (PS) adsorption and conversion, oxygen reduction reaction (ORR), and oxygen evolution reaction (OER) [173].
The emergence of wearable devices has led to the development of flexible and stretchable batteries due to reasonable energy densities, being lightweight, portable, wearable, and implantable [174,175]. Carbon nanomaterials such as graphene, CNTs, and their composites have demonstrated potential for modifying the composition and fabrication of flexible electrodes [174]. Nanomaterial-based electrodes in Li-ion and lithium-sulphur batteries such as graphene/MXene heterostructures exhibit outstanding performance in improving the electrochemical performance of fast charging batteries [176,177]. The incorporation of safe and rechargeable zinc/manganese dioxide chemistry into biocompatible poly(styrene-isobutylene-styrene) (SIBS) polymer and carbon nanofiber has resulted in the development of washable rechargeable batteries. Nanomaterial-based flexible, stretchable, and washable batteries have attracted significant interest for powering systems in wearable devices, electronic skin, strain sensors, and implantable medical devices [178].

7. Safety and Toxicity, the Biocompatibility of Nanomaterials-Based bioNEMS/MEMS

7.1. Safety

Numerous regulatory bodies that evaluated the effects of nanomaterials on occupational safety and health have published their official guidelines [179,180,181,182,183,184]. Potential dangers may include the following: (i) inhaled nanomaterials may deposit in the respiratory tract and harm lung cells and tissues, (ii) certain nanomaterials have the potential to breach cell membranes and harm cellular structures and functions, (iii) risk of explosions and fires may be present in some nanomaterials because they may be pyrophoric or easily combustible.
Inhalation, dermal contact, unintentional injection and ingestion can all result in exposures, and the risk grows as exposure time and nanoparticle concentrations in the sample or air rise. The greatest exposure hazard is inhalation. Sonication, shaking, stirring, pouring, or spraying can also cause inhalation exposure, while nanomaterials suspended in a solution or slurry present a lower risk. The least dangerous nanoparticles are those that are fixed inside the matrix. Risk assessment, engineering and administrative control are recommended work practices [179]. To identify the necessary control measures and ascertain whether the implemented controls are successful in reducing exposure, a thorough risk assessment should be carried out. It is advised to use high-efficiency particulate absorbing (HEPA) filter-equipped ventilation enclosures or local exhaust ventilation that operate at negative pressure. Administrative controls consist of developing protocols for decontaminating surfaces and cleaning [184].
The term “nano” should be included in the descriptor, for example, “nano materials”, and should be stored in labelled containers that indicate their chemical content and form.
Building validated models that can forecast the emission, passage, modification, deposition, and uptake of nanomaterials in the environment is necessary for scientists and researchers to evaluate the safety of nanomaterials [185,186].
Because of their small size, nanoparticles found in bioNEMS/MEMS structures may be able to interact with different biological barriers in the body. This interaction could cause a toxic effect. On the other hand, drug delivery could benefit from this. However, it is crucial to comprehend whether the nanoscale carriers have any negative effects. The fate of nanomaterial during diagnosis or treatment, as well as how the particles are excreted, biodegraded, or accumulated, is crucial because accumulation can have negative long-term effects, and therefore the safety profile of nanomaterials in humans is still a significant concern [187,188].
Two categories can be used to describe safety and health hazards associated with the use of nanomaterials in bioNEMS/MEMS: (i) potential medical applications for treating diseases using nanotechnological innovations; and (ii) additional health risks associated with nanomaterial exposure [189,190].
It has been speculated recently that it is time for a shift in perspective when it comes to the safety evaluation of nanomaterials in bioengineering (Figure 7). The suggestion presented recently is that we should stop worrying about insurmountable problems and instead pay attention to the significant progress that has been made in past few years as new and advanced approaches have been integrated into nanosafety research [191]. To use the newly developed techniques for the risk and hazard evaluation of biological nanomaterials, we need a paradigm shift in the way that nanosafety assessment is carried out.

7.2. Toxicity

There are many worries about the potential toxicity of nanoparticles. Suitable regulations for their use and evaluation are regularly amended and the recently adopted ISO standard 10993-22 is devoted to the biocompatibility assessment of nanomaterials used in medical devices [192]. Cell survival has been found to be influenced by nanoparticle features such as size, surface charge, dispersion state, matrix composition, surface functionalization, and protein corona formation [193]. These parameters must be mastered in order to conduct a biocompatibility assessment using the appropriate methodology. Numerous assays are used to determine the toxicity of the nanostructures because there are more nanomaterials and nanoparticles every day [185]. These tests are categorised into two main groups: in vitro and in vivo tests. Table 6 summarises the cytotoxicity methods recently used in nanomaterials toxicity assessment along with the main effects observed during evaluation in specific cell lines and tissues.

7.2.1. In Vitro Assessment of Nanomaterials Toxicity

Cytotoxicity Assays

In vitro cytotoxicity assays are among the gold standard technique to assess the cytotoxicity of nanomaterials.
  • Microculture tetrazolium assay
The colorimetric microculture tetrazolium assay is intended and widely used for a qualitative evaluation of the nanomaterial cytotoxicity [208]. It is a non-radioactive assessment technique that measures cell viability using the metabolic activity of the mitochondria. When it comes to cytotoxicity testing of nanomaterials-based NEMS/MEMS MTT test has been employed in various systems. Nano-crystalline Zr x -Cu100x thin films with thicknesses ranging from 50 to 185 nm were assessed in terms of their cytotoxicity. As determined by the MTT assay, Zr-Cu thin film is suitable as a bio-coating for minimally invasive medical devices because it did not cause cytotoxicity in osteoblast cells [194]. A graphene field-effect transistor-based artificial synapse was used for the MTT test of the bioMEMS system, and the results showed that the cell viability (against HK-2 cells) shows no decrease with the increased concentration, which suggests that the silver gel is a biocompatible material [195].
The MTT test has been employed in the initial biocompatibility testing of a peristaltic micropump that is wirelessly controlled in murine inner ear drug delivery, where a silicon MEMS-based 32-kHz oscillator was used [209]. A biocompatibility study was conducted, and the outcomes showed that the micropump parts passed important biocompatibility tests after a month a micropump prototype was implanted without any signs of infection or inflammation. According to these findings, these particular micropump parts passed important biocompatibility tests, indicating that they were appropriate for translational applications such as subcutaneous implantation in both humans and animals.
2.
Trypan blue exclusion assay
Trypan blue testing has recently been employed to examine the cytotoxicity of various coatings used with resin 3D printing. It has been demonstrated how multiple procedures can be coupled to offer better biocompatibility, which in turn increases accessibility to 3D-printed BioMEMS manufacturing. Any combination of the post-process treatments resulted in viable cultures (above the 85.00% threshold) for HL-1 cells. Similar performances of above the 85% threshold were observed for all of the coatings (PDMS, PS, SU-8, Au, and Medco/PET), with PDMS outperforming the others [197]. Cytotoxicity in encapsulated particles in aqueous ferrofluid droplets was examined, and a magnetic field was used to separate the encapsulated particles from the empty droplets. Trypan blue test results showed that 86% of cells were still alive after 2 h when compared to the control, showing that the ferrofluid used in the experiments had little impact on the viability of the cells. These findings suggest that the proposed method for sorting empty droplets in biochemical assays is appropriate [210]. Trypan blue testing demonstrates also that both the silicone-modified polyurethane films and nanofibers caused less toxicity in contact with cells. In contrast to the positive control, the cells can maintain their spreading shape and discrete intracytoplasmic granules without experiencing cell lysis, which can be considered as survival and the morphological grade of cytotoxicity is supposed to be zero [196].
3.
Clonogenic assay
The carbon nanoparticles’ relative toxicities are described in a recent review [198]. It has been shown that the small size of nanoparticles permits them to penetrate through harmful biological barriers. Depending on the type of analysis and models, negative consequences are highlighted.
4.
Apoptosis assay
Apoptosis, or programmed cell death, has found significant application potential in the evaluation of nanotoxicity. Analysis of the DNA damage caused by polysaccharide-surface functionalised (coated) and non-functionalised (uncoated) silver nanoparticles in mammalian cells, including mouse embryonic stem cells and mouse embryonic fibroblasts, revealed that severe DNA damage could cause the cells to undergo apoptosis. The enhanced activity of the apoptosis markers caspase-3 and caspase-9 provide further proof that silver nanoparticle therapy activated the apoptosis pathway in Drosophila melanogaster larval tissues [199].
5.
DNA laddering
Various genotoxic effects of graphene nanoparticles were hypothesised based on the recent literature findings [200,201,202]. There are not many investigations on graphene genotoxicity caused by direct interaction with DNA as of now [200,202,203]. It has been well documented that DNA damage results from oxidative stress brought on by graphene nanoparticles [200,203]. Studies on other forms of indirect genotoxicity, including epigenetic toxicity, inflammation, as well as autophagy, mostly concentrate on the genotoxic effects brought on by graphene. There is not much data on the mechanisms underlying this impact [211]. The inherent physicochemical characteristics of graphene nanoparticles (such as surface functionalization or coatings), exposure dose or durations, and their destination in organisms or the surroundings will all affect how genotoxic they are [200,212].
6.
Caspase assay
Caspase assay has recently been used for determining the molecular basis of hepatic toxicity brought on by mesoporous silica nanoparticles (MSNPs) both in vitro and in vivo. The findings showed that these nanoparticles caused liver inflammation, hepatic cell pyroptosis, and hepatotoxicity. These findings shed new light on the hepatotoxicity caused by MSNPs and show that the NLRP3 inflammasome, pyroptosis, and reactive oxygen species (ROS) are effective targets for raising biocompatibility and lowering the potential toxicity of MSNPs. To avoid the adverse effects of MSNPs in future biomedical applications, more study is required to assess the association between hepatotoxicity and the sizes or surface chemical modifications of MSNPs in vivo [204].
7.
Comet assay
The possibility for DNA damage by nanomaterials is the most significant of their possible impacts. The development of standard techniques for genotoxicity detection is a key objective of the EU project NANoREG. One of the proposed techniques mentions the use of comet assay as a tool to find DNA strand breaks that have been induced. Using two different human lung epithelial cell lines, eight different nanoparticles—titanium dioxide (TiO2) NPs, silica dioxide (SiO2) NPs, zinc oxide (ZnO) NPs, cerium dioxide (CeO2) NPs, silver NPs, and multi-walled carbon nanotubes were examined [205]. The outcomes supported the usefulness of the comet assay to identify nanomaterial’s potential for genotoxicity. The findings suggest that the majority of the nanomaterials had mild to considerable genotoxic effects, demonstrating the applicability of the cell line to assess a material’s genotoxic potential.
8.
TUNEL assay
Rat liver and intestinal NLRP3 inflammasome, oxidative stress, and apoptosis were studied in vivo and in vitro using TiO2 NPs [206]. Titanium dioxide nanoparticles increased cytotoxicity, oxidative stress, and apoptosis rate in a dose-dependent manner. Additionally, TiO2 NPs increased the gene expression of the inflammation and apoptotic pathways in the liver and gut. The liver and gut experienced morphological alterations as a result of TiO2 NPs. TiO2 NPs may cause oxidative stress, inflammation, and apoptosis, which may have deleterious effects on the structure and operation of the liver and the intestines.
9.
Annexin V and Propidium iodide (PI)
From the standpoint of cytotoxicity, graphene oxide–silver nanoparticles (GO-AgNPs) have attracted a lot of interest for their potential in biomedical applications. However, it is unknown if GO-AgNPs are hazardous to people and animals. In caprine foetal fibroblast cells, GO-AgNP-mediated cytotoxicity and epigenetic modification status were found. It was completed using the annexin V/PI assay [207]. The results showed that GO-AgNPs cytotoxicity is dose-dependent. By decreasing cell viability, producing ROS, boosting lactate dehydrogenase and malondialdehyde leakage, and enhancing the expression of pro-apoptotic genes, GO-AgNPs significantly increased cytotoxicity. DNA hypomethylation and DNMT3A expression were both induced by GO-AgNPs. The prospective applications of GO-AgNPs in biomedicine should be reassessed because they increase the production of ROS, induce apoptosis, and result in DNA hypomethylation.

Oxidative Stress Assays

  • 2,7-Dichlorodihydrofluorescein (DCFH) assay
When DCFH is used in an assay, ROS transforms it into the fluorescent substance 2,7-dichlorofluorescein, which is then identified by fluorimetry. Since DCFH is readily oxidised by several ROS functional groups, it is highly favoured as a cytotoxicity assay. This fluorogenic dye assesses the activity of ROS, including hydroxyl, peroxyl, and others. When a dye enters cells, esterases deacetylate it into a non-fluorescent substance. The latter is transformed into dichloro-dihydro-fluorescein by reactive oxygen species, which can be detected with a fluorometer, a flow cytometer, or a fluorescence microscope with an excitation spectrum at 485 nm and an emission spectrum at 535 nm [213]. For quantitative determination of H2O2 and other hydroperoxide group members such as tert-butyl hydroperoxide, DCFH is a regularly used assay. For nanoscale zinc oxide particles, used in the human lung adenocarcinoma cell line DCFH assay demonstrated that these NP cause apoptosis by raising intracellular ROS levels [199].
2.
Lipid peroxidation assay
Lipid peroxidation has been connected in some cases to the cytotoxicity of carbon-based nanomaterials (CBNMs) [198]. The small size of CBNMs permits them to penetrate harmful biological barriers. One of the most significant toxicity mechanisms of NPs is the generation of ROS, which causes oxidative stress, inflammation, lipid peroxidation, and damage to the proteins, cell membrane, and DNA. Oxidative stress is defined as an imbalance between the biological system’s capacity to remove reactive intermediates (superoxide radical anions and hydroxide ions) and the amount of ROS produced. This imbalance may be caused by either an increase in ROS production, a decrease in the cell nucleus defence mechanisms, or a combination of both.

