Photothermal therapy

Last updated

Photothermal therapy (PTT) refers to efforts to use electromagnetic radiation (most often in infrared wavelengths) for the treatment of various medical conditions, including cancer. This approach is an extension of photodynamic therapy, in which a photosensitizer is excited with specific band light. This activation brings the sensitizer to an excited state where it then releases vibrational energy (heat), which is what kills the targeted cells.

Contents

Unlike photodynamic therapy, photothermal therapy does not require oxygen to interact with the target cells or tissues. Current studies also show that photothermal therapy is able to use longer wavelength light, which is less energetic and therefore less harmful to other cells and tissues.

Nanoscale materials

Most materials of interest currently being investigated for photothermal therapy are on the nanoscale. One of the key reasons behind this is the enhanced permeability and retention effect observed with particles in a certain size range (typically 20 - 300 nm). [1] Molecules in this range have been observed to preferentially accumulate in tumor tissue. When a tumor forms, it requires new blood vessels in order to fuel its growth; these new blood vessels in/near tumors have different properties as compared to regular blood vessels, such as poor lymphatic drainage and a disorganized, leaky vasculature. These factors lead to a significantly higher concentration of certain particles in a tumor as compared to the rest of the body.[ citation needed ]

Gold NanoRods (AuNR)

Huang et al. investigated the feasibility of using gold nanorods for both cancer cell imaging as well as photothermal therapy. [2] The authors conjugated antibodies (anti-EGFR monoclonal antibodies) to the surface of gold nanorods, allowing the gold nanorods to bind specifically to certain malignant cancer cells (HSC and HOC malignant cells). After incubating the cells with the gold nanorods, an 800 nm Ti:sapphire laser was used to irradiate the cells at varying powers. The authors reported successful destruction of the malignant cancer cells, while nonmalignant cells were unharmed.[ citation needed ]

When AuNRs are exposed to NIR light, the oscillating electromagnetic field of the light causes the free electrons of the AuNR to collectively coherently oscillate. [3] Changing the size and shape of AuNRs changes the wavelength that gets absorbed. A desired wavelength would be between 700-1000 nm because biological tissue is optically transparent at these wavelengths. [4] While all AuNP are sensitive to change in their shape and size, Au nanorods properties are extremely sensitive to any change in any of their dimensions regarding their length and width or their aspect ratio. When light is shone on a metal NP, the NP forms a dipole oscillation along the direction of the electric field. When the oscillation reaches its maximum, this frequency is called the surface plasmon resonance (SPR). [3] AuNR have two SPR spectrum bands: one in the NIR region caused by its longitudinal oscillation which tends to be stronger with a longer wavelength and one in the visible region caused by the transverse electronic oscillation which tends to be weaker with a shorter wavelength. [5] The SPR characteristics account for the increase in light absorption for the particle. [3] As the AuNR aspect ratio increases, the absorption wavelength is redshifted [5] and light scattering efficiency is increased. [3] The electrons excited by the NIR lose energy quickly after absorption via electron-electron collisions, and as these electrons relax back down, the energy is released as a phonon that then heats the environment of the AuNP which in cancer treatments would be the cancerous cells. This process is observed when a laser has a continuous wave onto the AuNP. Pulsed laser light beams generally results in the AuNP melting or ablation of the particle. [3] Continuous wave lasers take minutes rather than a single pulse time for a pulsed laser, continues wave lasers are able to heat larger areas at once. [3]

Gold Nanoshells

Gold nanoshells, coated silica nanoparticles with a thin layer of gold. [6] have been conjugated to antibodies (anti-HER2 or anti-IgG) via PEG linkers. After incubation of SKBr3 cancer cells with the gold nanoshells, an 820 nm laser was used to irradiate the cells. Only the cells incubated with the gold nanoshells conjugated with the specific antibody (anti-HER2) were damaged by the laser. Another category of gold nanoshells are gold layer on liposomes, as soft template. In this case, drug can also be encapsulated inside and/or in bilayer and the release can be triggered by laser light. [7]

thermo Nano-Architectures (tNAs)

The failure of clinical translation of nanoparticles-mediated PTT is mainly ascribed to concerns about their persistence in the body. [8] Indeed, the optical response of anisotropic nanomaterials can be tuned in the NIR region by increasing their size to up to 150 nm. [9] On the other hand, body excretion of non-biodegradable noble metals nanomaterials above 10 nm occurs through the hepatobiliary route in a slow and inefficient manner. [10] A common approach to avoid metal persistence is to reduce the nanoparticles size below the threshold for renal clearance, i.e. ultrasmall nanoparticles (USNPs), meanwhile the maximum light-to-heat transduction is for < 5 nm nanoparticles. [11] On the other hand, the surface plasmon of excretable gold USNPs is in the UV/visible region (far from the first biological windows), severely limiting their potential application in PTT.

