Startseite Applications of nanomedicine-integrated phototherapeutic agents in cancer theranostics: A comprehensive review of the current state of research
Artikel Open Access

Applications of nanomedicine-integrated phototherapeutic agents in cancer theranostics: A comprehensive review of the current state of research

  • Ambreen Shoaib , Shamama Javed , Mohammad Tabish , Mohammad Ehtisham Khan EMAIL logo , Mehvash Zaki , Saad S. Alqahtani , Muhammad H. Sultan , Waquar Ahsan und Mohd Afzal EMAIL logo
Veröffentlicht/Copyright: 24. Mai 2024
Veröffentlichen auch Sie bei De Gruyter Brill

Abstract

Innovative approaches such as photodynamic therapy (PDT) and photothermal therapy (PTT) have made nanomedicines a promising frontier in cancer theranostics. The combination of nanocarriers with photothermal agents and photosensitizers (PSs) has shown excellent promise for the diagnosis and the treatment of cancer, primarily at the cellular, vascular, and tumor microenvironment level. Using nanocarriers in PDT has revolutionized precision and efficacy, allowing the drug to reach cancer cells faster and offering high enhancing PS accumulation. These agents are activated by light of specific wavelengths, leading to localized cytotoxicity, offering highly selective cancer therapy. Nanomaterials such as gold and silver nanoparticles have enabled remarkable progress in cancer hyperthermia using PTT. The unique optical properties of these nanoparticle-based nanomedicines make them ideal candidates for converting light energy into heat, selectively ablating the cancer cells. In this review, nanomedicine-integrated phototherapeutic agents are discussed and the most important recent developments in PDT and PTT are examined, as well as how nanoparticle-based formulations improve diagnosis and treatment. In addition, nanocarriers used in cancer phototherapy and their mode of action are discussed. Nanocarriers are useful for drug delivery as well as for imaging and diagnostic purposes during cancer treatment. In this review, we explore the role of nanoparticles in improving phototherapy precision and selectivity while minimizing collateral tissue damage. It specifies a comprehensive impression of the current research on cancer therapy, underscoring its potential to revolutionize the treatment paradigm by highlighting the current state of research.

1 Introduction

Cancer, one of the leading global causes of death in both men and women, remains a formidable global health challenge, necessitating continual exploration of novel therapeutic strategies to complement and surpass traditional treatment modalities [1,2]. The cancer treatment strategies include chemotherapy, photodynamic, photothermal, and hormonal therapies; however, a large number of patients remain at a high risk of disease recurrence or resistance [2,3]. Multidrug resistance (MDR), a developing issue in the treatment of cancer, further worsens the issue. Among the burgeoning avenues, the use of nanomedicines has emerged a transformative paradigm, offering an unprecedented precision and efficacy in cancer diagnosis and therapy [4]. Nanotechnology-based drug therapies have been extensively explored in recent years to address this issue, and the findings have shown remarkable potential in overcoming the drug resistance, with several nanocarrier-based pharmaceuticals being studied in the clinical settings [5]. Cancer nanomedicines aim to address the inherent limitations of traditional cancer diagnostics and therapy [6]. An integral part of cancer theranostics is the combination of cancer diagnosis and cancer treatment [4,7]. Many substances that can be incorporated into and released from theranostics, such as diagnostic and therapeutic ingredients [8]. The delivery can be triggered by an internal or external stimulus. Internal stimuli originate from the body, including pH, redox potential, oxidative stress, enzyme activity, and temperature [9]. An external stimulus is one that comes from outside the body, such as light, ultrasound, and magnetic fields. In addition to these factors, biomaterials that are able to respond to them have some advantages as well [9]. There are applications for nanoparticles in theranostics. Using nanoparticles, diagnostic materials, such as dyes, can be delivered to target tumors to allow imaging to be performed [10]. In addition, cancer drugs can be delivered to cancer cells using these devices [11]. By amalgamating the unique properties of nanoparticles with the principle of phototherapy, nanomedicines provide a versatile platform for targeted intervention, minimizing the systemic toxicity and maximizing the therapeutic outcomes [12]. The emerging area of nanotechnology has brought nanotherapeutics, and this technique has shown tremendous promise in treating cancer.

The conventional treatment of cancer, while often effective, is fraught with several limitations including nonspecificity, off-target effects, and resistance [13]. In response to these challenges, phototherapy has been proven to be an innovative and promising approach. Over the past few years, phototherapy has acquired regulatory approval for the treatment of many disorders, including cancer [14]. Solid tumors and other disorders are shown to be treated using phototherapy. Phototherapy is a photoactive technique that combines the use of PSs with particular light wavelengths. The global regulatory authorities have approved several photosensitizing medications and light applications for the therapy of cancer and microbial infections [15,16,17]. Hence, it garnered substantial interest from researchers worldwide, and as a result, it is now a more commonly used medical tool for the diagnosis and treatment of cancer. In the realm of phototherapy, two modalities, namely, photodynamic therapy (PDT) and photothermal therapy (PTT), have emerged as focal points of exploration. The PDT harnesses the cytotoxic effects of light-activated PSs to induce localized cell death, while PTT employs nanomaterials to convert light into heat causing cancer cell ablation.

Both modalities have benefited from the use of nanotechnology to increase their specificity and potency [18,19]. Target cells are killed by activated PSs by producing radicals or reactive oxygen species (ROS) in PDT and heat generation in PTT [16,20]. The integration of these phototherapeutic strategies with nanomedicine introduces a nuanced precision that addresses the shortcomings of conventional treatments. The intersection of nanomedicine, PDT, and PTT not only presents an opportunity to revolutionize cancer diagnosis and treatment but also challenges our understanding of the intricate interplay among light, nanoparticles, and cellular responses. However, despite the advantages and alluring properties of nanomedicines, there are still some hurdles associated with their therapeutic use, leading to an assert that they have not yet realized their full potential [17,21].

In clinical practice, targeting therapy through heightened permeability and retention (EPR) is not constantly effective, while the strength of the consequence alters depending on the type and the site of tumors, their blood perfusion status, and the properties of macromolecular anticancer agents [22,23]. Nanomedicine-based therapeutic regimes displayed better potential in targeting the cancer cells. Nanomedicines can specifically accumulate in solid tumors due to the EPR effect, which can improve the bioavailability of therapeutics at the target site [24,25]. This review aims to specify a comprehensive examination of the current landscape of nanomedicine-based phototherapy for cancer treatment, with a specific emphasis on the synergistic interplay between PDT and PTT. The intricate design of nanocarriers is tailored for these phototherapeutic applications, exploring their unique physicochemical properties that enhance drug delivery, imaging, and therapeutic efficacy. In addition, it aims to emphasize the recent progress in nanomedicine-based phototherapy with a comprehensive account of the current state of research that has been carried out using in vitro and in vivo studies in the treatment of cancer. We navigate through the evolving field of targeted delivery systems, addressing the critical balance between biocompatibility and therapeutic efficiency.

2 Photodynamic therapy using PS

2.1 Mechanisms underlying PDT

The PDT is a marginally invasive procedure of destroying cancer cells that employs a blend of light energy, a PS, and molecular oxygen present inside the cells. When all the three factors come into play, the photoproduction of ROS takes place resulting from the in vivo administration of the PS in that specific site showing cytotoxic effects on tumor cells [26]. To activate the PS, a certain wavelength of light is needed. The PS is less toxic to the cells until it is activated by light; however, upon activation, it leads to the formation of ROS, which renders toxicity to the tumor. PDT is a two-stage process, which is initiated upon the administration of a PS selectively in the cancerous tissue, followed by the use of an irradiation of a particular wavelength showing maximum absorption by the photoactive drugs [27].

During the photochemical process, the PS molecule absorbs incident light, changing it from the ground state (S0) to an excited singlet state (S1) for a short period (∼ns), which then undergoes intersystem crossing to a relatively more stable (∼ms) excited triplet state having a longer lifetime. From this triplet state (T1), the PS molecules return to the ground state (S0) by undergoing a type I or type II photodynamic reaction. In the type I reaction, there is a transfer of electrons from the PS to the biological substrate, resulting in the production of ROS such as O 2 ˙ and ˙OH [28]. However, in type II PDT, the transfer of energy occurs between PS and adjacent 3O2 directly for the production of cytotoxic 1O2 (Figure 1). The reactive species then activate several downstream biological events, including direct cytotoxicity, inflammation, and vascular events (Figure 2). Nevertheless, PDT involves oxygen in both these processes. The rate of type I PDT is less affected directly by the concentration of O2; however, in type II PDT, PS in the triplet state transfers energy to nearby 3O2 directly for the generation of cytotoxic singlet oxygen (1O2). This route needs the contribution of intratumoral O2 concentration for the formation of ROS. In both situations, the ROS is generated to target the cancerous cells in a localized manner [29,30,31].

Figure 1 
                  Type I and type II photochemical reactions in PDT. The PS changes from its ground state to an excited state after absorbing light and the activated PS generates free radicals (type I reaction) or oxidative substrates (type II reaction). Reprinted with permission from ref. [31], Copyright 2019, American Chemical Society.
Figure 1

Type I and type II photochemical reactions in PDT. The PS changes from its ground state to an excited state after absorbing light and the activated PS generates free radicals (type I reaction) or oxidative substrates (type II reaction). Reprinted with permission from ref. [31], Copyright 2019, American Chemical Society.

Figure 2 
                  Apoptosis, necrosis, and related biological cascades initiated by intracellular ROS produced upon the excitation of PS in PDT. Reprinted with permission from ref. [31], Copyright 2019, American Chemical Society.
Figure 2

Apoptosis, necrosis, and related biological cascades initiated by intracellular ROS produced upon the excitation of PS in PDT. Reprinted with permission from ref. [31], Copyright 2019, American Chemical Society.

2.2 Light sources for PDT

The PDT procedure represents an extremely promising approach for treating certain types of cancerous or precancerous lesions because of its efficiency and selectivity [32,33]. To accomplish the technique, light, oxygen, and a PS must exist simultaneously at a subcellular level [34]. In response to light with a particular wavelength, PS converts the nearest molecular oxygen/oxygenic species into highly cytotoxic ROS that initiate tumor necrosis [35]. The majority of PSs are nontoxic in the dark, so PDT action strikes simply when and where PS-containing tissues are illuminated, therefore ensuring high selectivity toward cancerous tissues [36].

The preference of light source in PDT rests on the target spot, PS used, and light dose to be delivered. The conventional PDT utilizes short-range UV-Vis light (400−700 nm) to excite the PS; however, it fails to achieve deep tissue penetration [37]. The absorption wavelength of the PS should be taken into consideration while selecting the wavelength of the light source. Generally, optical fiber-based devices are utilized to deliver the light adjacent to the target. The distribution of the treatment light is also affected by the optical properties of the tissue [16]. It is expected that during treatment, highly pigmented areas absorb the light, while the majority of light gets scattered by the localized tissue. Light-emitting diodes with various illumination spectra choices have been widely used for this purpose; however, poor coupling efficacy with the optical fiber distribution restricts their application [38]. On the other hand, lasers use optical fiber coupling to provide high optical power to a distal irradiation area [39]. Light-transmitting devices have been constantly explored in the clinical setting as an opportunity to deliver light directly to the PS, although this has often resulted in more invasive methods of delivery [40]. There are limitations to both of these improvements when it comes to tackling deep-seated lesions and metastases. Thus, alternative approaches may include introducing molecules, nano-objects, or materials that function as internal lights [41].

2.3 Role of oxygen in PDT

Oxygen is actively involved in the photosensitization process that yields oxygen-derived ROS, and hence, its presence is necessary to obtain a preferred effect. Under hypoxic conditions, PDT treatment is challenging for most solid tumors, and there are chances of recurrence, as the hypoxic cells are resistant to the therapy [42]. To enhance the performance of O2 delivery, nanomaterials and O2 carriers such as perfluorocarbon, hemoglobin (Hb), and metal-organic frameworks (MOFs) are often combined to develop a new O2 delivery system [43,44,45]. Moreover, nanoparticles coated with pH-sensitive polymer enable the controlled production of molecular oxygen as a function of external stimuli (temperature and pH). Several nanostructures of manganese oxide (MnO2) have been utilized to release tumor hypoxia through the decomposition of endogenous hydrogen peroxide (H2O2) and producing O2 within the cancer cells [46]. Recently, a nanosystem-mediated PDT capable of improving oxygen to accelerate 1O2 release, resulting in tumor inhibition was described [24]. Similarly, nanosized carriers such as micelles and vesicles allow homogeneous delivery of O2 in the tumor [47,48]. The Hb-loaded NPs exhibited considerable support for ROS production during ROS-enhanced therapy. MOFs have also been explored as oxygen carriers, which showed good potential as sorbent materials for oxygen molecules [45,49,50]. Due to this role, nanoformulations of zirconium(iv)-based MOF (UiO-66) were encapsulated inside red blood cells (RBCs) [45]. The released O2 can increase the 1O2 production and intensify the PDT outcomes for hypoxic conditions.

2.4 Features of PS

PS are the high-level conjugated molecular systems that transfer their energy from the incident light into another nearby molecule. During this photochemical reaction, a series of ROS are formed that can cause damage to the cells [42]. An ideal PS should fulfill several requirements such as being easy to obtain in the pure form, less toxicity in the dark with minimal side effects, and having a high quantum yield with maximum light absorption capacity in the far-red/NIR region [51]. Nevertheless, it should be cost-effective, dissolve easily in the body fluids, and get quickly eliminated from the body. Moreover, the biomolecules (Hb) absorb light in a shorter range (<650 nm), considerably decreasing the amount of light that can enter deep tissues. Therefore, NIR light is opted to expose the PS to improve deeper tissue penetration [52]. According to their structural makeup, PS can be divided into organic and inorganic types. The organic PS are categorized into three stages: the first stage corresponds to the porphyrin derivatives, which are usually hydrophobic. Variations of the porphyrin’s structure yield the second-generation PS such as phthalocyanines (chlorine e6; Ce6) with improved tumor discrimination. However, the use of these PS is limited owing to the photoinduced degradation and dark phototoxicity. Therefore, investigations aiming to find PS with better tumor accumulation properties as the third-generation PS (nanocarrier-assisted PS) are underway [53]. Despite their versatility and innovative nature, nanocarriers offer distinct advantages and characteristics for drug delivery [54]. They are promising choices for improving the accuracy and efficiency of drug therapy in diverse medical applications because of their nanoscale size, controlled release kinetics, biocompatibility, individualized drug delivery, and long-term stability [55]. In addition to controlled release kinetics, nanocarriers provide other key features. The precision of these systems allows them to deliver drugs sustained for a prolonged period of time, making them ideal for persistent drug delivery [56]. A nontoxic approach is used throughout these systems, which minimizes any adverse effects on the body [56]. The immune response is reduced by this characteristic, which makes nanocarriers a safe method for delivering drugs. Several factors that can affect nanocarrier biodistribution, which can be divided into nanoscale properties, physiological factors, blood circulation times, tumor microenvironments (TMEs), and administration routes [57]. Recent research published by Sultan et al. demonstrated that targeted delivery formulations for cancer treatment have made significant progress [58]. This study was intensive on the characterization of chitosan nanoparticles surface linked to rituximab with cisplatin (mAbCCNP) and with cisplatin-loaded chitosan nanoparticles with cisplatin (CCNP). There were notable physicochemical differences among these formulations, with CCNP exhibiting a zetapotential (ZP) value of 30.50 ± 5.64 mV and a particle size of 308.10 ± 1.10 nm, while mAbCCNP had a ZP value of 26.90 ± 9.09 mV and a slightly larger particle size of 349.40 ± 3.20 nm. It is important to note that CCNP and mAbCCNP showed controlled delivery kinetics of cisplatin, which indicates they have potential as effective delivery systems [58].

These PS bear unique optical absorption features and high quantum yield in the triplet-excited state allowing them to generate their own ROS. Particularly, nanomaterials of inorganic origin showed inherent chemical constancy and attracted much attention in modulating the second-generation PS [59]. The photosensitive materials exhibited energy bands, which deliberated them with light-activated ROS generation. Owing to their phototoxicity, TiO2 nanoparticles may cause glioblastoma cancer cells to undergo programmed cell death. The mechanistic investigations showed the formation of electron-hole pairs as a result of the shift of electrons from the valence band to the conduction band of TiO2 under UV light irradiation, which might additionally interact with the adjacent O2 and H2O molecules to produce ROS [60]. In this direction, several PS based on inorganic nanomaterials such as the Si nanoparticles, SnWO4 nanoparticles, and CdSe quantum dots (QDs) have been utilized as PS in the PDT [27,61,62,63].

2.5 Comparative studies between the PS and nanoformulated PS

Traditionally, dye molecules and aromatic hydrocarbons have been used as PSs, such as xanthane-derived dyes (Rose Bengal and eosin) and methylene blue [64]. During the electronic transitions between different excited states of the dye, ROS is produced and monitored by electron transfer to oxygen [64]. In a light reactive species, there can be at least two excited states: a singlet and a triplet. As a consequence of radiation absorption, photosensitive species typically undergo electronic transitions between the ground state (S0) and singlet excited states (S1 or S2), although a subsequent transition to a triplet excited state (T1) may also occur, which is forbidden in principle. Intersystem crossing is the term used to describe such a phenomenon. It is possible to cross intersystems when there is an applicable energy change between the excited states and there are heavy atoms present in the PS [65]. Two mechanisms may occur when the electrons are in a triplet state: (i) they can be shifted to other species, forming free radicals, which can react with oxygen to produce ROS, including superoxide radical anions (O2), H2O2, and hydroxyl radicals (OH), as described earlier; and (ii) they can be transported to triplet oxygen (3O2) to form singlet oxygen (1O2), as described earlier. As with both mechanisms, ROS production is more efficient when the intersystem crossing is more probable.

The PS summarizes different structures with descriptions of their chemistry and physical properties, as well as their combination into nanoparticles/nanostructures [66]. In general, porphyrins are hydrophobic compounds composed of four interconnected transformed pyrrole subunits [67]. In addition to absorption of visible light, they are well known for having high levels of conjugation [68]. These molecules can also contain metallic atoms, resulting in natural chromoproteins like hemoglobin and chlorophyll. These compounds have high quantum yields (F) and a rich chemistry, resulting in relatively high ROS production [69]. It is actually possible to modify the original porphyrin structure, which can impact its optical properties such as its absorbance spectrum. A major drawback of these filters is photobleaching, low absorption in the IR or NIR spectral window, and poor selectivity [70]. Developments in nanotechnology have provided new opportunities and implementations. The majority of biomolecular interactions occur in nanostructures, which have a size of 1–100 nm. In particular, PSs have been conjugated or incorporated into nanostructures to enable their use in nanomedicine [71]. In addition to multifunctionality, second-generation PSs have the potential to deliver drugs directly to tumor cells with increased efficiency and selectivity in terms of intracellular delivery [36]. Recently, PSs have been conjugated with different nanostructures, including inorganic nanoparticles, micelles, and vesicles, especially liposomes, that have already been commercially developed [36]. Four generations of PSs use porous carriers like mesoporous silica and MOFs. In these structures, there is the possibility of incorporating a large number of sensitizer molecules [72]. Pharmacological formulations of PSs are also referred to as third- and fourth-generation PSs from a pharmaceutical perspective.

3 PTT using PTA

3.1 Mechanisms underlying PTT

The PTT is a method of heat ablation in which the PTA utilizes light energy of a longer wavelength. Therefore, PTT is capable of damaging the pathologic cells selectively at more penetration depth. In this procedure, the conversion of photon energy to thermal energy takes place [73]. However, an excess dose of irradiation may cause tissue burning and inflammation in the adjacent tissues. The mechanism of cancer cell death depends on the temperature generated as necrosis occurs at ≥50°C, whereas apoptosis ensues between 43 and 50°C. It is therefore necessary to maintain the temperature for apoptosis as necrosis is associated with a risk of cancer metastasis [74]. The strategies applied for selective PTT using PTA are shown in Figure 3.

Figure 3 
                  Strategies to improve selective PTT using PTA; adapted from ref. [74], under Creative Commons License.
Figure 3

Strategies to improve selective PTT using PTA; adapted from ref. [74], under Creative Commons License.

Recent investigations are now being directed toward using heat to lessen the undesirable side effects and increase the effectiveness of PTT [75]. Nanoparticles play a crucial role in PTT because of their characteristic properties making them exceptional PTA [76]. They not only have unique shape, size, and optical properties but also possess good photothermal efficiency that allows easy tumor penetration [77]. The improvement in radioactive absorption and scattering properties of metal nanoparticles (Au, Ag, and Cu) due to their distinctive surface plasmon resonance (SPR) phenomenon makes them suitable for PTT [77,78]. The nanoparticles derived from these metal ions show high densities of free electrons, which is revealed during the plasmon resonance in the visible region suitable for PT applications [79]. The SPR phenomena appear from the collective oscillation of free charge carriers in nanoparticles steered by the electromagnetic field of incident light. It has garnered great attention for its role in heightened optical phenomena and optoelectronic control [80,81]. Nevertheless, their outstanding radioactive scattering nature plays a pivotal role in imaging-guided PTT.

3.2 Features of PTA

Based on their highly effective photothermal conversion efficiency (PCE), PTA can outpace any absorption disturbance from the biological chromophores [82]. The currently used laser in PTT is NIR light (biological window = 750–1,350 nm), resulting in deeper tissue penetration. This wavelength range can be divided into two subwindows: the NIR-I (750–1,000 nm) and the NIR-II (1,000–1,350 nm). The NIR-II has a deeper tissue penetration, and fewer biological interferences as compared to the NIR-I, which has a short tissue penetration depth. Nanosized photothermal agents in this situation can not only achieve larger tumor accumulation than small molecules but also offer numerous imaging routes and advantageous features [82].

4 Advantages and drawbacks of phototherapy

When compared to conventional treatments, phototherapy has several advantages. First, by altering the irradiation site, intensity, and power, phototherapy may accurately manage the area, time, and effectiveness of the treatment. It is simple to swiftly modify the treatment plan as per clinical requirements. An effective treatment plan is ensured by the constrained irradiation area, low risk of adverse effects, and high energy output. Phototherapy has shown an increase in the life span and a decrease in the size of tumor in murine model [59]. Nevertheless, despite these benefits, single-mode phototherapy has limited therapeutic use. Tumor relapse, one of the major problems, which cost thousands of lives every year, cannot be solved by phototherapy, although it has shown promising therapeutic results in solid tumors in animal models.