7.2.2. In Vivo Assessment of Nanomaterials Toxicity

A variety of techniques, including biodistribution, clearance, serum chemistry, and histopathology, animal models such as mice and rats are frequently used.

Biodistribution

The biodistribution and nanotoxicity of NPs are governed by their physiochemical properties. Following the administration of nanomaterials, the presence of nanomaterials in key organs or tissues such as the lung, liver, kidneys, heart, brain, pancreas, fat, and muscle is estimated. Then, physical detection techniques can be used to monitor the distribution of NPs in these organs. By using instrumental neutron activation analyses for gold composite NPs in animal tumour models, it was demonstrated that composite nanodevices might target specific organs based on their size and/or surface charge [199]. The biokinetics and biodistribution of nanomaterials delivered through the respiratory system differ from those following intravenous exposure. The biokinetics of intravenously administered nanomaterials does not serve as a substitute for exposure through the lungs or mouth [214]. Rats were used to evaluate the biokinetics and biodistribution in diverse organs over the course of 1 h to 28 days using radiolabelled TiO2 NPs. The liver accumulated the most titanium following intravenous injection, followed by the spleen, skeleton, and blood after 1 h. Thereafter, the blood’s volume rapidly shrank, but the distribution of titanium in the other organs and tissues remained stable until day 28 [215].

Clearance

By analysing the excretion and metabolism of NPs at a certain time point after NP therapy, the clearance of NPs is investigated. Their transit to the liver and kidney was discovered to be significantly constrained [199]. Nanomaterials that enter the vascular system connect with blood cells and are then disseminated to the organs on the periphery. Their size, surface structure, and changes all affect how they are distributed to peripheral organs and how they are cleared. Due to their prolonged circulation duration, nanomaterials that are not effectively eliminated from the body are more likely to interact with cells, tissue, and organs and accumulate there. Different pathways might be used by the body to remove nanoparticles. Although renal clearance is the most efficient excretion method, many nanomaterials cannot be cleared via this method due to size limits (6–8 nm) [214]. Instead, they undergo biliary excretion, where they are broken down by the liver and eliminated through the gastrointestinal tract. As nanoparticles trapped in the mucus are normally carried to the pharynx and then swallowed, mucociliary clearances in the respiratory system can be closely associated with gastrointestinal clearance. Nanomaterials that have been ingested by mononuclear macrophages can stay for a very long time, trapped within the reticuloendothelial system, while elimination via the kidneys and liver can take place over a timescale ranging from 30 min to a few days.

Serum Chemistry

To evaluate nanotoxicity, alterations in serum chemistry after NPs administration are analysed. Blood urea nitrogen, creatinine, glucose, total protein, albumin, globulin, albumin to globulin ratio, sorbitol dehydrogenase, alanine aminotransferase, alkaline phosphatase, and creatine kinase are the parameters that are determined during serum chemistry tests. Red blood cell count, haemoglobin, packed cell volume, mean red cell volume, mean red cell haemoglobin, mean red cell haemoglobin concentration, white blood cell count, differential white blood cell count, and platelet count are all variables that must be determined during a haematological analysis [199].

Histopathology

Histopathological analysis of NP-exposed organs, including the heart, lungs, spleen, liver, kidneys, and eyes, is another method for determining nanotoxicity. After exposure to nano-ferric oxide, follicular lymphoid hyperplasia with inflammatory cells gathered around bronchia was discovered. Alveolar walls thickened after NP therapy, indicating the establishment of fibrosis. The pulmonary alveoli were discovered to contain the macrophage-phagocytosed particles. The pulmonary alveolus contained inflammatory cells such as neutrophils, lymphocytes, and eosinophils [199].

7.3. Biocompatibility

The biological risk has to be calculated as part of risk management, by standards, for the medical device to be regarded as biocompatible [216]. The manufacturer is in charge of assessing the biocompatibility of a finished device, and they should do so as early in the product life cycle as possible to plan a strategy to meet regulatory requirements. An updated version of ISO 10993-1 that stressed the value of learning more about the device was released in August 2018 [217]. Not all biological effects should be evaluated by biological testing, according to the new version of the standard. If a valid reason can be provided, some tests can now be waived. Although there is a truly impressive number of articles describing bioNEMS/MEMS systems for various medical indications, not all of them provide biocompatibility data, much less according to the ISO 10993 standard. However, some studies accurately reported the biocompatibility tests that were actually conducted with the design of NEMS/MEMS systems.
In the literature, less optimistic views on this topic can be found regarding the rapid translation of bioNEMS/MEMS systems into clinical practice. In a recent review, Burton [218] suggests that findings demonstrate that clinical translatability is hampered by insufficient consideration of the biocompatibility of nanorobots with human in-vivo environments in upstream development processes. The design focus is undoubtedly amplified by the absence of meaningful integration with multidisciplinary downstream knowledge.
Utilising MEMS technology, the vaccine-coated solid microneedle patch was created via the process of silicon microneedle arrays using wet chemical etching, inductively coupled plasma reactive ion etching and UV lithography [219]. This combination of vaccine formulation and microneedle to fabricate the delivery device was evaluated in vitro and the material under test exhibited no cytotoxic effects and complied with ISO 10993-5 biocompatibility requirements.
In general, we can take parts of cells and partially make them function outside the organism using nanotechnology. Adenosine triphosphate, a high-energy phosphate particle used to store and deliver energy for work inside living organisms, expands as the engines move. The devices might be on a scale that is comparable to a single molecule. Such devices would malfunction as a result of wear. Particularly for man-made nanomachinery used in physiological conditions, tribological considerations, together with biocompatibility concerns in the design are critical [220].
For implants and encapsulations, biocompatibility should be assessed from two different perspectives [221]. The first is surface biocompatibility, which states that the coating must be chosen to reduce the foreign-body reaction, prevent any significant post-operative inflammation, and prevent the diffusion of harmful materials. In addition to cytotoxicity testing, non-biocompatible chemical groups can be detected by changes in cell shape and metabolism. The criteria for assessing a medical device’s biocompatibility are part of the ISO 10993 standards. The mechanical compatibility between the encapsulated implant and the surrounding tissue is the focus of the second form of biocompatibility, known as structural biocompatibility. To lessen implant insertion injury, particularly to the central nervous system, new materials and techniques are required. In vivo tests need to be performed to evaluate this property.
Another aspect of biocompatibility that plays a vital role in biomedical applications, as well as the repurposing of food waste to fabricate nanomaterials for potential applications in bioNEMS and MEMS is non-toxic and high nutritional values. As discussed previously, nanomaterials are one of the most researched materials in drug delivery. Because of their adaptability and numerous intriguing qualities, including controlled release, blood stability, non-immunogenicity and non-toxic nature, biodegradable polymer-based nanoparticles are used in innovative drug delivery methods [222,223]. Colloidal drug carriers such as liposomes, emulsions, micelles and nanoparticles have shown promising characteristics as future medications that can cross the blood-brain barrier. Similar to this, colloidal systems are employed to control the rate of medication release at targeted areas (i.e., tissues or organs). There are myriad nanomaterials (e.g., silver nanoparticles, gold nanoparticles, carbon nanotubes, laponite nanoplates, polymer-based nanoparticles) that paved the way towards greener MEMS and NEMS [224,225].
Recently, the World Health Organisation also created some guidelines with advice on ethics and good laboratory practices for how to better protect workers from the possible risks of dangerous nanomaterials. Anyone can share their view of the ethics of nanotechnology in several different forums. The opinions of these populations tend not to be based on the vocabulary of the professional ethicists. Researchers, engineers, philosophers and policymakers should communicate and provide their ideas in a similar and simple language that is clearer and more effective [226].

8. Challenges and Conclusions

This review summarises the most recent advancements in bioNEMS and MEMS based on nanomaterials for biomedical applications. We discussed the critical nanomaterial selections and desirable properties (for example, mechanical, optical, and electrical) as biosensors, actuator systems, drug nanocarriers, and other biomedical applications. The evolution of fabrication strategies from traditional techniques (such as bulk and surface micromachining) to more advanced techniques such as photolithography and electrospinning was thoroughly discussed. According to the most recent literature review, the incorporation of nanotechnology into NEMS and MEMS has demonstrated undeniable potential in clinical applications (i.e., diagnostic, monitoring, therapeutic applications, cell manipulation) with dependable functionality in vitro and in vivo. As a result, more groundbreaking discoveries and innovative bioNEMS/MEMS-based platforms are expected to be translated from bench to bedside in the future. Despite thriving research in the field of nanomaterials-based bioNEMS/MEMS in recent years, bioNEMS/MEMS for real-world human applications is still in its infancy. Many ideas remain unexplored, and numerous challenges in this field remain unsolved. As such, some of the major challenges are highlighted as follows. One of the major challenges associated with this technology is the scaling up of materials for commercialisation. In most cases, high-quality nanomaterials involved precise synthesis routes under harsh conditions with heavy reliance on sophisticated instrumentations [9]. Furthermore, expensive chemicals required for nanomaterial synthesis could further hinder the scaling-up process due to projected increased production costs. An attempt to reduce production costs by switching to cheaper materials may jeopardise the quality of nanomaterials, thus the overall functionality and reliability of the bioNEMS/MEMS platforms.
A paradigm shift towards upcycling food waste to create innovative nanomaterials for future bioNEMS/MEMS devices will be a crucial step for sustainable food waste management. The food waste-to-nanomaterials conversion strategy is anticipated to reduce carbon footprints and greenhouse gas emissions (e.g., methane and carbon dioxide) to the atmosphere. Food wastes are rich in valuable nutrients from organic to inorganic biomolecules (e.g., flavonoids, phenolic and alkaloids compounds) with a proven sustainable synthesis of food waste-mediated nanomaterials such as silver and gold nanoparticles. These high-value engineered nanoparticles have shown potential for various biomedical applications due to their non-toxicity, better biocompatibility, and green synthesis properties. Furthermore, the conversion of food waste materials into useful nanomaterials could potentially reduce the cost of source materials, eventually controlling the end-user price [28,44]. Regardless, pre-processing of food waste could contribute to higher overhead costs, thus requiring deliberate strategies to overcome it. Food waste valorisation and zero-waste strategies have opened up exciting research avenues in the NEMS and MEMS applications with the foreseeable future of green device technology.
Taking a glimpse into integrating nanotechnology and NEMS/MEMS technology to IoT applications, limitless opportunities and challenges lie ahead of this exceptional idea. The current state-of-art knowledge proved that there is potential for further research and scientific breakthroughs [168]. Despite the opportunities of IoT technology in improving human health and longevity through rapid disease diagnosis and real-time treatment monitoring. Upcoming research direction is anticipated to delve into proof-of-concept experiments and application research in the next decades is of paramount importance. Above all, challenges associated with nano biodevices into IoT systems can be classified as the followings [140]:
  • Connectivity
In a telehealth system, any disruption in data transmission could have devastating patient consequences. IoT device connectivity depends on the transmission coverage of its wireless modules such as RF module, Bluetooth, and Wi-Fi. For that reason, wireless and internet connectivity in the healthcare setting must be strengthened with ultra-high reliability and low-latency communication networks to prevent connectivity problems during an emergency situation.
2.
Data continuity
Nanodevices such as wearable and medical implants are highly dependent on the battery’s life to ensure their functionalities, efficiency, and performance [227]. Frequently charging the devices may disturb data continuity while direct charging of wearable devices exposes patients to an electrical hazard. The incorporation of nanomaterials in maximised nanobattery runtime and the emergence of wireless energy harvesting through nanoantennas potentially provide uninterrupted access to essential patient data.
3.
Compliance
Healthcare IoT is subject to evolving regulations that vary by country. For example, US Food Drug Administration (FDA), CE marking for European Union, Therapeutic Goods Administration (TGA) Australia, and Medical Device Authority (MDA) Malaysia are compliance bodies that outline the need for device makers, pharmaceutical companies, and other players to ensure the safety, efficiency, and reliability of medical devices including IoT device for healthcare applications.
4.
Coexistence
Other than IoT nanodevices for healthcare applications, there are many existing IoT devices currently available that potentially compete for connectivity. The existence of many IoT nanodevices in specific and limited spaces potentially leads to signal interference between devices that causes connectivity failures or corrupted data.
5.
Cybersecurity
Health Insurance Portability and Accountability Act (HIPAA) sets the standard for sensitive patient data protection. Under HIPAA, companies that deal with protected health information (PHI) must comply with the physical, network, and process security measures outlined in the HIPAA. Although data collection using IoT devices commonly contain basic information such as patient ID and physiological information, the health and information privacy of patients, and security concerns remain important. Security breaches potentially expose patient information to irresponsible parties.
Regardless, prospects of NEMS and MEMS for engineering-based applications have been positive leading to rapid translation of this technology to biomedical fields. In closing, it is anticipated that this comprehensive review article will enlighten novel research directions and inspire ideas for further exploration of nanomaterials-based bioNEMS/MEMS for biomedical applications in the near future.

Author Contributions

Conceptualization, F.I., N.F.J. and A.T.; writing—original draft preparation, N.A.D., F.I., L.M., S.M., N.F.J., B.P. and S.K.; writing—review and editing, F.I., S.K. and G.M.S.; funding acquisition, F.I. and G.M.S. All authors have read and agreed to the published version of the manuscript.

Funding

We thank and acknowledge financial support from the European Union’s Horizon 2020 research and innovation programme under the Marie Skłodowska-Curie grant agreement No. 872370, Universiti Malaya-Horizon 2020 Partnership Grant: RK006-2021 and Ministry of Science and Technology (MOSTI) Technology Development Fund 1 (REF No: TDF07211415).

Data Availability Statement

Not applicable.