Excretion of metals has been combined with NIR-triggered PTT by employing ultrasmall-in-nano architectures composed by metal USNPs embedded in biodegradable silica nanocapsules. [12] tNAs are the first reported NIR-absorbing plasmonic ultrasmall-in-nano platforms that jointly combine: i) photothermal conversion efficacy suitable for hyperthermia, ii) multiple photothermal sequences and iii) renal excretion of the building blocks after the therapeutic action. [12] [13] [14] Nowadays, tNAs therapeutic effect has been assessed on valuable 3D models of human pancreatic adenocarcinoma. [12]

Graphene and graphene oxide

Graphene is viable for photothermal therapy. [15] An 808 nm laser at a power density of 2 W/cm2 was used to irradiate the tumor sites on mice for 5 minutes. As noted by the authors, the power densities of lasers used to heat gold nanorods range from 2 to 4 W/cm2. Thus, these nanoscale graphene sheets require a laser power on the lower end of the range used with gold nanoparticles to photothermally ablate tumors.[ citation needed ]

In 2012, Yang et al. incorporated the promising results regarding nanoscale reduced graphene oxide reported by Robinson et al. into another in vivo mice study. [16] < [17] The therapeutic treatment used in this study involved the use of nanoscale reduced graphene oxide sheets, nearly identical to the ones used by Robinson et al. (but without any active targeting sequences attached). Nanoscale reduced graphene oxide sheets were successfully irradiated in order to completely destroy the targeted tumors. Most notably, the required power density of the 808 nm laser was reduced to 0.15 W/cm2, an order of magnitude lower than previously required power densities. This study demonstrates the higher efficacy of nanoscale reduced graphene oxide sheets as compared to both nanoscale graphene sheets and gold nanorods.[ citation needed ]

Conjugated polymers (CPs)

PTT utilizes photothermal transduction agents (PTAs) which can transform light energy to heat through photothermal effect to raise the temperature of tumor area and thus cause the ablation of tumor cells. [18] [19] Specifically, ideal PTAs should have high photothermal conversion efficiency (PCE), excellent optical stability and biocompatibility, and strong light adsorption in the near-infrared (NIR) region (650-1350 nm) due to the deep-tissue penetration and minimal absorption of NIR light in the biological tissues. [18] [19] PTAs mainly include inorganic materials and organic materials. [19] Inorganic PTAs, such as noble metal materials, carbon-based nanomaterials, and other 2D materials, have high PCE and excellent photostability, but they are not biodegradable and thus have potential long-term toxicity in vivo. [19] [20] Organic PTAs including small molecule dyes and conjugated polymers (CPs) have good biocompatibility and biodegradability, but poor photostability. [19] Among them, small molecule dyes, such as cyanine, porphyrin, phthalocyanine, are limited in the field of cancer treatment because of their susceptibility to photobleaching and poor tumor enrichment ability. [19] Conjugated polymers with large π−π conjugated skeleton and a high electron delocalization structure show potential for PTT due to their strong NIR absorption, excellent photostability, low cytotoxicity, outstanding PCE, good dispersibility in aqueous medium, increased accumulation at tumor site, and long blood circulation time. [18] [19] [20] [21] Moreover, conjugated polymers can be easily combined with other imaging agents and drugs to construct multifunctional nanomaterials for selective and synergistic cancer therapy. [18]

The CPs used for tumor PTT mainly include polyaniline (PANI), polypyrrole (PPy), polythiophene (PTh), polydopamine (PDA), donor−acceptor (D-A) conjugated polymers, and poly(3,4-ethylenedioxythiophene):poly(4-styrenesulfonate) (PEDOT:PSS). [18] [19]

Photothermal conversion mechanism

The nonradiative process for heat generation of organic PTAs is different from that of inorganic PTAs such as metals and semiconductors which is related with surface plasmon resonance. [22] As shown in the figure, conjugated polymers are first activated to the excited state (S1) under light irradiation and then excited state (S1) decays back to the ground state (S0) via three processes: (I) emitting a photon (fluorescence), (II) intersystem crossing, and (III) nonradiative relaxation (heat generation). [22] Because these three pathways of the S1 decaying back to the S0 are usually competitive in photosensitive materials, light emitting and intersystem crossing must be efficiently reduced in order to increase the heat generation and improve the photothermal conversion efficiency. [18] [22] For conjugated polymers, on the one hand, their unique structures lead to closed stacking of the molecular sensitizers with highly frequent intermolecular collisions which can efficiently quench the fluorescence and intersystem crossing, and thus enhance the yield of nonradiative relaxation. [22] On the other hand, compared with monomeric phototherapeutic molecules, conjugated polymers possess higher stability in vivo against disassembly and photobleaching, longer blood circulation time, and more accumulation at tumor site due to the enhanced permeability and retention (EPR) effect. [22] Therefore, conjugated polymers have high photothermal conversion efficiency and a large amount of heat generation. One of the most widely used equations to calculate photothermal conversion efficiency (η) of organic PTAs is as follows:

η = (hAΔΤmax-Qs)/I(1-10-Aλ)

where h is the heat transfer coefficient, A is the container surface area, ΔΤmax means the maximum temperature change in the solution, Aλ means the light absorbance, I is the laser power density, and Qs is the heat associated with the light absorbance of the solvent. [23]

Furthermore, various efficient methods, especially donor-acceptor (D-A) strategy, have been designed to enhance the photothermal conversion efficiency and heat generation of conjugated polymers. [18] The D-A assembly system in the conjugated polymers contributes to strong intermolecular electron transfer from the donor to the acceptor, thus bringing efficient fluorescence and intersystem crossing quenching, and improved heat generation. [22] In addition, the HOMO-LUMO gap of the D−A conjugated polymers can be easily tuned through changing the selection of electron donor (ED) and electron acceptor (EA) moieties, and thus D−A structured polymers with extremely low band gap can be developed to improve the NIR absorption and photothermal conversion efficiency of CPs. [19] [21]