In addition, tissues underlying the tumor also suffer some sort of damage as a result of the heat produced during PTT. Eventually, since lasers can only penetrate small and superficial tumors, the range of phototherapy is constrained. As a result, phototherapy does not result as well in clinical settings as it does in animal studies. The PDT is also associated with several practical issues including those related to light penetration depth, delivery and localization of PS, photodynamics, and photochemistry of PS, all of which are important factors for the precise generation of ROS. These issues are being investigated to develop a mechanistic approach that may solve the majority of PDT-related issues, starting at the molecular level to their biological consequences. As a result, the studies are being focused on how cellular death and vascular blockage can work well, and how ROS can stimulate the immune system following PDT. Finally, antioxidants, inorganic salts, and nanoparticles are utilized to enhance the ROS generation and eventual improvement of the therapeutic efficacy [83].

There are some hurdles identified in clinical practices associated with phototherapy for cancer. The immunological responses, hemolysis, and thrombogenicity are the major hurdles, which should be considered in clinical settings. Although cancer phototherapies can have several advantages such as minimal invasiveness and use of nonionizing radiations; however, major concerns revolving around PDA and PTT are their ability to prevent cancer relapse and limiting collateral damage to healthy tissues. It is therefore important to carefully investigate the specificity and selectivity of nanocarrier-based phototherapy in the treatment of cancers for maximized laser tissue penetrations.

5 Nanocarrier-assisted phototherapy

5.1 Nanocarrier-assisted PDT

The nanomaterial-based delivery technologies have been applied recently in the PDT that can alter biodistribution as well as the pharmacokinetics (PK) and pharmacodynamics (PD) of PS. The nanostructured materials generated using nanomedicines pose as an intriguing replacement for the conventional PDT since they allow PS to be transported and absorbed well, thereby improving their anticancer potential. Nanoparticle holds unique physiochemical and photoconverting properties that can convert exogenous NIR light into self-illuminating substances that can cause tumor tissues to produce their light. The advantage of taking nanoscale particles (1–100 nm), particularly known as upconverting luminescent nanoparticles (UCNPs) is that they can show anti-Stokes luminescent, which can tune the NIR light (spectral range in 710−1,100 nm) into visible light and thus to trigger PS via FRET. In this context, several PS have also been employed with UCNPs to build composite nanoplatforms for deep PDT [39].

Photogenerated ROS targets the tumor tissue through a photosensitization process leaving behind the surrounding tissue unaffected [84]. Based on the remarkable therapeutic outcomes, nanomaterial-based PDT has emerged as a favorable therapy for a variety of cancer forms (Figure 4). Previous investigations revealed that the nanocarrier-modified materials exhibited considerable interest in assembling next-generation PS owing to their intrinsic chemical stability. Other benefits of inorganic nanomaterials include their unique energy band, which enables them to produce ROS when exposed to light for further therapeutic usage [85].

Figure 4 
                  Applications of nanomedicine in the generation or depletion of ROS and their therapeutic and diagnostic implications. Reprinted with permission from ref. [31] Copyright 2019, American Chemical Society.
Figure 4

Applications of nanomedicine in the generation or depletion of ROS and their therapeutic and diagnostic implications. Reprinted with permission from ref. [31] Copyright 2019, American Chemical Society.

5.2 Nanocarrier-assisted PTT

Various nanoparticles based on inorganic, organic, and composite materials have been explored till date for their photothermal applications. These materials exhibit distinct features such as easier to synthesize, robust NIR absorption, and improved photostability [86]. Nevertheless, their clinical applications are hindered owing to their poor biodegradability, although organic nanoparticles, dyes, and polymers have shown better biocompatibility and biodegradability [25]. However, poor photostability and tedious synthetic methods are still associated with organic PTAs, which limit their uses in phototherapy. Therefore, new organic–inorganic composite materials are being developed to overcome such drawbacks, which are achieved through chemical conjugation and display better photoelectric properties in PTT [87].

Several forms of nanoparticles have been investigated for photothermal applications including the ferromagnetic nanoparticles, single-walled carbon nanotubes, multiwalled carbon nanotubes (MWCNTs), and polymer-based materials [88,89]. However, their use is still limited as PTA owing to their low biocompatibility, water insolubility, and elevated toxicity. Recently, gold nanoparticles (AuNPs) have shown good promise as PTA in PTT. Colloidal AuNPs have been reported to have localized plasmon surface resonance (LPSR), resulting in photoacoustic (PA) and hyperthermic properties useful for selective cancer targeting and medical applications in imaging [90]. By tuning the shapes of AuNPs (nanorods, nanocages, and nanostars) and size, the LPSR photochemical properties can be tuned. Consequently, the photothermal and PA properties of the material can be changed by using varied light wavelengths in the NIR area [91].

The AuNPs showed several advantages over other nanoparticles as they can be delivered efficiently into the local tumor site, activated through NIR laser with the possibility for deep penetration, and can be tempered for complicated cancer PTT and drug delivery systems [92]. Commonly used light-based therapeutic candidates in targeted cancer PTT are gold-silica nanoshells (GSNs). These GSNs (AuroShells) bear a diameter of ∼150 nm, maximally absorb the NIR light and convert it to heat [93]. Once, the nanoparticles accumulate in the tumor tissue, they undergo photothermal heating under the influence of NIR laser, resulting in hyperthermic cell death, while healthy tissue remains unaffected. The mechanism associated with cell death, mainly necrosis, is the high-energy irradiation that leads to a change in protein and lipid structure. On the other hand, apoptosis, which is typically brought on by low-energy radiation, has no impact on immune or inflammatory responses [94]. Therefore, with the use of AuNPs, a unique photothermal mechanism for selective cancer therapy can be envisaged.

6 Types of nanocarriers used in phototherapy

The PDT and PTT individually employ light as radiation, which helps achieve their therapeutic outcomes [95,96]. These therapeutic approaches gained much attention recently owing to the noninvasiveness of light and partial adversative consequences associated with these treatments. The nanoscale-based carriers could be helpful to simplify the collective treatment and can be utilized to achieve necessary developments in the area of photo-chemotherapy and photo-radiotherapy, improved PK, co-loading multiple agents, reducing toxicities, and sustained release [97100]. The nanoscale-based carriers are the materials that possess nano-dimensions and which are capable of carrying several imaging agents and drug molecules. These systems could be in the form of metal nanoparticles [97], polymers [98], graphene or graphene-based materials [99], as well as non-metals, such as silica nanoparticles [100]. These nanomaterials have numerous applications in the field of novel drug delivery systems, gene delivery, imaging, and phototherapy [101103].

The target-based mechanism of nano-entities depends on the improved EPR effects that are supposed to take place when the tumors influence a specified size, and allow the formation of blood vessels that repeatedly seem to be leaking [104]. The accumulation of substantial elements is further enhanced by the absence of lymphatic drainage in certain cancers. The EPR effect is frequently raised as an inactive pointing in divergence to active targets that employ targeting vectors, such as peptides or antibodies [105]. The EPR outcome was confirmed in several preclinical trials; however, the trial results on human volunteers have been extensively uncertain. One of the reasons for the conflict might be the multiplicity of tumors of the same type. There are various factors such as tumor interstitial fluid pressure, degree of angiogenesis, and lymph angiogenesis, which might vary from tumor to tumor, resulting in relatively diverse consequences [104107]. PTT is generally inadequate to exterminate the tumor completely, and combining it with chemotherapy can be more efficient than the individual therapy. The use of nanoscale-based carriers can permit the integration of both therapies effectively. The nanocarrier systems utilized for photo-based thermal therapy and chemotherapy are divided into three major classes: (i) gold (Au)-based materials, (ii) carbon/carbon-based materials, and (iii) other inorganic materials.

6.1 Gold (Au)-based nanomaterials

The gold (Au) nanodimensional materials have been utilized in diverse biomedical applications owing to their good biocompatibility and the feasibility to fabricate the nanostrcutures in adaptable shapes and sizes [108,109]. The main advantage of AuNPs is their special SPR, which is quite effective in biomedical applications [110]. These optical characteristics and absorbance of photon-based light in Au nanoparticles might possibly be shifted to the anticipated wavelength. The AuNPs generally display two SPR emission modes, the first is initiation from the transverse mode (∼520 nm) and another is from the longitudinal mode [111]. The position can be contingent on the characteristic ratio and could be shifted toward the NIR region [112]. Principally in the field of phototherapy photon-based absorption, ∼800 nm wavelength is required for deeper tissue penetration [113]. The fabrication and functionalization of AuNPs and their utilization in imaging, diagnostics, and medicine have been reported previously [114]. The biomedical applications of AuNPs owing to their immunological properties have also been described [115]. The biomedical applications of AuNPs include early-stage diagnosis of tumors and their treatment. The shape and morphology-dependent fabrication of AuNPs were achieved successfully for the precise tumor analysis and treatment. The expected properties of AuNPs ought to be scientifically and intensely understood [116]. The applications of AuNPs can be divided into two major categories: physical and chemical, as shown in Figure 5.

Figure 5 
                  Applications of AuNPs in the tumor diagnosis and its treatment, adapted from ref. [116] under Creative Commons license.
Figure 5

Applications of AuNPs in the tumor diagnosis and its treatment, adapted from ref. [116] under Creative Commons license.

6.2 Carbon/carbon-based materials

A variety of carbon and carbon-based materials such as carbon-dots, carbon nanotubes, and fullerenes have been considered for biomedical applications [117119]. Both graphene and carbon nanotubes are low-dimensional sp2-hybridized carbon-based materials that exhibit unique physical and chemical characteristics making them useful in a wide range of applications including nanomedicine [120]. Among them, carbon nanotubes and nanoscale graphene showed maximum absorption of NIR light and are consequently being considered as nanoscale carriers for PTT and as drug delivery vehicles for the intracellular transfer of genes, proteins, and chemotherapeutic drugs [121]. The carbon nanotubes are classified into single-walled and MWCNTs and are being considered owing to their high surface area, resulting in unmatched biodistribution effects and PK [122]. The carbon nanotubes, like other carbon-based materials, require to be functionalized to improve their dispersion in aqueous media and allow them to be attached to the target-based vectors. A recent study on carbon nanotube structure is reported where QDs-based MWCNT nanocomposites were fabricated successfully for photothermal applications [123]. The effect of intratumoral injection of the prepared nanocomposites was examined in the A549 cell line-induced tumor-bearing nude mice, and the results revealed successful tumor inhibition.

The nanosized graphene and graphene oxide chemically functionalized with other materials have also been considered efficient carriers for the delivery of drugs including the bio-inert materials owing to their high loading efficiency [124]. In addition, nanosized graphene oxides act as photothermal agents, which can absorb NIR light in its compact form. In most of the cases, an ∼808 nm NIR laser is employed for the photoactivation. The PEGylation of the graphene nanosized particles is generally anticipated to improve the circulation time, further enhancing the tumor uptake and delivery of anticancer drugs [125]. The transferrin-coated nanosized graphene oxide was also able to penetrate the blood–brain barrier (BBB) and attack glioblastoma tumors. This specified that the transferrin simplified the permeation of nanoparticles over the BBB [126].

The following categories can be used to categorize photothermal agents in general [127]: i) organic dyes, including indocyanine green and heptamethine cyanine; ii) organic nanoparticles, such as porphyrin–lipid conjugate porphysomes and organic semiconducting polymeric nanoparticles; iii) gold nanomaterials; iv) single-walled/multiwalled carbon nanotubes and graphene oxide, and v) other inorganic nanoparticles such as metal oxide and QDs [128]. Photothermal agents such as organic nanomaterials are frequently biocompatible and biodegradable. However, they suffer from inherent constraints such as poor photothermal stability, poor PCE, and complicated synthesis [87]. The intrinsic optical properties of inorganic nanomaterials typically provide excellent NIR light absorption, high photothermal competence, and high photostability compared to organic nanomaterials. Among the benefits of localized SPR (e.g. gold nanomaterials), narrow emission spectra, and structural characteristics are the ease of synthesis and the ability to modify surfaces with functionalization, such as carboxylic, hydroxyl, and epoxy groups (e.g., carbon nanomaterials) [129].

6.3 Other inorganic and organic materials

These include the nanomaterials, where their physicochemical properties can be ascribed to the presence of inorganic materials such as metals [130]. The term inorganic nanoparticles is comparatively newer as this class was established a few decades ago and their biomedical applications have only been identified recently [131,132]. The inorganic nanoparticles consist of two parts: a core, which comprises metals such as in AuNPs, QDs, and iron oxide nanoparticles, and a shell, which comprises organic polymers or metals that shield the core from chemical interactions or assist as a substrate for coupling with biomolecules [133135]. The utilization of inorganic nanoparticles offers numerous advantages such as the target-based release, improved stability, and increased solubility of the drugs. The additional advantage that is relatively significant is that the remarkable penetrability of nanoparticles makes them particularly useful for the treatment of cancer [136]. In general, organic phototherapy agents are lipophilic; thus, co-precipitation is a simple method of achieving biological applications [137]. However, for water-soluble phototherapy agents such as cyanine dyes, it is challenging to encapsulate them into nanoparticles by co-precipitation approach [137,138]. At present, the preparation of water-soluble phototherapy agents into liposomes could be a better alternative [139]. In addition, liposomes can be designed to release drugs thermally when photothermal agents are trapped within them, which is a common strategy for designing smart phototherapy materials. Organic dyes and photothermal nanoagents can be used to improve the photothermal effect, including metallic nanoparticles, nanocarbons, and metal oxide nanomaterials [140,141]. An effective PTT system has been constructed with several kinds of organic materials. In addition to their superior PCE, graphene quantum dots (GQDs) also have an incomparable morphology and are simple to functionalize [142]. The chemotherapeutic drugs can be delivered inside cancer cells by pH-sensitive GQDs [143]. Upon laser irradiation and acidification of the intracellular environment, the nanocarriers released doxorubicin (DOX) [144].

Several inorganic nanomaterial-based drugs are in the clinical phase and are being tested for their application in the treatment of various cancer forms, and a few of them are summarized in Table 1.

Table 1

Inorganic nanoparticle-based formulations in clinical stages utilized for the treatment of cancer

S. No. Formulations Nanoparticles used Applications Clinical phase Ref.
1 NK105 Micellar nanoparticles Breast cancer Phase II [145]
2 AuroLase AuNPs Neck cancer and lung cancer Phase I [146]
3 Docetaxel PNP Polymeric nanoparticles Advanced solid malignancies Phase I [147]
4 Carbon dots or C-dots PEG-coated SiO2 NPs Melanoma Phase I [148]
5 CYT-6091 AuNPs Breast cancer and pancreatic cancer Phase I/II [149]
6 Combidex Iron oxide nanoparticle Imaging of tumor Not available [150]

QDs are semiconductor nanoparticles with inimitable photonic/optical possessions, owing to their quantum effect and size effect [151]. These are ideal for drug delivery applications owing to various intrinsic properties such as high loading capacity of drugs, no adverse drug reactions reported, good biocompatibility and low toxicity, prolonged residence time as shown in the in vivo studies, appropriate particle size and shape ideal for the delivery of drugs, and improved stability [152]. The QDs are signified by inorganic nanoparticles such as CdS, CdTe PbS, and ZnS; however, the frequently used QDs system contains the inner semiconductor core consisting of CdSe, which is covered with the ZnS outer shell [153]. Owing to their inimitable properties such as resistive behavior to photo-based bleaching, powerful and stable fluorescence for a longer period, high sensitivity, and precision, QDs are considered a novel class of biosensors, which can be utilized for the early-stage detection of cancer [154]. In a previous study, QDs were prepared, and the emission spectrum of scanning multiphoton microscopy was utilized to investigate the tumor cell eruption in animals and the cells labeled with QDs intravenously inoculated into mice. It was reported that the QDs and spectral imaging permitted the instant recognition of five diverse types of cells with multiphoton laser excitation [155]. In conclusion, nanoparticles from organic or inorganic materials are promising carriers for the delivery of drugs in cancer therapy and can be further utilized for clinical applications. The nanoparticle-based drug delivery system proves to be superior to the conventional systems for the reason that they can reduce the general side effects, which the patients undergo during chemotherapy, by delivering the same cytotoxic levels of drugs at the tumor site.

7 Phototherapeutic nanomedicines for the diagnosis and treatment of cancer

7.1 For cancer imaging

In line with the recent advancements in the individualized cancer treatments, more advanced imaging techniques for cancer diagnosis must be developed. Further, the relevant clinical studies appear to be trending toward the effective integration of diagnosis and treatment of cancer. Phototheranostic nanomedicines are especially well suited to bridge the gaps between the diagnosis and therapy of cancer. To improve medical imaging quality, numerous contrast materials have also been developed using NPs. Imaging technologies such as fluorescence imaging, MRI, PA imaging, computed tomography, and positron emission tomography have been effectively playing crucial roles in cancer therapy in conjunction with advanced nanoplatforms. Fluorescence imaging, which utilizes the excitation characteristics of fluorophores, has advanced rapidly in recent years as the most cost-effective imaging technique. Because many phototherapeutic agents are fluorescent, NPs containing these components (for example, NIR dyes, SPNs, and QDs) may perhaps be used as fluorescence contrast compounds for cancer imaging in real-time greater spatial resolution fluorescence imaging [42,156,157]. Examples of phototheranostic nanoplatforms developed so far are summarized in Table 2.

Table 2

Treatment and diagnosis of cancer using phototherapeutic nanoplatforms

S. No. Types of nanoplatforms Nanomaterials features Treatment Application Techniques Ref.
1 Inorganic nanoparticles Featuring optical, thermal, and electrical conductivity, as well as magnetic and catalytic properties, SPR and photothermal materials may exhibit long-term circulation in vivo and toxicity issues Cancer imaging and therapy Drug carriers, imaging tools, therapy agents, and functional coatings that are effective against cancer Photodynamic therapy and hyperthermia [150]
2 Lipid-based nanoparticles Biocompatible, biodegradable, and amphiphilic drug colloidal carriers Enhancing the antitumor activity of several chemotherapeutic agents Drug delivery in nanocarriers controlled and modified, preventing degradation of hydrophilic and hydrophobic drugs ME cold dilution [158]
3 Protein nanoparticles Biocompatible, biodegradable, and low immunogenic structures; FDA-approved drugs; easy functionalization Breast cancer with metastatic spread Treatment and imaging of cancer Genetic recombination [159]
4 ZnO QDs conjugated with gold NPs Unique physicochemical properties, porous structure, and biodegradability Delivery of drugs targeted at tumors Platform for delivery of PDT, PTT, imaging, PA imaging, radiotherapy, and phototherapy Electrochemical [160]
5 Mesoporous ZnO nanofibers (ZnOnFs) Biocompatible, biodegradable, and unique physicochemical properties Breast cancer Targeted and sustained delivery of bioactive and chemotherapeutic anticancer drugs; specific toxicity via reactive oxygen species generation Electrospinning [161]
6 AuNPs High photostability and photoluminescence in a one-dimensional, SPR effect Therapy for breast cancer It has excellent plasmonic materials, with adjustable energy regulation, bio-receptor immobilization, improved analyte loading, and strong catalytic properties Photothermal [162]
7 Silver nanoparticles (AgNPs) A one-dimensional, unique physicochemical property, including optical, thermal, and electrical conductivity, as well as resistance to viruses, fungi, and bacteria Antitumor agents Bioreceptors are immobilized, the analyte is loaded better, and the catalytic properties of the enzyme are strong Imaging in biomedicine [163]
8 AuNPs/graphene oxides The SPR effect, the strong absorption of NIR light, the high surface area, and the high photostability and photoluminescence of the material Therapy for breast cancer Photothermal imaging and drug carriers that immobilize bioreceptors, improve analyte loading, and demonstrate good catalytic properties NIR-activated PTT [164]
9 Ag−Au nanostructure Effects of SPR and photothermal radiation Therapy for breast cancer Immobilization of bioreceptors, radiology, phototherapy, and improved presence of analytes in PA imaging and Raman spectroscopy imaging NIR PTT [165]
10 Polymeric nanoparticles Easy to modify and biocompatible Cancer therapy Intelligent drug responses, functional coatings Chemotherapy [166]

In contrast to the NIR-I fluorescence imaging, NIR-II fluorescence imaging is favored for having a higher spatial and temporal resolution, which inhibits autofluorescence in the corpora. As a result, researchers sought to include NIR-II fluorescent dyes in phototherapeutic NPs to achieve the desired NIR-II fluorescence [167]. For MRI to work, water protons’ magnetic dipoles must be able to align when subjected to a strong magnetic field. In previous studies, the related physical principles, picture capture, and processing have been explored [167,168]. Similarly, MRI is recognized among imaging modalities for its dominance in soft tissue contrast and ability to offer more information on tissue function, structure, blood perfusion, and many more. Because of their magnetic inhomogeneity, super-paramagnetic iron oxide nanoparticles (SPIONs), which have been approved by the FDA, can be used as MRI contrast agents for medical diagnostics and rehabilitation [169]. Table 2 summarizes the application of various nanomaterials in targeted cancer therapy and imaging by incorporating gold and AgNPs into the phototherapeutic nanosystems. Metal ion–chelated MRI contrast agents have also garnered a lot of interest recently. As a result, gadolinium(iii)-based contrast agents (GBCAs) are utilized in over 40% of all MRI examinations, accounting for approximately 40 million GBCA administrations annually. The function of phototherapeutic nanostructured materials for MRI is achieved by chelating Gd(iii) into phototherapeutics or nanocarrier materials [170].