Acknowledgments

The authors acknowledge the financial support provided by the European Union’s Horizon 2020 research and innovation programme under the Marie Skłodowska-Curie grant agreement No. 872370. The project was also funded by Universiti Malaya-Horizon 2020 Partnership Grant: RK006-2021 and Ministry of Science and Technology (MOSTI) Technology Development Fund 1 (REF No: TDF07211415).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

2D—two-dimensional, 3D—three-dimensional, 4G—fourth generation, 5G—fifth generation, 6G—sixth generation, AChE—acetylcholinesterase, Ag—silver, AuNPs—gold nanoparticles, AuNs—gold nanostructures, BAN—body area network, bioNEMS/MEMS—biomedical nano-/micro-electro-mechanical systems, CBNMs—carbon-based nanomaterials, CeO2—cerium oxide, CNT—carbon nanotube, CNTFET—carbon nanotube field-effect transistor, C-MEMS—carbon micro-electro-mechanical systems, CMOS—complementary metal-oxide semiconductor, COOH—carboxyl groups, Cr—chromium, Cu—copper, DCFH—2,7-dichlorodihydrofluorescein, DDS—drug delivery system, DEP—dielectrophoresis, DNA—deoxyribonucleic acid, DNMT3A—DNA (cytosine-5)-methyltransferase, DOX—doxorubicin, EBL—electron beam lithography, ECG—electrocardiogram, EDM—electro-discharge machining, EHS—environmental, health and safety, EMG—electromyography, FDA—Food Drug Administration, FET—field-effect transistor, GaN/AIN—gallium nitride/aluminium nitride, GFET—graphene field-effect transistor, GO—graphene oxide, H2O2—hydrogen peroxide, HEPA—high-efficiency particulate absorbing, HIPAA—Health Insurance Portability and Accountability Act, HK-2—human kidney-2, HL-1—mouse cardiac cell line, ICP-RIE—inductively coupled plasma-reactive ion etching, ID—identity, IL-6—interleukin-6, IoB—Internet of Bodies, IOP—intraocular pressure, IoT—Internet of Things, ISM—Industrial, scientific and medical, ITO—indium-doped tin oxide, ISO—International Organisation for Standardisation, JFETs—junction field-effect transistors, Li—lithium, LOC—lab-on-a-chip, LOD—limit of detection, MDA—Medical Device Authority, MEA—microelectrode arrays, MEMS—micro-electro-mechanical systems, MWCNT—multi-welled carbon nanotube, MOSFET—metal-oxide semiconductor field-effect transistor, MTT—(3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, MXene—titanium carbide, NEMS—nano-electro-mechanical systems, MSNPs—mesoporous silica nanoparticles, NM—nanomaterial, OER—oxygen evolution reaction, ORR—oxygen reduction reaction, PDMS—polydimethylsiloxane, PEG—poly(ethylene glycol), PHI—protected health information, PLA—polylactide, PS -polysulfide, SIBS—poly(styrene-isobutylene-styrene), m-SPL—mechanical scanning probe lithography, NIL—nanoimprint lithography, RF—radio-frequency, rGO—reduced graphene oxide, RIU—refractive index unit, ROS—reactive oxygen species, SAG—self-assembly graphene, SARS-CoV-2—severe acute respiratory syndrome coronavirus 2, Si—silica, SiO2—silica dioxide, SPL—scanning probe lithography, SPR—surface plasma resonance, TGA—Therapeutic Goods Administration, TiO2—titanium dioxide, t-SPL—thermal scanning probe lithography, UV—ultraviolet, UWB—ultra-wideband, WO3—tungsten trioxide, ZnO—zinc oxide, Zr—zirconium.