Polyaniline (PANI)

Polyaniline (PANI) is one of the earliest types of conjugated polymers reported for tumor PTT. [19] [24] [20] [21] [25] [26]

Polypyrrole (PPy)

Polypyrrole (PPy) is suited for PTT applications because of its strong NIR absorbance, large PCE, stability, and biocompatibility. [21] In vivo experiments show that tumors treated with PPy NPs could be effectively eliminated under the irradiation of an 808 nm laser (1 W cm−2, 5 min). [27] PPy nanosheets exhibit promising photothermal ablation ability toward cancer cells in the NIR II window for deep-tissue PTT. [28]

PPy nanoparticles and its derivative nanomaterials can also be combined with imaging contrast agents and diverse drugs to construct multifunctional theranostic applications in imaging-guided PTT and synergistic treatment, including fluorescent imaging, magnetic resonance imaging (MRI), photoacoustic imaging (PA), computed tomography (CT), photodynamic therapy (PDT), chemotherapy, etc. [19] For example, PPy has been used to encapsulate ultrasmall iron oxide nanoparticles (IONPs) and finally develop IONP@PPy NPs for in vivo MR and PA imaging-guided PTT. [29] Polypyrrole (I-PPy) nanocomposites have been investigated for CT imaging-guided tumor PTT. [30]

Polythiophene (PTh)

Polythiophene (PTh) and its derivatives-based polymers are also one kind of conjugated polymers for PTT. Polythiophene-based polymers usually exhibit excellent photostability, large light-harvesting ability, easy synthesis, and facile functionalization with different substituents. [21]

Conjugated copolymer (C3) with promising photothermal properties can be prepared by linking 2-N,N′-bis(2-(ethyl)hexyl)-perylene-3,4,9,10-tetra-carboxylic acid bis-imide to a thienylvinylene oligomer. C3 was coprecipitated with PEG-PCL and indocyanine green (ICG) to obtain PEG-PCL-C3-ICG nanoparticles for fluorescence-guided photothermal/photodynamic therapy against oral squamous cell carcinoma (OSCC). [31] A biodegradable PLGA-PEGylated DPPV (poly{2,2′-[(2,5-bis(2-hexyldecyl)-3,6-dioxo-2,3,5,6-tetrahydropyrrolo[3,4-c]-pyrrole-1,4-diyl)-dithiophene]-5,5′-diyl-alt-vinylene) conjugated polymer for PA-guided PTT with PCE 71% (@ 808 nm, 0.3 W cm−2). The vinylene bonds in the main chain improves the biodegradability, biocompatibility and photothermal conversion efficiency of CPs. [32]

Polydopamine (PDA)

Dopamine is one of neurotransmitters in the body which helps cells send impulses. Polydopamine (PDA) is obtained through the self-aggregation of dopamine to form a melanin-like substance under mild alkaline conditions. [33] PDA has strong NIR absorption, good photothermal stability, excellent biocompatibility and biodegradability, and high photothermal conversion efficiency. [34] Furthermore, with π conjugated structure and different active groups, PDA can be easily combined with various materials to achieve multifunction, such as fluorescence imaging, MRI, CT, PA, targeted therapy etc. [19] In view of this, PDA and its composite nanomaterials have a broad application prospect in the biomedical field.[ citation needed ]

Dopamine-melanin colloidal nanospheres is an efficient near-infrared photothermal therapeutic agent for in vivo cancer therapy. [23] PDA can also be modified on the surface of other PTAs, such as gold nanorods, carbon-based materials, to enhance the photothermal stability and efficiency in vivo. [19] For example, PDA-modified spiky gold nanoparticles (SGNP@PDAs) have been investigated for chemo-photothermal therapy. [35]

Donor−Acceptor (D−A) CPs

Donor−acceptor (D−A) conjugated polymers have been investigated for the medicinal purposes. Nano-PCPDTBT CPs have two moieties: 2-ethylhexyl cyclopentadithiophene and 2,1,3-benzothiadiazole. When the PCPDTBT nanoparticle solution (0.115 mg/mL) was exposed to an 808 nm NIR laser (0.6 W/cm2), the temperature could be increased by more than 30 °C. [36] Wang et al. designed four NIR-absorbing D-A structured conjugated polymer dots (Pdots) containing diketopyrrolo-pyrrole (DPP) and thiophene units as effective photothermal materials with the PCE up to 65% for in vivo cancer therapy. [37] Zhang et al. constructed PBIBDF-BT D-A CPs by using isoindigo derivative (BIBDF) and bithiophene (BT) as EA and ED respectively. PBIBDF-BT was further modified with poly(ethylene glycol)-block-poly(hexyl ethylene phosphate) (mPEG-b-PHEP) to obtain PBIBDF-BT@NP PPE with PCE of 46.7% and high stability in physiological environment. [38] Yang’s group designed PBTPBF-BT CPs, in which the bis(5-oxothieno[3,2-b]pyrrole-6-ylidene)-benzodifurandione (BTPBF) and the 3,3′-didodecyl-2,2′-bithiophene (BT) units acting as EA and ED respectively. The D-A CPs have a maximum absorption peak at 1107 nm and a relative high photothermal conversion efficiency (66.4%). [39] Pu et al. synthesized PC70BM-PCPDTBT D-A CPs via nanoprecipitation of EA (6,6)-phenyl-C71-butyric acid methyl ester (PC70BM) and ED PCPDTBT (SPs) for PA-guided PTT. [40] Wang et al. developed D-A CPs TBDOPV-DT containing thiophene-fused benzodifurandione-based oligo(p-phenylenevinylene) (TBDOPV) as EA unit and 2,2′-bithio-phene (DT) as ED unit. TBDOPV-DT CPs have a strong absorption at 1093 nm and achieve highly efficient NIR-II photothermal conversion. [41]