The PA imaging is a novel hybrid imaging technology that combines optical and ultrasonic imaging to increase the image depth to many centimeters [171]. The short-pulsed laser first stimulates the intrinsic or exogenously supplied light absorber at the target spot, which then partially transforms the energy into heat via vibrational relaxation during the PA imaging process. Thermoelastic expansion creates sound waves, which an ultrasonic transducer captures for three-dimensional reconstruction. Consequently, the PA images are produced based on the arrival timings of sound waves at the transducer. Unlike photons, phonons are not widely distributed in biological tissues. Furthermore, the scattered photons produced during PA imaging may aid in the creation of PA waves [172]. Therefore, PA imaging utilizes both high-resolution acoustic imaging and high-contrast optical imaging and has been used clinically to detect prostate and breast cancer [173], and tumor metastases [173,174]. It also finds applications in therapeutic monitoring and endoscopic gastrointestinal imaging [175]. PA imaging can be performed without contrast media, for instance, endogenous Hb and melanin, which may produce PA signals; however, most biological tissues have intrinsically low contrast due to weak NIR absorption. External PA imaging compounds with higher adsorption coefficients and tumor selectivity, notably NPs absorbing light in BWs, have been developed to expand their usage in disease diagnostics [176].

Gold nanoclusters are typical PTT agents for PA imaging [177], and other than this, CNTs [178], reduced graphene oxide [179], UCNPs [180], QDs [181], small-molecule dyes [182], SPNs [183], and melanin [184] are also developed. PA agents efficiently convert light to heat for the PTT. For example, GNRs have outstanding and tunable optical characteristics making them suitable for PA imaging [185,186]. Recently, a new type of tiny GNRs was disclosed in a study, which was found to improve the PA contrast when compared to regular-sized GNRs [186]. The seedless technique was employed to manufacture micro GNRs with absorption in the NIR-II region, having smaller size than the conventional ones with the same aspect ratio (length/width). Miniaturized nanorods are remarkably more stable and can create a robust PA signal when illuminated by a nanosecond pulsed laser than the conventional GNRs. Smaller GNRs improved drug delivery by 30% and elicited greater PA signals in tumor-bearing mice. The theoretical and numerical analysis concluded that the PA signal affects GNR absorption and surface-to-volume ratio.

Owing to the advantages, such as their adaptability to change, high biocompatibility, and relatively low cost, organic PA agents, particularly small molecule dyes, have received much attention [187,188]. Their weak photostability, however, precludes future utilization. SPNs have also gained much interest as a different type of PA agent due to their great photostability and carefully controlled optical characteristics [184,189]. A recent study concentrated on developing SNP-based NIR-II absorbing compounds as PA agents to lessen the tissue absorption and light scattering while performing PA imaging [183]. Notably, PA cavitation could be employed to cause mechanical damage to target tissues, which would have a therapeutic effect. To overcome the challenges of treating glioblastoma, a dendrimer-based NP with high red absorbance and efficient BBB penetration in tumor tissues was developed [190]. Adenosine receptor on the BBB may promptly be activated by the presence of 4-[2-[[[6-amino-9-(N-ethyl-a-d-ribofuranuronamidosyl)-9H-purin-2-yl]-amino] ethyl] benzene propanoic acid hydrochloride (CGS) on the surface of NPs to promote self-accumulation in the tumor (Figure 6). In addition, the NPs used PA to convert pulsed laser light into a shockwave, creating a targeted antitumor impact [42]. More importantly, NP-mediated PA could reveal the tumor depth, shape, and vascular architecture.

Figure 6 
                  (a) Preparation of the Den-RGD/CGS/Cy5.5 nanoparticles and (b) utilization of Den-RGD/CGS/Cy5.5 in PA precision glioblastoma therapy. Reprinted with permission from ref. [190], Copyright 2019 WILEY‐VCH Verlag GmbH & Co.
Figure 6

(a) Preparation of the Den-RGD/CGS/Cy5.5 nanoparticles and (b) utilization of Den-RGD/CGS/Cy5.5 in PA precision glioblastoma therapy. Reprinted with permission from ref. [190], Copyright 2019 WILEY‐VCH Verlag GmbH & Co.

The NPs were injected into murine models through their tail veins and after the brain inoculation phase, animals were subjected to pulsed laser energy to initiate PA treatment. The brain glioblastoma arteries prevalent in the tumor periphery were overexpressed with the protein v3 integrin and were the target for the developed nanoparticles. The connection between the agonist, CGS, and the A2A adenosine receptor (A2AAR) on the vascular endothelial cells was made feasible by increasing the local concentration of NPs. The BBB was made permeable by the CGS present on the NPs for a sufficient period to allow a large number of NPs to enter the glioblastoma. Since the v3 integrin is also widely expressed in tumor cells, the NPs directly targeted the glioblastoma once they crossed the BBB. The developed NPs absorbed the light energy from the pulsed laser application to create a PA shockwave, causing local mechanical injury to the tumor cell. The PA image produced by the pulsed laser application is displayed in Figure 6 on the bottom left-hand side. This image contained data about the shape of the vasculature and the depth of the tumor [190].

7.2 For cancer stem cells (CSCs)

Genetic mutations are one of the root causes of cancer at the molecular level. Cells can resist many common cell cycle processes, most notably cell cycle arrest, due to mistakes in DNA instructions. Still, CSCs can develop tumors. These CSCs originate from healthy stem cells that have changed genetically, epigenetically, and de-differentiated from somatic tumor cells. These cells make up around 1% of the tumor tissues. In addition, it has been shown that these CSCs, which exhibit great propensities for self-renewal and multiplication, are in charge of the development, upkeep, spread, and recurrence of tumors. The CSCs possess several distinct characteristics that enable them to endure standard cancer treatments including radiotherapy and chemotherapy [191]. As a result, other recently developed therapeutic modalities, such as phototherapy, have served as an alternative strategy for the treatment of CSCs [192]. In a previous study, ionizing radiation (IR) and PTT treatment of patient-derived xenografts were combined, and the results showed that mild hyperthermia (42°C) via AuroLase therapy could sensitize the breast cancer stem cells (bCSCs) to IR [193]. This might be because PTT altered the expression of heat shock protein (HSP) in bCSC and reduced their ability to repair double-stranded breaks in the DNA due to the IR radiation. In addition, another study was performed to assess the PTT’s ability to prevent metastasis following initial tumor treatment by inhibiting bCSCs via highly crystalline iron oxide NPs [194]. It was reported that the PTT mediated by nano agents had the potential to increase the destruction of CSCs, enhance breast cancer survivorship, and offer an alternative therapy for patients with metastatic cancer who can no longer be treated.

7.3 For cancer immunotherapy

Various approaches for cancer immunotherapy, particularly immune checkpoint inhibition and development of anticancer vaccines, have now been investigated extensively [195198]. Yet, a long journey awaits before the single-cancer immunotherapy could be used to cure both primary and distant cancers. This is mostly because it is difficult to find the most appropriate biomarkers and because each patient is different from the other. Combining two or more therapies to achieve a synergistic therapeutic outcome is a promising strategy to further enhance the therapeutic effect [199], in which the phototherapy-synergized cancer immunotherapy has been identified as a key approach [200,201]. The activation of immune cells’ memories and the promotion of the antitumor immune response have both been linked to the phototherapy. Understanding the PDT/PTT-assisted tumor immunotherapy mechanism is important as it could lead to an immune cell cascade impact in the TME [202]. PDT/PTT would cause a wide variety of antigens to be produced from tumor lesions, which could then enter the bloodstream. It is vital to consider, nevertheless, that the amount of laser radiation also affects the immune system cells and damages the nearby tissues. It is preferable to execute PTT at a relatively low temperature (41–47°C) by adjusting the laser irradiation dose as hyperthermia (>50°C) generated by PTT can potentially cause inflammatory disease and heat damage to neighboring normal tissues [196,203]. To lessen the impact of these immunosuppressive substances, immunosuppressive inhibitors must typically be administered [196]. Several new combinatorial techniques have been adopted using phototheranostic nanomedicines for the phototherapy-synergized cancer treatment resulting in promising immunotherapeutic outcomes.

7.3.1 PTT-synergized cancer immunotherapy

Synergistic outcomes were observed when the PTT was combined with cancer immunotherapeutics. Tumor ablation induced by the photothermal heating during PTT-mediated cancer immunotherapy may make it easier for the immune cells to reach the TME, which is crucial for an antitumor immune response. It is also important to note that the best temperature range for PTT cancer immunotherapy is between 39 and 45°C, as higher temperatures can hasten cell necrosis, which can impair the production of HSPs and other immunostimulatory factors [204]. PTT-synergized immunoadjuvant treatment, among other approaches, could promote lymphocyte recruitment, resulting in improved cancer immunotherapeutic efficacy [205207]. For instance, IFN promotes MHC I expression in antigen-presenting cells (APCs), while also having a detrimental impact on cancer cell growth and angiogenesis. As a result, a pertinent study using CpG-encoded structured DNA-conjugated GNP nanogels was conducted to boost splenocyte IFN production during PTT [207].

The cargo-free photothrombotic nanomedicine for PTT-synergized immune adjuvant therapy was created using an innovative method to encapsulate PTT agent ICG inside the immune adjuvant OVA in addition to the inorganic NPs [208]. As shown in Figure 7, the OVA-ICG nano vaccines were produced with high antigen-loading efficiency (80.8%) and ICG-loading content (19.2%) through simple mixing of ICG and OVA [42]. In addition, the formulation OVA-ICG did not affect the PCE of ICG. According to the findings, immature DC 2.4 cells were stimulated to secrete IL-6 and TNF-a in vitro, indicating a successful immunostimulant. OVA-ICG injection followed with 808 nm laser irradiation reduced the melanoma tumors in vivo.

Figure 7 
                     Preparation of OVA-ICG nanovaccines and their role in DC stimulation/tracking, photothermal-immunotherapy, and tumor prevention. Reprinted with permission from ref. [208], Copyright 2018 WILEY‐VCH Verlag GmbH & Co.
Figure 7

Preparation of OVA-ICG nanovaccines and their role in DC stimulation/tracking, photothermal-immunotherapy, and tumor prevention. Reprinted with permission from ref. [208], Copyright 2018 WILEY‐VCH Verlag GmbH & Co.

Immune checkpoint inhibitors were also combined with the PTT, and the results have been promising so far [209212]. In one such study, PEGylated SWNCTs were developed and utilized as PTT agents to heat the tumor cells [212]. Tumor cells generated immunostimulatory mediators such as tumor-associated antigens, HSPs, and inflammatory cytokines upon exposure to an 808 nm NIR laser light. These immunostimulatory mediators caused dendritic cells to mature, which then attracted the tumor-specific CD8+ T lymphocytes. In addition, by including anti-CTLA-4 blocking therapy in the therapeutic process, the suppressive function of Treg cells was effectively suppressed, improving the ratios of CD4 and CD8 cells to Treg cells. Notably, CD20 tumor-infiltrating B lymphocytes may favorably influence the facilitation of tumor-specific antigen presentation. As anticipated, the animals receiving PTT-synergized immune checkpoint inhibitor therapy showed improved tumor growth inhibition.

7.3.2 PDT-synergized cancer immunotherapy

Similar to PTT, PDT may also trigger potent antitumor immune responses through a related mechanism. To actualize the PDT-synergized immunoadjuvant therapy, immunoadjuvants (such as GM-CSF, OVA, and CpG) were initially linked to the photothermal nanosystems. In addition, certain immunogenic substitutes for the aforementioned immune adjuvants have been reported by various studies to support the PDT’s ability to elicit an immunological response. For instance, proapoptotic cells often produce the Ca2+-binding protein, calreticulin (CRT), which in the lumina of the endoplasmic reticulum may move to the cell surface acting as a phagocytic signal as the cell undergoes apoptosis [213,214]. To enhance the anticancer effects of phototherapy, UCNP-based antigen-capturing nanosystems (UCNP/ICG/RB-mal-NPs) were developed, and the findings suggested that the tumor-derived protein antigens, in particular, CRT and phototherapy-treated tumor cells, could be stopped and kept, thereby improving the APC tumor antigen absorption and presentation [215]. Another new class of immune adjuvants called synthetic long peptides (SLPs) with immunotherapeutic effects have been developed as cancer vaccines for various cancer forms. The SLP and bremachlorin-mediated PDT were combined in a study to improve the percentage of CD8+ T cells and the effectiveness of immunotherapy [216]. Over 30% of primary and distant cancer forms were successfully treated by the SLP-PDT vaccination, and it outperformed the single SLP vaccine (20%) and PDT (0%), indicating that systemic immunity can be acquired.

The therapeutic nanosystems for PDT-synergized immune checkpoint inhibitors are also being studied [217221]. With the help of siRNA silencing, PD-L1 was found to be downregulated in the POP micelle, a multifunctional micelleplex nanoplatform [217]. The proinflammatory cytokines and the attraction of cytotoxic CD8+ T cells were made feasible by the efficient induction of the adaptive immune response. Although PDT or PD-L1 siRNA silencing alone only suppressed around 73 or 65% of the relevant tumor development, respectively, PD-L1 immune checkpoint blockade therapy combined with PDT was able to completely eradicate the tumor. Indoleamine-2,3 dioxygenase (IDO), an intracellular enzyme that is overexpressed in the TME of many malignancies, has recently been identified as another intriguing immune checkpoint modulator [222]. Tryptophan can be converted to kynurenine in the presence of IDO, which causes cytotoxic T cells to become “starved” and activates T regulatory cells. As an alternative strategy for immune checkpoint inhibition, numerous IDO inhibitors have been discovered [223]. PpIX-1MT-NPs were reported to have self-assembled using the amphiphilic chimeric peptide PpIX-1MT [224]. To create the molecular structure of PpIX-1MT, 1-methyltryptophan (1-MT), an inhibitor of IDO, was first linked to the C-terminal of a peptide that activates caspase-responsive peptide. The PS PpIX was then further linked to the N-terminal of the peptide via a PEG segment and palmitic acid. According to the findings, the 1-MT release behavior of PDT-induced apoptotic cells treated with PpIX-1MT-NPs in the presence of caspase-3 was consistent and quick, reaching up to 83% over 50 h (Figure 8) [42]. The flow cytometry results indicated that the immune response was activated during PDT because the exposure to CRT was increased after PDT. Meanwhile, the serum and spleen showed a superior ratio of CD8+ T cells/CD4+ T cells. In a CT26 metastatic murine model, the researchers further demonstrated the efficacy of the therapy of primary and secondary CT26 tumors.

Figure 8 
                     (a) The EPR effect allows the chimeric peptide PpIX-1MT to concentrate in the tumor, activates CD8+ T lymphocytes through cascade activations, and reduces the initial tumor and lung metastasis; (b) In situ PDT in the primary tumor resulted in caspase-3 synthesis, 1MT release from PpIX-1MT nanoparticles, and death of tumor cells. Reprinted with permission from ref. [224], Copyright 2018, American Chemical Society.
Figure 8

(a) The EPR effect allows the chimeric peptide PpIX-1MT to concentrate in the tumor, activates CD8+ T lymphocytes through cascade activations, and reduces the initial tumor and lung metastasis; (b) In situ PDT in the primary tumor resulted in caspase-3 synthesis, 1MT release from PpIX-1MT nanoparticles, and death of tumor cells. Reprinted with permission from ref. [224], Copyright 2018, American Chemical Society.

PS and other drugs have low tumor penetration, although the PDT can boost immune function and is a viable method for combining various treatments. Thus, their therapeutic efficacy was underutilized. To implement the use of PDT and immunotherapy together, as study was performed where a hyaluronidase-sensitive size-reducible vehicle (mCAuNCs@HA) coated with RBC membranes (RBCm) was developed. In this approach, the PS pheophorbide A (PheoA), ROS-responsive prodrug PXTK, and anti-PD-L1 peptide were loaded onto the inner NPs, which had the ideal size of 150 nm, when the original vehicle got to the cancer cells. By combining PDT, immunotherapy, and chemotherapy, this technique could remarkably improve antitumor and antimetastatic efficiency owing to its increased penetration capacity [42].

8 Recent developments in nanomaterials for cancer theranostics

The ever-evolving nature of cancer, TME, and other complexities such as biological barriers, and multi-drug resistance limits the efficacy of antitumor drugs and has made cancer research a topic of extensive investigation. Over the last two decades, a steep rise in the cancer-related research, particularly in the development of theranostic nanoparticles for effective cancer treatments, has been observed. The idea of integrating nanomaterials with phototherapies including PDT and PTT is an innovative emerging strategy in cancer nanotheranostics. This might be owing to the advantages associated with PTT such as noninvasiveness, high specificity, strong anticancer activity, and fewer side effects to the normal tissues. The combination of nanotechnology with phototherapy results in synergistic anticancer effects; however, despite all the advantages associated with phototherapeutics, their ability to cause phototoxicity is still a challenge [225]. The nanomaterials used in the phototherapy for cancer theranostics can be divided according to their mode of action.

8.1 Deep tissue penetrating nanomaterials

Recently redox-responsive polyethyleneimine/tetrahedral DNA/DOX nanocomplexes (NCs) were prepared to achieve deep tumor tissue penetration and overcome the issues of MDR. In xenografted drug-resistant tumor murine models of breast (MCF-7/R) and ovarian (SKOV3/R) cancer, the prepared NCs displayed greater tumor penetration and therapeutic potential [226]. Reverse nanomicelles of azobenzene-functionalized interfacial cross-linked loaded with 5-fluorouracil (5-FU) were developed, which could balance the aggregation and penetration of 5-FU in the tumor [227]. A black phosphorous QDs-based DNA-gel nanoparticle (NP) formulation allowed particles to penetrate the cells and was found safe for cancer treatment [228]. Biodegradable amphiphilic gelatin-wrapped NCs comprising DOX and copper sulfide-loaded dendrimers showed dual cell-tissue penetrability and effective deep-tissue penetration in chemo-phototherapy [229]. A biomimetic upconversion nanosystem delivered glucose oxide (GOx) to tumor areas via homotypic targeting to selectively starve cancer cells and generate H2O2. This starvation-phototherapy cascade was regarded as a smart and simple multimodal cancer therapy [230].

The IR-780 and GOx-based polylactic-co-glycolic acid nanospheres penetrated in 3D tumors and selectively accumulated in mitochondria at the same time, suggesting a synergistic treatment approach of phototherapy and enzyme-induced starvation therapy under dual-imaging guidance, making these nanospheres a promising nanocarrier for cancer theranostic applications [231]. Nano-scintillator-mediated X-ray-induced PDT could cure deep-seated cancers [232]. Two-photon lasers and X-ray irradiators increased the tissue penetration for deep PDT [233]. X-ray-excited theranostics help overcome the light penetration and tissue attenuation issues [234]. Due to its deep tissue penetration capabilities, NIR light-detonated phototherapy has garnered much attention in the treatment of cancer. Because of low tissue autofluorescence and deep tissue penetration, NIR imaging proved to be a promising MDR monitoring method. MDR cancer molecular NIR imaging required stable biomarker probes with excellent specificity and affinity [235]. Gliomas are very infiltrative and difficult to remove; however, a biomimetic catalase-integrated albumin phototheranostic nanoprobe could accurately guide glioma excision [236]. A recent approach, wherein organic phototheranostic nanomedicines with optimized NIR biological transparent window (700–900 nm) were found to be desirable for multimodal cancer therapy [237]. A novel polymer encapsulated sorafenib and chlorin e6 NPs were designed for efficient tumor treatment by accumulating in the tumor as a result of the EPR effect postintravenous administration in vivo [238]. For the diagnosis and treatment of cancer, novel IR808 dye-sensitized glutathione cladded Au-Bi bimetallic NPs were prepared to enhance the inhibition effect of tumors. The in vitro and in vivo results proved its excellent ablation effects on cancer cells [239]. Another recent magneto-thermodynamic strategy for deep-seated tumors was reported in the literature, wherein this novel design triggered free radical generation and exhibited significant therapeutic efficacy for orthotopic liver tumors in a rat model [240].

8.2 Target-specific nanomaterials

Conventional PDT is associated with problems in delivering the PS to tumor locations. A novel approach to overcome the problem is the development of a multifunctional drug delivery system of two or more NPs for efficient cancer therapy. Recently, photothermally active polydopamine NPs were formulated for loading chemotherapeutic drugs and targeting cancer cells. These NPs were first functionalized with polyamidoamine dendrimers followed by the conjugation with polyethylene glycol and folic acid targeting moieties, and finally, DOX was absorbed on the surface of the particles. The nanosystems demonstrated high NIR PCE [241]. For targeted PDT, nanosheets of the iodo-BODIPY-biotin conjugate as a PS showed a high NIR extinction coefficient, high singlet oxygen efficiency, and a surface-targeting ligand [242]. With high singlet oxygen generation and a short sensitivity time, NPs loaded with porphyrin sensitizers are the most widely used PS for PDT of ovarian cancer [243]. Among various recent theranostics, indocyanine/polydopamine-coated laponite nanosystems modified with polyethylene glycol-arginine-glycine-aspartic acid DOX-loaded nanoplatforms (ICG/LAP-PDA-PEG-RGD/DOX) showed high effectiveness PA imaging-guided chemotherapy of cancer cells overexpressing αvβ3 integrin.

Synergetic chemo-phototherapy under NIR laser irradiation was observed in vivo in a 4T1 tumor-bearing mouse model [244]. Fucoidan and photosensitive polypyrrole NP complexation was found to be an effective P-selectin-mediated delivery strategy with significant ROS/photothermal combinatorial therapeutic effects on lung cancer cells and tumors [245]. Apoferritin-conjugated cypate nanoprobes fabricated for NIR PA imaging and fluorescence imaging emerged as a versatile theranostic platform for solid tumor imaging and therapy [246]. When paclitaxel and the PS IR780 iodide were co-loaded into micelles with a particle size of ∼150 nm, a multimodal method for the treatment of malignant tumors was identified. Outstanding stability and photothermal/photodynamic efficacy were exhibited by these nanomicelles [247]. Aptamer-targeted PDT where aptamers could act as ligands that recognize and bind specifically to tumor cells or their membrane proteins [248]. The imaging-guided theranostics and aptamer-targeted treatment platforms including PTT, chemotherapy, and PDT were also developed. These aptamer-targeted photodynamic systems for tumor therapy have excellent potential for the future. For improved PDT for cancer, TME-responsive and efficient NPs were developed [249]. A unique melanin nanoprobe (PMNs-II-813) and extremely specialized prostrate-specific membrane antigen-specific molecular inhibitor were developed for targeted prostrate treatment [250].