References

  1. Kulinsky, L.; Madou, M.J. BioMEMs for drug delivery applications. In MEMS for Biomedical Applications; Bhansali, S., Vasudev, A., Eds.; Woodhead Publishing: Philadelphia, PA, USA, 2012; pp. 218–268. [Google Scholar]
  2. Madou, M.J. Historical note: The ascent of silicon, MEMS, and NEMS. In Solid-State Physics, Fluidics, and Analytical Techniques in Micro-and Nanotechnology; Madou, M.J., Ed.; CRC Press: Boca Raton, FL, USA, 2011; pp. 19–50. [Google Scholar]
  3. Lu, Y.; Palanikumar, L.; Choi, E.S.; Huskens, J.; Ryu, J.-H.; Wang, Y.; Pang, W.; Duan, X. Hypersound-enhanced intracellular delivery of drug-loaded mesoporous silica nanoparticles in a non-endosomal pathway. ACS Appl. Mater. Interfaces 2019, 11, 19734–19742. [Google Scholar] [CrossRef] [PubMed]
  4. Wang, L.; Wang, X.; Wu, Y.; Guo, M.; Gu, C.; Dai, C.; Kong, D.; Wang, Y.; Zhang, C.; Qu, D.; et al. Rapid and ultrasensitive electromechanical detection of ions, biomolecules and SARS-CoV-2 RNA in unamplified samples. Nat. Biomed. Eng. 2022, 6, 276–285. [Google Scholar] [CrossRef] [PubMed]
  5. Anderson, W.D.; Wilson, S.L.M.; Holdsworth, D.W. Development of a wireless telemetry sensor device to measure load and deformation in orthopaedic applications. Sensors 2020, 20, 6772. [Google Scholar] [CrossRef] [PubMed]
  6. Fujiwara, S.; Morikawa, K.; Endo, T.; Hisamoto, H.; Sueyoshi, K. Size sorting of exosomes by tuning the thicknesses of the electric double layers on a micro-nanofluidic device. Micromachines 2020, 11, 458. [Google Scholar] [CrossRef] [PubMed]
  7. Sant, S.; Tao, S.L.; Fisher, O.Z.; Xu, Q.; Peppas, N.A.; Khademhosseini, A. Microfabrication technologies for oral drug delivery. Adv. Drug. Deliv. Rev. 2012, 64, 496–507. [Google Scholar] [CrossRef] [Green Version]
  8. Sharma, D.; Kanchi, S.; Bisetty, K.; Nuthalapati, V.N. Perspective on analytical sciences and nanotechnology. In Advanced Environmental Analysis: Applications of Nanomaterials; The Royal Society of Chemistry: London, UK, 2017; Volume 1, pp. 1–34. [Google Scholar]
  9. Baig, N.; Kammakakam, I.; Falath, W. Nanomaterials: A review of synthesis methods, properties, recent progress, and challenges. Mater. Adv. 2021, 2, 1821–1871. [Google Scholar] [CrossRef]
  10. Khan, I.; Saeed, K.; Khan, I. Nanoparticles: Properties, applications and toxicities. Arab. J. Chem. 2019, 12, 908–931. [Google Scholar] [CrossRef]
  11. Barkam, S.; Saraf, S.; Seal, S. Fabricated micro-nano devices for in vivo and in vitro biomedical applications. WIREs Naomed. Nanobiotechnol. 2013, 5, 544–568. [Google Scholar] [CrossRef]
  12. Chauhan, N.; Saxena, K.; Tikadar, M.; Jain, U. Recent advances in the design of biosensors based on novel nanomaterials: An insight. Nanotechnol. Precis. Eng. 2021, 4, 045003. [Google Scholar] [CrossRef]
  13. Ansari, A.A.; Alhoshan, M.; Alsalhi, M.S.; Aldwayyan, A.S. Prospects of nanotechnology in clinical immunodiagnostics. Sensors 2010, 10, 6535–6581. [Google Scholar] [CrossRef]
  14. Song, J.-H.; Min, S.-H.; Kim, S.-G.; Cho, Y.; Ahn, S.-H. Multi-functionalization strategies using nanomaterials: A review and case study in sensing applications. Int. J. Precis. Eng. Manuf.-Green Technol. 2022, 9, 323–347. [Google Scholar] [CrossRef]
  15. Bhushan, B. Nanotribology and materials characterization of MEMS/NEMS and bioMEMS/bioNEMS materials and devices. In Springer Handbook of Nanotechnology; Bhushan, B., Ed.; Springer: Berlin/Heidelberg, Germany, 2007; pp. 1575–1638. [Google Scholar] [CrossRef]
  16. Bhushan, B. MEMS/NEMS and bioMEMS/bioNEMS: Materials, devices, and biomimetics. In Nanotribology and Nanomechanics II: Nanotribology, Biomimetics, and Industrial Applications; Bhushan, B., Ed.; Springer: Berlin/Heidelberg, Germany, 2011; pp. 833–945. [Google Scholar] [CrossRef]
  17. Jyotsna; Stanley Abraham, L.; Hanumant Singh, R.; Panda, R.C.; Senthilvelan, T. Biomedical applications of carbon-based nanomaterials. In Nanomaterials and Their Biomedical Applications; Santra, T.S., Mohan, L., Eds.; Springer: Singapore, 2021; pp. 157–174. [Google Scholar] [CrossRef]
  18. Illath, K.; Wankhar, S.; Mohan, L.; Nagai, M.; Santra, T.S. Metallic nanoparticles for biomedical applications. In Nanomaterials and Their Biomedical Applications; Santra, T.S., Mohan, L., Eds.; Springer: Singapore, 2021; pp. 29–81. [Google Scholar] [CrossRef]
  19. Wang, H.; Xiang, Z.; Hu, C.F.; Pastorin, G.; Fang, W.; Lee, C. Microneedle array integrated with CNT nanofilters for controlled and selective drug delivery. J. Microelectromec. Syst. 2014, 23, 1036–1044. [Google Scholar] [CrossRef]
  20. Moore, T.L.; Pitzer, J.E.; Podila, R.; Wang, X.; Lewis, R.L.; Grimes, S.W.; Wilson, J.R.; Skjervold, E.; Brown, J.M.; Rao, A.; et al. Multifunctional polymer-coated carbon nanotubes for safe drug delivery. Part. Part. Syst. Charact. 2013, 30, 365–373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Sharma, D.; Lee, J.; Seo, J.; Shin, H. Development of a sensitive electrochemical enzymatic reaction-based cholesterol biosensor using nano-sized carbon interdigitated electrodes decorated with gold nanoparticles. Sensors 2017, 17, 2128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Abdelbasir, S.M.; McCourt, K.M.; Lee, C.M.; Vanegas, D.C. Waste-Derived Nanoparticles: Synthesis Approaches, Environmental Applications, and Sustainability Considerations. Front. Chem. 2020, 8, 782. [Google Scholar] [CrossRef]
  23. Aswathi, V.P.; Meera, S.; Maria, C.G.A.; Nidhin, M. Green synthesis of nanoparticles from biodegradable waste extracts and their applications: A critical review. Nanotechnol. Environ. Eng. 2022. [Google Scholar] [CrossRef]
  24. Schneider, F. Review of food waste prevention on an international level. Proc. Inst. Civ. Eng. Waste Resour. Manag. 2013, 166, 187–203. [Google Scholar] [CrossRef]
  25. Sahakian, M.; Shenoy, M.; Soma, T.; Watabe, A.; Yagasa, R.; Premakumara, D.G.J.; Liu, C.; Favis, A.M.; Saloma, C. Apprehending food waste in Asia: Policies, practices and promising trends. In Routledge Handbook of Food Waste; Routledge: London, UK, 2020; pp. 187–206. [Google Scholar]
  26. Food Wastage Foodprint: Full-Cost Accounting; Food and Agriculture Organization of the United Nations: Rome, Italy, 2014.
  27. Food Waste Index Report 2021; United Nations Environment Programme: Nairobi, Kenya, 2021.
  28. Levchenko, I.; Mandhakini, M.; Prasad, K.; Bazaka, O.; Ivanova, E.P.; Jacob, M.V.; Baranov, O.; Riccardi, C.; Roman, H.E.; Xu, S.; et al. Functional nanomaterials from waste and low-value natural products: A technological approach level. Adv. Mater. Technol. 2022, 2101471. [Google Scholar] [CrossRef]
  29. Cai, S.; Li, W.; Xu, P.; Xia, X.; Yu, H.; Zhang, S.; Li, X. In situ construction of metal–organic framework (MOF) UiO-66 film on Parylene-patterned resonant microcantilever for trace organophosphorus molecules detection. Analyst 2019, 144, 3729–3735. [Google Scholar] [CrossRef]
  30. Zhang, S.; Pei, X.; Gao, H.; Chen, S.; Wang, J. Metal-organic framework-based nanomaterials for biomedical applications. Chin. Chem. Lett. 2020, 31, 1060–1070. [Google Scholar] [CrossRef]
  31. Elkodous, M.A.; El-Husseiny, H.M.; El-Sayyad, G.S.; Hashem, A.H.; Doghish, A.S.; Elfadil, D.; Radwan, Y.; El-Zeiny, H.M.; Bedair, H.; Ikhdair, O.A.; et al. Recent advances in waste-recycled nanomaterials for biomedical applications: Waste-to-wealth. Nanotechnol. Rev. 2021, 10, 1662–1739. [Google Scholar] [CrossRef]
  32. Wang, Z.; Shen, D.; Wu, C.; Gu, S. State-of-the-art on the production and application of carbon nanomaterials from biomass. Green Chem. 2018, 20, 5031–5057. [Google Scholar] [CrossRef] [Green Version]
  33. Chen, W.; Meng, X.-T.; Wang, H.-H.; Zhang, X.-Q.; Wei, Y.; Li, Z.-Y.; Li, D.; Zhang, A.-P.; Liu, C.-F. A feasible way to produce carbon nanofiber by electrospinning from sugarcane bagasse. Polymers 2019, 11, 1968. [Google Scholar] [CrossRef] [Green Version]
  34. Gaud, B.; Singh, A.; Jaybhaye, S.; Narayan, H. Synthesis of carbon nano fiber from organic waste and activation of its surface area. Int. J. Phys. Res. Appl. 2019, 2766, 2748. [Google Scholar] [CrossRef] [Green Version]
  35. Yusefi, M.; Shameli, K.; Ali, R.R.; Pang, S.-W.; Teow, S.-Y. Evaluating anticancer activity of plant-mediated synthesized iron oxide nanoparticles using Punica granatum fruit peel extract. J. Mol. Struct. 2020, 1204, 127539. [Google Scholar] [CrossRef]
  36. Joshi, P.; Joshi, H.C.; Sanghi, S.K.; Kundu, S. Immobilization of monoamine oxidase on eggshell membrane and its application in designing an amperometric biosensor for dopamine. Microchim. Acta 2010, 169, 383–388. [Google Scholar] [CrossRef]
  37. Tamilselvi, R.; Ramesh, M.; Lekshmi, G.S.; Bazaka, O.; Levchenko, I.; Bazaka, K.; Mandhakini, M. Graphene oxide—Based supercapacitors from agricultural wastes: A step to mass production of highly efficient electrodes for electrical transportation systems. Renew. Energy 2020, 151, 731–739. [Google Scholar] [CrossRef]
  38. Purkait, T.; Singh, G.; Singh, M.; Kumar, D.; Dey, R.S. Large area few-layer graphene with scalable preparation from waste biomass for high-performance supercapacitor. Sci. Rep. 2017, 7, 15239. [Google Scholar] [CrossRef] [Green Version]
  39. Manfredi, S.; Cristobal, J. Towards more sustainable management of European food waste: Methodological approach and numerical application. Waste Manag. Res. 2016, 34, 957–968. [Google Scholar] [CrossRef]
  40. Auría-Soro, C.; Nesma, T.; Juanes-Velasco, P.; Landeira-Viñuela, A.; Fidalgo-Gomez, H.; Acebes-Fernandez, V.; Gongora, R.; Almendral Parra, M.J.; Manzano-Roman, R.; Fuentes, M. Interactions of nanoparticles and biosystems: Microenvironment of nanoparticles and biomolecules in nanomedicine. Nanomaterials 2019, 9, 1365. [Google Scholar] [CrossRef] [Green Version]
  41. Villanueva-Flores, F.; Castro-Lugo, A.; Ramírez, O.T.; Palomares, L.A. Understanding cellular interactions with nanomaterials: Towards a rational design of medical nanodevices. Nanotechnology 2020, 31, 132002. [Google Scholar] [CrossRef] [PubMed]
  42. Kaneko, A.; Mori, A.; Kanada, K.; Sumin, C.; Ke, W. Micro/nano-printing of metal and nanoparticle thin film and its application to MEMS device. In Proceedings of JSME 2020 Conference on Leading Edge Manufacturing/Materials and Processing; American Society of Mechanical Engineers: New York, NY, USA, 2021. [Google Scholar]
  43. Bani-Yaseen, A.D.; Kawaguchi, T.; Jankowiak, R. Electrochemically deposited metal nanoparticles for enhancing the performance of microfluidic MEMS in biochemical analysis. Int. J. Nanomanuf. 2009, 4, 99–107. [Google Scholar] [CrossRef]
  44. Anik, M.I.; Mahmud, N.; Al Masud, A.; Hasan, M. Gold nanoparticles (GNPs) in biomedical and clinical applications: A review. Nano Sel. 2022, 3, 792–828. [Google Scholar] [CrossRef]
  45. Shah, M.; Badwaik, V.D.; Dakshinamurthy, R. Biological applications of gold nanoparticles. J. Nanosci. Nanotechnol. 2014, 14, 344–362. [Google Scholar] [CrossRef] [PubMed]
  46. Hu, T.; Fang, K.; Zhang, Z.; Jiang, X.; Zhao, Y. The hybrid fabrication process of metal/silicon composite structure for MEMS S&A Device. Micromachines 2019, 10, 469. [Google Scholar] [CrossRef] [Green Version]
  47. Schlicke, H.; Battista, D.; Kunze, S.; Schröter, C.J.; Eich, M.; Vossmeyer, T. Freestanding membranes of cross-linked gold nanoparticles: Novel functional materials for electrostatic actuators. ACS Appl. Mater. Interfaces 2015, 7, 15123–15128. [Google Scholar] [CrossRef]
  48. Bhushan, B. Micro/nanotribology of MEMS/NEMS materials and devices. In Springer Handbook of Nanotechnology; Bhushan, B., Ed.; Springer: Berlin/Heidelberg, Germany, 2004; pp. 983–1021. [Google Scholar] [CrossRef]
  49. Liao, M.; Koide, Y. Carbon-based materials: Growth, properties, MEMS/NEMS technologies, and MEM/NEM switches. Crit. Rev. Solid State Mater. Sci. 2011, 36, 66–101. [Google Scholar] [CrossRef]
  50. Neuville, S. Selective carbon material engineering for improved MEMS and NEMS. Micromachines 2019, 10, 539. [Google Scholar] [CrossRef] [Green Version]
  51. Cho, J.H.; Cayll, D.; Behera, D.; Cullinan, M. Towards repeatable, scalable graphene integrated micro-nano electromechanical systems (MEMS/NEMS). Micromachines 2022, 13, 27. [Google Scholar] [CrossRef]
  52. Zhang, Y.; Zhang, L.; Zhou, C. Review of chemical vapor deposition of graphene and related applications. Acc. Chem. Res. 2013, 46, 2329–2339. [Google Scholar] [CrossRef]
  53. Chen, C.; Hone, J. Graphene nanoelectromechanical systems. Proc. IEEE 2013, 101, 1766–1779. [Google Scholar] [CrossRef]
  54. Rahman, S.H.A.; Soin, N.; Ibrahim, F. Design of graphene-based MEMS intracranial pressure sensor. In Proceedings of the 2016 IEEE International Symposium on Medical Measurements and Applications (MeMeA), Benevento, Italy, 15–18 May 2016; pp. 1–5. [Google Scholar]
  55. Khan, N.I.; Song, E. Detection of an IL-6 biomarker using a GFET platform developed with a facile organic solvent-free aptamer immobilization approach. Sensors 2021, 21, 1335. [Google Scholar] [CrossRef] [PubMed]