PEDOT:PSS

Poly(3,4-ethylenedioxythiophene):poly(4-styrenesulfonate) (PEDOT:PSS) is often used in organic electrons and have strong NIR absorption. In 2012, Liu’s group first reported PEGylated PEDOT:PSS polymeric nanoparticle (PEDOT:PSS-PEG) for near-infrared photothermal therapy of cancer. PEDOT:PSS-PEG nanoparticles have high stability in vivo and long blood circulation half-life of 21.4 ± 3.1 h. The PTT in animals showed no appreciable side effects for the tested dose and an excellent therapeutic efficacy under the 808 nm laser irradiation. [42] Kang et al. synthesized magneto-conjugated polymer core−shell MNP@PEDOT:PSS nanoparticles for multimodal imaging-guided PTT. [43] Furthermore, PEDOT:PSS NPs can not only serve as PTAs but also as a drug carrier to load various types of drugs, such as SN38, chemotherapy drugs DOX and photodynamic agent chlorin e6 (Ce6), thus achieving synergistic cancer therapy. [44]

See also

Related Research Articles

Nanomedicine is the medical application of nanotechnology. Nanomedicine ranges from the medical applications of nanomaterials and biological devices, to nanoelectronic biosensors, and even possible future applications of molecular nanotechnology such as biological machines. Current problems for nanomedicine involve understanding the issues related to toxicity and environmental impact of nanoscale materials.

<span class="mw-page-title-main">Nanomaterials</span> Materials whose granular size lies between 1 and 100 nm

Nanomaterials describe, in principle, materials of which a single unit is sized between 1 and 100 nm.

<span class="mw-page-title-main">Nanoshell</span>

A nanoshell, or rather a nanoshell plasmon, is a type of spherical nanoparticle consisting of a dielectric core which is covered by a thin metallic shell. These nanoshells involve a quasiparticle called a plasmon which is a collective excitation or quantum plasma oscillation where the electrons simultaneously oscillate with respect to all the ions.

<span class="mw-page-title-main">Colloidal gold</span> Suspension of gold nanoparticles in a liquid

Colloidal gold is a sol or colloidal suspension of nanoparticles of gold in a fluid, usually water. The colloid is coloured usually either wine red or blue-purple . Due to their optical, electronic, and molecular-recognition properties, gold nanoparticles are the subject of substantial research, with many potential or promised applications in a wide variety of areas, including electron microscopy, electronics, nanotechnology, materials science, and biomedicine.

<span class="mw-page-title-main">Photosensitizer</span> Type of molecule reacting to light

Photosensitizers are light absorbers that alter the course of a photochemical reaction. They usually are catalysts. They can function by many mechanisms, sometimes they donate an electron to the substrate, sometimes they abstract a hydrogen atom from the substrate. At the end of this process, the photosensitizer returns to its ground state, where it remains chemically intact, poised to absorb more light. One branch of chemistry which frequently utilizes photosensitizers is polymer chemistry, using photosensitizers in reactions such as photopolymerization, photocrosslinking, and photodegradation. Photosensitizers are also used to generate prolonged excited electronic states in organic molecules with uses in photocatalysis, photon upconversion and photodynamic therapy. Generally, photosensitizers absorb electromagnetic radiation consisting of infrared radiation, visible light radiation, and ultraviolet radiation and transfer absorbed energy into neighboring molecules. This absorption of light is made possible by photosensitizers' large de-localized π-systems, which lowers the energy of HOMO and LUMO orbitals to promote photoexcitation. While many photosensitizers are organic or organometallic compounds, there are also examples of using semiconductor quantum dots as photosensitizers.

Gold Nanocages are hollow, porous gold nanoparticles ranging in size from 10 to over 150 nm. They are created by reacting silver nanoparticles with chloroauric acid (HAuCl4) in boiling water. Whereas gold nanoparticles absorb light in the visible spectrum of light (at about 550 nm), gold nanocages absorb light in the near-infrared, where biological tissues absorb the least light. Because they are also biocompatible, gold nanocages are promising as a contrast agent for optical coherence tomography. Gold nanocages also absorb light and heat up (Photothermal effect), killing surrounding cancer cells. Nanocages have been functionalized with cancer-specific antibodies.

<span class="mw-page-title-main">Artificial enzyme</span>

An artificial enzyme is a synthetic organic molecule or ion that recreates one or more functions of an enzyme. It seeks to deliver catalysis at rates and selectivity observed in naturally occurring enzymes.

Carbon nanotubes (CNTs) are very prevalent in today's world of medical research and are being highly researched in the fields of efficient drug delivery and biosensing methods for disease treatment and health monitoring. Carbon nanotube technology has shown to have the potential to alter drug delivery and biosensing methods for the better, and thus, carbon nanotubes have recently garnered interest in the field of medicine.