Integrated dual targeting with controlled chemotherapy and PTT for cancer was recently achieved by Fe3O4@carbon(C)/ZnO-DOX-folic acid (FA) nano drug delivery systems, and the in vitro and in vivo results were promising [251]. Redox-responsive docetaxel (DTX)-loaded hyaluronic acid-cystamine-docosahexaenoic acid and choline e6 NPs were fabricated for the management of breast cancer with accurate cellular absorption and enough release of drugs in tumor tissues [252]. Dual-modal nano bipyramids for targeted cancer imaging and PTT are the latest entrants in this area, which have emerged as an effective platform for various biomedical applications [253]. The double targeting of nedaplatin-carboxyl-functionalized magnetic mesoporous silica-galactosylated chitosan NPs revealed that the NPs combined with PTT exhibited good anticancer effects [254]. The cerium oxide (CeO2) catalase nanozyme-loaded hyaluronic acid nanovesicles were made with recycled CeO2 to deal with the lack of oxygen at the tumor site, which makes it difficult for PS to act for a long time.

Targeted delivery of the PS Indocyanine green to the tumor was improved [255]. Magnetic molybdenum disulfide–chitosan/carboxymethylcellulose functionalized nanocomposites loaded with DOX (mMoS2-CS/CMC-DOX) were prepared for excellent photothermal effects exhibiting tumor targeting properties than plain mMoS2 [256]. Synergistic effects of combining PTT with immunotherapy were recently investigated to treat and target tumors [257]. Nanographene oxide covalently conjugated with FA, followed by the hydrothermal deposition of CuS nanoflowers was developed recently. The NGO-FA-CuS cytotoxicity toward cancer cells was enhanced, and the photothermal effects of the complex were found to be dependent on its concentration and power intensity of the laser source [258]. Targeted PDT for prostate-specific membrane antigen (PSMA) helped get more PSMA to tumors by making the tumors’ blood vessels more permeable [259]. Combining photothermal and IR in the targeted treatment of triple-negative breast cancer using silver NPs is another recent development [260].

8.3 Photoactivatable nanomaterials

Recently, an organic semiconducting pro-nanostimulant with NIR photoactivable immunotherapeutic action for synergistic cancer therapy was revealed. In a mouse xenograft model, it stopped the growth of both primary/distant tumors and lung metastasis, which could not be achieved with phototherapy alone [261]. A photoactivable pro-therapeutic was designed and proposed for metastasis-inhibited cancer therapy. A semiconducting polymer nanoblocker was developed to inhibit intracellular protein synthesis upon NIR photoactivation to synergize PDT for metastasis-inhibited cancer therapy [262]. In yet another interesting study, a photoactivable prodrug-backboned polymeric NP system (CNPPtCP/si(c-fos)) was developed for light-controlled delivery and synergistic photoactivated chemotherapy. Promising results for gene/drug co-delivery nanosystems were obtained for various cancer types [262]. A photoactivable RNAi system was developed, which fought cancer better when gene therapy and PTT were used together, in both in vitro and in vivo investigations [263]. A new strategy for drug/gene co-delivery was developed when controllable endo/lysosomal escaping versatile nanoplatforms such as photoactivated poly-prodrug NPs for effective light-controlled Pt(iv) and siRNA were fabricated for the treatment of various cancers [264]. Self-assembled nanostructures for protecting ruthenium moieties improved their stability, and these assemblies could get activated by red light, making them aptly suitable for in vivo cancer phototherapy [265]. A ROS-activatable thioketal connection between cabazitaxel and TKdC prodrug produced a colloidal-stable nanoassemble with low systemic toxicity and improved efficacy [266]. Photoactivable nanomicelles self-assembled from polyethylene glycol-stearamine conjugate with ROS-sensitive thioketal linker and co-loaded with DOX and pheophorbide A were developed for enhanced chemo-PDT [267].

8.4 Multifunctional nanomaterials

A multifunctional PMSA-targeted melanin-like polydopamine nanocarrier was developed for prostate cancer holding potential in early cancer theranostics [268]. In another recent study, a photothermally active polydopamine NP-based platform was designed for loading the chemotherapeutic drugs and targeting cancer cells as a promising approach to cancer therapy [241]. For the treatment of hypoxic cancer therapy, to increase drug loading and to control the release of radicals into the environment, a CuFeSe2-based system was created by layering a MIL-100(Fe) shell with a polymerization initiator and phase change material. The remarkable biocompatibility and anticancer properties of this potential multifunctional nanomaterial were demonstrated by in vitro and in vivo investigations [269]. Combinatorial chemotherapy and PDT were developed to augment antitumor responses of α-PD1 through core-shell metal ion drug NPs [270]. The one-step strategy simplified the complex synthesis of multifunctional NPs and created an “all-in-one” theranostic agent. In the development of an iodinated polyaniline NPs, integration of iodine doping with chemical oxidative polymerization for computed tomography imaging and PA imaging-guided PTT was carried out [271]. Cathepsin B-responsive multifunctional peptide-conjugated nanorods were fabricated for mitochondrial targeting [269]. A multifunctional, programmable DNA nanotrain with mitochondrion targeting was formulated that responded to azoreductase for highly effective photodynamic treatment and activatable hypoxia imaging [272]. Imaging-guided chemo-photodynamic combination treatment was observed with graphitic carbon nitride QDs [273,274]. Nanohybrids combining chemo-photo-gene therapy were developed for effective tumor growth reduction, and no adverse effects were observed [275]. The co-biomembrane-coated Fe3O4/MnO2 multifunctional NPs were also fabricated recently for enhanced chemo-dynamic therapy [276]. The use of multifunctional nano-biosensors based on MOFs was reported for improved fluorescence imaging of intracellular miRNA-122 for simultaneous chemo-photothermal treatment [277].

9 Future perspectives

The benefits of PDT in treating some cancers and pre-cancers over traditional chemotherapy and radiation therapy have made this treatment a mainstream cancer therapy despite significant advancements over the last decade. A number of weaknesses need to be overcome before these nanostructures can be incorporated into clinical practice. The main issues are as follows:

  • For large or deep-seated tumors, PDT is not recommended because light is unable to penetrate deeply into tissues. Therefore, PSs excited with NIR light will penetrate deeper into tissues than PSs excited with UV light or visible light.

  • PDT is only useful for treating metastatic cancers because it is a local treatment.

  • In most cases, patients considered with PDT report high sensitivity to light after treatment, requiring special precautions after PDT. Currently, efforts to address these constraints can be categorized into either of two approaches: finding increased efficacy in PSs or developing nanostructures that can improve PS localization.

First, intensive research aims to optimize the properties of PSs, either by preparing new compounds or by altering the core of existing compounds. Specifically, we will obtain PSs whose optical window absorption is greater, whose extinction coefficients are greater, whose singlet oxygen production is more efficient, and whose chemical and physical properties, especially solubility, are better. In addition, the targeting ability of the nanostructured system plays an important role in controlling the PS’s location. Various targeting molecules attach to the surface of the nanostructured carrier system in order to realize active PS targeting, such as carbohydrates, proteins, peptides, and antibodies. Furthermore, after the nanostructures have completed their therapeutic functions, they should be removed from the body to minimize side effects. The majority of current NPs used in PDT are nonbiodegradable and may gather in the body if they are not excreted, which confines their clinical application. The development of biodegradable nanostructures with high phototherapeutic properties should be the focus of future research.

10 Conclusion

Resistance to chemotherapeutic drugs resembles the resistance for infectious diseases, which is mediated by the reduction of apoptosis-related proteins, an increase of DNA repair, drug inactivation, efflux protein overexpression, and miRNAs. Thus, research on cancer are now aimed to overcome this resistance. Nanotechnology is increasingly utilized to combat MDR in the treatment of numerous diseases including cancer by the use of passive or active targeting. Nanocarriers to pool siRNAs, nano-based chemotherapeutic drug combinations, and antibody-mediated target action have been explored in many malignancies. Nanomedicines surpass traditional drugs in having better biocompatibility, target selectivity/specificity, PK, and stability apart from reducing the systemic toxicity and MDR. Although the nanotechnology-based anticancer products have shown great promise in preclinical studies, advanced clinical studies are still underway. Application of nanotechnology in phototherapy and immunotherapy are the key advancements in recent decades. Integration of nanotechnology with phototherapy proved to be an ideal approach to achieve target specificity and deeper tumor penetration which could cure metastatic malignancies with low incidence of side effects. Therefore, it can be concluded that nanomedicine-based phototherapy can serve as a novel and effective approach for the theranostics of various cancer forms, and further advanced studies are warranted to establish their role in clinical settings.



Acknowledgments

The authors extend their appreciation to the Deanship of Scientific Research at King Khalid University for funding this work through Small Groups Project under grant number (RGP. 1/247/44).

  1. Funding information: The authors extend their appreciation to the Deanship of Scientific Research at King Khalid University for funding this work through Small Groups Project under grant number (RGP. 1/247/44).

  2. Author contributions: Conceptualization: AS, SJ, MT, MEK, and MA; writing – original draft: AS, SJ, MT, MEK, and MA; writing – review and editing: WA, MZ, SSA, STM, and MHS; validation: AS, SJ, MT, MEK, and MA; funding acquisition: SSA and MHS; supervision: SSA and MHS. All authors have accepted responsibility for the entire content of this manuscript and approved its submission.

  3. Conflict of interest: The authors state no conflict of interest.

References

[1] Saeed M, Shoaib A, Kandimalla R, Javed S, Almatroudi A, Gupta R, et al. Microbe-based therapies for colorectal cancer: Advantages and limitations. Semin Cancer Biol. 2022;86(3):652–65.10.1016/j.semcancer.2021.05.018Suche in Google Scholar PubMed

[2] Nirmala MJ, Kizhuveetil U, Johnson A, Balaji G, Nagarajan R, Muthuvijayan V. Cancer nanomedicine: a review of nano-therapeutics and challenges ahead. RSC Adv. 2023;13:8606–29.10.1039/D2RA07863ESuche in Google Scholar

[3] Shoaib A, Tabish M, Ali S, Arafah A, Wahab S, Almarshad FM, et al. Dietary phytochemicals in cancer signalling pathways: Role of miRNA targeting. Curr Med Chem. 2021;28:8036–67.10.2174/0929867328666210420101605Suche in Google Scholar PubMed

[4] Wang C, Zhang S. Advantages of nanomedicine in cancer therapy: A review. ACS Appl Nano Mater. 2023;6:22594–610.10.1021/acsanm.3c04487Suche in Google Scholar

[5] Mir SA, Hamid L, Bader GN, Shoaib A, Rahamathulla M, Alshahrani MY, et al. Role of nanotechnology in overcoming the multidrug resistance in cancer therapy: A review. Molecules. 2022;27:6608.10.3390/molecules27196608Suche in Google Scholar PubMed PubMed Central

[6] Ji B, Wei M, Yang B. Recent advances in nanomedicines for photodynamic therapy (PDT)-driven cancer immunotherapy. Theranostics. 2022;12:434.10.7150/thno.67300Suche in Google Scholar PubMed PubMed Central

[7] Bhushan A, Gonsalves A, Menon JU. Current state of breast cancer diagnosis, treatment, and theranostics. Pharmaceutics. 2021;13:723.10.3390/pharmaceutics13050723Suche in Google Scholar PubMed PubMed Central

[8] Cook AB, Decuzzi P. Harnessing endogenous stimuli for responsive materials in theranostics. ACS Nano. 2021;15:2068–98.10.1021/acsnano.0c09115Suche in Google Scholar PubMed PubMed Central

[9] Mohapatra A, Uthaman S, Park I-K. External and internal stimuli-responsive metallic nanotherapeutics for enhanced anticancer therapy. Front Mol Biosci. 2021;7:597634.10.3389/fmolb.2020.597634Suche in Google Scholar PubMed PubMed Central

[10] He J, Li C, Ding L, Huang Y, Yin X, Zhang J, et al. Tumor targeting strategies of smart fluorescent nanoparticles and their applications in cancer diagnosis and treatment. Adv Mater. 2019;31:1902409.10.1002/adma.201902409Suche in Google Scholar PubMed

[11] Raj S, Khurana S, Choudhari R, Kesari KK, Kamal MA, Garg N, et al. Specific targeting cancer cells with nanoparticles and drug delivery in cancer therapy. Semin Cancer Biol. 2021;69:166–77.10.1016/j.semcancer.2019.11.002Suche in Google Scholar PubMed

[12] Li J, Wang S, Fontana F, Tapeinos C, Shahbazi M-A, Han H, et al. Nanoparticles-based phototherapy systems for cancer treatment: Current status and clinical potential. Bioact Mater. 2023;23:471–507.10.1016/j.bioactmat.2022.11.013Suche in Google Scholar PubMed PubMed Central

[13] Yang M, Chen Y, Zhu L, You L, Tong H, Meng H, et al. Harnessing nanotechnology: Emerging strategies for multiple myeloma therapy. Biomolecules. 2024;14:83.10.3390/biom14010083Suche in Google Scholar PubMed PubMed Central

[14] Paszko E, Ehrhardt C, Senge MO, Kelleher DP, Reynolds JV. Nanodrug applications in photodynamic therapy. Photodiagn Photodyn Ther. 2011;8:14–29.10.1016/j.pdpdt.2010.12.001Suche in Google Scholar PubMed

[15] Garg T, Jain NK, Rath G, Goyal A. Nanotechnology-based photodynamic therapy: concepts, advances, and perspectives. Crit Reviews™ Ther Drug Carr Syst. 2015;32(5):389–439.10.1615/CritRevTherDrugCarrierSyst.2015011645Suche in Google Scholar PubMed

[16] Yoo SW, Oh G, Ahn JC, Chung E. Non-oncologic applications of nanomedicine-based photo-therapy. Biomedicines. 2021;9:113.10.3390/biomedicines9020113Suche in Google Scholar PubMed PubMed Central

[17] He X, Zhang S, Tian Y, Cheng W, Jing H. Research progress of nanomedicine-based mild photothermal therapy in tumor. Int J Nanomed. 2023;18:1433–68.10.2147/IJN.S405020Suche in Google Scholar PubMed PubMed Central

[18] Li Y, Li X, Zhou F, Doughty A, Hoover AR, Nordquist RE, et al. Nanotechnology-based photoimmunological therapies for cancer. Cancer Lett. 2019;442:429–38.10.1016/j.canlet.2018.10.044Suche in Google Scholar PubMed PubMed Central

[19] Liu Z, Xie Z, Li W, Wu X, Jiang X, Li G, et al. Photodynamic immunotherapy of cancers based on nanotechnology: Recent advances and future challenges. J Nanobiotechnology. 2021;19:1–18.10.1186/s12951-021-00903-7Suche in Google Scholar PubMed PubMed Central

[20] Zhou Z, Song J, Nie L, Chen X. Reactive oxygen species generating systems meeting challenges of photodynamic cancer therapy. Chem Soc Rev. 2016;45:6597–626.10.1039/C6CS00271DSuche in Google Scholar PubMed PubMed Central

[21] Bhatia SN, Chen X, Dobrovolskaia MA, Lammers T. Cancer nanomedicine. Nat Rev Cancer. 2022;22:550–6.10.1038/s41568-022-00496-9Suche in Google Scholar PubMed PubMed Central

[22] Subhan MA, Yalamarty SSK, Filipczak N, Parveen F, Torchilin VP. Recent advances in tumor targeting via EPR effect for cancer treatment. J Personalized Med. 2021;11:571.10.3390/jpm11060571Suche in Google Scholar PubMed PubMed Central

[23] Fang J, Islam W, Maeda H. Exploiting the dynamics of the EPR effect and strategies to improve the therapeutic effects of nanomedicines by using EPR effect enhancers. Adv Drug Delivery Rev. 2020;157:142–60.10.1016/j.addr.2020.06.005Suche in Google Scholar PubMed

[24] Zhang C, Wang X, Du J, Gu Z, Zhao Y. Reactive oxygen species‐regulating strategies based on nanomaterials for disease treatment. Adv Sci. 2021;8:2002797.10.1002/advs.202002797Suche in Google Scholar PubMed PubMed Central

[25] Duong T, Li X, Yang B, Schumann C, Albarqi HA, Taratula O, et al. Phototheranostic nanoplatform based on a single cyanine dye for image-guided combinatorial phototherapy. Nanomed: Nanotechnol Biol Med. 2017;13:955–63.10.1016/j.nano.2016.11.005Suche in Google Scholar PubMed

[26] Tabish TA, Zhang S, Winyard PG. Developing the next generation of graphene-based platforms for cancer therapeutics: The potential role of reactive oxygen species. Redox Biol. 2018;15:34–40.10.1016/j.redox.2017.11.018Suche in Google Scholar PubMed PubMed Central

[27] Ge J, Lan M, Zhou B, Liu W, Guo L, Wang H, et al. A graphene quantum dot photodynamic therapy agent with high singlet oxygen generation. Nat Commun. 2014;5:4596.10.1038/ncomms5596Suche in Google Scholar PubMed PubMed Central

[28] Dąbrowski JM. Reactive oxygen species in photodynamic therapy: mechanisms of their generation and potentiation. Adv Inorg Chem. 2017;70:343–94.10.1016/bs.adioch.2017.03.002Suche in Google Scholar

[29] Agostinis P, Berg K, Cengel KA, Foster TH, Girotti AW, Gollnick SO, et al. Photodynamic therapy of cancer: an update. CA: A Cancer J Clinicians. 2011;61:250–81.10.3322/caac.20114Suche in Google Scholar PubMed PubMed Central

[30] Wan Y, Fu LH, Li C, Lin J, Huang P. Conquering the hypoxia limitation for photodynamic therapy. Adv Mater. 2021;33:2103978.10.1002/adma.202103978Suche in Google Scholar PubMed

[31] Yang B, Chen Y, Shi J. Reactive oxygen species (ROS)-based nanomedicine. Chem Rev. 2019;119:4881–985.10.1021/acs.chemrev.8b00626Suche in Google Scholar PubMed

[32] Gunaydin G, Gedik ME, Ayan S. Photodynamic therapy for the treatment and diagnosis of cancer–a review of the current clinical status. Front Chem. 2021;9:686303.10.3389/fchem.2021.686303Suche in Google Scholar PubMed PubMed Central

[33] Hu T, Wang Z, Shen W, Liang R, Yan D, Wei M. Recent advances in innovative strategies for enhanced cancer photodynamic therapy. Theranostics. 2021;11:3278.10.7150/thno.54227Suche in Google Scholar PubMed PubMed Central

[34] Das S, Tiwari M, Mondal D, Sahoo BR, Tiwari DK. Growing tool-kit of photosensitizers for clinical and non-clinical applications. J Mater Chem B. 2020;8:10897–940.10.1039/D0TB02085KSuche in Google Scholar

[35] Hu J-J, Lei Q, Zhang X-Z. Recent advances in photonanomedicines for enhanced cancer photodynamic therapy. Prog Mater Sci. 2020;114:100685.10.1016/j.pmatsci.2020.100685Suche in Google Scholar

[36] Plekhova N, Shevchenko O, Korshunova O, Stepanyugina A, Tananaev I, Apanasevich V. Development of novel tetrapyrrole structure photosensitizers for cancer photodynamic therapy. Bioengineering. 2022;9:82.10.3390/bioengineering9020082Suche in Google Scholar PubMed PubMed Central

[37] Fan W, Huang P, Chen X. Overcoming the Achilles’ heel of photodynamic therapy. Chem Soc Rev. 2016;45:6488–519.10.1039/C6CS00616GSuche in Google Scholar

[38] Tugaoen HON, Garcia-Segura S, Hristovski K, Westerhoff P. Compact light-emitting diode optical fiber immobilized TiO2 reactor for photocatalytic water treatment. Sci Total Environ. 2018;613:1331–8.10.1016/j.scitotenv.2017.09.242Suche in Google Scholar PubMed

[39] Hou Z, Zhang Y, Deng K, Chen Y, Li X, Deng X, et al. UV-emitting upconversion-based TiO2 photosensitizing nanoplatform: near-infrared light mediated in vivo photodynamic therapy via mitochondria-involved apoptosis pathway. ACS Nano. 2015;9:2584–99.10.1021/nn506107cSuche in Google Scholar PubMed

[40] Li S, Yang F, Wang Y, Du T, Hou X. Emerging nanotherapeutics for facilitating photodynamic therapy. Chem Eng J. 2023;451:138621.10.1016/j.cej.2022.138621Suche in Google Scholar

[41] Bessière A, Durand J-O, Noûs C. Persistent luminescence materials for deep photodynamic therapy. Nanophotonics. 2021;10:2999–3029.10.1515/nanoph-2021-0254Suche in Google Scholar

[42] Gao D, Guo X, Zhang X, Chen S, Wang Y, Chen T, et al. Multifunctional phototheranostic nanomedicine for cancer imaging and treatment. Mater Today Bio. 2020;5:100035.10.1016/j.mtbio.2019.100035Suche in Google Scholar PubMed PubMed Central

[43] Yu M, Xu X, Cai Y, Zou L, Shuai X. Perfluorohexane-cored nanodroplets for stimulations-responsive ultrasonography and O2-potentiated photodynamic therapy. Biomaterials. 2018;175:61–71.10.1016/j.biomaterials.2018.05.019Suche in Google Scholar PubMed

[44] Wang P, Li X, Yao C, Wang W, Zhao M, El-Toni AM, et al. Orthogonal near-infrared upconversion co-regulated site-specific O2 delivery and photodynamic therapy for hypoxia tumor by using red blood cell microcarriers. Biomaterials. 2017;125:90–100.10.1016/j.biomaterials.2017.02.017Suche in Google Scholar PubMed

[45] Gao S, Zheng P, Li Z, Feng X, Yan W, Chen S, et al. Biomimetic O2-Evolving metal-organic framework nanoplatform for highly efficient photodynamic therapy against hypoxic tumor. Biomaterials. 2018;178:83–94.10.1016/j.biomaterials.2018.06.007Suche in Google Scholar PubMed

[46] Lin T, Zhao X, Zhao S, Yu H, Cao W, Chen W, et al. O2-generating MnO2 nanoparticles for enhanced photodynamic therapy of bladder cancer by ameliorating hypoxia. Theranostics. 2018;8:990.10.7150/thno.22465Suche in Google Scholar PubMed PubMed Central