  56. Irani, F.S.; Shafaghi, A.H.; Tasdelen, M.C.; Delipinar, T.; Kaya, C.E.; Yapici, G.G.; Yapici, M.K. Graphene as a piezoresistive material in strain sensing applications. Micromachines 2022, 13, 119. [Google Scholar] [CrossRef] [PubMed]
  57. Chen, Z.; Wang, Z.; Li, X.; Lin, Y.; Luo, N.; Long, M.; Zhao, N.; Xu, J.-B. Flexible piezoelectric-induced pressure sensors for static measurements based on nanowires/graphene heterostructures. ACS Nano 2017, 11, 4507–4513. [Google Scholar] [CrossRef]
  58. Kis, A.; Zettl, A. Nanomechanics of carbon nanotubes. Philos. Trans. R. Sci. A 2008, 366, 1591–1611. [Google Scholar] [CrossRef]
  59. Ajayan, P.M.; Zhou, O.Z. Applications of carbon nanotubes. In Carbon Nanotubes: Synthesis, Structure, Properties, and Applications; Dresselhaus, M.S., Dresselhaus, G., Avouris, P., Eds.; Springer: Berlin/Heidelberg, Germany, 2001; pp. 391–425. [Google Scholar] [CrossRef]
  60. Xi, S.; Shi, T.; Liu, D.; Xu, L.; Long, H.; Lai, W.; Tang, Z. Integration of carbon nanotubes to three-dimensional C-MEMS for glucose sensors. Sens. Actuator A Phys. 2013, 198, 15–20. [Google Scholar] [CrossRef]
  61. Xu, H.; Wang, L.; Luo, J.; Song, Y.; Liu, J.; Zhang, S.; Cai, X. Selective recognition of 5-hydroxytryptamine and dopamine on a multi-walled carbon nanotube-chitosan hybrid film-modified microelectrode array. Sensors 2015, 15, 1008–1021. [Google Scholar] [CrossRef] [Green Version]
  62. Sepúlveda, A.T.; Fachin, F.; Villoria, R.G.d.; Wardle, B.L.; Viana, J.C.; Pontes, A.J.; Rocha, L.A. Nanocomposite flexible pressure sensor for biomedical applications. Procedia Eng. 2011, 25, 140–143. [Google Scholar] [CrossRef] [Green Version]
  63. Hanniet, Q.; Petit, E.; Calas-Etienne, S.; Etienne, P.; Aissou, K.; Gervais, C.; Charlot, B.; Salameh, C. Rational design of Si (B) Cn microstructures using direct photolithography of patternable preceramic photoresists. SSRN Electron. J. 2022, 223, 111234. [Google Scholar] [CrossRef]
  64. Londe, G.; Han, A.; Cho, H.J. MEMS for nanotechnology: Top-down perspective. In Functional Nanostructures: Processing, Characterization, and Applications; Seal, S., Ed.; Springer: New York, NY, USA, 2008; pp. 107–167. [Google Scholar] [CrossRef]
  65. Mitu, G.; Bejinaru Mihoc, A. Applications of photolithography for the manufacture of solid MEMS bodies. Transilv. Univ. Press Bras. 2019, 1, 65–68. [Google Scholar]
  66. Verma, G.; Mondal, K.; Gupta, A. Si-based MEMS resonant sensor: A review from microfabrication perspective. Microelectron. J. 2021, 118, 105210. [Google Scholar] [CrossRef]
  67. Sui, W.; Zheng, X.Q.; Lin, J.T.; Alphenaar, B.W.; Feng, P.X.L. Thermal response and TC f of GaN/AlN heterostructure multimode micro string resonators from −10 °C up to 325 °C. J. Microelectromec. Syst. 2021, 30, 521–529. [Google Scholar] [CrossRef]
  68. Jithin, M.A.; Ganapathi, K.L.; Ambresh, M.; Nukala, P.; Udayashankar, N.K.; Mohan, S. Development of titanium nitride thin film microheaters using laser micromachining. Vacuum 2022, 197, 110795. [Google Scholar] [CrossRef]
  69. Tricinci, O.; Carlotti, M.; Desii, A.; Meder, F.; Mattoli, V. Two-step MEMS microfabrication via 3D direct laser lithography. In Proceedings of the Advanced Fabrication Technologies for Micro/Nano Optics and Photonics XIV, San Francisco, CA, USA, 6–12 March 2021; pp. 34–40. [Google Scholar]
  70. Udara, S.; Hadimani, H.C.; Revankar, P.K. Sensitivity and Selectivity Enhancement of MEMS/NEMS Cantilever by Coating of Polyvinylpyrrolidone. Mater. Today: Proc. 2019, 18, 1610–1619. [Google Scholar] [CrossRef]
  71. Chai, H.; Wang, J.; Song, Y. MEMS, NEMS, AEMS, and quantum films for the next generation of computing and information technology. In Inorganic and Organic Thin Films: Fundamentals, Fabrication and Applications; Song, Y., Ed.; Wiley-VCH: Weinheim, Germany, 2021; Volume 1, pp. 199–219. [Google Scholar]
  72. Randall, J.; Owen, J.H.; Fuchs, E.; Saini, R.; Santini, R.; Moheimani, S.O.R. Atomically Precise Digital E-Beam Lithography; SPIE: San Jose, CA, USA, 2020; Volume 11324. [Google Scholar]
  73. Kumar, D.; Singh, N.K.; Bajpai, V. Achieving nano-patterned features by micro-EDM process using vertically aligned ZnO nanorods grown on microprobe tip: A scaling approach. Microelectron. Eng. 2022, 260, 111792. [Google Scholar] [CrossRef]
  74. Kono, T.; Hatano, M.; Tokue, H.; Kato, H.; Fukuhara, K.; Nakasugi, T. Half-pitch 14nm direct patterning with nanoimprint lithography. In Proceedings of the Novel Patterning Technologies for Semiconductors, MEMS/NEMS, and MOEMS 2019, San Francisco, CA, USA, 1 March 2019; p. 109580H. [Google Scholar]
  75. Supreeti, S.; Kirchner, J.; Hofmann, M.; Mastylo, R.; Rangelow, I.; Manske, E.; Hoffmann, M.; Sinzinger, S. Integrated Soft UV-nanoimprint Lithography in a Nanopositioning and Nanomeasuring Machine for Accurate Positioning of Stamp to Substrate; SPIE: San Jose, CA, USA, 2019; Volume 10958. [Google Scholar]
  76. Jiang, Y.; Dodds, K.; Dunare, C.; Lomax, P.; Cheung, R. Thermal scanning probe lithography using Parylene C as thermal resist. Micro Nano Lett. 2022, 17, 96–99. [Google Scholar] [CrossRef]
  77. Howell, S.T.; Grushina, A.; Holzner, F.; Brugger, J. Thermal scanning probe lithography—A review. Microsyst. Nanoeng. 2020, 6, 21. [Google Scholar] [CrossRef] [Green Version]
  78. Chen, P.; Wu, L.; Gao, J.; Feng, C.; Yu, B.; Qian, L. An improved scanning probe-based lithography: Site-controlled formation of self-assembled film as an etch mask. Microelectron. Eng. 2021, 244–246, 111560. [Google Scholar] [CrossRef]
  79. Yang, Z. Advanced MEMS/NEMS Fabrication and Sensors; Springer: Cham, Switzerland, 2021. [Google Scholar]
  80. Zhu, J.; Liu, X.; Shi, Q.; He, T.; Sun, Z.; Guo, X.; Liu, W.; Sulaiman, O.B.; Dong, B.; Lee, C. Development trends and perspectives of future sensors and MEMS/NEMS. Micromachines 2020, 11, 7. [Google Scholar] [CrossRef] [Green Version]
  81. Hassanin, H.; Essa, K.; Elshaer, A.; Imbaby, M.; El-Mongy, H.H.; El-Sayed, T.A. Micro-fabrication of ceramics: Additive manufacturing and conventional technologies. J. Adv. Ceram. 2021, 10, 1–27. [Google Scholar] [CrossRef]
  82. Mizui, K.; Hanabata, M.; Takei, S. Gas Permeable Mold for Defect Reduction in Nanoimprint Lithography; SPIE: San Jose, CA, USA, 2019; Volume 10958. [Google Scholar]
  83. Cherala, A.; Im, S.-H.; Meissl, M.; Simpson, L.; Thompson, E.; Choi, J.; Hiura, M.; Iino, S. Addressing Nanoimprint Lithography Mix and Match Overlay Using Drop Pattern Compensation. In Proceedings of the Novel Patterning Technologies for Semiconductors, MEMS/NEMS and MOEMS 2020, San Jose, CA, USA, 23–27 February 2020; pp. 39–48. [Google Scholar]
  84. Hamdana, G.; Puranto, P.; Langfahl-Klabes, J.; Li, Z.; Pohlenz, F.; Xu, M.; Granz, T.; Bertke, M.; Wasisto, H.S.; Brand, U.; et al. Nanoindentation of crystalline silicon pillars fabricated by soft UV nanoimprint lithography and cryogenic deep reactive ion etching. Sens. Actuator A Phys. 2018, 283, 65–78. [Google Scholar] [CrossRef]
  85. Shao, J.; Chen, X.; Li, X.; Tian, H.; Wang, C.; Lu, B. Nanoimprint lithography for the manufacturing of flexible electronics. Sci. China Technol. Sci. 2019, 62, 175–198. [Google Scholar] [CrossRef]
  86. Huff, M. Recent advances in reactive ion etching and applications of high-aspect-ratio microfabrication. Micromachines 2021, 12, 991. [Google Scholar] [CrossRef] [PubMed]
  87. Tahmasebipour, M.; Vafaie, A. A novel single axis capacitive MEMS accelerometer with double-sided suspension beams fabricated using μWEDM. Sens. Actuator A Phys. 2020, 309, 112003. [Google Scholar] [CrossRef]
  88. Ashok, A.; Gangele, A.; Pal, P.; Pandey, A.K. An analysis of stepped trapezoidal-shaped microcantilever beams for MEMS-based devices. J. Micromec. Microeng. 2018, 28, 075009. [Google Scholar] [CrossRef]
  89. Xu, J.; Bertke, M.; Li, X.; Mu, H.; Zhou, H.; Yu, F.; Hamdana, G.; Schmidt, A.; Bremers, H.; Peiner, E. Fabrication of ZnO nanorods and Chitosan@ZnO nanorods on MEMS piezoresistive self-actuating silicon microcantilever for humidity sensing. Sens. Actuator B Chem. 2018, 273, 276–287. [Google Scholar] [CrossRef]
  90. Fraga, M.; Pessoa, R. Progresses in synthesis and application of SiC films: From CVD to ALD and from MEMS to NEMS. Micromachines 2020, 11, 799. [Google Scholar] [CrossRef] [PubMed]
  91. Swarnalatha, V.; Purohit, S.; Pal, P.; Sharma, R.K. Enhanced etching characteristics of Si{100} in NaOH-based two-component solution. Micro Nano Syst. Lett. 2022, 10, 10. [Google Scholar] [CrossRef]
  92. Pal, P.; Swarnalatha, V.; Rao, A.V.N.; Pandey, A.K.; Tanaka, H.; Sato, K. High speed silicon wet anisotropic etching for applications in bulk micromachining: A review. Micro Nano Syst. Lett. 2021, 9, 4. [Google Scholar] [CrossRef]
  93. Kim, S.; Kim, J.; Joung, Y.-H.; Choi, J.; Koo, C. Bonding strength of a glass microfluidic device fabricated by femtosecond laser micromachining and direct welding. Micromachines 2018, 9, 639. [Google Scholar] [CrossRef] [Green Version]
  94. Kanwal, A.; Ilic, B.R.; Ray, C.H.; Siebein, K.; Liddle, J.A. Chromium oxide—A novel sacrificial layer material for MEMS/NEMS and micro/nanofluidic device fabrication. Micro Nano Eng. 2022, 16, 100145. [Google Scholar] [CrossRef]
  95. Gong, X.; Zhao, R.; Yu, X. A 3-D-silicon nanowire FET biosensor based on a novel hybrid process. J. Microelectromec. Syst. 2018, 27, 164–170. [Google Scholar] [CrossRef]
  96. Racka-Szmidt, K.; Stonio, B.; Żelazko, J.; Filipiak, M.; Sochacki, M. A review: Inductively coupled plasma reactive ion etching of silicon carbide. Materials 2022, 15, 123. [Google Scholar] [CrossRef] [PubMed]
  97. Chen, G.; Rinaldi, M. Aluminum nitride combined overtone resonators for the 5G high frequency bands. J. Microelectromec. Syst. 2020, 29, 148–159. [Google Scholar] [CrossRef]
  98. Ma, Z.; Wang, Y.; Shen, Q.; Zhang, H.; Guo, X. Key processes of silicon-on-glass MEMS fabrication technology for gyroscope application. Sensors 2018, 18, 1240. [Google Scholar] [CrossRef] [Green Version]
  99. Pradhan, B.B.; Masanta, M.; Sarkar, B.R.; Bhattacharyya, B. Investigation of electro-discharge micro-machining of titanium super alloy. Int. J. Adv. Manuf. Technol. 2008, 41, 1094. [Google Scholar] [CrossRef]
  100. Shiu, P.-P.; Knopf, G.K.; Ostojic, M. Fabrication of metallic micromolds by laser and electro-discharge micromachining. Microsyst. Technol. 2009, 16, 477. [Google Scholar] [CrossRef]
  101. Liu, K.; Wu, H.; Huang, R.; Tan, N.Y.J. Ultra-precision machining of micro-step pillar array using a straight-edge milling tool. Nanomanuf. Metrol. 2020, 3, 260–268. [Google Scholar] [CrossRef]
  102. Vafaie, A.; Tahmasebipour, M.; Tahmasebipour, Y. A novel capacitive micro-accelerometer made of steel using micro wire electrical discharge machining method. J. Micromec. Microeng. 2019, 29, 125018. [Google Scholar] [CrossRef]
  103. Karazi, S.; Moradi, M.; Benyounis, K. Statistical and numerical approaches for modelling and optimising laser micromachining process-Review. Refer. Modul. Mater. Sci. Eng. 2019. [Google Scholar] [CrossRef]
  104. Li, X.-X.; He, J.-H. Nanoscale adhesion and attachment oscillation under the geometric potential. Part 1: The formation mechanism of nanofiber membrane in the electrospinning. Results Phys. 2019, 12, 1405–1410. [Google Scholar] [CrossRef]
  105. George, D.; Garcia, A.; Pham, Q.; Perez, M.R.; Deng, J.; Nguyen, M.T.; Zhou, T.; Martinez-Chapa, S.O.; Won, Y.; Liu, C.; et al. Fabrication of patterned graphitized carbon wires using low voltage near-field electrospinning, pyrolysis, electrodeposition, and chemical vapor deposition. Microsyst. Nanoeng. 2020, 6, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Lin, Y.; Gao, C.; Gritsenko, D.; Zhou, R.; Xu, J. Soft lithography based on photolithography and two-photon polymerization. Microfluid. Nanofluidics 2018, 22, 97. [Google Scholar] [CrossRef]
  107. Alfaifi, A.; Alhomoudi, I.A.; Nabki, F.; El-Gamal, M.N. In-plane high-sensitivity capacitive accelerometer in a 3-D CMOS-compatible surface micromachining process. J. Microelectromec. Syst. 2019, 28, 14–24. [Google Scholar] [CrossRef]
  108. Afandi, Y.; Parish, G.; Keating, A. Surface micromachining multilayer porous silicon for spectral filtering applications. Mater. Sci. Semicond. Process. 2022, 138, 106314. [Google Scholar] [CrossRef]
  109. Puoza, J.C.; Hua, X.; Liu, Q.; Kang, Z.; Zhang, P. Manufacturing of micro-textures on metals by nanosecond laser micromachining. Adv. Mater. Process. Technol. 2018, 4, 86–99. [Google Scholar] [CrossRef]
  110. Havlíček, K.; Svobodová, L.; Bakalova, T.; Lederer, T. Influence of electrospinning methods on characteristics of polyvinyl butyral and polyurethane nanofibres essential for biological applications. Mater. Des. 2020, 194, 108898. [Google Scholar] [CrossRef]
  111. Wan Yahya, W.N.; Ibrahim, F.; Thiha, A.; Jamaluddin, N.F.; Madou, M. Fabrication of a 3D carbon electrode for potential dielectrophoresis-based hepatic cell patterning application using carbon micro-electrical-mechanical system (CMEMS). J. Micromec. Microeng. 2022, 32, 055005. [Google Scholar] [CrossRef]