Multiple layered plasmonics use electronically responsive media to change and manipulate the plasmonic properties of plasmons. The properties typically being manipulated can include the directed scattering of light and light absorption. The use of these to use “changeable” plasmonics is currently undergoing development in the academic community by allowing them to have multiple sets of functions that are dependent on how they are being manipulated or excited. Under these new manipulations, such as multiple layers that respond to different resonant frequencies, their new functions were designed to accomplish multiple objectives in a single application.

<span class="mw-page-title-main">Plasmonic nanoparticles</span>

Plasmonic nanoparticles are particles whose electron density can couple with electromagnetic radiation of wavelengths that are far larger than the particle due to the nature of the dielectric-metal interface between the medium and the particles: unlike in a pure metal where there is a maximum limit on what size wavelength can be effectively coupled based on the material size.

Photoimmunotherapy (PIT) is an oncological treatment that combines photodynamic therapy of tumor with immunotherapy treatment. Combining photodynamic therapy with immunotherapy enhances the immunostimulating response and has synergistic effects for metastatic cancer treatment.

The applications of nanotechnology, commonly incorporate industrial, medicinal, and energy uses. These include more durable construction materials, therapeutic drug delivery, and higher density hydrogen fuel cells that are environmentally friendly. Being that nanoparticles and nanodevices are highly versatile through modification of their physiochemical properties, they have found uses in nanoscale electronics, cancer treatments, vaccines, hydrogen fuel cells, and nanographene batteries.

<span class="mw-page-title-main">Localized surface plasmon</span>

A localized surface plasmon (LSP) is the result of the confinement of a surface plasmon in a nanoparticle of size comparable to or smaller than the wavelength of light used to excite the plasmon. When a small spherical metallic nanoparticle is irradiated by light, the oscillating electric field causes the conduction electrons to oscillate coherently. When the electron cloud is displaced relative to its original position, a restoring force arises from Coulombic attraction between electrons and nuclei. This force causes the electron cloud to oscillate. The oscillation frequency is determined by the density of electrons, the effective electron mass, and the size and shape of the charge distribution. The LSP has two important effects: electric fields near the particle's surface are greatly enhanced and the particle's optical absorption has a maximum at the plasmon resonant frequency. Surface plasmon resonance can also be tuned based on the shape of the nanoparticle. The plasmon frequency can be related to the metal dielectric constant. The enhancement falls off quickly with distance from the surface and, for noble metal nanoparticles, the resonance occurs at visible wavelengths. Localized surface plasmon resonance creates brilliant colors in metal colloidal solutions.

<span class="mw-page-title-main">Nanocarrier</span>

A nanocarrier is nanomaterial being used as a transport module for another substance, such as a drug. Commonly used nanocarriers include micelles, polymers, carbon-based materials, liposomes and other substances. Nanocarriers are currently being studied for their use in drug delivery and their unique characteristics demonstrate potential use in chemotherapy. This class of materials was first reported by a team of researchers of University of Évora, Alentejo in early 1960's, and grew exponentially in relevance since then.

<span class="mw-page-title-main">Gold nanoparticles in chemotherapy</span> Drug delivery technique using gold nanoparticles as vectors

Gold nanoparticles in chemotherapy and radiotherapy is the use of colloidal gold in therapeutic treatments, often for cancer or arthritis. Gold nanoparticle technology shows promise in the advancement of cancer treatments. Some of the properties that gold nanoparticles possess, such as small size, non-toxicity and non-immunogenicity make these molecules useful candidates for targeted drug delivery systems. With tumor-targeting delivery vectors becoming smaller, the ability to by-pass the natural barriers and obstacles of the body becomes more probable. To increase specificity and likelihood of drug delivery, tumor specific ligands may be grafted onto the particles along with the chemotherapeutic drug molecules, to allow these molecules to circulate throughout the tumor without being redistributed into the body.

<span class="mw-page-title-main">Carbon quantum dot</span> Type of carbon nanoparticle

Carbon quantum dots also commonly called carbon nano dots are carbon nanoparticles which are less than 10 nm in size and have some form of surface passivation.

Hadiyah-Nicole Green (1981) is an American medical physicist, known for the development of a method using laser-activated nanoparticles as a potential cancer treatment. She is one of 66 black women to earn a Ph.D. in physics in the United States between 1973 and 2012, and is the second black woman and the fourth black person ever to earn a doctoral degree in physics from The University of Alabama at Birmingham.

Upconverting nanoparticles (UCNPs) are nanoscale particles that exhibit photon upconversion. In photon upconversion, two or more incident photons of relatively low energy are absorbed and converted into one emitted photon with higher energy. Generally, absorption occurs in the infrared, while emission occurs in the visible or ultraviolet regions of the electromagnetic spectrum. UCNPs are usually composed of rare-earth based lanthanide- or actinide-doped transition metals and are of particular interest for their applications in in vivo bio-imaging, bio-sensing, and nanomedicine because of their highly efficient cellular uptake and high optical penetrating power with little background noise in the deep tissue level. They also have potential applications in photovoltaics and security, such as infrared detection of hazardous materials.

Nanoparticle drug delivery systems are engineered technologies that use nanoparticles for the targeted delivery and controlled release of therapeutic agents. The modern form of a drug delivery system should minimize side-effects and reduce both dosage and dosage frequency. Recently, nanoparticles have aroused attention due to their potential application for effective drug delivery.