[47] Wang S, Yuan F, Chen K, Chen G, Tu K, Wang H, et al. Synthesis of hemoglobin conjugated polymeric micelle: a ZnPc carrier with oxygen self-compensating ability for photodynamic therapy. Biomacromolecules. 2015;16:2693–700.10.1021/acs.biomac.5b00571Suche in Google Scholar PubMed

[48] Song X, Feng L, Liang C, Yang K, Liu Z. Ultrasound triggered tumor oxygenation with oxygen-shuttle nanoperfluorocarbon to overcome hypoxia-associated resistance in cancer therapies. Nano Lett. 2016;16:6145–53.10.1021/acs.nanolett.6b02365Suche in Google Scholar PubMed

[49] Xie Z, Cai X, Sun C, Liang S, Shao S, Huang S, et al. O2-loaded pH-responsive multifunctional nanodrug carrier for overcoming hypoxia and highly efficient chemo-photodynamic cancer therapy. Chem Mater. 2018;31:483–90.10.1021/acs.chemmater.8b04321Suche in Google Scholar

[50] Cai X, Xie Z, Ding B, Shao S, Liang S, Pang M, et al. Monodispersed copper(I)‐based nano metal–organic framework as a biodegradable drug carrier with enhanced photodynamic therapy efficacy. Adv Sci. 2019;6:1900848.10.1002/advs.201900848Suche in Google Scholar PubMed PubMed Central

[51] Yoon I, Li JZ, Shim YK. Advance in photosensitizers and light delivery for photodynamic therapy. Clin Endoscopy. 2013;46:7–23.10.5946/ce.2013.46.1.7Suche in Google Scholar PubMed PubMed Central

[52] Benov L. Photodynamic therapy: current status and future directions. Med Princ Pract. 2014;24:14–28.10.1159/000362416Suche in Google Scholar PubMed PubMed Central

[53] Escudero A, Carrillo-Carrión C, Castillejos MC, Romero-Ben E, Rosales-Barrios C, Khiar N. Photodynamic therapy: photosensitizers and nanostructures. Mater Chem Front. 2021;5:3788–812.10.1039/D0QM00922ASuche in Google Scholar

[54] Khizar S, Alrushaid N, Khan FA, Zine N, Jaffrezic-Renault N, Errachid A, et al. Nanocarriers based novel and effective drug delivery system. Int J Pharmaceutics. 2023;632:122570.10.1016/j.ijpharm.2022.122570Suche in Google Scholar PubMed

[55] Adepu S, Ramakrishna S. Controlled drug delivery systems: current status and future directions. Molecules. 2021;26:5905.10.3390/molecules26195905Suche in Google Scholar PubMed PubMed Central

[56] Lavan DA, McGuire T, Langer R. Small-scale systems for in vivo drug delivery. Nat Biotechnol. 2003;21:1184–91.10.1038/nbt876Suche in Google Scholar PubMed

[57] Zhao Z, Ukidve A, Krishnan V, Mitragotri S. Effect of physicochemical and surface properties on in vivo fate of drug nanocarriers. Adv Drug Delivery Rev. 2019;143:3–21.10.1016/j.addr.2019.01.002Suche in Google Scholar PubMed

[58] Sultan MH, Moni SS, Madkhali OA, Bakkari MA, Alshahrani S, Alqahtani SS, et al. Characterization of cisplatin-loaded chitosan nanoparticles and rituximab-linked surfaces as target-specific injectable nano-formulations for combating cancer. Sci Rep. 2022;12:468.10.1038/s41598-021-04427-wSuche in Google Scholar PubMed PubMed Central

[59] Alsaab HO, Alghamdi MS, Alotaibi AS, Alzhrani R, Alwuthaynani F, Althobaiti YS, et al. Progress in clinical trials of photodynamic therapy for solid tumors and the role of nanomedicine. Cancers. 2020;12:2793.10.3390/cancers12102793Suche in Google Scholar PubMed PubMed Central

[60] Rozhkova EA. Nanoscale materials for tackling brain cancer: Recent progress and outlook. Adv Mater. 2011;23:H136–50.10.1002/adma.201004714Suche in Google Scholar PubMed

[61] Xiao L, Gu L, Howell SB, Sailor MJ. Porous silicon nanoparticle photosensitizers for singlet oxygen and their phototoxicity against cancer cells. ACS Nano. 2011;5:3651–9.10.1021/nn1035262Suche in Google Scholar PubMed PubMed Central

[62] Seidl C, Ungelenk J, Zittel E, Bergfeldt T, Sleeman JP, Schepers U, et al. Tin tungstate nanoparticles: a photosensitizer for photodynamic tumor therapy. ACS Nano. 2016;10:3149–57.10.1021/acsnano.5b03060Suche in Google Scholar PubMed

[63] Samia AC, Chen X, Burda C. Semiconductor quantum dots for photodynamic therapy. J Am Chem Soc. 2003;125:15736–37.10.1021/ja0386905Suche in Google Scholar PubMed

[64] Vazquez-Ortega F, Lagunes I, Trigos A. Cosmetic dyes as potential photosensitizers of singlet oxygen generation. Dye Pigment. 2020;176:108248.10.1016/j.dyepig.2020.108248Suche in Google Scholar

[65] Cui G, Fang WH. State-specific heavy-atom effect on intersystem crossing processes in 2-thiothymine: A potential photodynamic therapy photosensitizer. J Chem Phys. 2013;138:044315.10.1063/1.4776261Suche in Google Scholar PubMed

[66] Borah R, Ag KR, Minja AC, Verbruggen SW. A review on self‐assembly of colloidal nanoparticles into clusters, patterns, and films: emerging synthesis techniques and applications. Small Methods. 2023;7(6):2201536.10.1002/smtd.202201536Suche in Google Scholar PubMed

[67] Lee H, Hong K-I, Jang W-D. Design and applications of molecular probes containing porphyrin derivatives. Coord Chem Rev. 2018;354:46–73.10.1016/j.ccr.2017.06.008Suche in Google Scholar

[68] Dai C, Liu B. Conjugated polymers for visible-light-driven photocatalysis. Energy Environ Sci. 2020;13:24–52.10.1039/C9EE01935ASuche in Google Scholar

[69] Zhang D, Yan S, Song W. Photochemically induced formation of reactive oxygen species (ROS) from effluent organic matter. Environ Sci & Technol. 2014;48:12645–53.10.1021/es5028663Suche in Google Scholar PubMed

[70] Vankayala R, Hwang KC. Near‐infrared‐light‐activatable nanomaterial‐mediated phototheranostic nanomedicines: an emerging paradigm for cancer treatment. Adv Mater. 2018;30:1706320.10.1002/adma.201706320Suche in Google Scholar PubMed

[71] Park W, Na K. Advances in the synthesis and application of nanoparticles for drug delivery. Wiley Interdiscip Reviews: Nanomed Nanobiotechnol. 2015;7:494–508.10.1002/wnan.1325Suche in Google Scholar PubMed

[72] Zheng Q, Liu X, Zheng Y, Yeung KW, Cui Z, Liang Y, et al. The recent progress on metal–organic frameworks for phototherapy. Chem Soc Rev. 2021;50:5086–125.10.1039/D1CS00056JSuche in Google Scholar PubMed

[73] Liang C, Xu L, Song G, Liu Z. Emerging nanomedicine approaches fighting tumor metastasis: animal models, metastasis-targeted drug delivery, phototherapy, and immunotherapy. Chem Soc Rev. 2016;45:6250–69.10.1039/C6CS00458JSuche in Google Scholar

[74] Zhao L, Zhang X, Wang X, Guan X, Zhang W, Ma J. Recent advances in selective photothermal therapy of tumor. J Nanobiotechnol. 2021;19:1–15.10.1186/s12951-021-01080-3Suche in Google Scholar PubMed PubMed Central

[75] Bhole R, Bonde C, Kadam P, Wavwale R. A comprehensive review on photodynamic therapy (PDT) and photothermal therapy (PTT) for cancer treatment. Turkish J Oncol. 2021;36(1):125–32.10.5505/tjo.2020.2400Suche in Google Scholar

[76] Shi Y, Liu M, Deng F, Zeng G, Wan Q, Zhang X, et al. Recent progress and development on polymeric nanomaterials for photothermal therapy: a brief overview. J Mater Chem B. 2017;5:194–206.10.1039/C6TB02249ASuche in Google Scholar PubMed

[77] Yang W, Liang H, Ma S, Wang D, Huang J. Gold nanoparticle based photothermal therapy: Development and application for effective cancer treatment. Sustain Mater Technol. 2019;22:e00109.10.1016/j.susmat.2019.e00109Suche in Google Scholar

[78] Taheri-Ledari R, Ganjali F, Zarei-Shokat S, Dinmohammadi R, Asl FR, Emami A, et al. Plasmonic porous micro-and nano-materials based on Au/Ag nanostructures developed for photothermal cancer therapy: challenges in clinicalization. Nanoscale Adv. 2023;5:6768–86.10.1039/D3NA00763DSuche in Google Scholar PubMed PubMed Central

[79] Zada A, Muhammad P, Ahmad W, Hussain Z, Ali S, Khan M, et al. Surface plasmonic‐assisted photocatalysis and optoelectronic devices with noble metal nanocrystals: design, synthesis, and applications. Adv Funct Mater. 2020;30:1906744.10.1002/adfm.201906744Suche in Google Scholar

[80] Krishchenko I, Manoilov É, Kravchenko S, Snopok B. Resonant optical phenomena in heterogeneous plasmon nanostructures of noble metals: a review. Theor Exp Chem. 2020;56:67–110.10.1007/s11237-020-09642-6Suche in Google Scholar

[81] Ding X, Liow CH, Zhang M, Huang R, Li C, Shen H, et al. Surface plasmon resonance enhanced light absorption and photothermal therapy in the second near-infrared window. J Am Chem Soc. 2014;136:15684–93.10.1021/ja508641zSuche in Google Scholar PubMed

[82] Chen J, Gong M, Fan Y, Feng J, Han L, Xin HL, et al. Collective plasmon coupling in gold nanoparticle clusters for highly efficient photothermal therapy. ACS Nano. 2022;16:910–20.10.1021/acsnano.1c08485Suche in Google Scholar PubMed

[83] Hemmer E, Benayas A, Légaré F, Vetrone F. Exploiting the biological windows: current perspectives on fluorescent bioprobes emitting above 1,000 nm. Nanoscale Horiz. 2016;1:168–84.10.1039/C5NH00073DSuche in Google Scholar PubMed

[84] Taratula O, Patel M, Schumann C, Naleway MA, Pang AJ, He H, et al. Phthalocyanine-loaded graphene nanoplatform for imaging-guided combinatorial phototherapy. Int J Nanomed. 2015;2347–62.10.2147/IJN.S81097Suche in Google Scholar PubMed PubMed Central

[85] Yang B, Chen Y, Shi J. Nanocatalytic medicine. Adv Mater. 2019;31:1901778.10.1002/adma.201901778Suche in Google Scholar PubMed

[86] Devi P, Saini S, Kim K-H. The advanced role of carbon quantum dots in nanomedical applications. Biosens Bioelectron. 2019;141:111158.10.1016/j.bios.2019.02.059Suche in Google Scholar PubMed

[87] Li J, Zhang W, Ji W, Wang J, Wang N, Wu W, et al. Near infrared photothermal conversion materials: Mechanism, preparation, and photothermal cancer therapy applications. J Mater Chem B. 2021;9:7909–26.10.1039/D1TB01310FSuche in Google Scholar

[88] Hwang Y, Park S-H, Lee JW. Applications of functionalized carbon nanotubes for the therapy and diagnosis of cancer. Polymers. 2017;9:13.10.3390/polym9010013Suche in Google Scholar PubMed PubMed Central

[89] Chen Y, Zhou F, Wang C, Hu L, Guo P. Nanostructures as photothermal agents in tumor treatment. Molecules. 2022;28:277.10.3390/molecules28010277Suche in Google Scholar PubMed PubMed Central

[90] Shao J, Griffin RJ, Galanzha EI, Kim J-W, Koonce N, Webber J, et al. Photothermal nanodrugs: potential of TNF-gold nanospheres for cancer theranostics. Sci Rep. 2013;3:1293.10.1038/srep01293Suche in Google Scholar PubMed PubMed Central

[91] Vines JB, Yoon J-H, Ryu N-E, Lim D-J, Park H. Gold nanoparticles for photothermal cancer therapy. Front Chem. 2019;7:167.10.3389/fchem.2019.00167Suche in Google Scholar PubMed PubMed Central

[92] Kennedy LC, Bickford LR, Lewinski NA, Coughlin AJ, Hu Y, Day ES, et al. A new era for cancer treatment: gold‐nanoparticle‐mediated thermal therapies. Small. 2011;7:169–83.10.1002/smll.201000134Suche in Google Scholar PubMed

[93] Rastinehad AR, Anastos H, Wajswol E, Winoker JS, Sfakianos JP, Doppalapudi SK, et al. Gold nanoshell-localized photothermal ablation of prostate tumors in a clinical pilot device study. Proc Natl Acad Sci. 2019;116:18590–96.10.1073/pnas.1906929116Suche in Google Scholar PubMed PubMed Central

[94] Han HS, Choi KY. Advances in nanomaterial-mediated photothermal cancer therapies: toward clinical applications. Biomedicines. 2021;9:305.10.3390/biomedicines9030305Suche in Google Scholar PubMed PubMed Central

[95] Wang H, Naghavi M, Allen C, Barber RM, Bhutta ZA, Carter A, et al. Global, regional, and national life expectancy, all-cause mortality, and cause-specific mortality for 249 causes of death, 1980–2015: a systematic analysis for the Global Burden of Disease Study 2015. Lancet. 2016;388:1459–1544.10.1016/S0140-6736(16)31012-1Suche in Google Scholar PubMed PubMed Central

[96] Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA: A Cancer J Clinicians. 2011;61:69–90.10.3322/caac.20107Suche in Google Scholar PubMed

[97] Ahmad MZ, Akhter S, Jain GK, Rahman M, Pathan SA, Ahmad FJ, et al. Metallic nanoparticles: technology overview & drug delivery applications in oncology. Expert Opin Drug Delivery. 2010;7:927–42.10.1517/17425247.2010.498473Suche in Google Scholar PubMed

[98] Liechty WB, Kryscio DR, Slaughter BV, Peppas NA. Polymers for drug delivery systems. Annu Rev Chem Biomol Eng. 2010;1:149–73.10.1146/annurev-chembioeng-073009-100847Suche in Google Scholar PubMed PubMed Central

[99] McCallion C, Burthem J, Rees-Unwin K, Golovanov A, Pluen A. Graphene in therapeutics delivery: Problems, solutions and future opportunities. Eur J Pharm Biopharm. 2016;104:235–50.10.1016/j.ejpb.2016.04.015Suche in Google Scholar PubMed

[100] Slowing II, Vivero-Escoto JL, Wu CW, Lin VSY. Mesoporous silica nanoparticles as controlled release drug delivery and gene transfection carriers. Adv Drug Delivery Rev. 2008;60:1278–88.10.1016/j.addr.2008.03.012Suche in Google Scholar PubMed

[101] Foote RS, Lee JW. Micro and nano technologies in bioanalysis: Methods and protocols. Springer; 2009. pp. 43–52.10.1007/978-1-59745-483-4Suche in Google Scholar

[102] Babaei M, Ganjalikhani M. The potential effectiveness of nanoparticles as radio sensitizers for radiotherapy. BioImpacts: BI. 2014;4:15.Suche in Google Scholar

[103] Nune SK, Gunda P, Thallapally PK, Lin Y-Y, Laird Forrest M, Berkland CJ. Nanoparticles for biomedical imaging. Expert Opin Drug Delivery. 2009;6:1175–94.10.1517/17425240903229031Suche in Google Scholar PubMed PubMed Central

[104] Kobayashi H, Watanabe R, Choyke PL. Improving conventional enhanced permeability and retention (EPR) effects; what is the appropriate target? Theranostics. 2014;4:81.10.7150/thno.7193Suche in Google Scholar PubMed PubMed Central

[105] Zhang L, Su H, Cai J, Cheng D, Ma Y, Zhang J, et al. A multifunctional platform for tumor angiogenesis-targeted chemo-thermal therapy using polydopamine-coated gold nanorods. ACS Nano. 2016;10:10404–17.10.1021/acsnano.6b06267Suche in Google Scholar PubMed

[106] Zhang W, Li Y, Sun J-H, Tan C-P, Ji L-N, Mao Z-W. Supramolecular self-assembled nanoparticles for chemo-photodynamic dual therapy against cisplatin resistant cancer cells. Chem Commun. 2015;51:1807–10.10.1039/C4CC08583CSuche in Google Scholar PubMed

[107] Lammers T, Kiessling F, Hennink WE, Storm G. Drug targeting to tumors: principles, pitfalls and (pre-) clinical progress. Journal of Control Rel. 2012;161(2):175–87.10.1016/j.jconrel.2011.09.063Suche in Google Scholar PubMed

[108] Chakraborty S, Mal S, Halder A, Das S, Sen KK, Mukherjee A, et al. Nano-dimensional gold synthesis for biomedical applications: upscaling and challenges. Part Sci Technol. 2024;42:145–63.10.1080/02726351.2023.2211954Suche in Google Scholar

[109] Crapnell RD, Banks CE. Electroanalytical overview: utilising micro-and nano-dimensional sized materials in electrochemical-based biosensing platforms. Microchim Acta. 2021;188:1–23.10.1007/s00604-021-04913-ySuche in Google Scholar PubMed PubMed Central

[110] Sarfraz N, Khan I. Plasmonic gold nanoparticles (AuNPs): properties, synthesis and their advanced energy, environmental and biomedical applications. Chem–An Asian J. 2021;16:720–42.10.1002/asia.202001202Suche in Google Scholar PubMed

[111] Hang Y, Wang A, Wu N. Plasmonic silver and gold nanoparticles: shape-and structure-modulated plasmonic functionality for point-of-caring sensing, bio-imaging and medical therapy. Chem Soc Rev. 2024;53:2932–71.10.1039/D3CS00793FSuche in Google Scholar

[112] Abadeer NS, Murphy CJ. Recent progress in cancer thermal therapy using gold nanoparticles. Nanomater Neoplasms. 2021;143–217.10.1201/9780429027819-3Suche in Google Scholar

[113] Riley RS, Day ES. Gold nanoparticle‐mediated photothermal therapy: applications and opportunities for multimodal cancer treatment. Wiley Interdiscip Reviews: Nanomed Nanobiotechnol. 2017;9:e1449.10.1002/wnan.1449Suche in Google Scholar PubMed PubMed Central

[114] Dreaden EC, Alkilany AM, Huang X, Murphy CJ, El-Sayed MA. The golden age: gold nanoparticles for biomedicine. Chem Soc Rev. 2012;41:2740–79.10.1039/C1CS15237HSuche in Google Scholar PubMed PubMed Central

[115] Dykman L, Khlebtsov N. Gold nanoparticles in biomedical applications: recent advances and perspectives. Chem Soc Rev. 2012;41:2256–82.10.1039/C1CS15166ESuche in Google Scholar PubMed

[116] Bai X, Wang Y, Song Z, Feng Y, Chen Y, Zhang D, et al. The basic properties of gold nanoparticles and their applications in tumor diagnosis and treatment. Int J Mol Sci. 2020;21:2480.10.3390/ijms21072480Suche in Google Scholar PubMed PubMed Central

[117] Cha C, Shin SR, Annabi N, Dokmeci MR, Khademhosseini A. Carbon-based nanomaterials: multifunctional materials for biomedical engineering. ACS Nano. 2013;7:2891–7.10.1021/nn401196aSuche in Google Scholar PubMed PubMed Central

[118] Devi N, Kumar R, Chen Y-S, Singh RK. Carbon-based nanomaterials: carbon nanotube, fullerene, and carbon dots. In: Nanomaterials: Advances and applications. Springer; 2023. p. 27–57.10.1007/978-981-19-7963-7_2Suche in Google Scholar

[119] Gaur M, Misra C, Yadav AB, Swaroop S, Maolmhuaidh FÓ, Bechelany M, et al. Biomedical applications of carbon nanomaterials: fullerenes, quantum dots, nanotubes, nanofibers, and graphene. Materials. 2021;14:5978.10.3390/ma14205978Suche in Google Scholar PubMed PubMed Central

[120] Paul S, Mondal S, Saha A, Roy S. Fundamentals and functionalization of CNTs and other carbon nanomaterials. In: Functionalized carbon nanomaterials for theranostic applications. Elsevier; 2023. p. 77–90.10.1016/B978-0-12-824366-4.00008-XSuche in Google Scholar

[121] Qi K, Sun B, Liu SY, Zhang M. Research progress on carbon materials in tumor photothermal therapy. Biomed Pharmacother. 2023;165:115070.10.1016/j.biopha.2023.115070Suche in Google Scholar PubMed

[122] Ruggiero A, Villa CH, Bander E, Rey DA, Bergkvist M, Batt CA, et al. Paradoxical glomerular filtration of carbon nanotubes. Proc Natl Acad Sci. 2010;107:12369–74.10.1073/pnas.0913667107Suche in Google Scholar PubMed PubMed Central

[123] Zhang M, Wang W, Wu F, Yuan P, Chi C, Zhou N. Magnetic and fluorescent carbon nanotubes for dual modal imaging and photothermal and chemo-therapy of cancer cells in living mice. Carbon. 2017;123:70–83.10.1016/j.carbon.2017.07.032Suche in Google Scholar

[124] Zhang L, Xia J, Zhao Q, Liu L, Zhang Z. Functional graphene oxide as a nanocarrier for controlled loading and targeted delivery of mixed anticancer drugs. Small. 2010;6:537–44.10.1002/smll.200901680Suche in Google Scholar PubMed

[125] Dong H, Jin M, Liu Z, Xiong H, Qiu X, Zhang W, et al. In vitro and in vivo brain-targeting chemo-photothermal therapy using graphene oxide conjugated with transferrin for Gliomas. Lasers Med Sci. 2016;31:1123–31.10.1007/s10103-016-1955-2Suche in Google Scholar PubMed