  112. Marvi, F.; Jafari, K. An ultra-sensitive 1D photonic crystal bioNEMS platform for label free detection of biomolecules. IEEE Sens. J. 2021, 21, 22927–22933. [Google Scholar] [CrossRef]
  113. Hemanth, S.; Halder, A.; Caviglia, C.; Chi, Q.; Keller, S.S. 3D carbon microelectrodes with bio-functionalized graphene for electrochemical biosensing. Biosensors 2018, 8, 70. [Google Scholar] [CrossRef] [Green Version]
  114. Forouzanfar, S.; Pala, N.; Wang, C. In-situ integration of 3D C-MEMS microelectrodes with bipolar exfoliated graphene for label-free electrochemical cancer biomarkers aptasensor. Micromachines 2022, 13, 104. [Google Scholar] [CrossRef] [PubMed]
  115. Forouzanfar, S.; Alam, F.; Pala, N.; Wang, C. Highly sensitive label-free electrochemical aptasensors based on photoresist derived carbon for cancer biomarker detection. Biosens. Bioelectron. 2020, 170, 112598. [Google Scholar] [CrossRef] [PubMed]
  116. Sharma, R.; Grover, A. Myocilin-associated glaucoma: A historical perspective and recent research progress. Mol. Vis. 2021, 27, 480–493. [Google Scholar] [PubMed]
  117. Liu, Z.; Wang, G.; Ye, C.; Sun, H.; Pei, W.; Wei, C.; Dai, W.; Dou, Z.; Sun, Q.; Lin, C.-T.; et al. An ultrasensitive contact lens sensor based on self-assembly graphene for continuous intraocular pressure monitoring. Adv. Funct. Mater. 2021, 31, 2010991. [Google Scholar] [CrossRef]
  118. Zhang, H.; Li, Y.; Zhang, P.; Ren, C.; Xu, X.; Sun, B.; Yang, Z. A flexible implantable polyimide catheter device for targeted treatment of cardiovascular diseases by aggregating magnetic nanoparticles. IEEE Trans. Compon. Packag. Manuf. Technol. 2021, 11, 911–917. [Google Scholar] [CrossRef]
  119. Xuan, X.; Yoon, H.S.; Park, J.Y. A wearable electrochemical glucose sensor based on simple and low-cost fabrication supported micro-patterned reduced graphene oxide nanocomposite electrode on flexible substrate. Biosens. Bioelectron. 2018, 109, 75–82. [Google Scholar] [CrossRef]
  120. Kim, S.; Jeon, H.-J.; Park, S.; Lee, D.Y.; Chung, E. Tear glucose measurement by reflectance spectrum of a nanoparticle embedded contact lens. Sci. Rep. 2020, 10, 8254. [Google Scholar] [CrossRef]
  121. Sharma, D.; Lee, J.; Shin, H. An electrochemical immunosensor based on a 3D carbon system consisting of a suspended mesh and substrate-bound interdigitated array nanoelectrodes for sensitive cardiac biomarker detection. Biosens. Bioelectron. 2018, 107, 10–16. [Google Scholar] [CrossRef]
  122. Sharma, D.; Lee, J.; Shin, H. Gold nanostructure decorated 3D porous carbon architectures as a non-enzymatic glucose sensor. In Proceedings of the 2019 IEEE 32nd International Conference on Micro Electro Mechanical Systems (MEMS), Seoul, Korea, 27–31 January 2019; pp. 580–583. [Google Scholar]
  123. Mahboob Alam, T.; Iqbal, M.A.; Ali, Y.; Wahab, A.; Ijaz, S.; Imtiaz Baig, T.; Hussain, A.; Malik, M.A.; Raza, M.M.; Ibrar, S.; et al. A model for early prediction of diabetes. Inform. Med. Unlocked 2019, 16, 100204. [Google Scholar] [CrossRef]
  124. Khor, S.M.; Choi, J.; Won, P.; Ko, S.H. Challenges and strategies in developing an enzymatic wearable sweat glucose biosensor as a practical point-of-care monitoring tool for type II Diabetes. Nanomaterials 2022, 12, 221. [Google Scholar] [CrossRef]
  125. Martín, A.; Kim, J.; Kurniawan, J.F.; Sempionatto, J.R.; Moreto, J.R.; Tang, G.; Campbell, A.S.; Shin, A.; Lee, M.Y.; Liu, X.; et al. Epidermal microfluidic electrochemical detection system: Enhanced sweat sampling and metabolite detection. ACS Sens. 2017, 2, 1860–1868. [Google Scholar] [CrossRef] [PubMed]
  126. Iyengar, K.P.; Kariya, A.D.; Botchu, R.; Jain, V.K.; Vaishya, R. Significant capabilities of SMART sensor technology and their applications for Industry 4.0 in trauma and orthopaedics. Sens. Int. 2022, 3, 100163. [Google Scholar] [CrossRef]
  127. Farra, R.; Sheppard, N.F.; McCabe, L.; Neer, R.M.; Anderson, J.M.; Santini, J.T.; Cima, M.J.; Langer, R. First-in-human testing of a wirelessly controlled drug delivery microchip. Sci. Transl. Med. 2012, 4, 122ra121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Delcassian, D.; Patel, A.K.; Cortinas, A.B.; Langer, R. Drug delivery across length scales. J. Drug Target. 2019, 27, 229–243. [Google Scholar] [CrossRef] [PubMed]
  129. Li, X.; Wang, J.; Li, S.; Liu, Z.; Zheng, Z.; Zhang, Y. Development and evaluation of multifunctional poly(lactic-co-glycolic acid) nanoparticles embedded in carboxymethyl β-glucan porous microcapsules as a novel drug delivery system for gefitinib. Pharmaceutics 2019, 11, 469. [Google Scholar] [CrossRef] [Green Version]
  130. Chae, M.-S.; Yoo, Y.K.; Kim, J.; Kim, T.G.; Hwang, K.S. Graphene-based enzyme-modified field-effect transistor biosensor for monitoring drug effects in Alzheimer’s disease treatment. Sens. Actuator B Chem. 2018, 272, 448–458. [Google Scholar] [CrossRef]
  131. Bruno, G.; Di Trani, N.; Hood, R.L.; Zabre, E.; Filgueira, C.S.; Canavese, G.; Jain, P.; Smith, Z.; Demarchi, D.; Hosali, S.; et al. Unexpected behaviors in molecular transport through size-controlled nanochannels down to the ultra-nanoscale. Nat. Commun. 2018, 9, 1682. [Google Scholar] [CrossRef] [Green Version]
  132. Di Trani, N.; Jain, P.; Chua, C.Y.X.; Ho, J.S.; Bruno, G.; Susnjar, A.; Pons-Faudoa, F.P.; Sizovs, A.; Hood, R.L.; Smith, Z.W.; et al. Nanofluidic microsystem for sustained intraocular delivery of therapeutics. Nanomed. Nanotechnol. Biol. Med. 2019, 16, 1–9. [Google Scholar] [CrossRef]
  133. NanoMedical Systems. Nanotechnology Transforming Drug Delivery. Available online: https://nanomedsys.com (accessed on 18 August 2022).
  134. Panwala, F.C.; Kumar, R. Asymmetric sifter-shaped microchannel network in biological MEMS for size- and mass-based mammalian cell sorting and separation using hydrodynamic technique. J. Supercomput. 2020, 76, 3814–3846. [Google Scholar] [CrossRef]
  135. Kumar, M.; Yadav, S.; Kumar, A.; Sharma, N.N.; Akhtar, J.; Singh, K. MEMS impedance flow cytometry designs for effective manipulation of micro entities in health care applications. Biosens. Bioelectron. 2019, 142, 111526. [Google Scholar] [CrossRef]
  136. Catarino, S.O.; Rodrigues, R.O.; Pinho, D.; Miranda, J.M.; Minas, G.; Lima, R. Blood cells separation and sorting techniques of passive microfluidic devices: From fabrication to applications. Micromachines 2019, 10, 593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Yin, J.; Deng, J.; Du, C.; Zhang, W.; Jiang, X. Microfluidics-based approaches for separation and analysis of circulating tumor cells. TrAC-Trends Anal. Chem. 2019, 117, 84–100. [Google Scholar] [CrossRef]
  138. Nieuwenhuis, J.H.; Jachimowicz, A.; Svasek, P.; Vellekoop, M.J. Optimization of microfluidic particle sorters based on dielectrophoresis. IEEE Sens. J. 2005, 5, 810–816. [Google Scholar] [CrossRef]
  139. Khan, T.; Civas, M.; Cetinkaya, O.; Abbasi, N.A.; Akan, O.B. Nanosensor networks for smart health care. In Nanosensors for Smart Cities; Han, B., Tomer, V.K., Nguyen, T.A., Farmani, A., Kumar Singh, P., Eds.; Elsevier: Cambridge, MA, USA, 2020; pp. 387–403. [Google Scholar] [CrossRef]
  140. Bhattacharjee, I.; Maiti, D.K. Nanotools and nanodevices in geothermal energy. In Nano Tools and Devices for Enhanced Renewable Energy; Devasahayam, S., Hussain, C.M., Eds.; Elsevier: Cambridge, MA, USA, 2021; pp. 507–518. [Google Scholar] [CrossRef]
  141. John, S.A.; Chattree, A.; Ramteke, P.W.; Shanthy, P.; Nguyen, T.A.; Rajendran, S. Nanosensors for plant health monitoring. In Nanosensors for Smart Agriculture; Denizli, A., Nguyen, T.A., Rajendran, S., Yasin, G., Nadda, A.K., Eds.; Elsevier: Cambridge, MA, USA, 2022; pp. 449–461. [Google Scholar] [CrossRef]
  142. Singh, P.; Yadava, R.D.S. Nanosensors for health care. In Nanosensors for Smart Cities; Han, B., Tomer, V.K., Nguyen, T.A., Farmani, A., Kumar Singh, P., Eds.; Elsevier: Cambridge, MA, USA, 2020; pp. 433–450. [Google Scholar] [CrossRef]
  143. Tang, X.; Zhu, Y.; Guan, W.; Zhou, W.; Wei, P. Advances in nanosensors for cardiovascular disease detection. Life Sci. 2022, 305, 120733. [Google Scholar] [CrossRef]
  144. Yin, R.; Xu, Z.; Mei, M.; Chen, Z.; Wang, K.; Liu, Y.; Tang, T.; Priydarshi, M.K.; Meng, X.; Zhao, S.; et al. Soft transparent graphene contact lens electrodes for conformal full-cornea recording of electroretinogram. Nat. Commun. 2018, 9, 2334. [Google Scholar] [CrossRef] [Green Version]
  145. Javaid, M.; Haleem, A.; Singh, R.P.; Rab, S.; Suman, R. Exploring the potential of nanosensors: A brief overview. Sens. Int. 2021, 2, 100130. [Google Scholar] [CrossRef]
  146. Kaisti, M.; Panula, T.; Leppänen, J.; Punkkinen, R.; Jafari Tadi, M.; Vasankari, T.; Jaakkola, S.; Kiviniemi, T.; Airaksinen, J.; Kostiainen, P.; et al. Clinical assessment of a non-invasive wearable MEMS pressure sensor array for monitoring of arterial pulse waveform, heart rate and detection of atrial fibrillation. NPJ Digit. Med. 2019, 2, 39. [Google Scholar] [CrossRef] [Green Version]
  147. Lee, S.-H.; Lee, D.; Choi, M.H.; Son, J.-H.; Seo, M. Highly sensitive and selective detection of steroid hormones using terahertz molecule-specific sensors. Anal. Chem. 2019, 91, 6844–6849. [Google Scholar] [CrossRef]
  148. Lee, M.A.; Wang, S.; Jin, X.; Bakh, N.A.; Nguyen, F.T.; Dong, J.; Silmore, K.S.; Gong, X.; Pham, C.; Jones, K.K.; et al. Implantable nanosensors for human steroid hormone sensing in vivo using a self-templating corona phase molecular recognition. Adv. Healthc. Mater. 2020, 9, 2000429. [Google Scholar] [CrossRef]
  149. Ahmadi, H.; Moradi, H.; Pastras, C.J.; Abolpour Moshizi, S.; Wu, S.; Asadnia, M. Development of ultrasensitive biomimetic auditory hair cells based on piezoresistive hydrogel nanocomposites. ACS Appl. Mater. Interfaces 2021, 13, 44904–44915. [Google Scholar] [CrossRef]
  150. Cho, S.; Chang, T.; Yu, T.; Lee, C.H. Smart electronic textiles for wearable sensing and display. Biosensors 2022, 12, 222. [Google Scholar] [CrossRef] [PubMed]
  151. Borandeh, S.; Alimardani, V.; Abolmaali, S.S.; Seppälä, J. Graphene family nanomaterials in ocular applications: Physicochemical properties and toxicity. Chem. Res. Toxicol. 2021, 34, 1386–1402. [Google Scholar] [CrossRef] [PubMed]
  152. Duarte, F.; Torres, J.P.N.; Baptista, A.; Marques Lameirinhas, R.A. Optical nanoantennas for photovoltaic applications. Nanomaterials 2021, 11, 422. [Google Scholar] [CrossRef] [PubMed]
  153. Chowdhury, M.S.U.; Rahman, M.A.; Hossain, M.A.; Mobashsher, A.T. Design of an ITO based CNT coated transparent nano patch antenna assisted by characteristic mode analysis. In Proceedings of the 2019 5th International Conference on Advances in Electrical Engineering (ICAEE), Dhaka, Bangladesh, 26–28 September 2019; pp. 134–138. [Google Scholar]
  154. Razzari, L. Resonant nanoantennas for enhancing the interaction of terahertz radiation with nanomaterials. In Proceedings of the 2018 International Applied Computational Electromagnetics Society Symposium (ACES), Denver, CO, USA, 25–29 March 2018; pp. 1–2. [Google Scholar]
  155. Sangeeta, T.R.; Deny, J. A study on optical interconnects to improve on-chip wireless communication using plasmonic nanoantennas and seeking dielectric nanoantenna as an alternative. In Proceedings of the 2022 IEEE International Conference on Nanoelectronics, Nanophotonics, Nanomaterials, Nanobioscience & Nanotechnology (5NANO), Kottayam, India, 28–29 April 2022; pp. 1–5. [Google Scholar]
  156. Sarycheva, A.; Polemi, A.; Liu, Y.; Dandekar, K.; Anasori, B.; Gogotsi, Y. 2D titanium carbide (MXene) for wireless communication. Sci. Adv. 2018, 4, eaau0920. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Vergütz, A.; Prates, N.G.; Henrique Schwengber, B.; Santos, A.; Nogueira, M. An architecture for the performance management of smart healthcare applications. Sensors 2020, 20, 5566. [Google Scholar] [CrossRef] [PubMed]
  158. Ergul, O.; Isiklar, G.; Cetin, I.C.; Algun, M. Design and analysis of nanoantenna arrays for imaging and sensing applications at optical frequencies. Adv. Electromagn. 2019, 8, 18–27. [Google Scholar] [CrossRef]
  159. Ranga, R.; Kalra, Y.; Kishor, K. Petal shaped nanoantenna for solar energy harvesting. J. Opt. 2020, 22, 035001. [Google Scholar] [CrossRef]
  160. Costa, K.; Dmitriev, V.; Souza, J.; Silvano, G. Analysis of nanodipoles in optical nanocircuits fed by gaussian beam. Int. J. Antennas Propag. 2014, 2014, 429425. [Google Scholar] [CrossRef]
  161. Xin, H.; Namgung, B.; Lee, L.P. Nanoplasmonic optical antennas for life sciences and medicine. Nat. Rev. Mater. 2018, 3, 228–243. [Google Scholar] [CrossRef]
  162. Sindhu, B.; Adepu, V.; Sahatiya, P.; Nandi, S. An MXene based flexible patch antenna for pressure and level sensing applications. FlatChem 2022, 33, 100367. [Google Scholar] [CrossRef]