<span class="mw-page-title-main">Ji-Xin Cheng</span> Academic, inventor, and entrepreneur

Ji-Xin Cheng is an academic, inventor, and entrepreneur. He holds the Moustakas Chair Professorship in Optoelectronics and Photonics at Boston University. His inventions span optical imaging, cancer diagnosis, neuromodulation, and phototherapy of infectious diseases. He holds positions of co-founder of Vibronic and of Pulsethera. He is also the scientific advisor of Photothermal Spectroscopy and Axorus.

References

  1. Maeda H, Wu J, Sawa T, Matsumura Y, Hori K (March 2000). "Tumor vascular permeability and the EPR effect in macromolecular therapeutics: a review". Journal of Controlled Release. 65 (1–2): 271–84. doi:10.1016/s0168-3659(99)00248-5. PMID   10699287.
  2. Huang X, El-Sayed IH, Qian W, El-Sayed MA (February 2006). "Cancer cell imaging and photothermal therapy in the near-infrared region by using gold nanorods". Journal of the American Chemical Society. 128 (6): 2115–20. doi:10.1021/ja057254a. PMID   16464114.
  3. 1 2 3 4 5 6 Huang X, El-Sayed MA (January 2010). "Gold nanoparticles: Optical properties and implementations in cancer diagnosis and photothermal therapy". Journal of Advanced Research. 1 (1): 13–28. doi: 10.1016/j.jare.2010.02.002 .
  4. Hauck TS, Jennings TL, Yatsenko T, Kumaradas JC, Chan WC (October 2008). "Enhancing the Toxicity of Cancer Chemotherapeutics with Gold Nanorod Hyperthermia". Advanced Materials. 20 (20): 3832–3838. doi: 10.1002/adma.200800921 . ISSN   1521-4095. S2CID   137257403.
  5. 1 2 Huang X, Jain PK, El-Sayed IH, El-Sayed MA (July 2008). "Plasmonic photothermal therapy (PPTT) using gold nanoparticles". Lasers in Medical Science. 23 (3): 217–28. doi:10.1007/s10103-007-0470-x. PMID   17674122. S2CID   207053590.
  6. Loo C, Lowery A, Halas N, West J, Drezek R (April 2005). "Immunotargeted nanoshells for integrated cancer imaging and therapy". Nano Letters. 5 (4): 709–11. Bibcode:2005NanoL...5..709L. doi:10.1021/nl050127s. PMID   15826113.
  7. Abbasi A, Park K, Bose A, Bothun GD (May 2017). "Near-Infrared Responsive Gold-Layersome Nanoshells". Langmuir. 33 (21): 5321–5327. doi:10.1021/acs.langmuir.7b01273. PMID   28486807.
  8. Chen F, Cai W (January 2015). "Nanomedicine for targeted photothermal cancer therapy: where are we now?". Nanomedicine. 10 (1): 1–3. doi:10.2217/nnm.14.186. PMC   4299941 . PMID   25597770.
  9. Riley RS, Day ES (July 2017). "Gold nanoparticle-mediated photothermal therapy: applications and opportunities for multimodal cancer treatment". Wiley Interdisciplinary Reviews. Nanomedicine and Nanobiotechnology. 9 (4): e1449. doi:10.1002/wnan.1449. PMC   5474189 . PMID   28160445.
  10. Cassano D, Pocoví-Martínez S, Voliani V (January 2018). "Ultrasmall-in-Nano Approach: Enabling the Translation of Metal Nanomaterials to Clinics". Bioconjugate Chemistry. 29 (1): 4–16. doi: 10.1021/acs.bioconjchem.7b00664 . PMID   29186662.
  11. Jiang K, Smith DA, Pinchuk A (2013-12-27). "Size-Dependent Photothermal Conversion Efficiencies of Plasmonically Heated Gold Nanoparticles". The Journal of Physical Chemistry C. 117 (51): 27073–27080. doi:10.1021/jp409067h.
  12. 1 2 3 Cassano D, Santi M, D'Autilia F, Mapanao AK, Luin S, Voliani V (2019). "Photothermal effect by NIR-responsive excretable ultrasmall-in-nano architectures" (PDF). Materials Horizons. 6 (3): 531–537. doi: 10.1039/C9MH00096H .
  13. Cassano D, Summa M, Pocovíd-Martínez S, Mapanao AK, Catelani T, Bertorelli R, Voliani V (February 2019). "Biodegradable Ultrasmall-in-Nano Gold Architectures: Mid-Period In Vivo Distribution and Excretion Assessment". Particle & Particle Systems Characterization. 36 (2): 1800464. doi:10.1002/ppsc.201800464. S2CID   104434042.
  14. Cassano D, Mapanao AK, Summa M, Vlamidis Y, Giannone G, Santi M, Guzzolino E, Pitto L, Poliseno L, Bertorelli R, Voliani V (2019-10-21). "Biosafety and Biokinetics of Noble Metals: The Impact of Their Chemical Nature". ACS Applied Bio Materials. 2 (10): 4464–4470. doi:10.1021/acsabm.9b00630. ISSN   2576-6422. PMID   35021406. S2CID   204266885.
  15. Yang K, Zhang S, Zhang G, Sun X, Lee ST, Liu Z (September 2010). "Graphene in mice: ultrahigh in vivo tumor uptake and efficient photothermal therapy". Nano Letters. 10 (9): 3318–23. Bibcode:2010NanoL..10.3318Y. doi:10.1021/nl100996u. PMID   20684528.
  16. Robinson JT, Tabakman SM, Liang Y, Wang H, Casalongue HS, Vinh D, Dai H (May 2011). "Ultrasmall reduced graphene oxide with high near-infrared absorbance for photothermal therapy". Journal of the American Chemical Society. 133 (17): 6825–31. doi:10.1021/ja2010175. PMID   21476500.
  17. Yang K, Wan J, Zhang S, Tian B, Zhang Y, Liu Z (March 2012). "The influence of surface chemistry and size of nanoscale graphene oxide on photothermal therapy of cancer using ultra-low laser power". Biomaterials. 33 (7): 2206–14. doi:10.1016/j.biomaterials.2011.11.064. PMID   22169821.
  18. 1 2 3 4 5 6 7 Wang Y, Meng HM, Song G, Li Z, Zhang XB (August 2020). "Conjugated-Polymer-Based Nanomaterials for Photothermal Therapy". ACS Applied Polymer Materials. 2 (10): 4258–4272. doi:10.1021/acsapm.0c00680. ISSN   2637-6105. S2CID   225217380.
  19. 1 2 3 4 5 6 7 8 9 10 11 12 13 Yu C, Xu L, Zhang Y, Timashev PS, Huang Y, Liang XJ (September 2020). "Polymer-Based Nanomaterials for Noninvasive Cancer Photothermal Therapy". ACS Applied Polymer Materials. 2 (10): 4289–4305. doi:10.1021/acsapm.0c00704. ISSN   2637-6105. S2CID   225312270.
  20. 1 2 3 Xu L, Cheng L, Wang C, Peng R, Liu Z (2014). "Conjugated polymers for photothermal therapy of cancer". Polym. Chem. 5 (5): 1573–1580. doi:10.1039/C3PY01196H. ISSN   1759-9954.
  21. 1 2 3 4 5 Pierini F, Nakielski P, Urbanek O, Pawłowska S, Lanzi M, De Sio L, Kowalewski TA (November 2018). "Polymer-Based Nanomaterials for Photothermal Therapy: From Light-Responsive to Multifunctional Nanoplatforms for Synergistically Combined Technologies". Biomacromolecules. 19 (11): 4147–4167. doi:10.1021/acs.biomac.8b01138. hdl: 11573/1178237 . PMID   30230317. S2CID   52293861.
  22. 1 2 3 4 5 6 Zhao L, Liu Y, Chang R, Xing R, Yan X (November 2018). "Supramolecular Photothermal Nanomaterials as an Emerging Paradigm toward Precision Cancer Therapy". Advanced Functional Materials. 29 (4): 1806877. doi:10.1002/adfm.201806877. ISSN   1616-301X. S2CID   106028103.
  23. 1 2 Liu Y, Ai K, Liu J, Deng M, He Y, Lu L (March 2013). "Dopamine-melanin colloidal nanospheres: an efficient near-infrared photothermal therapeutic agent for in vivo cancer therapy". Advanced Materials. 25 (9): 1353–9. doi:10.1002/adma.201204683. PMID   23280690. S2CID   5241524.
  24. Yang J, Choi J, Bang D, Kim E, Lim EK, Park H, et al. (January 2011). "Convertible organic nanoparticles for near-infrared photothermal ablation of cancer cells". Angewandte Chemie. 50 (2): 441–4. doi:10.1002/anie.201005075. PMID   21132823.
  25. Wang J, Yan R, Guo F, Yu M, Tan F, Li N (July 2016). "Targeted lipid-polyaniline hybrid nanoparticles for photoacoustic imaging guided photothermal therapy of cancer". Nanotechnology. 27 (28): 285102. Bibcode:2016Nanot..27B5102W. doi:10.1088/0957-4484/27/28/285102. PMID   27255659.
  26. Tian Q, Li Y, Jiang S, An L, Lin J, Wu H, et al. (October 2019). "Tumor pH-Responsive Albumin/Polyaniline Assemblies for Amplified Photoacoustic Imaging and Augmented Photothermal Therapy". Small. 15 (42): e1902926. doi:10.1002/smll.201902926. PMID   31448572. S2CID   201750011.
  27. Chen M, Fang X, Tang S, Zheng N (September 2012). "Polypyrrole nanoparticles for high-performance in vivo near-infrared photothermal cancer therapy". Chemical Communications. 48 (71): 8934–6. doi:10.1039/c2cc34463g. PMID   22847451.
  28. Wang X, Ma Y, Sheng X, Wang Y, Xu H (April 2018). "Ultrathin Polypyrrole Nanosheets via Space-Confined Synthesis for Efficient Photothermal Therapy in the Second Near-Infrared Window". Nano Letters. 18 (4): 2217–2225. Bibcode:2018NanoL..18.2217W. doi:10.1021/acs.nanolett.7b04675. PMID   29528661.
  29. Song X, Gong H, Yin S, Cheng L, Wang C, Li Z, et al. (September 2013). "Ultra-Small Iron Oxide Doped Polypyrrole Nanoparticles for In Vivo Multimodal Imaging Guided Photothermal Therapy". Advanced Functional Materials. 24 (9): 1194–1201. doi:10.1002/adfm.201302463. ISSN   1616-301X. S2CID   97828466.
  30. Zou Q, Huang J, Zhang X (November 2018). "One-Step Synthesis of Iodinated Polypyrrole Nanoparticles for CT Imaging Guided Photothermal Therapy of Tumors". Small. 14 (45): e1803101. doi:10.1002/smll.201803101. PMID   30300473. S2CID   52946295.
  31. Ren S, Cheng X, Chen M, Liu C, Zhao P, Huang W, et al. (September 2017). "Hypotoxic and Rapidly Metabolic PEG-PCL-C3-ICG Nanoparticles for Fluorescence-Guided Photothermal/Photodynamic Therapy against OSCC". ACS Applied Materials & Interfaces. 9 (37): 31509–31518. doi:10.1021/acsami.7b09522. PMID   28858474.
  32. Lyu Y, Zeng J, Jiang Y, Zhen X, Wang T, Qiu S, et al. (February 2018). "Enhancing Both Biodegradability and Efficacy of Semiconducting Polymer Nanoparticles for Photoacoustic Imaging and Photothermal Therapy". ACS Nano. 12 (2): 1801–1810. doi:10.1021/acsnano.7b08616. hdl: 10356/91064 . PMID   29385336.
  33. Wang X, Zhang J, Wang Y, Wang C, Xiao J, Zhang Q, Cheng Y (March 2016). "Multi-responsive photothermal-chemotherapy with drug-loaded melanin-like nanoparticles for synergetic tumor ablation". Biomaterials. 81: 114–124. doi:10.1016/j.biomaterials.2015.11.037. PMID   26731575.
  34. Cheng W, Zeng X, Chen H, Li Z, Zeng W, Mei L, Zhao Y (August 2019). "Versatile Polydopamine Platforms: Synthesis and Promising Applications for Surface Modification and Advanced Nanomedicine". ACS Nano. 13 (8): 8537–8565. doi:10.1021/acsnano.9b04436. PMID   31369230. S2CID   199380635.
  35. Nam J, Son S, Ochyl LJ, Kuai R, Schwendeman A, Moon JJ (March 2018). "Chemo-photothermal therapy combination elicits anti-tumor immunity against advanced metastatic cancer". Nature Communications. 9 (1): 1074. Bibcode:2018NatCo...9.1074N. doi:10.1038/s41467-018-03473-9. PMC   5852008 . PMID   29540781.
  36. MacNeill CM, Coffin RC, Carroll DL, Levi-Polyachenko NH (January 2013). "Low band gap donor-acceptor conjugated polymer nanoparticles and their NIR-mediated thermal ablation of cancer cells". Macromolecular Bioscience. 13 (1): 28–34. doi:10.1002/mabi.201200241. PMID   23042788.
  37. Li S, Wang X, Hu R, Chen H, Li M, Wang J, et al. (December 2016). "Near-Infrared (NIR)-Absorbing Conjugated Polymer Dots as Highly Effective Photothermal Materials for In Vivo Cancer Therapy". Chemistry of Materials. 28 (23): 8669–8675. doi:10.1021/acs.chemmater.6b03738. ISSN   0897-4756.
  38. Zhang G, Li P, Tang L, Ma J, Wang X, Lu H, et al. (March 2014). "A bis(2-oxoindolin-3-ylidene)-benzodifuran-dione containing copolymer for high-mobility ambipolar transistors". Chemical Communications. 50 (24): 3180–3. doi:10.1039/c3cc48695h. PMID   24519589.
  39. Cao Z, Feng L, Zhang G, Wang J, Shen S, Li D, Yang X (February 2018). "Semiconducting polymer-based nanoparticles with strong absorbance in NIR-II window for in vivo photothermal therapy and photoacoustic imaging". Biomaterials. 155: 103–111. doi:10.1016/j.biomaterials.2017.11.016. PMID   29175079.
  40. Lyu Y, Fang Y, Miao Q, Zhen X, Ding D, Pu K (April 2016). "Intraparticle Molecular Orbital Engineering of Semiconducting Polymer Nanoparticles as Amplified Theranostics for in Vivo Photoacoustic Imaging and Photothermal Therapy". ACS Nano. 10 (4): 4472–81. doi:10.1021/acsnano.6b00168. hdl: 10220/42127 . PMID   26959505.
  41. Cao Y, Dou JH, Zhao NJ, Zhang S, Zheng YQ, Zhang JP, et al. (January 2017). "Highly Efficient NIR-II Photothermal Conversion Based on an Organic Conjugated Polymer". Chemistry of Materials. 29 (2): 718–725. doi:10.1021/acs.chemmater.6b04405. ISSN   0897-4756.
  42. Cheng L, Yang K, Chen Q, Liu Z (June 2012). "Organic stealth nanoparticles for highly effective in vivo near-infrared photothermal therapy of cancer". ACS Nano. 6 (6): 5605–13. doi:10.1021/nn301539m. PMID   22616847.
  43. Yan H, Zhao L, Shang W, Liu Z, Xie W, Qiang C, et al. (February 2017). "General synthesis of high-performing magneto-conjugated polymer core–shell nanoparticles for multifunctional theranostics". Nano Research. 10 (2): 704–717. doi:10.1007/s12274-016-1330-4. ISSN   1998-0124. S2CID   100521646.
  44. Gong H, Cheng L, Xiang J, Xu H, Feng L, Shi X, Liu Z (December 2013). "Near-Infrared Absorbing Polymeric Nanoparticles as a Versatile Drug Carrier for Cancer Combination Therapy". Advanced Functional Materials. 23 (48): 6059–6067. doi:10.1002/adfm.201301555. S2CID   137636106.

Further reading