[126] Song J, Yang X, Jacobson O, Lin L, Huang P, Niu G, et al. Sequential drug release and enhanced photothermal and photoacoustic effect of hybrid reduced graphene oxide-loaded ultrasmall gold nanorod vesicles for cancer therapy. ACS Nano. 2015;9:9199–209.10.1021/acsnano.5b03804Suche in Google Scholar PubMed PubMed Central

[127] Li L, Han X, Wang M, Li C, Jia T, Zhao X. Recent advances in the development of near-infrared organic photothermal agents. Chem Eng J. 2021;417:128844.10.1016/j.cej.2021.128844Suche in Google Scholar

[128] Tufani A, Qureshi A, Niazi JH. Iron oxide nanoparticles based magnetic luminescent quantum dots (MQDs) synthesis and biomedical/biological applications: A review. Mater Sci Eng: C. 2021;118:111545.10.1016/j.msec.2020.111545Suche in Google Scholar PubMed

[129] Peng C, Zhang X. Chemical functionalization of graphene nanoplatelets with hydroxyl, amino, and carboxylic terminal groups. Chemistry. 2021;3:873–88.10.3390/chemistry3030064Suche in Google Scholar

[130] Anastasiadis SH, Chrissopoulou K, Stratakis E, Kavatzikidou P, Kaklamani G, Ranella A. How the physicochemical properties of manufactured nanomaterials affect their performance in dispersion and their applications in biomedicine: A review. Nanomaterials. 2022;12:552.10.3390/nano12030552Suche in Google Scholar PubMed PubMed Central

[131] Jiao M, Zhang P, Meng J, Li Y, Liu C, Luo X, et al. Recent advancements in biocompatible inorganic nanoparticles towards biomedical applications. Biomater Sci. 2018;6:726–45.10.1039/C7BM01020FSuche in Google Scholar

[132] Wang X, Zhong X, Li J, Liu Z, Cheng L. Inorganic nanomaterials with rapid clearance for biomedical applications. Chem Soc Rev. 2021;50:8669–8742.10.1039/D0CS00461HSuche in Google Scholar

[133] Yezhelyev M, Yacoub R, O’Regan R. Inorganic nanoparticles for predictive oncology of breast cancer. Nanomed (Lond). 2009 Jan;4(1):83–103.10.2217/17435889.4.1.83Suche in Google Scholar PubMed

[134] Chiozzi V, Rossi F. Inorganic–organic core/shell nanoparticles: progress and applications. Nanoscale Adv. 2020;2:5090–5105.10.1039/D0NA00411ASuche in Google Scholar PubMed PubMed Central

[135] Rashid EU, Nawaz S, Munawar J, Sarker A, Hussain S, Iqbal HM, et al. Organic and inorganic nanoparticles. Micro and Nano Technologies. 2023;13:93–119.10.1016/B978-0-323-91611-0.00014-1Suche in Google Scholar

[136] Lytton-Jean AK, Kauffman KJ, Kaczmarek JC, Langer R. Cancer nanotherapeutics in clinical trials. Nanotechnol-Based Precis Tools Detect Treat Cancer. 2015;136:293–322.10.1007/978-3-319-16555-4_13Suche in Google Scholar PubMed

[137] Xie Q, Tang J, Guo S, Zhao Q, Li S. Recent progress of preparation strategies in organic nanoparticles for cancer phototherapeutics. Molecules. 2023;28:6038.10.3390/molecules28166038Suche in Google Scholar PubMed PubMed Central

[138] Rejinold NS, Choi G, Choy J-H. Recent developments on semiconducting polymer nanoparticles as smart photo-therapeutic agents for cancer treatments – A review. Polymers. 2021;13:981.10.3390/polym13060981Suche in Google Scholar PubMed PubMed Central

[139] Fuse T, Tagami T, Tane M, Ozeki T. Effective light-triggered contents release from helper lipid-incorporated liposomes co-encapsulating gemcitabine and a water-soluble photosensitizer. Int J Pharmaceutics. 2018;540:50–6.10.1016/j.ijpharm.2018.01.040Suche in Google Scholar PubMed

[140] Mendes R, Pedrosa P, Lima JC, Fernandes AR, Baptista PV. Photothermal enhancement of chemotherapy in breast cancer by visible irradiation of gold nanoparticles. Sci Rep. 2017;7:10872.10.1038/s41598-017-11491-8Suche in Google Scholar PubMed PubMed Central

[141] Zhao Y, He Z, Zhang Q, Wang J, Jia W, Jin L, et al. 880 nm NIR-triggered organic small molecular-based nanoparticles for photothermal therapy of tumor. Nanomaterials. 2021;11:773.10.3390/nano11030773Suche in Google Scholar PubMed PubMed Central

[142] Cheng X, Sun R, Yin L, Chai Z, Shi H, Gao M. Light‐triggered assembly of gold nanoparticles for photothermal therapy and photoacoustic imaging of tumors in vivo. Adv Mater. 2017;29:1604894.10.1002/adma.201604894Suche in Google Scholar PubMed

[143] Karami MH, Abdouss M, Rahdar A, Pandey S. Graphene quantum dots: Background, synthesis methods, and applications as nanocarrier in drug delivery and cancer treatment: An updated review. Inorg Chem Commun. 2024;112032.10.1016/j.inoche.2024.112032Suche in Google Scholar

[144] Wen K, Zhou M, Lu H, Bi Y, Ruan L, Chen J, et al. Near-infrared/pH dual-sensitive nanocarriers for enhanced intracellular delivery of doxorubicin. ACS Biomater Sci Eng. 2018;4:4244–54.10.1021/acsbiomaterials.8b01051Suche in Google Scholar PubMed

[145] Kato K, Chin K, Yoshikawa T, Yamaguchi K, Tsuji Y, Esaki T, et al. Phase II study of NK105, a paclitaxel-incorporating micellar nanoparticle, for previously treated advanced or recurrent gastric cancer. Invest New Drugs. 2012;30:1621–7.10.1007/s10637-011-9709-2Suche in Google Scholar PubMed

[146] Egusquiaguirre SP, Igartua M, Hernández RM, Pedraz JL. Nanoparticle delivery systems for cancer therapy: advances in clinical and preclinical research. Clin Transl Oncol. 2012;14:83–93.10.1007/s12094-012-0766-6Suche in Google Scholar PubMed

[147] Jung KH, Kim K-P, Yoon DH, Hong YS, Choi C-M, Ahn J-H, et al. A phase I trial to determine the maximum tolerated dose and evaluate the safety and pharmacokinetics (PK) of docetaxel-PNP, polymeric nanoparticle formulation of docetaxel, in subjects with advanced solid malignancies. Journal of Clinical Oncology. 2012;30:15–23.10.1200/jco.2012.30.15_suppl.e13104Suche in Google Scholar

[148] Friedman R. Nano dot technology enters clinical trials. Journal of the National Cancer Institute. 2011;103(19):1428–9.10.1093/jnci/djr400Suche in Google Scholar PubMed

[149] Libutti SK, Paciotti GF, Byrnes AA, Alexander Jr HR, Gannon WE, Walker M, et al. Phase I and pharmacokinetic studies of CYT-6091, a novel PEGylated colloidal gold-rhTNF nanomedicine. Clin Cancer Res. 2010;16:6139–49.10.1158/1078-0432.CCR-10-0978Suche in Google Scholar PubMed PubMed Central

[150] Bhattacharyya S, Kudgus RA, Bhattacharya R, Mukherjee P. Inorganic nanoparticles in cancer therapy. Pharm Res. 2011;28:237–59.10.1007/s11095-010-0318-0Suche in Google Scholar PubMed PubMed Central

[151] Srinivasan M, Rajabi M, Mousa SA. Multifunctional nanomaterials and their applications in drug delivery and cancer therapy. Nanomaterials. 2015;5:1690–703.10.3390/nano5041690Suche in Google Scholar PubMed PubMed Central

[152] Zhao M-X, Zhu B-J. The research and applications of quantum dots as nano-carriers for targeted drug delivery and cancer therapy. Nanoscale Res Lett. 2016;11:1–9.10.1186/s11671-016-1394-9Suche in Google Scholar PubMed PubMed Central

[153] Vashist SK, Tewari R, Bajpai RP, Bharadwaj LM, Raiteri R. Review of quantum dot technologies for cancer detection and treatment. AZojono J Nanotechnol Online. 2006;2:1–14.Suche in Google Scholar

[154] Malik P, Gulia S, Kakkar R. Quantum dots for diagnosis of cancers. Adv Mater Lett. 2013;4:811–22.10.5185/amlett.2013.3437Suche in Google Scholar

[155] Voura EB, Jaiswal JK, Mattoussi H, Simon SM. Tracking metastatic tumor cell extravasation with quantum dot nanocrystals and fluorescence emission-scanning microscopy. Nat Med. 2004;10:993–8.10.1038/nm1096Suche in Google Scholar PubMed

[156] Wang K, He X, Yang X, Shi H. Functionalized silica nanoparticles: a platform for fluorescence imaging at the cell and small animal levels. Acc Chem Res. 2013;46:1367–76.10.1021/ar3001525Suche in Google Scholar PubMed

[157] Jokerst JV, Gambhir SS. Molecular imaging with theranostic nanoparticles. Acc Chem Res. 2011;44:1050–60.10.1021/ar200106eSuche in Google Scholar PubMed PubMed Central

[158] García-Pinel B, Porras-Alcalá C, Ortega-Rodríguez A, Sarabia F, Prados J, Melguizo C, et al. Lipid-based nanoparticles: application and recent advances in cancer treatment. Nanomaterials. 2019;9:638.10.3390/nano9040638Suche in Google Scholar PubMed PubMed Central

[159] Miao Y, Yang T, Yang S, Yang M, Mao C. Protein nanoparticles directed cancer imaging and therapy. Nano Convergence. 2022;9:2.10.1186/s40580-021-00293-4Suche in Google Scholar PubMed PubMed Central

[160] Chen T, Zhao T, Wei D, Wei Y, Li Y, Zhang H. Core–shell nanocarriers with ZnO quantum dots-conjugated Au nanoparticle for tumor-targeted drug delivery. Carbohydr Polym. 2013;92:1124–32.10.1016/j.carbpol.2012.10.022Suche in Google Scholar PubMed

[161] Ali MA, Mondal K, Singh C, Malhotra BD, Sharma A. Anti-epidermal growth factor receptor conjugated mesoporous zinc oxide nanofibers for breast cancer diagnostics. Nanoscale. 2015;7:7234–45.10.1039/C5NR00194CSuche in Google Scholar

[162] Xu W, Qian J, Hou G, Wang Y, Wang J, Sun T, et al. A dual-targeted hyaluronic acid-gold nanorod platform with triple-stimuli responsiveness for photodynamic/photothermal therapy of breast cancer. Acta Biomater. 2019;83:400–13.10.1016/j.actbio.2018.11.026Suche in Google Scholar PubMed

[163] Zhang X-F, Shen W, Gurunathan S. Silver nanoparticle-mediated cellular responses in various cell lines: an in vitro model. Int J Mol Sci. 2016;17:1603.10.3390/ijms17101603Suche in Google Scholar PubMed PubMed Central

[164] Yang L, Tseng Y-T, Suo G, Chen L, Yu J, Chiu W-J, et al. Photothermal therapeutic response of cancer cells to aptamer–gold nanoparticle-hybridized graphene oxide under NIR illumination. ACS Appl Mater Interfaces. 2015;7:5097–106.10.1021/am508117eSuche in Google Scholar PubMed

[165] Wu P, Gao Y, Zhang H, Cai C. Aptamer-guided silver–gold bimetallic nanostructures with highly active surface-enhanced raman scattering for specific detection and near-infrared photothermal therapy of human breast cancer cells. Anal Chem. 2012;84:7692–9.10.1021/ac3015164Suche in Google Scholar PubMed

[166] Li B, Li Q, Mo J, Dai H. Drug-loaded polymeric nanoparticles for cancer stem cell targeting. Front Pharmacology. 2017;8:51.10.3389/fphar.2017.00051Suche in Google Scholar PubMed PubMed Central

[167] Li T, Li C, Ruan Z, Xu P, Yang X, Yuan P, et al. Polypeptide-conjugated second near-infrared organic fluorophore for image-guided photothermal therapy. Acs Nano. 2019;13:3691–3702.10.1021/acsnano.9b00452Suche in Google Scholar PubMed

[168] Plewes DB, Kucharczyk W. Physics of MRI: a primer. J Magn Reson Imaging. 2012;35:1038–54.10.1002/jmri.23642Suche in Google Scholar PubMed

[169] Yoon HY, Jeon S, You DG, Park JH, Kwon IC, Koo H, et al. Inorganic nanoparticles for image-guided therapy. Bioconjugate Chem. 2017;28:124–34.10.1021/acs.bioconjchem.6b00512Suche in Google Scholar PubMed

[170] Wahsner J, Gale EM, Rodríguez-Rodríguez A, Caravan P. Chemistry of MRI contrast agents: current challenges and new frontiers. Chem Rev. 2018;119:957–1057.10.1021/acs.chemrev.8b00363Suche in Google Scholar PubMed PubMed Central

[171] Pu K, Mei J, Jokerst JV, Hong G, Antaris AL, Chattopadhyay N, et al. Diketopyrrolopyrrole‐based semiconducting polymer nanoparticles for in vivo photoacoustic imaging. Adv Mater. 2015;27:5184–90.10.1002/adma.201502285Suche in Google Scholar PubMed PubMed Central

[172] Wang X, Pang Y, Ku G, Xie X, Stoica G, Wang LV. Noninvasive laser-induced photoacoustic tomography for structural and functional in vivo imaging of the brain. Nat Biotechnol. 2003;21:803–6.10.1038/nbt839Suche in Google Scholar PubMed

[173] Kim J, Park S, Jung Y, Chang S, Park J, Zhang Y, et al. Programmable real-time clinical photoacoustic and ultrasound imaging system. Sci Rep. 2016;6:35137.10.1038/srep35137Suche in Google Scholar PubMed PubMed Central

[174] Zackrisson S, Van De Ven S, Gambhir S. Light in and sound out: emerging translational strategies for photoacoustic imaging. Cancer Res. 2014;74:979–1004.10.1158/0008-5472.CAN-13-2387Suche in Google Scholar PubMed PubMed Central

[175] Upputuri PK, Pramanik M. Recent advances toward preclinical and clinical translation of photoacoustic tomography: a review. J Biomed Opt. 2017;22:041006.10.1117/1.JBO.22.4.041006Suche in Google Scholar PubMed

[176] Fu Q, Zhu R, Song J, Yang H, Chen X. Photoacoustic imaging: contrast agents and their biomedical applications. Adv Mater. 2019;31:1805875.10.1002/adma.201805875Suche in Google Scholar PubMed

[177] Chen Y-S, Frey W, Kim S, Kruizinga P, Homan K, Emelianov S. Silica-coated gold nanorods as photoacoustic signal nanoamplifiers. Nano Lett. 2011;11:348–54.10.1021/nl1042006Suche in Google Scholar PubMed PubMed Central

[178] Zerda AD, Liu Z, Bodapati S, Teed R, Vaithilingam S, Khuri-Yakub BT, et al. Ultrahigh sensitivity carbon nanotube agents for photoacoustic molecular imaging in living mice. Nano Lett. 2010;10:2168–72.10.1021/nl100890dSuche in Google Scholar PubMed PubMed Central

[179] Moon H, Kumar D, Kim H, Sim C, Chang J-H, Kim J-M, et al. Amplified photoacoustic performance and enhanced photothermal stability of reduced graphene oxide coated gold nanorods for sensitive photoacoustic imaging. ACS Nano. 2015;9:2711–9.10.1021/nn506516pSuche in Google Scholar PubMed

[180] Liu Y, Kang N, Lv J, Zhou Z, Zhao Q, Ma L, et al. Deep photoacoustic/luminescence/magnetic resonance multimodal imaging in living subjects using high‐efficiency upconversion nanocomposites. Adv Mater. 2016;28:6411–9.10.1002/adma.201506460Suche in Google Scholar PubMed

[181] Dirheimer L, Pons T, Marchal F, Bezdetnaya L. Quantum dots mediated imaging and phototherapy in cancer spheroid models: State of the art and perspectives. Pharmaceutics. 2022;14:2136.10.3390/pharmaceutics14102136Suche in Google Scholar PubMed PubMed Central

[182] Kim G, Huang S-W, Day KC, O’Donnell M, Agayan RR, Day MA, et al. Indocyanine-green-embedded PEBBLEs as a contrast agent for photoacoustic imaging. J Biomed Opt. 2007;12:044020.10.1117/1.2771530Suche in Google Scholar PubMed

[183] Jiang Y, Upputuri PK, Xie C, Zeng Z, Sharma A, Zhen X, et al. Metabolizable semiconducting polymer nanoparticles for second near‐infrared photoacoustic imaging. Adv Mater. 2019;31:1808166.10.1002/adma.201808166Suche in Google Scholar PubMed

[184] Gujrati V, Prakash J, Malekzadeh-Najafabadi J, Stiel A, Klemm U, Mettenleiter G, et al. Bioengineered bacterial vesicles as biological nano-heaters for optoacoustic imaging. Nat Commun. 2019;10:1114.10.1038/s41467-019-09034-ySuche in Google Scholar PubMed PubMed Central

[185] Xu C, Chen F, Valdovinos HF, Jiang D, Goel S, Yu B, et al. Bacteria-like mesoporous silica-coated gold nanorods for positron emission tomography and photoacoustic imaging-guided chemo-photothermal combined therapy. Biomaterials. 2018;165:56–65.10.1016/j.biomaterials.2018.02.043Suche in Google Scholar PubMed PubMed Central

[186] Chen Y-S, Zhao Y, Yoon SJ, Gambhir SS, Emelianov S. Miniature gold nanorods for photoacoustic molecular imaging in the second near-infrared optical window. Nat Nanotechnol. 2019;14:465–72.10.1038/s41565-019-0392-3Suche in Google Scholar PubMed PubMed Central

[187] Wang Z, Zhen X, Upputuri PK, Jiang Y, Lau J, Pramanik M, et al. Redox-activatable and acid-enhanced nanotheranostics for second near-infrared photoacoustic tomography and combined photothermal tumor therapy. ACS Nano. 2019;13:5816–25.10.1021/acsnano.9b01411Suche in Google Scholar PubMed

[188] Hu D, Zhong L, Wang M, Li H, Qu Y, Liu Q, et al. Perfluorocarbon‐loaded and redox‐activatable photosensitizing agent with oxygen supply for enhancement of fluorescence/photoacoustic imaging guided tumor photodynamic therapy. Adv Funct Mater. 2019;29:1806199.10.1002/adfm.201806199Suche in Google Scholar

[189] Cao Z, Feng L, Zhang G, Wang J, Shen S, Li D, et al. Semiconducting polymer-based nanoparticles with strong absorbance in NIR-II window for in vivo photothermal therapy and photoacoustic imaging. Biomaterials. 2018;155:103–11.10.1016/j.biomaterials.2017.11.016Suche in Google Scholar PubMed

[190] Liu L, Chen Q, Wen L, Li C, Qin H, Xing D. Photoacoustic therapy for precise eradication of glioblastoma with a tumor site blood–brain barrier permeability upregulating nanoparticle. Adv Funct Mater. 2019;29:1808601.10.1002/adfm.201808601Suche in Google Scholar

[191] Jiang Y, Pu K. Advanced photoacoustic imaging applications of near‐infrared absorbing organic nanoparticles. Small. 2017;13:1700710.10.1002/smll.201700710Suche in Google Scholar PubMed

[192] Yang Z, Sun N, Cheng R, Zhao C, Liu Z, Li X, et al. pH multistage responsive micellar system with charge-switch and PEG layer detachment for co-delivery of paclitaxel and curcumin to synergistically eliminate breast cancer stem cells. Biomaterials. 2017;147:53–67.10.1016/j.biomaterials.2017.09.013Suche in Google Scholar PubMed

[193] Atkinson RL, Zhang M, Diagaradjane P, Peddibhotla S, Contreras A, Hilsenbeck SG, et al. Thermal enhancement with optically activated gold nanoshells sensitizes breast cancer stem cells to radiation therapy. Sci Transl Med. 2010;2:55ra79.10.1126/scitranslmed.3001447Suche in Google Scholar PubMed PubMed Central

[194] Paholak HJ, Stevers NO, Chen H, Burnett JP, He M, Korkaya H, et al. Elimination of epithelial-like and mesenchymal-like breast cancer stem cells to inhibit metastasis following nanoparticle-mediated photothermal therapy. Biomaterials. 2016;104:145–57.10.1016/j.biomaterials.2016.06.045Suche in Google Scholar PubMed PubMed Central

[195] Banchereau J, Palucka K. Cancer vaccines on the move. Nat Rev Clin Oncol. 2018;15:9–10.10.1038/nrclinonc.2017.149Suche in Google Scholar PubMed

[196] Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12:252–64.10.1038/nrc3239Suche in Google Scholar PubMed PubMed Central

[197] Evans ER, Bugga P, Asthana V, Drezek R. Metallic nanoparticles for cancer immunotherapy. Mater Today. 2018;21:673–85.10.1016/j.mattod.2017.11.022Suche in Google Scholar PubMed PubMed Central

[198] Kobayashi H, Choyke PL. Near-infrared photoimmunotherapy of cancer. Acc Chem Res. 2019;52:2332–9.10.1021/acs.accounts.9b00273Suche in Google Scholar PubMed PubMed Central

[199] Mahoney KM, Rennert PD, Freeman GJ. Combination cancer immunotherapy and new immunomodulatory targets. Nat Rev Drug Discovery. 2015;14:561–84.10.1038/nrd4591Suche in Google Scholar PubMed

[200] Ghassemi S, Carreno BM. Heating up cancer vaccines. Sci Immunol. 2017;2:eaap9419.10.1126/sciimmunol.aap9419Suche in Google Scholar PubMed

[201] Liu X, Liu C, Zheng Z, Chen S, Pang X, Xiang X, et al. Vesicular antibodies: a bioactive multifunctional combination platform for targeted therapeutic delivery and cancer immunotherapy. Adv Mater. 2019;31:1808294.10.1002/adma.201808294Suche in Google Scholar PubMed