  163. Dash, S.; Patnaik, A. Sub-wavelength graphene planar nanoantenna for THz application. Mater. Today Proc. 2019, 18, 1336–1341. [Google Scholar] [CrossRef]
  164. Dash, S.; Patnaik, A. Impact of silicon-based substrates on graphene THz antenna. Phys. E Low-Dimens. Syst. Nanostruct. 2021, 126, 114479. [Google Scholar] [CrossRef]
  165. Sergiyenko, A.; Molchanov, O.; Orlova, M. Nano-processor for the small tasks. In Proceedings of the 2019 IEEE 39th International Conference on Electronics and Nanotechnology (ELNANO), Kyiv, Ukraine, 16–18 April 2019; pp. 674–677. [Google Scholar]
  166. Yan, H.; Choe, H.S.; Nam, S.; Hu, Y.; Das, S.; Klemic, J.F.; Ellenbogen, J.C.; Lieber, C.M. Programmable nanowire circuits for nanoprocessors. Nature 2011, 470, 240–244. [Google Scholar] [CrossRef] [PubMed]
  167. Ali, O.; Ishak, M.K.; Bhatti, M.K.L. Emerging IoT domains, current standings and open research challenges: A review. PeerJ Comput. Sci. 2021, 7, e659. [Google Scholar] [CrossRef] [PubMed]
  168. Hills, G.; Lau, C.; Wright, A.; Fuller, S.; Bishop, M.D.; Srimani, T.; Kanhaiya, P.; Ho, R.; Amer, A.; Stein, Y.; et al. Modern microprocessor built from complementary carbon nanotube transistors. Nature 2019, 572, 595–602. [Google Scholar] [CrossRef] [PubMed]
  169. Li, L.; Wong, H.P. Integrating Graphene into Future Generations of Interconnect Wires. In Proceedings of the 2018 IEEE International Electron Devices Meeting (IEDM), San Francisco, CA, USA, 1–5 December 2018; pp. 5.5.1–5.5.4. [Google Scholar]
  170. Aziz, T.; Wei, S.; Sun, Y.; Ma, L.-P.; Pei, S.; Dong, S.; Ren, W.; Liu, Q.; Cheng, H.-M.; Sun, D.-M. High-performance flexible resistive random access memory devices based on graphene oxidized with a perpendicular oxidation gradient. Nanoscale 2021, 13, 2448–2455. [Google Scholar] [CrossRef]
  171. Gilardi, C.; Pedrinazzi, P.; Patel, K.A.; Anzi, L.; Luo, B.; Booth, T.J.; Bøggild, P.; Sordan, R. Graphene–Si CMOS oscillators. Nanoscale 2019, 11, 3619–3625. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Yasin, G.; Muhammad, N.; Nguyen, T.A.; Nguyen-Tri, P. Nanobattery: An introduction. In Nanobatteries and Nanogenerators; Song, H., Venkatachalam, R., Nguyen, T.A., Wu, H.B., Nguyen-Tri, P., Eds.; Elsevier: Cambridge, MA, USA, 2021; pp. 3–9. [Google Scholar] [CrossRef]
  173. AbdelHamid, A.A.; Mendoza-Garcia, A.; Ying, J.Y. Advances in and prospects of nanomaterials’ morphological control for lithium rechargeable batteries. Nano Energy 2022, 93, 106860. [Google Scholar] [CrossRef]
  174. Wu, Z.; Wang, Y.; Liu, X.; Lv, C.; Li, Y.; Wei, D.; Liu, Z. Carbon-nanomaterial-based flexible batteries for wearable electronics. Adv. Mater. 2019, 31, 1800716. [Google Scholar] [CrossRef]
  175. Kim, S.D.; Sarkar, A.; Ahn, J.-H. Graphene-based nanomaterials for flexible and stretchable batteries. Small 2021, 17, 2006262. [Google Scholar] [CrossRef]
  176. Du, Y.-T.; Kan, X.; Yang, F.; Gan, L.-Y.; Schwingenschlögl, U. MXene/graphene heterostructures as high-performance electrodes for Li-ion batteries. ACS Appl. Mater. Interfaces 2018, 10, 32867–32873. [Google Scholar] [CrossRef] [PubMed]
  177. Yuanzheng, L.; Zhicheng, Y.; Lianghao, M.; Buyin, L.; Shufa, L. A freestanding nitrogen-doped MXene/graphene cathode for high-performance Li–S batteries. Nanoscale Adv. 2022, 4, 2189–2195. [Google Scholar] [CrossRef] [PubMed]
  178. Nguyen, T.N.; Iranpour, B.; Cheng, E.; Madden, J.D.W. Washable and stretchable Zn–MnO2 rechargeable cell. Adv. Energy Mater. 2022, 12, 2103148. [Google Scholar] [CrossRef]
  179. Managing Nanomaterials in the Workplace. Available online: https://osha.europa.eu/en/emerging-risks/nanomaterials (accessed on 6 August 2022).
  180. Valic, M.S.; Halim, M.; Schimmer, P.; Zheng, G. Guidelines for the experimental design of pharmacokinetic studies with nanomaterials in preclinical animal models. J. Control. Release 2020, 323, 83–101. [Google Scholar] [CrossRef] [PubMed]
  181. Harshitha, C.; Bhattacharyya, S. A brief review on nanotechnology as a challenging field in pharmaceuticals and their regulatory approval. J. Crit. Rev. 2020, 7, 963–968. [Google Scholar] [CrossRef]
  182. Nielsen, M.B.; Baun, A.; Mackevica, A.; Thit, A.; Odnevall Wallinder, I.; Gallego, J.A.; Westergaard Clausen, L.P.; Rissler, J.; Skjolding, L.; Castro Nilsson, A.; et al. Nanomaterials in the European chemicals legislation—Methodological challenges for registration and environmental safety assessment. Environ. Sci. Nano 2021, 8, 731–747. [Google Scholar] [CrossRef]
  183. Rasmussen, K.; Rauscher, H.; Kearns, P.; González, M.; Riego Sintes, J. Developing OECD test guidelines for regulatory testing of nanomaterials to ensure mutual acceptance of test data. Regul. Toxicol. Pharmacol. 2019, 104, 74–83. [Google Scholar] [CrossRef]
  184. Beaucham, C.; Hodson, L. General Safe Practices for Working with Engineered Nanomaterials in Research Laboratories; Publication No. 2012-147; Department of Health and Human Services, Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health (NIOSH): Washington, DC, USA, 2012.
  185. Daima, H.K.; Kothari, S.L.; Kumar, B.S. Nanotoxicology: Toxicity Evaluation of Nanomedicine Applications; CRC Press: Boca Raton, FL, USA, 2021. [Google Scholar]
  186. Kumari, P.; Seth, R.; Meena, A. Consequences of nanomaterials on human health and ecosystem. In Nanotoxicology; Daima, H.K., Kothari, S.L., Bhargava, S.K., Eds.; CRC Press: Boca Raton, FL, USA, 2021; pp. 157–200. [Google Scholar]
  187. Khan, A.U.; Khan, M.; Cho, M.H.; Khan, M.M. Selected nanotechnologies and nanostructures for drug delivery, nanomedicine and cure. Bioprocess Biosyst. Eng. 2020, 43, 1339–1357. [Google Scholar] [CrossRef] [PubMed]
  188. Singh, D.; Dilnawaz, F.; Sahoo, S.K. Challenges of moving theranostic nanomedicine into the clinic. Nanomedicine 2020, 15, 111–114. [Google Scholar] [CrossRef] [Green Version]
  189. Dugershaw, B.B.; Aengenheister, L.; Hansen, S.S.K.; Hougaard, K.S.; Buerki-Thurnherr, T. Recent insights on indirect mechanisms in developmental toxicity of nanomaterials. Part. Fibre Toxicol. 2020, 17, 31. [Google Scholar] [CrossRef]
  190. Foulkes, R.; Man, E.; Thind, J.; Yeung, S.; Joy, A.; Hoskins, C. The regulation of nanomaterials and nanomedicines for clinical application: Current and future perspectives. Biomater. Sci. 2020, 8, 4653–4664. [Google Scholar] [CrossRef] [PubMed]
  191. Fadeel, B.; Farcal, L.; Hardy, B.; Vázquez-Campos, S.; Hristozov, D.; Marcomini, A.; Lynch, I.; Valsami-Jones, E.; Alenius, H.; Savolainen, K. Advanced tools for the safety assessment of nanomaterials. Nat. Nanotechnol. 2018, 13, 537–543. [Google Scholar] [CrossRef] [PubMed]
  192. Biological Evaluation of Medical Devices—Part 22: Guidance on Nanomaterials (ISO/TR 10993-22: 2017); International Organization for Standardization: Geneva, Switzerland, 2017.
  193. Bernard, M.; Jubeli, E.; Pungente, M.D.; Yagoubi, N. Biocompatibility of polymer-based biomaterials and medical devices—Regulations, in vitro screening and risk-management. Biomater. Sci. 2018, 6, 2025–2053. [Google Scholar] [CrossRef]
  194. Badhirappan, G.P.; Nallasivam, V.; Varadarajan, M.; Leobeemrao, V.P.; Bose, S.; Venugopal, E.; Rajendran, S.; Angleo, P.C. Phase competition induced bio-electrochemical resistance and bio-compatibility effect in nanocrystalline Zr x–Cu100− x thin films. J. Nanosci. Nanotechnol. 2018, 18, 4534–4543. [Google Scholar] [CrossRef]
  195. Han, C.Y.; Li, Y.; Zhang, Z.X.; Liu, W.H.; Li, X.; Guo, X.; Wan, J.; Zhang, G.H.; Wang, X.L.; Lin, Q. An artifical synapse based on graphene field-effect transistor with silver gel/polarized-aptamer gate. Org. Electron. 2021, 92, 106118. [Google Scholar] [CrossRef]
  196. Chuan, Y. Study on Electrospun Silicone Modified Polyurethane Nanofibers. Doctoral Thesis, Shinshu University, Nagano, Japan, 2019. [Google Scholar]
  197. Hart, C.; Didier, C.M.; Sommerhage, F.; Rajaraman, S. Biocompatibility of blank, post-processed and coated 3D printed resin structures with electrogenic cells. Biosensors 2020, 10, 152. [Google Scholar] [CrossRef] [PubMed]
  198. Madannejad, R.; Shoaie, N.; Jahanpeyma, F.; Darvishi, M.H.; Azimzadeh, M.; Javadi, H. Toxicity of carbon-based nanomaterials: Reviewing recent reports in medical and biological systems. Chem. Biol. Interact. 2019, 307, 206–222. [Google Scholar] [CrossRef]
  199. Das, B.; Chabukdhara, M.; Patra, S.; Gogoi, M. Current knowledge on toxicity of nanomaterials: Toxicity assessment and impact. In Nanotoxicology; Daima, H.K., Kothari, S.L., Bhargava, S.K., Eds.; CRC Press: Boca Raton, FL, USA, 2021; pp. 339–375. [Google Scholar]
  200. Wu, K.; Zhou, Q.; Ouyang, S. Direct and indirect genotoxicity of graphene family nanomaterials on DNA—A review. Nanomaterials 2021, 11, 2889. [Google Scholar] [CrossRef]
  201. Pelin, M.; Sosa, S.; Prato, M.; Tubaro, A. Occupational exposure to graphene based nanomaterials: Risk assessment. Nanoscale 2018, 10, 15894–15903. [Google Scholar] [CrossRef]
  202. Burgum, M.J.; Clift, M.J.D.; Evans, S.J.; Hondow, N.; Tarat, A.; Jenkins, G.J.; Doak, S.H. Few-layer graphene induces both primary and secondary genotoxicity in epithelial barrier models in vitro. J. Nanobiotechnol. 2021, 19, 24. [Google Scholar] [CrossRef]
  203. Mortezaee, K.; Najafi, M.; Samadian, H.; Barabadi, H.; Azarnezhad, A.; Ahmadi, A. Redox interactions and genotoxicity of metal-based nanoparticles: A comprehensive review. Chem. Biol. Interact. 2019, 312, 108814. [Google Scholar] [CrossRef] [PubMed]
  204. Zhang, X.; Luan, J.; Chen, W.; Fan, J.; Nan, Y.; Wang, Y.; Liang, Y.; Meng, G.; Ju, D. Mesoporous silica nanoparticles induced hepatotoxicity via NLRP3 inflammasome activation and caspase-1-dependent pyroptosis. Nanoscale 2018, 10, 9141–9152. [Google Scholar] [CrossRef] [PubMed]
  205. García-Rodríguez, A.; Rubio, L.; Vila, L.; Xamena, N.; Velázquez, A.; Marcos, R.; Hernández, A. The comet assay as a tool to detect the genotoxic potential of nanomaterials. Nanomaterials 2019, 9, 1385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Abbasi-Oshaghi, E.; Mirzaei, F.; Pourjafar, M. NLRP3 inflammasome, oxidative stress, and apoptosis induced in the intestine and liver of rats treated with titanium dioxide nanoparticles: In vivo and in vitro study. Int. J. Nanomed. 2019, 14, 1919–1936. [Google Scholar] [CrossRef] [Green Version]
  207. Yuan, Y.-G.; Cai, H.-Q.; Wang, J.-L.; Mesalam, A.; Md Talimur Reza, A.M.; Li, L.; Chen, L.; Qian, C. Graphene oxide–silver nanoparticle nanocomposites induce oxidative stress and aberrant methylation in caprine fetal fibroblast cells. Cells 2021, 10, 682. [Google Scholar] [CrossRef]
  208. Ganguly, P.; Breen, A.; Pillai, S.C. Toxicity of nanomaterials: Exposure, pathways, assessment, and recent advances. ACS Biomater. Sci. Eng. 2018, 4, 2237–2275. [Google Scholar] [CrossRef]
  209. Forouzandeh, F.; Zhu, X.; Alfadhel, A.; Ding, B.; Walton, J.P.; Cormier, D.; Frisina, R.D.; Borkholder, D.A. A nanoliter resolution implantable micropump for murine inner ear drug delivery. J. Control. Release 2019, 298, 27–37. [Google Scholar] [CrossRef]
  210. Banerjee, U.; Jain, S.K.; Sen, A.K. Particle encapsulation in aqueous ferrofluid drops and sorting of particle-encapsulating drops from empty drops using a magnetic field. Soft Matter 2021, 17, 6020–6028. [Google Scholar] [CrossRef]
  211. Xie, Y.; Wan, B.; Yang, Y.; Cui, X.; Xin, Y.; Guo, L.-H. Cytotoxicity and autophagy induction by graphene quantum dots with different functional groups. J. Environ. Sci. 2019, 77, 198–209. [Google Scholar] [CrossRef]
  212. Barrios, A.C.; Wang, Y.; Gilbertson, L.M.; Perreault, F. Structure–property–toxicity relationships of graphene oxide: Role of surface chemistry on the mechanisms of interaction with bacteria. Environ. Sci. Technol. 2019, 53, 14679–14687. [Google Scholar] [CrossRef]
  213. Patel, G.; Patra, C.; Srinivas, S.P.; Kumawat, M.; Navya, P.N.; Daima, H.K. Methods to evaluate the toxicity of engineered nanomaterials for biomedical applications: A review. Environ. Chem. Lett. 2021, 19, 4253–4274. [Google Scholar] [CrossRef]
  214. Bourquin, J.; Milosevic, A.; Hauser, D.; Lehner, R.; Blank, F.; Petri-Fink, A.; Rothen-Rutishauser, B. Biodistribution, clearance, and long-term fate of clinically relevant nanomaterials. Adv. Mater. 2018, 30, 1704307. [Google Scholar] [CrossRef] [PubMed]
  215. Kreyling, W.G.; Holzwarth, U.; Schleh, C.; Kozempel, J.; Wenk, A.; Haberl, N.; Hirn, S.; Schäffler, M.; Lipka, J.; Semmler-Behnke, M.; et al. Quantitative biokinetics of titanium dioxide nanoparticles after oral application in rats: Part 2. Nanotoxicology 2017, 11, 443–453. [Google Scholar] [CrossRef] [Green Version]
  216. Cambiaghi, A. Biological Evaluation of Medical Devices as an Essential Part of the Risk Management Process: Updates and Challenges of ISO 10993-1: 2018; Eurofins Medical Device Testing: Lancaster, PA, USA, 2018. [Google Scholar]
  217. Biological Evaluation of Medical Devices—Part 1: Evaluation and Testing Within a Risk Management Process (ISO 10993-1: 2018); International Organization for Standardization: Geneva, Switzerland, 2018.