[202] Rajendrakumar SK, Uthaman S, Cho C-S, Park I-K. Nanoparticle-based phototriggered cancer immunotherapy and its domino effect in the tumor microenvironment. Biomacromolecules. 2018;19:1869–87.10.1021/acs.biomac.8b00460Suche in Google Scholar PubMed

[203] Yang Y, Zhu W, Dong Z, Chao Y, Xu L, Chen M, et al. 1D coordination polymer nanofibers for low‐temperature photothermal therapy. Adv Mater. 2017;29:1703588.10.1002/adma.201703588Suche in Google Scholar PubMed

[204] Moy AJ, Tunnell JW. Combinatorial immunotherapy and nanoparticle mediated hyperthermia. Adv Drug Delivery Rev. 2017;114:175–83.10.1016/j.addr.2017.06.008Suche in Google Scholar PubMed

[205] Guo L, Yan DD, Yang D, Li Y, Wang X, Zalewski O, et al. Combinatorial photothermal and immuno cancer therapy using chitosan-coated hollow copper sulfide nanoparticles. ACS Nano. 2014;8:5670–81.10.1021/nn5002112Suche in Google Scholar PubMed PubMed Central

[206] Yata T, Takahashi Y, Tan M, Nakatsuji H, Ohtsuki S, Murakami T, et al. DNA nanotechnology-based composite-type gold nanoparticle-immunostimulatory DNA hydrogel for tumor photothermal immunotherapy. Biomaterials. 2017;146:136–45.10.1016/j.biomaterials.2017.09.014Suche in Google Scholar PubMed

[207] Ye Y, Wang C, Zhang X, Hu Q, Zhang Y, Liu Q, et al. A melanin-mediated cancer immunotherapy patch. Sci Immunol. 2017;2:eaan5692.10.1126/sciimmunol.aan5692Suche in Google Scholar PubMed

[208] Pan J, Wang Y, Zhang C, Wang X, Wang H, Wang J, et al. Antigen‐directed fabrication of a multifunctional nanovaccine with ultrahigh antigen loading efficiency for tumor photothermal‐immunotherapy. Adv Mater. 2018;30:1704408.10.1002/adma.201704408Suche in Google Scholar PubMed

[209] Nahhas AF, Webster TJ. A review of treating viral outbreaks with self-assembled nanomaterial-like peptides: From Ebola to the Marburg virus. OpenNano. 2022;8:100094.10.1016/j.onano.2022.100094Suche in Google Scholar

[210] Wang C, Xu L, Liang C, Xiang J, Peng R, Liu Z. Immunological responses triggered by photothermal therapy with carbon nanotubes in combination with anti‐CTLA‐4 therapy to inhibit cancer metastasis. Adv Mater. 2014;26:8154–62.10.1002/adma.201402996Suche in Google Scholar PubMed

[211] Chen Q, Xu L, Liang C, Wang C, Peng R, Liu Z. Photothermal therapy with immune-adjuvant nanoparticles together with checkpoint blockade for effective cancer immunotherapy. Nat Commun. 2016;7:13193.10.1038/ncomms13193Suche in Google Scholar PubMed PubMed Central

[212] Liu WL, Zou MZ, Liu T, Zeng JY, Li X, Yu WY, et al. Expandable immunotherapeutic nanoplatforms engineered from cytomembranes of hybrid cells derived from cancer and dendritic cells. Adv Mater. 2019;31:1900499.10.1002/adma.201900499Suche in Google Scholar PubMed

[213] Panaretakis T, Kepp O, Brockmeier U, Tesniere A, Bjorklund AC, Chapman DC, et al. Mechanisms of pre‐apoptotic calreticulin exposure in immunogenic cell death. EMBO J. 2009;28:578–90.10.1038/emboj.2009.1Suche in Google Scholar PubMed PubMed Central

[214] Obeid M, Tesniere A, Ghiringhelli F, Fimia GM, Apetoh L, Perfettini J-L, et al. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med. 2007;13:54–61.10.1038/nm1523Suche in Google Scholar PubMed

[215] Wang M, Song J, Zhou F, Hoover AR, Murray C, Zhou B, et al. Nir‐triggered phototherapy and immunotherapy via an antigen‐capturing nanoplatform for metastatic cancer treatment. Adv Sci. 2019;6:1802157.10.1002/advs.201802157Suche in Google Scholar PubMed PubMed Central

[216] Kleinovink JW, van Driel PB, Snoeks TJ, Prokopi N, Fransen MF, Cruz LJ, et al. Combination of photodynamic therapy and specific immunotherapy efficiently eradicates established tumors. Clin Cancer Res. 2016;22:1459–68.10.1158/1078-0432.CCR-15-0515Suche in Google Scholar PubMed

[217] Wang D, Wang T, Liu J, Yu H, Jiao S, Feng B, et al. Acid-activatable versatile micelleplexes for PD-L1 blockade-enhanced cancer photodynamic immunotherapy. Nano Lett. 2016;16:5503–13.10.1021/acs.nanolett.6b01994Suche in Google Scholar PubMed

[218] He C, Duan X, Guo N, Chan C, Poon C, Weichselbaum RR, et al. Core-shell nanoscale coordination polymers combine chemotherapy and photodynamic therapy to potentiate checkpoint blockade cancer immunotherapy. Nat Commun. 2016;7:12499.10.1038/ncomms12499Suche in Google Scholar PubMed PubMed Central

[219] Lan G, Ni K, Xu Z, Veroneau SS, Song Y, Lin W. Nanoscale metal–organic framework overcomes hypoxia for photodynamic therapy primed cancer immunotherapy. J Am Chem Soc. 2018;140:5670–3.10.1021/jacs.8b01072Suche in Google Scholar PubMed PubMed Central

[220] Xu J, Xu L, Wang C, Yang R, Zhuang Q, Han X, et al. Near-infrared-triggered photodynamic therapy with multitasking upconversion nanoparticles in combination with checkpoint blockade for immunotherapy of colorectal cancer. ACS Nano. 2017;11:4463–74.10.1021/acsnano.7b00715Suche in Google Scholar PubMed

[221] Fan Q, Chen Z, Wang C, Liu Z. Toward biomaterials for enhancing immune checkpoint blockade therapy. Adv Funct Mater. 2018;28:1802540.10.1002/adfm.201802540Suche in Google Scholar

[222] Chen W. IDO: more than an enzyme. Nat Immunol. 2011;12:809–11.10.1038/ni.2088Suche in Google Scholar PubMed

[223] Amobi A, Qian F, Lugade AA, Odunsi K. Tryptophan catabolism and cancer immunotherapy targeting IDO mediated immune suppression. Tumor Immune Microenviron Cancer Progression Cancer Ther. 2017;136:129–44.10.1007/978-3-319-67577-0_9Suche in Google Scholar PubMed

[224] Song W, Kuang J, Li C-X, Zhang M, Zheng D, Zeng X, et al. Enhanced immunotherapy based on photodynamic therapy for both primary and lung metastasis tumor eradication. ACS Nano. 2018;12:1978–89.10.1021/acsnano.7b09112Suche in Google Scholar PubMed

[225] Kirsanova DY, Gadzhimagomedova ZM, Maksimov AY, Soldatov AV. Nanomaterials for deep tumor treatment. Mini Rev Med Chem. 2021;21:677–88.10.2174/1389557520666201111161705Suche in Google Scholar PubMed

[226] Yan J, Zhang N, Zhang Z, Zhu W, Li B, Li L, et al. Redox-responsive polyethyleneimine/tetrahedron DNA/doxorubicin nanocomplexes for deep cell/tissue penetration to overcome multidrug resistance. J Controlled Rel. 2021;329:36–49.10.1016/j.jconrel.2020.11.050Suche in Google Scholar PubMed

[227] Yao Y, Dai X, Tan Y, Chen Y, Liao C, Yang T, et al. Deep drug penetration of nanodrug aggregates at tumor tissues by fast extracellular drug release. Adv Healthcare Mater. 2021;10:2001430.10.1002/adhm.202001430Suche in Google Scholar PubMed

[228] Zhou L, Pi W, Hao M, Li Y, An H, Li Q, et al. An injectable and biodegradable nano-photothermal DNA hydrogel enhances penetration and efficacy of tumor therapy. Biomater Sci. 2021;9:4904–21.10.1039/D1BM00568ESuche in Google Scholar PubMed

[229] Pan X, Li P, Bai L, Ma J, Li S, Zhang F, et al. Biodegradable nanocomposite with dual cell‐tissue penetration for deep tumor chemo‐phototherapy. Small. 2020;16:2000809.10.1002/smll.202000809Suche in Google Scholar PubMed

[230] Wang H, Wang Z, Tu Y, Li Y, Xu T, Yang M, et al. Homotypic targeting upconversion nano-reactor for cascade cancer starvation and deep-tissue phototherapy. Biomaterials. 2020;235:119765.10.1016/j.biomaterials.2020.119765Suche in Google Scholar PubMed

[231] Wang Y, Wang B, Zhang L, Huang J, Li P, Zhao Y, et al. Mitochondria-targeted nanospheres with deep tumor penetration for photo/starvation therapy. J Mater Chem B. 2020;8:7740–54.10.1039/D0TB00001ASuche in Google Scholar

[232] Sun W, Zhou Z, Pratx G, Chen X, Chen H. Nanoscintillator-mediated X-ray induced photodynamic therapy for deep-seated tumors: from concept to biomedical applications. Theranostics. 2020;10:1296.10.7150/thno.41578Suche in Google Scholar PubMed PubMed Central

[233] Sivasubramanian M, Chuang YC, Lo L-W. Evolution of nanoparticle-mediated photodynamic therapy: from superficial to deep-seated cancers. Molecules. 2019;24:520.10.3390/molecules24030520Suche in Google Scholar PubMed PubMed Central

[234] Fan W, Tang W, Lau J, Shen Z, Xie J, Shi J, et al. Breaking the depth dependence by nanotechnology‐enhanced X‐ray‐excited deep cancer theranostics. Adv Mater. 2019;31:1806381.10.1002/adma.201806381Suche in Google Scholar PubMed

[235] Wang Z, Meng Q, Li S. The role of NIR fluorescence in MDR cancer treatment: from targeted imaging to phototherapy. Curr Med Chem. 2020;27:5510–29.10.2174/0929867326666190627123719Suche in Google Scholar PubMed

[236] Yang Z, Du Y, Sun Q, Peng Y, Wang R, Zhou Y, et al. Albumin-based nanotheranostic probe with hypoxia alleviating potentiates synchronous multimodal imaging and phototherapy for glioma. ACS Nano. 2020;14:6191–212.10.1021/acsnano.0c02249Suche in Google Scholar PubMed

[237] Pan J, Ouyang A, Fang W, Cheng G, Liu W, Wang F, et al. cis-Silicon phthalocyanine conformation endows J-aggregated nanosphere with unique near-infrared absorbance and fluorescence enhancement: a tumor sensitive phototheranostic agent with deep tissue penetrating ability. J Mater Chem B. 2020;8:2895–908.10.1039/D0TB00192ASuche in Google Scholar PubMed

[238] Shu M, Tang J, Chen L, Zeng Q, Li C, Xiao S, et al. Tumor microenvironment triple-responsive nanoparticles enable enhanced tumor penetration and synergetic chemo-photodynamic therapy. Biomaterials. 2021;268:120574.10.1016/j.biomaterials.2020.120574Suche in Google Scholar PubMed

[239] Jia P, Ji H, Liu S, Zhang R, He F, Zhong L, et al. Integration of IR-808 and thiol-capped Au–Bi bimetallic nanoparticles for NIR light mediated photothermal/photodynamic therapy and imaging. J Mater Chem B. 2021;9:101–11.10.1039/D0TB02378GSuche in Google Scholar

[240] Huang G, Qiu Y, Yang F, Xie J, Chen X, Wang L, et al. Magnetothermally triggered free-radical generation for deep-seated tumor treatment. Nano Lett. 2021;21:2926–31.10.1021/acs.nanolett.1c00009Suche in Google Scholar PubMed

[241] Grześkowiak BF, Maziukiewicz D, Kozłowska A, Kertmen A, Coy E, Mrówczyński R. Polyamidoamine dendrimers decorated multifunctional polydopamine nanoparticles for targeted chemo-and photothermal therapy of liver cancer model. Int J Mol Sci. 2021;22:738.10.3390/ijms22020738Suche in Google Scholar PubMed PubMed Central

[242] Perumal D, Golla M, Pillai KS, Raj G, PK AK, Varghese R. Biotin-decorated NIR-absorbing nanosheets for targeted photodynamic cancer therapy. Org Biomol Chem. 2021;19:2804–10.10.1039/D1OB00002KSuche in Google Scholar

[243] Liu R, Gao Y, Liu N, Suo Y. Nanoparticles loading porphyrin sensitizers in improvement of photodynamic therapy for ovarian cancer. Photodiagn Photodyn Ther. 2021;33:102156.10.1016/j.pdpdt.2020.102156Suche in Google Scholar PubMed

[244] Liu R, Xu F, Wang L, Liu M, Cao X, Shi X, et al. Polydopamine-coated laponite nanoplatforms for photoacoustic imaging-guided chemo-phototherapy of breast cancer. Nanomaterials. 2021;11:394.10.3390/nano11020394Suche in Google Scholar PubMed PubMed Central

[245] Lu K-Y, Jheng P-R, Lu L-S, Rethi L, Mi F-L, Chuang E-Y. Enhanced anticancer effect of ROS-boosted photothermal therapy by using fucoidan-coated polypyrrole nanoparticles. Int J Biol Macromol. 2021;166:98–107.10.1016/j.ijbiomac.2020.10.091Suche in Google Scholar PubMed

[246] Gu Y, Zhou Y, Wu Y, Ma J, Wang Y, Wang Q, et al. Apoferritin-engineered nanoprobe for tumor-targeted triple-NIR imaging and phototherapy. Anal Chem. 2021;93:8835–45.10.1021/acs.analchem.1c00730Suche in Google Scholar PubMed

[247] Yang Y, Yun K, Li Y, Zhang L, Zhao W, Zhu Z, et al. Self-assembled multifunctional polymeric micelles for tumor-specific bioimaging and synergistic chemo-phototherapy of cancer. Int J Pharmaceutics. 2021;602:120651.10.1016/j.ijpharm.2021.120651Suche in Google Scholar PubMed

[248] Yan J, Gao T, Lu Z, Yin J, Zhang Y, Pei R. Aptamer-targeted photodynamic platforms for tumor therapy. ACS Appl Mater Interfaces. 2021;13:27749–73.10.1021/acsami.1c06818Suche in Google Scholar PubMed

[249] Tao Y-k, Hou X-y, Gao H, Zhang X, Zuo F-m, Wang Y, et al. Grade-targeted nanoparticles for improved hypoxic tumor microenvironment and enhanced photodynamic cancer therapy. Nanomedicine. 2021;16:221–35.10.2217/nnm-2020-0096Suche in Google Scholar PubMed

[250] Xia L, Meng X, Wen L, Zhou N, Liu T, Xu X, et al. A highly specific multiple enhancement theranostic nanoprobe for PET/MRI/PAI image‐guided radioisotope combined photothermal therapy in prostate cancer. Small. 2021;17:2100378.10.1002/smll.202100378Suche in Google Scholar PubMed

[251] Liu X, Wang C, Wang X, Tian C, Shen Y, Zhu M. A dual-targeting Fe3O4@C/ZnO-DOX-FA nanoplatform with pH-responsive drug release and synergetic chemo-photothermal antitumor in vitro and in vivo. Mater Sci Eng: C. 2021;118:111455.10.1016/j.msec.2020.111455Suche in Google Scholar PubMed

[252] Wang R, Yang H, Khan AR, Yang X, Xu J, Ji J, et al. Redox-responsive hyaluronic acid-based nanoparticles for targeted photodynamic therapy/chemotherapy against breast cancer. J Colloid Interface Sci. 2021;598:213–28.10.1016/j.jcis.2021.04.056Suche in Google Scholar PubMed

[253] Wang Y, Li M, Luo T, Jiao M, Jin S, Dou P, et al. Development of FL/MR dual-modal Au nanobipyramids for targeted cancer imaging and photothermal therapy. Mater Sci Eng: C. 2021;127:112190.10.1016/j.msec.2021.112190Suche in Google Scholar PubMed

[254] Kong Y, Li X, Liu X, Pang J, Mu X, Liu W. Galactosylated chitosan modified magnetic mesoporous silica nanoparticles loaded with nedaplatin for the targeted chemo-photothermal synergistic therapy of cancer. J Nanosci Nanotechnol. 2021;21:4553–64.10.1166/jnn.2021.19142Suche in Google Scholar PubMed

[255] Zeng L, Cheng H, Dai Y, Su Z, Wang C, Lei L, et al. In vivo regenerable cerium oxide nanozyme-loaded pH/H₂O₂-responsive nanovesicle for tumor-targeted photothermal and photodynamic therapies. ACS Appl Mater Interfaces. 2021;13(1): 233–44.10.1021/acsami.0c19074Suche in Google Scholar PubMed

[256] Xie M, Li J, Deng T, Yang N, Yang M. Modification of magnetic molybdenum disulfide by chitosan/carboxymethylcellulose with enhanced dispersibility for targeted photothermal-/chemotherapy of cancer. J Mater Chem B. 2021;9:1833–45.10.1039/D0TB01664KSuche in Google Scholar

[257] Gao Y, Zhao Q, Xiao M, Huang X, Wu X. A versatile photothermal vaccine based on acid-responsive glyco-nanoplatform for synergistic therapy of cancer. Biomaterials. 2021;273:120792.10.1016/j.biomaterials.2021.120792Suche in Google Scholar PubMed PubMed Central

[258] Neelgund GM, Oki A, Bandara S, Carson L. Photothermal effect and cytotoxicity of CuS nanoflowers deposited over folic acid conjugated nanographene oxide. J Mater Chem B. 2021;9:1792–803.10.1039/D0TB02366CSuche in Google Scholar

[259] Ngen EJ, Chen Y, Azad BB, Boinapally S, Jacob D, Lisok A, et al. Prostate-specific membrane antigen (PSMA)-targeted photodynamic therapy enhances the delivery of PSMA-targeted magnetic nanoparticles to PSMA-expressing prostate tumors. Nanotheranostics. 2021;5:182.10.7150/ntno.52361Suche in Google Scholar PubMed PubMed Central

[260] Sears J, Swanner J, Fahrenholtz CD, Snyder C, Rohde M, Levi-Polyachenko N, et al. Combined photothermal and ionizing radiation sensitization of triple-negative breast cancer using triangular silver nanoparticles. Int J Nanomed. 2021;16:851–65.10.2147/IJN.S296513Suche in Google Scholar PubMed PubMed Central

[261] Li J, Cui D, Huang J, He S, Yang Z, Zhang Y, et al. Organic semiconducting pro‐nanostimulants for near‐infrared photoactivatable cancer immunotherapy. Angew Chem Int Ed. 2019;58:12680–87.10.1002/anie.201906288Suche in Google Scholar PubMed

[262] Li J, Cui D, Jiang Y, Huang J, Cheng P, Pu K. Near‐infrared photoactivatable semiconducting polymer nanoblockaders for metastasis‐inhibited combination cancer therapy. Adv Mater. 2019;31:1905091.10.1002/adma.201905091Suche in Google Scholar PubMed

[263] Ren X, Lin J, Wang X, Liu X, Meng E, Zhang R, et al. Photoactivatable RNAi for cancer gene therapy triggered by near-infrared-irradiated single-walled carbon nanotubes. Int J Nanomed. 2017;7885–96.10.2147/IJN.S141882Suche in Google Scholar PubMed PubMed Central

[264] Zhang Q, Kuang G, Zhou D, Qi Y, Wang M, Li X, et al. Photoactivated polyprodrug nanoparticles for effective light-controlled Pt (IV) and siRNA codelivery to achieve synergistic cancer therapy. J Mater Chem B. 2020;8:5903–11.10.1039/D0TB01103GSuche in Google Scholar

[265] Chen M, Sun W, Kretzschmann A, Butt H-J, Wu S. Nanostructured polymer assemblies stabilize photoactivatable anticancer ruthenium complexes under physiological conditions. J Inorg Biochem. 2020;207:111052.10.1016/j.jinorgbio.2020.111052Suche in Google Scholar PubMed

[266] Huang L, Chen X, Bian Q, Zhang F, Wu H, Wang H, et al. Photosensitizer-stabilized self-assembling nanoparticles potentiate chemo/photodynamic efficacy of patient-derived melanoma. J Controlled Rel. 2020;328:325–38.10.1016/j.jconrel.2020.08.062Suche in Google Scholar PubMed

[267] Uthaman S, Pillarisetti S, Mathew AP, Kim Y, Bae WK, Huh KM, et al. Long circulating photoactivable nanomicelles with tumor localized activation and ROS triggered self-accelerating drug release for enhanced locoregional chemo-photodynamic therapy. Biomaterials. 2020;232:119702.10.1016/j.biomaterials.2019.119702Suche in Google Scholar PubMed

[268] Dai L, Shen G, Wang Y, Yang P, Wang H, Liu Z. PSMA-targeted melanin-like nanoparticles as a multifunctional nanoplatform for prostate cancer theranostics. J Mater Chem B. 2021;9:1151–61.10.1039/D0TB02576CSuche in Google Scholar PubMed

[269] Jin X, Yang H, Mao Z, Wang B. Cathepsin B-responsive multifunctional peptide conjugated gold nanorods for mitochondrial targeting and precise photothermal cancer therapy. J Colloid Interface Sci. 2021;601:714–26.10.1016/j.jcis.2021.05.135Suche in Google Scholar PubMed

[270] Geng Z, Chen F, Wang X, Wang L, Pang Y, Liu J. Combining anti-PD-1 antibodies with Mn2+ -drug coordinated multifunctional nanoparticles for enhanced cancer therapy. Biomaterials. 2021;275:120897.10.1016/j.biomaterials.2021.120897Suche in Google Scholar PubMed

[271] Fu L, Yang S, Jiang S, Zhou X, Sha Z, He C. One-step synthesis of multifunctional nanoparticles for CT/PA imaging guided breast cancer photothermal therapy. Colloids Surf B: Biointerfaces. 2021;201:111630.10.1016/j.colsurfb.2021.111630Suche in Google Scholar PubMed

[272] Liu J, Ding G, Chen S, Xue C, Chen M, Wu X, et al. Multifunctional programmable DNA nanotrain for activatable hypoxia imaging and mitochondrion-targeted enhanced photodynamic therapy. ACS Appl Mater Interfaces. 2021;13:9681–90.10.1021/acsami.0c21681Suche in Google Scholar PubMed

[273] Zhang W, Dang G, Dong J, Li Y, Jiao P, Yang M, et al. A multifunctional nanoplatform based on graphitic carbon nitride quantum dots for imaging-guided and tumor-targeted chemo-photodynamic combination therapy. Colloids Surf B: Biointerfaces. 2021;199:111549.10.1016/j.colsurfb.2020.111549Suche in Google Scholar PubMed

[274] Khan ME, Mohammad A, Yoon T. State-of-the-art developments in carbon quantum dots (CQDs): Photo-catalysis, bio-imaging, and bio-sensing applications. Chemosphere. 2022;302:134815.10.1016/j.chemosphere.2022.134815Suche in Google Scholar PubMed

[275] Wang X, Liu Z, Jin R, Cai B, Liu S, Bai Y, et al. Multifunctional hierarchical nanohybrids perform triple antitumor theranostics in a cascaded manner for effective tumor treatment. Acta Biomater. 2021;128:408–19.10.1016/j.actbio.2021.04.019Suche in Google Scholar PubMed

[276] Guo Y, Zheng X, Gai T, Wei Z, Zhang S. Co-biomembrane-coated Fe3O4/MnO2 multifunctional nanoparticles for targeted delivery and enhanced chemodynamic/photothermal/chemo therapy. Chem Commun. 2021;57:5754–7.10.1039/D1CC01375KSuche in Google Scholar PubMed

[277] Tang K, Wang W, Song Z, Luo X. Multifunctional nano-biosensor based on metal-organic framework for enhanced fluorescence imaging of intracellular miRNA-122 and synergistic chemo-photothermal therapy of tumor cells. Anal Chim Acta. 2021;1176:338779.10.1016/j.aca.2021.338779Suche in Google Scholar PubMed

Received: 2023-11-27
Revised: 2024-03-24
Accepted: 2024-04-08
Published Online: 2024-05-24

© 2024 the author(s), published by De Gruyter

This work is licensed under the Creative Commons Attribution 4.0 International License.

Artikel in diesem Heft

  1. Research Articles
  2. Tension buckling and postbuckling of nanocomposite laminated plates with in-plane negative Poisson’s ratio
  3. Polyvinylpyrrolidone-stabilised gold nanoparticle coatings inhibit blood protein adsorption
  4. Energy and mass transmission through hybrid nanofluid flow passing over a spinning sphere with magnetic effect and heat source/sink
  5. Surface treatment with nano-silica and magnesium potassium phosphate cement co-action for enhancing recycled aggregate concrete
  6. Numerical investigation of thermal radiation with entropy generation effects in hybrid nanofluid flow over a shrinking/stretching sheet
  7. Enhancing the performance of thermal energy storage by adding nano-particles with paraffin phase change materials
  8. Using nano-CaCO3 and ceramic tile waste to design low-carbon ultra high performance concrete
  9. Numerical analysis of thermophoretic particle deposition in a magneto-Marangoni convective dusty tangent hyperbolic nanofluid flow – Thermal and magnetic features
  10. Dual numerical solutions of Casson SA–hybrid nanofluid toward a stagnation point flow over stretching/shrinking cylinder
  11. Single flake homo p–n diode of MoTe2 enabled by oxygen plasma doping
  12. Electrostatic self-assembly effect of Fe3O4 nanoparticles on performance of carbon nanotubes in cement-based materials
  13. Multi-scale alignment to buried atom-scale devices using Kelvin probe force microscopy
  14. Antibacterial, mechanical, and dielectric properties of hydroxyapatite cordierite/zirconia porous nanocomposites for use in bone tissue engineering applications
  15. Time-dependent Darcy–Forchheimer flow of Casson hybrid nanofluid comprising the CNTs through a Riga plate with nonlinear thermal radiation and viscous dissipation
  16. Durability prediction of geopolymer mortar reinforced with nanoparticles and PVA fiber using particle swarm optimized BP neural network
  17. Utilization of zein nano-based system for promoting antibiofilm and anti-virulence activities of curcumin against Pseudomonas aeruginosa
  18. Antibacterial effect of novel dental resin composites containing rod-like zinc oxide
  19. An extended model to assess Jeffery–Hamel blood flow through arteries with iron-oxide (Fe2O3) nanoparticles and melting effects: Entropy optimization analysis
  20. Comparative study of copper nanoparticles over radially stretching sheet with water and silicone oil
  21. Cementitious composites modified by nanocarbon fillers with cooperation effect possessing excellent self-sensing properties
  22. Confinement size effect on dielectric properties, antimicrobial activity, and recycling of TiO2 quantum dots via photodegradation processes of Congo red dye and real industrial textile wastewater
  23. Biogenic silver nanoparticles of Moringa oleifera leaf extract: Characterization and photocatalytic application
  24. Novel integrated structure and function of Mg–Gd neutron shielding materials
  25. Impact of multiple slips on thermally radiative peristaltic transport of Sisko nanofluid with double diffusion convection, viscous dissipation, and induced magnetic field
  26. Magnetized water-based hybrid nanofluid flow over an exponentially stretching sheet with thermal convective and mass flux conditions: HAM solution
  27. A numerical investigation of the two-dimensional magnetohydrodynamic water-based hybrid nanofluid flow composed of Fe3O4 and Au nanoparticles over a heated surface
  28. Development and modeling of an ultra-robust TPU-MWCNT foam with high flexibility and compressibility
  29. Effects of nanofillers on the physical, mechanical, and tribological behavior of carbon/kenaf fiber–reinforced phenolic composites
  30. Polymer nanocomposite for protecting photovoltaic cells from solar ultraviolet in space
  31. Study on the mechanical properties and microstructure of recycled concrete reinforced with basalt fibers and nano-silica in early low-temperature environments
  32. Synergistic effect of carbon nanotubes and polyvinyl alcohol on the mechanical performance and microstructure of cement mortar
  33. CFD analysis of paraffin-based hybrid (Co–Au) and trihybrid (Co–Au–ZrO2) nanofluid flow through a porous medium
  34. Forced convective tangent hyperbolic nanofluid flow subject to heat source/sink and Lorentz force over a permeable wedge: Numerical exploration
  35. Physiochemical and electrical activities of nano copper oxides synthesised via hydrothermal method utilising natural reduction agents for solar cell application
  36. A homotopic analysis of the blood-based bioconvection Carreau–Yasuda hybrid nanofluid flow over a stretching sheet with convective conditions
  37. In situ synthesis of reduced graphene oxide/SnIn4S8 nanocomposites with enhanced photocatalytic performance for pollutant degradation
  38. A coarse-grained Poisson–Nernst–Planck model for polyelectrolyte-modified nanofluidic diodes
  39. A numerical investigation of the magnetized water-based hybrid nanofluid flow over an extending sheet with a convective condition: Active and passive controls of nanoparticles
  40. The LyP-1 cyclic peptide modified mesoporous polydopamine nanospheres for targeted delivery of triptolide regulate the macrophage repolarization in atherosclerosis
  41. Synergistic effect of hydroxyapatite-magnetite nanocomposites in magnetic hyperthermia for bone cancer treatment
  42. The significance of quadratic thermal radiative scrutinization of a nanofluid flow across a microchannel with thermophoretic particle deposition effects
  43. Ferromagnetic effect on Casson nanofluid flow and transport phenomena across a bi-directional Riga sensor device: Darcy–Forchheimer model
  44. Performance of carbon nanomaterials incorporated with concrete exposed to high temperature
  45. Multicriteria-based optimization of roller compacted concrete pavement containing crumb rubber and nano-silica
  46. Revisiting hydrotalcite synthesis: Efficient combined mechanochemical/coprecipitation synthesis to design advanced tunable basic catalysts
  47. Exploration of irreversibility process and thermal energy of a tetra hybrid radiative binary nanofluid focusing on solar implementations
  48. Effect of graphene oxide on the properties of ternary limestone clay cement paste
  49. Improved mechanical properties of graphene-modified basalt fibre–epoxy composites
  50. Sodium titanate nanostructured modified by green synthesis of iron oxide for highly efficient photodegradation of dye contaminants
  51. Green synthesis of Vitis vinifera extract-appended magnesium oxide NPs for biomedical applications
  52. Differential study on the thermal–physical properties of metal and its oxide nanoparticle-formed nanofluids: Molecular dynamics simulation investigation of argon-based nanofluids
  53. Heat convection and irreversibility of magneto-micropolar hybrid nanofluids within a porous hexagonal-shaped enclosure having heated obstacle
  54. Numerical simulation and optimization of biological nanocomposite system for enhanced oil recovery
  55. Laser ablation and chemical vapor deposition to prepare a nanostructured PPy layer on the Ti surface
  56. Cilostazol niosomes-loaded transdermal gels: An in vitro and in vivo anti-aggregant and skin permeation activity investigations towards preparing an efficient nanoscale formulation
  57. Linear and nonlinear optical studies on successfully mixed vanadium oxide and zinc oxide nanoparticles synthesized by sol–gel technique
  58. Analytical investigation of convective phenomena with nonlinearity characteristics in nanostratified liquid film above an inclined extended sheet
  59. Optimization method for low-velocity impact identification in nanocomposite using genetic algorithm
  60. Analyzing the 3D-MHD flow of a sodium alginate-based nanofluid flow containing alumina nanoparticles over a bi-directional extending sheet using variable porous medium and slip conditions
  61. A comprehensive study of laser irradiated hydrothermally synthesized 2D layered heterostructure V2O5(1−x)MoS2(x) (X = 1–5%) nanocomposites for photocatalytic application
  62. Computational analysis of water-based silver, copper, and alumina hybrid nanoparticles over a stretchable sheet embedded in a porous medium with thermophoretic particle deposition effects
  63. A deep dive into AI integration and advanced nanobiosensor technologies for enhanced bacterial infection monitoring
  64. Effects of normal strain on pyramidal I and II 〈c + a〉 screw dislocation mobility and structure in single-crystal magnesium
  65. Computational study of cross-flow in entropy-optimized nanofluids
  66. Significance of nanoparticle aggregation for thermal transport over magnetized sensor surface
  67. A green and facile synthesis route of nanosize cupric oxide at room temperature
  68. Effect of annealing time on bending performance and microstructure of C19400 alloy strip
  69. Chitosan-based Mupirocin and Alkanna tinctoria extract nanoparticles for the management of burn wound: In vitro and in vivo characterization
  70. Electrospinning of MNZ/PLGA/SF nanofibers for periodontitis
  71. Photocatalytic degradation of methylene blue by Nd-doped titanium dioxide thin films
  72. Shell-core-structured electrospinning film with sequential anti-inflammatory and pro-neurogenic effects for peripheral nerve repairment
  73. Flow and heat transfer insights into a chemically reactive micropolar Williamson ternary hybrid nanofluid with cross-diffusion theory
  74. One-pot fabrication of open-spherical shapes based on the decoration of copper sulfide/poly-O-amino benzenethiol on copper oxide as a promising photocathode for hydrogen generation from the natural source of Red Sea water
  75. A penta-hybrid approach for modeling the nanofluid flow in a spatially dependent magnetic field
  76. Advancing sustainable agriculture: Metal-doped urea–hydroxyapatite hybrid nanofertilizer for agro-industry
  77. Utilizing Ziziphus spina-christi for eco-friendly synthesis of silver nanoparticles: Antimicrobial activity and promising application in wound healing
  78. Plant-mediated synthesis, characterization, and evaluation of a copper oxide/silicon dioxide nanocomposite by an antimicrobial study
  79. Effects of PVA fibers and nano-SiO2 on rheological properties of geopolymer mortar
  80. Investigating silver and alumina nanoparticles’ impact on fluid behavior over porous stretching surface
  81. Potential pharmaceutical applications and molecular docking study for green fabricated ZnO nanoparticles mediated Raphanus sativus: In vitro and in vivo study
  82. Effect of temperature and nanoparticle size on the interfacial layer thickness of TiO2–water nanofluids using molecular dynamics
  83. Characteristics of induced magnetic field on the time-dependent MHD nanofluid flow through parallel plates
  84. Flexural and vibration behaviours of novel covered CFRP composite joints with an MWCNT-modified adhesive
  85. Experimental research on mechanically and thermally activation of nano-kaolin to improve the properties of ultra-high-performance fiber-reinforced concrete
  86. Analysis of variable fluid properties for three-dimensional flow of ternary hybrid nanofluid on a stretching sheet with MHD effects
  87. Biodegradability of corn starch films containing nanocellulose fiber and thymol
  88. Toxicity assessment of copper oxide nanoparticles: In vivo study
  89. Some measures to enhance the energy output performances of triboelectric nanogenerators
  90. Reinforcement of graphene nanoplatelets on water uptake and thermomechanical behaviour of epoxy adhesive subjected to water ageing conditions
  91. Optimization of preparation parameters and testing verification of carbon nanotube suspensions used in concrete
  92. Max-phase Ti3SiC2 and diverse nanoparticle reinforcements for enhancement of the mechanical, dynamic, and microstructural properties of AA5083 aluminum alloy via FSP
  93. Advancing drug delivery: Neural network perspectives on nanoparticle-mediated treatments for cancerous tissues
  94. PEG-PLGA core–shell nanoparticles for the controlled delivery of picoplatin–hydroxypropyl β-cyclodextrin inclusion complex in triple-negative breast cancer: In vitro and in vivo study
  95. Conduction transportation from graphene to an insulative polymer medium: A novel approach for the conductivity of nanocomposites
  96. Review Articles
  97. Developments of terahertz metasurface biosensors: A literature review
  98. Overview of amorphous carbon memristor device, modeling, and applications for neuromorphic computing
  99. Advances in the synthesis of gold nanoclusters (AuNCs) of proteins extracted from nature
  100. A review of ternary polymer nanocomposites containing clay and calcium carbonate and their biomedical applications
  101. Recent advancements in polyoxometalate-functionalized fiber materials: A review
  102. Special contribution of atomic force microscopy in cell death research
  103. A comprehensive review of oral chitosan drug delivery systems: Applications for oral insulin delivery
  104. Cellular senescence and nanoparticle-based therapies: Current developments and perspectives
  105. Cyclodextrins-block copolymer drug delivery systems: From design and development to preclinical studies
  106. Micelle-based nanoparticles with stimuli-responsive properties for drug delivery
  107. Critical assessment of the thermal stability and degradation of chemically functionalized nanocellulose-based polymer nanocomposites
  108. Research progress in preparation technology of micro and nano titanium alloy powder
  109. Nanoformulations for lysozyme-based additives in animal feed: An alternative to fight antibiotic resistance spread
  110. Incorporation of organic photochromic molecules in mesoporous silica materials: Synthesis and applications
  111. A review on modeling of graphene and associated nanostructures reinforced concrete
  112. A review on strengthening mechanisms of carbon quantum dots-reinforced Cu-matrix nanocomposites
  113. Review on nanocellulose composites and CNFs assembled microfiber toward automotive applications
  114. Nanomaterial coating for layered lithium rich transition metal oxide cathode for lithium-ion battery
  115. Application of AgNPs in biomedicine: An overview and current trends
  116. Nanobiotechnology and microbial influence on cold adaptation in plants
  117. Hepatotoxicity of nanomaterials: From mechanism to therapeutic strategy
  118. Applications of micro-nanobubble and its influence on concrete properties: An in-depth review
  119. A comprehensive systematic literature review of ML in nanotechnology for sustainable development
  120. Exploiting the nanotechnological approaches for traditional Chinese medicine in childhood rhinitis: A review of future perspectives
  121. Twisto-photonics in two-dimensional materials: A comprehensive review
  122. Current advances of anticancer drugs based on solubilization technology
  123. Recent process of using nanoparticles in the T cell-based immunometabolic therapy
  124. Future prospects of gold nanoclusters in hydrogen storage systems and sustainable environmental treatment applications
  125. Preparation, types, and applications of one- and two-dimensional nanochannels and their transport properties for water and ions
  126. Microstructural, mechanical, and corrosion characteristics of Mg–Gd–x systems: A review of recent advancements
  127. Functionalized nanostructures and targeted delivery systems with a focus on plant-derived natural agents for COVID-19 therapy: A review and outlook
  128. Mapping evolution and trends of cell membrane-coated nanoparticles: A bibliometric analysis and scoping review
  129. Nanoparticles and their application in the diagnosis of hepatocellular carcinoma
  130. In situ growth of carbon nanotubes on fly ash substrates
  131. Structural performance of boards through nanoparticle reinforcement: An advance review
  132. Reinforcing mechanisms review of the graphene oxide on cement composites
  133. Seed regeneration aided by nanomaterials in a climate change scenario: A comprehensive review
  134. Surface-engineered quantum dot nanocomposites for neurodegenerative disorder remediation and avenue for neuroimaging
  135. Graphitic carbon nitride hybrid thin films for energy conversion: A mini-review on defect activation with different materials
  136. Nanoparticles and the treatment of hepatocellular carcinoma
  137. Special Issue on Advanced Nanomaterials and Composites for Energy Conversion and Storage - Part II
  138. Highly safe lithium vanadium oxide anode for fast-charging dendrite-free lithium-ion batteries
  139. Recent progress in nanomaterials of battery energy storage: A patent landscape analysis, technology updates, and future prospects
  140. Special Issue on Advanced Nanomaterials for Carbon Capture, Environment and Utilization for Energy Sustainability - Part II
  141. Calcium-, magnesium-, and yttrium-doped lithium nickel phosphate nanomaterials as high-performance catalysts for electrochemical water oxidation reaction
  142. Low alkaline vegetation concrete with silica fume and nano-fly ash composites to improve the planting properties and soil ecology
  143. Mesoporous silica-grafted deep eutectic solvent-based mixed matrix membranes for wastewater treatment: Synthesis and emerging pollutant removal performance
  144. Electrochemically prepared ultrathin two-dimensional graphitic nanosheets as cathodes for advanced Zn-based energy storage devices
  145. Enhanced catalytic degradation of amoxicillin by phyto-mediated synthesised ZnO NPs and ZnO-rGO hybrid nanocomposite: Assessment of antioxidant activity, adsorption, and thermodynamic analysis
  146. Incorporating GO in PI matrix to advance nanocomposite coating: An enhancing strategy to prevent corrosion
  147. Synthesis, characterization, thermal stability, and application of microporous hyper cross-linked polyphosphazenes with naphthylamine group for CO2 uptake
  148. Engineering in ceramic albite morphology by the addition of additives: Carbon nanotubes and graphene oxide for energy applications
  149. Nanoscale synergy: Optimizing energy storage with SnO2 quantum dots on ZnO hexagonal prisms for advanced supercapacitors
  150. Aging assessment of silicone rubber materials under corona discharge accompanied by humidity and UV radiation
  151. Tuning structural and electrical properties of Co-precipitated and Cu-incorporated nickel ferrite for energy applications
  152. Sodium alginate-supported AgSr nanoparticles for catalytic degradation of malachite green and methyl orange in aqueous medium
  153. An environmentally greener and reusability approach for bioenergy production using Mallotus philippensis (Kamala) seed oil feedstock via phytonanotechnology
  154. Micro-/nano-alumina trihydrate and -magnesium hydroxide fillers in RTV-SR composites under electrical and environmental stresses
  155. Mechanism exploration of ion-implanted epoxy on surface trap distribution: An approach to augment the vacuum flashover voltages
  156. Nanoscale engineering of semiconductor photocatalysts boosting charge separation for solar-driven H2 production: Recent advances and future perspective
  157. Excellent catalytic performance over reduced graphene-boosted novel nanoparticles for oxidative desulfurization of fuel oil
  158. Special Issue on Advances in Nanotechnology for Agriculture
  159. Deciphering the synergistic potential of mycogenic zinc oxide nanoparticles and bio-slurry formulation on phenology and physiology of Vigna radiata
  160. Nanomaterials: Cross-disciplinary applications in ornamental plants
  161. Special Issue on Catechol Based Nano and Microstructures
  162. Polydopamine films: Versatile but interface-dependent coatings
  163. In vitro anticancer activity of melanin-like nanoparticles for multimodal therapy of glioblastoma
  164. Poly-3,4-dihydroxybenzylidenhydrazine, a different analogue of polydopamine
  165. Chirality and self-assembly of structures derived from optically active 1,2-diaminocyclohexane and catecholamines
  166. Advancing resource sustainability with green photothermal materials: Insights from organic waste-derived and bioderived sources
  167. Bioinspired neuromelanin-like Pt(iv) polymeric nanoparticles for cancer treatment
  168. Special Issue on Implementing Nanotechnology for Smart Healthcare System
  169. Intelligent explainable optical sensing on Internet of nanorobots for disease detection
  170. Special Issue on Green Mono, Bi and Tri Metallic Nanoparticles for Biological and Environmental Applications
  171. Tracking success of interaction of green-synthesized Carbopol nanoemulgel (neomycin-decorated Ag/ZnO nanocomposite) with wound-based MDR bacteria
  172. Green synthesis of copper oxide nanoparticles using genus Inula and evaluation of biological therapeutics and environmental applications
  173. Biogenic fabrication and multifunctional therapeutic applications of silver nanoparticles synthesized from rose petal extract
  174. Metal oxides on the frontlines: Antimicrobial activity in plant-derived biometallic nanoparticles
  175. Controlling pore size during the synthesis of hydroxyapatite nanoparticles using CTAB by the sol–gel hydrothermal method and their biological activities
  176. Special Issue on State-of-Art Advanced Nanotechnology for Healthcare
  177. Applications of nanomedicine-integrated phototherapeutic agents in cancer theranostics: A comprehensive review of the current state of research
  178. Smart bionanomaterials for treatment and diagnosis of inflammatory bowel disease
  179. Beyond conventional therapy: Synthesis of multifunctional nanoparticles for rheumatoid arthritis therapy
Heruntergeladen am 6.9.2025 von https://www.degruyterbrill.com/document/doi/10.1515/ntrev-2024-0023/html
Button zum nach oben scrollen