  218. Burton, S.D. Not anytime soon: The clinical translation of nanorobots and its biocompatibility-interdisciplinarity critique. In Artificial Intelligence and Its Discontents: Critiques from the Social Sciences and Humanities; Hanemaayer, A., Ed.; Springer International Publishing: Cham, Switzerland, 2022; pp. 147–178. [Google Scholar] [CrossRef]
  219. Tang, Y.H.; Lin, Y.H.; Huang, T.T.; Wang, J.S.; Hu, Y.C.; Shiao, M.H. Development of micro-needle array for tumor vaccine patch applications. In Proceedings of the 2019 IEEE 19th International Conference on Nanotechnology (IEEE-NANO), Macao, China, 22–26 July 2019; pp. 129–132. [Google Scholar]
  220. Molino, J.; Araúz, B.; Nieto, C.; Reginensi, D.; Tristan, S.d. Advancements in MEMS and NEMS from bio-tribological perspective. J. Nanomed. Nanotechnol. Nanomater. 2021, 2, 133. [Google Scholar]
  221. Mariello, M.; Kim, K.; Wu, K.; Lacour, S.P.; Leterrier, Y. Recent advances in encapsulation of flexible bioelectronic implants: Materials, technologies, and characterization methods. Adv. Mater. 2022, 34, 2201129. [Google Scholar] [CrossRef]
  222. Deng, S.; Gigliobianco, M.R.; Censi, R.; Di Martino, P. Polymeric nanocapsules as nanotechnological alternative for drug delivery system: Current status, challenges and opportunities. Nanomaterials 2020, 10, 847. [Google Scholar] [CrossRef]
  223. Pushpamalar, J.; Meganathan, P.; Tan, H.L.; Dahlan, N.A.; Ooi, L.-T.; Neerooa, B.N.H.M.; Essa, R.Z.; Shameli, K.; Teow, S.-Y. Development of a polysaccharide-based hydrogel drug delivery system (DDS): An update. Gels 2021, 7, 153. [Google Scholar] [CrossRef]
  224. Muhammad, Z.; Raza, A.; Ghafoor, S.; Naeem, A.; Naz, S.S.; Riaz, S.; Ahmed, W.; Rana, N.F. PEG capped methotrexate silver nanoparticles for efficient anticancer activity and biocompatibility. Eur. J. Pharm. Sci. 2016, 91, 251–255. [Google Scholar] [CrossRef] [PubMed]
  225. Harish, V.; Tewari, D.; Gaur, M.; Yadav, A.B.; Swaroop, S.; Bechelany, M.; Barhoum, A. Review on nanoparticles and nanostructured materials: Bioimaging, biosensing, drug delivery, tissue engineering, antimicrobial, and agro-food applications. Nanomaterials 2022, 12, 457. [Google Scholar] [CrossRef]
  226. Toumey, C. Later voices on ethics in nanotechnology. Nat. Nanotechnol. 2019, 14, 636–637. [Google Scholar] [CrossRef]
  227. Selvaraj, S.; Sundaravaradhan, S. Challenges and opportunities in IoT healthcare systems: A systematic review. SN Appl. Sci. 2019, 2, 139. [Google Scholar] [CrossRef]
Figure 1. Development trends and clinical applications of nanomaterials-based bioNEMS/MEMS devices and future opportunities for integration with Internet of Things (IoT) technology.
Figure 1. Development trends and clinical applications of nanomaterials-based bioNEMS/MEMS devices and future opportunities for integration with Internet of Things (IoT) technology.
Nanomaterials 12 04025 g001
Figure 2. Types of inorganic nanomaterials developed using bioNEMS/MEMS techniques for biomedical applications.
Figure 2. Types of inorganic nanomaterials developed using bioNEMS/MEMS techniques for biomedical applications.
Nanomaterials 12 04025 g002
Figure 3. Household food waste in Asia and Europe countries published in Food Waste Index Report 2021 [27].
Figure 3. Household food waste in Asia and Europe countries published in Food Waste Index Report 2021 [27].
Nanomaterials 12 04025 g003
Figure 4. bioNEMS/MEMS fabrication strategies.
Figure 4. bioNEMS/MEMS fabrication strategies.
Nanomaterials 12 04025 g004
Figure 6. Schematic depiction of cell-sorting separation and cell manipulation adapting the NEMS and MEMS technology. Illustrations are adapted from mechanisms described by ref [6] and [111], and are not-to-scale.
Figure 6. Schematic depiction of cell-sorting separation and cell manipulation adapting the NEMS and MEMS technology. Illustrations are adapted from mechanisms described by ref [6] and [111], and are not-to-scale.
Nanomaterials 12 04025 g006
Figure 7. Platform for safety assessment of nanomaterials with focus on human health and the environment. EHS, environmental, health and safety; NM, nanomaterials.
Figure 7. Platform for safety assessment of nanomaterials with focus on human health and the environment. EHS, environmental, health and safety; NM, nanomaterials.
Nanomaterials 12 04025 g007
Table 1. Advantages and disadvantages of bioNEMS/MEMS fabrication strategies.
Table 1. Advantages and disadvantages of bioNEMS/MEMS fabrication strategies.
TechniqueAdvantagesDisadvantagesRef
Photolithography
Strong chemical stability
Adequate adhesion
Good photosensitivity
Resolution limited to the wavelength of the used light
[63,65]
Electron Beam
Lithography
No mask used
Precise control over energy and dose of the electron beam
Accurate registration over small areas on a wafer
Lower defect densities
Low exposure speed
Costly method
Resolution is limited to the diffraction of electron beam in solids
[64,73,74]
Scanning Probe
Lithography
High resolution method
Electrons have lower energy in comparison to EBL
Low cost
3D patterning
High quality patterning
Relatively slow method
[64,75,76]
Soft
Lithography
Cost-effective
Does not require much expertise or sophisticated equipment
Slow
Other methods are commonly preferred
[64,106]
Nanoimprint
Lithography
High resolution (better than 10 nm)
High efficiency
Mechanical misalignments are common
Defectivity and template life are difficult to control
[74]
Bulk
Micromachining
Low cost
Etched surface is smooth
Good for making holes in MEMS
No layering on the same wafer
[71,79,92]
Surface
Micromachining
Simple method
Low cost
Convenient formation of multilayer systems
Due to thin layers, bending is frequent
Post processing is needed for stress relieving
[64,107,108]
Combined
Micromachining
Decreased fabrication cost; improves yield
Low cost
Guarantees electrical reliability
Post processing is needed
[79]
Electron-discharge
Micromachining
Fabrication on any conductive material
Only used on conductive material
[99]
Laser
Micromachining
No mask required
No post fabrication steps
Precise, fast
Few heat-affected zones
Good for rapid prototyping
Micro cracks
Shock wave surface damage
[68,79,109]
Electrospinning
Suitable for testing
Simple set up
High modulative capability
High productivity
Great nanofiber diameter distribution
[105,110]
Table 3. Examples of nanomaterials-based nanosensors in wearable devices and implants.
Table 3. Examples of nanomaterials-based nanosensors in wearable devices and implants.
ApplicationTypes of NanosensorAdvantage(s)Ref
Wearable cardiovascular monitoring
MEMS pressure sensor array
Electrochemical
Optical
Pressure
Paper-based
IoT platforms in cardiovascular monitoring potentially reduce morbidity and mortality statistics due to early diagnosis and response
[143,146]
Human steroid
Hormone sensing
Nanoslot array-based terahertz molecule-specific sensor chip
Implantable sensor
Discrimination and quantification of trace amounts of steroid hormones in biological specimens to elucidate changing expression and prevention of endocrine disorders
[147,148]
Artificial auditory system
Piezoresistive
Hydrogel
Nanocomposites
Closely mimic the biological cochlea for hearing disorder due to ageing
[149]
Smart electronics textile for wearable body sensor
Carbon-based nanomaterials (CNT)
Reduced graphene oxide (rGO)
Real-time body physical, chemical and electrophysiological sensing (strain, pressure, temperature, sweat, electrocardiogram (ECG), electromyography (EMG) and etc.)
[150]
Ocular applications (contact lens)
Graphene contact lens electrodes
The graphene electrodes demonstrate high-efficacy measurements of various kinds of electroretinography without corneal irritation for ocular application
[144,151]
Table 4. Examples of nanomaterials-based nanoantenna and its advantages for wireless communication.
Table 4. Examples of nanomaterials-based nanoantenna and its advantages for wireless communication.
Types of
Nanoantenna
Nanomaterial(s)Advantages(s)Ref
Flexible and low-cost patch antenna
MXene (Ti3C2Tx)
A functional nanomaterial-based antenna that serves the dual purpose of sensing and communication
High potential for based wireless sensing
[162]
Circular patch
nanoantenna
Graphene
Graphene nanoantenna enables high miniaturization
Ultra-wideband (UWB) behaviour
Easy reconfiguration
[163]
Graphene plasmonic Terahertz (THz) antenna
Graphene
The silicon-based substrate on graphene THz antennas demonstrates good performance in reflection coefficient, impedance bandwidth, front-to-back ratio and directivity and promising for THz short-range communication, sensing and biomedical applications
[164]
ITO-based CNT coated transparent nano E-shape patch antenna
ITO-based CNT
ITO-based CNT nanoantenna has better performance in terms of broad bandwidths, reasonable radiation efficiency, sufficient gain, lightweight and compactness
Suitable for satellite communication that is potentially used for telehealth in rural or areas with limited mobile communication network
[153]
Half-wave dipole MXene antenna for RF devices
2D titanium carbide (MXene) coating
MXene coating offers a transparent antenna for various classes of RF for IoT applications, portable, flexible and wearable electronic devices
[156]
Optical dipole nanoantenna
Cadmium selenide quantum dots
Intensified Terahertz radiation for time-domain spectroscopy for medical imaging applications
[154]
Table 5. Nanomaterials and selected applications of nanoprocessors for future biomedical applications along with their advantages.
Table 5. Nanomaterials and selected applications of nanoprocessors for future biomedical applications along with their advantages.
Application(s)Nanomaterial(s)Advantage(s)Ref
Energy consumption
Carbon nanotube field-effect transistor (CNFET)-based digital circuits
The CNFET that designed and fabricated using industry-standard design flows and processes overcoming nanoscale imperfections at macroscopic scales across full water substrates
Offer energy-efficiency benefit
[168]
Interconnect wires in the processor core
Integrating graphene on the interconnect wires
Increase about 8% of speed boost and contribute to 12% of energy saving
[169]
High-performance resistive read access memory (RRAM)
Graphene oxidised with a perpendicular oxidation gradient
Offers a high on-off current ratio of ~105, long-term retention of ~106 s, reproducibility over 104 cycles and long-term flexibility at a bending strain of 0.6%
Demonstrate great potential in wearable smart data-storage devices
[170]
Complementary metal-oxide-semiconductor (CMOS) oscillators
Graphene- silica (Si) CMOS hybrid circuit
Introduces graphene in Si CMOS has ameliorated voltage swing and switch ability of Si CMOS circuit in a modern electronics system
[171]
Table 6. Nanomaterials and cytotoxicity assay together with the main results of toxicity assessment.
Table 6. Nanomaterials and cytotoxicity assay together with the main results of toxicity assessment.
TestNanomaterialToxicity AssessmentRef
MTTZr x -Cu100x No toxicity in osteoblast cells[194]
MTTGrapheneBiocompatible against HK-2 cells [195]
MTTSilicon MEMSNo signs of infection or inflammation[196]
Trypan blue exclusion assayPDMS, PS, SU-8>85% cells viability[197]
Clonogenic AssayCarbon>50 cell colony formation[198]
Apoptosis AssayAgDNA damage leading to apoptosis[199]
DNA ladderingGrapheneDNA damage due to oxidation stress[200,201,202,203]
Caspase AssayMesoporous SiLiver inflammation, hepatotoxicity[204]
Comet AssayTiO2, SiO2, ZnO, CeO2, Ag, MWCNTMild to considerable genotoxic effect[205]
Tunnel AssayTiO2Increased gene expression of the inflammation and apoptotic effect [206]
Annexin V and Propidium iodide GO-AgIncreased production of ROS[207]
Lipid peroxidation AssayCarbon basedGeneration of ROS, inflammation, damage to the proteins[198,199]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Dahlan, N.A.; Thiha, A.; Ibrahim, F.; Milić, L.; Muniandy, S.; Jamaluddin, N.F.; Petrović, B.; Kojić, S.; Stojanović, G.M. Role of Nanomaterials in the Fabrication of bioNEMS/MEMS for Biomedical Applications and towards Pioneering Food Waste Utilisation. Nanomaterials 2022, 12, 4025. https://doi.org/10.3390/nano12224025

AMA Style

Dahlan NA, Thiha A, Ibrahim F, Milić L, Muniandy S, Jamaluddin NF, Petrović B, Kojić S, Stojanović GM. Role of Nanomaterials in the Fabrication of bioNEMS/MEMS for Biomedical Applications and towards Pioneering Food Waste Utilisation. Nanomaterials. 2022; 12(22):4025. https://doi.org/10.3390/nano12224025

Chicago/Turabian Style

Dahlan, Nuraina Anisa, Aung Thiha, Fatimah Ibrahim, Lazar Milić, Shalini Muniandy, Nurul Fauzani Jamaluddin, Bojan Petrović, Sanja Kojić, and Goran M. Stojanović. 2022. "Role of Nanomaterials in the Fabrication of bioNEMS/MEMS for Biomedical Applications and towards Pioneering Food Waste Utilisation" Nanomaterials 12, no. 22: 4025. https://doi.org/10.3390/nano12224025

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop