<<

Advanced Delivery Reviews 171 (2021) 94–107

Contents lists available at ScienceDirect

Advanced Reviews

journal homepage: www.elsevier.com/locate/addr

Targeting drug delivery with light: A highly focused approach

Teresa L. Rapp a,ColeA.DeForesta,b,c,d,e,⁎ a Department of Chemical Engineering, University of Washington, Seattle, WA 98105, USA b Department of Bioengineering, University of Washington, Seattle, WA 98105, USA c Department of Chemistry, University of Washington, Seattle, WA 98105, USA d Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA 98109, USA e Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA 98105, USA article info abstract

Article history: Light is a uniquely powerful tool for controlling molecular events in biology. No other external input (e.g., heat, Received 16 October 2020 ultrasound, magnetic field) can be so tightly focused or so highly regulated as a clinical laser. Drug delivery vehi- Received in revised form 4 January 2021 cles that can be photonically activated have been developed across many platforms, from the simplest “caging” of Accepted 8 January 2021 therapeutics in a prodrug form, to more complex micelles and circulating liposomes that improve drug uptake Available online 22 January 2021 and efficacy, to large-scale hydrogel platforms that can be used to protect and deliver macromolecular agents in- cluding full-length proteins. In this Review, we discuss recent innovations in photosensitive drug delivery and Keywords: Photochemistry highlight future opportunities to engineer and exploit such light-responsive technologies in the clinical setting. Photoactivated ©2021ElsevierB.V.Allrightsreserved. Light-mediated delivery In vivo drug delivery Photodegradable hydrogels Photoresponsive nanoparticles Prodrugs Dual-action Photoswitches

Contents

1. Introduction...... 95 2. Circulatingdrugcarriers:small-moleculeprodrugs,micelles,andliposomes...... 96 2.1. Small-moleculeprodrugs...... 96 2.1.1. Photocagedsmallmoleculesforinvitrodiscovery...... 98 2.1.2. Cagedprodrugsfortargeteddelivery...... 99 2.2. Nanoparticledeliveryvehicles:micelles,liposomes,andnanoparticles...... 100 2.2.1. Disruptingnanostructureswithorganicphotosensitizers...... 100 2.2.2. Disruptingnanostructureswithinorganicphotosensitizers...... 101 3. Drug-loadeddepots:softbiomaterialsasdrugdeliveryplatforms...... 101 3.1. Phototriggereddrugreleasefromhydrogels...... 101 3.1.1. Photodegradablehydrogelsfordrugdelivery...... 101 3.1.2. Photocleavablelinkerstoreleasetetheredcargo...... 102 3.2. Directing cell growth In Vitro withlight...... 103 3.3. Light-responsivelivinghydrogels...... 103

Abbreviations: 4FAB, tetrafluoroazobenzene; ASO, antisense oligonucleotide; βCD, beta-cyclodextrin; bLA, beta-lactamase; BSA, bovine serum albumin; Ce6, chlorin e6; DOX, doxorubicin; dVio, deoxyviolacein; EGF, epidermal growth factor; FWHM, full width at half maximum; GABA, γ-aminobutyric acid; HA, hyaluronic acid; IR, infrared; LOV2, light-oxy- gen-voltage-sensing domain 2; MRI, magnetic resonance imaging; mRNA, messenger RNA; NHS, N-hydroxysuccinimide; NIR, near-infrared; NMT, N-myristoyltransferase; oNB, ortho- nitrobenzyl; PA-IPTG, photoactivatable isopropyl β-d-1-thiogalactopyranoside; PDT, photodynamic therapy; PEG, poly(ethylene glycol); POI, protein of interest; PTX, paclitaxel; RISC, RNA-Induced silencing complex; ROS, reactive oxygen species; siRNA, short interfering RNA; TGF-β1, transforming growth factor beta-1; UCNP, upconverting nanoparticle; UV, ultraviolet. ⁎ Corresponding author at: Department of Chemical Engineering, University of Washington, Seattle, WA 98105, USA. E-mail address: [email protected] (C.A. DeForest).

https://doi.org/10.1016/j.addr.2021.01.009 0169-409X/© 2021 Elsevier B.V. All rights reserved. T.L. Rapp and C.A. DeForest Advanced Drug Delivery Reviews 171 (2021) 94–107

4. Perspectives...... 103 Acknowledgements...... 104 References...... 104

1. Introduction most organic and inorganic chromophores have molar absorptivities (104–105 M−1 cm−1 at 500 nm) that exceed those of biological chromo- Engineering photoresponsive behavior into drug delivery vehicles phores such as human rhodopsin (~104 M−1 cm−1 at 500 nm), the com- has progressed remarkably since the first literature expounded on the paratively high concentration of the latter can necessitate fairly large benefits of light-based therapies. Early treatments of lupus vulgaris light dosages to phototrigger engineered material changes. Since quan- exploited ultraviolet (UV) light, a therapy option that in 1903 earned tum yields are often lower than desired for many traditional photo- Niels Ryberg Finsen one of the first awarded Nobel Prizes in Medicine chemical reactions, the flux of light required may near the range of [1]. Since this discovery, light has found consistent use in the delivery thermal tissue damage, especially in the use of high-energy light. For ad- and activation of in the body [2]. Despite challenges associated ditional commentary on photothermal tissue damage, we direct the with tissue opacity, light’s surprising tenacity over decades of drug de- reader to this excellent review [1]. livery advancements can be attributed to the uniquely enabling spatio- While challenges persist in its clinical application, light is unique temporal control that it affords (Fig. 1a). among all other endogenous and exogenous triggers in important re- The first and primary challenge facing the clinical application of spects: (1) Light as electromagnetic radiation occurs in a wide range photoresponsive drug delivery vehicles is the poor penetration depth of efficacious and relatively safe energies – nominally UV, visible, and of low-energy electromagnetic radiation. While X-rays or radio waves IR light – that can be used to achieve molecular changes to activate may pass through the body with relative ease, visible, UV, and infrared and/or release drugs in a fully cytocompatible manner [7]. These ener- (IR) light experience variable and high levels of absorption/scattering gies can be accessed selectively using different wavelengths of light, by living tissue. As such, the greatest depth of penetration is achieved allowing researchers to potentially trigger multiple events separately by low-energy IR light, with 750 nm light penetrating ~5 mm below and/or sequentially, compounding the efficacy of drug therapies. the skin’s surface (Fig. 1b). Due to high amounts of light scattering as (2) Light can be highly focused to yield localized response, permitting well as high absorption of hemoglobin and melanin, higher-energy precise spatiotemporal control over therapeutic release. Light- light fares much worse with 350 nm UV light penetrating less than a responsive drug constructs can be dispersed throughout the body but millimeter before complete attenuation [3,4]. This increased attenua- activated only within the targeted tissue or organ using a variety of tion of high-energy over low-energy light provides for the optical win- readily available light sources (Box 1). dow for in vivo photoregulation, the range of most effectively Owing to these intrinsic features, light has persisted as one of the penetrating wavelengths in the visible-IR regions, generally accepted most popular external triggers for therapeutic application to date. to be between 650 – 900 nm [5,6]. In this regard, photomediated reactive oxygen generation, small- Another consideration when using light to trigger biological events molecule activation, micelle disruption, and material degradation is the energy flux through tissue to the location desired. Though has dominated the literature for the past several decades [8–12].

Fig. 1. A. Methods for incorporation of photoresponse in drug delivery. Small-molecule prodrugs (red), nanostructures (blue), and hydrogel biomaterials (green) have all been modified with photoactivators to deliver drug cargo. B. Differently colored light affords different penetration into skin. Image depicts the maximum depth into human skin that >1% of incident photons will reach as a function of wavelength. (For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.)

95 T.L. Rapp and C.A. DeForest Advanced Drug Delivery Reviews 171 (2021) 94–107

Box 1 spatiotemporally governing cell fate, as scientists work towards the ul- Light sources for laboratory light delivery timate goal of culturing organoids and growing tissues on the benchtop. There are several options for expanding in-house access to light sources In an age when fully folded proteins are poised as the next generation of at a wide variety of wavelengths. The most readily available are listed therapeutics, biomaterials spanning several orders of magnitude in size below with their associated wavelength ranges. have been used to protect sensitive protein cargo. This Review surveys common approaches within photomediated Mercury Arc 250–600 nm Broad-spectra mercury-arc lamps drug delivery, highlighting recent reports and innovative applications lamps are available from several suppliers using systems across a wide range of size scales including small mole- (e.g., Excelitas) whose output can cules, nanostructures, and hydrogels. Recurring chemical moieties that be refined to desired wavelengths render material photoresponsiveness will be discussed, including new with bandpass filters (typically to ruthenium-based small-molecule photosensitizers and innovations in capture major outputs at 254, 365, the use of ortho-nitrobenzyl (oNB) groups, inorganic nanoparticles, 405, 436, 546, and/or 579 nm). and conjugated organic porphyrins (Table 1). We offer our near- and These light sources are compara- long-term vision of the field’s future, identifying technical innovations tively inexpensive and high that will need to be developed along the way so as to maximize utility powered. and clinical translation. LEDs 250–700 nm A wide variety of wavelengths and intensities are available from several 2. Circulating drug carriers: small-molecule prodrugs, micelles, and manufacturers. ThorLabs offers liposomes multiwavelength arrays of LEDs for use in microscopy that can be easily The primary motivation for the design of photoregulatable circulat- adapted for light delivery on the ing drug carriers is the minimization of off-targeting effects; ubiquitous benchtop. One can also purchase delivery through the circulatory system but local activation with light individual LEDs from companies affords precise control over therapeutic function. This benefit is particu- (e.g., LED Engin) for user control larly attractive in cancer treatment, where chemotherapeutics are cyto- over wavelength and power. While toxic by design and likely to harm healthy tissue if misdelivered. In this LEDs offer changeable intensity, regard, several photosensitizers for PDT have reached the clinic, with their emission can be broad, with photoactivatable biomolecules following closely behind. Photocaged the full-width half maximum species have been used extensively in vitro, where achieving light pen- (FWHM) value extending etration is less of an issue, and selectively in vivo, particularly in small ±10-20 nm beyond the reported animal models including zebrafish embryos. Abundant literature is wavelength. available on the use of large, photoresponsive caging groups for neuro- Lasers 250–900 nm Laser pointers may be repurposed transmitters that have helped drive a revolution in neurobiology and for laboratory research, and are the study of neuronal signaling on the benchtop [17,35,36]. often desirable for their highly “Prodrugs” have also been widely explored for localized therapeutic focused beam and narrow regulation. Such small molecules have little to no activity until they en- bandwidth; lasers are often counter a specific stimulus (e.g., light), gaining activity through a con- centered within ±1 nm of the formational change or generation of other reactive species. Of these, reported wavelength. Zbolt offers a the generation of ROS is the most common mode of action and is partic- selection of high powered, ularly effective when cell destruction is desired. Many metal-based che- constant-on laser pointers that are motherapeutics utilize light to yield a structural change, revealing useful for benchtop experiments species that can bind to DNA and trigger apoptosis [37,38]. and the irradiation of a small area. Other circulating, light-responsive designs include the incorporation Though highly focused, laser of photoactivators in micellar and liposomal structures leading to their pointers cannot readily change triggerable disruption and associated cargo release. These nanostruc- intensity, so experiments testing tures are often useful at solubilizing hydrophobic drugs, taking advan- variable light intensities are tage of current photoresponsive technologies both organic and challenging. inorganic in nature, and maintaining appreciable therapeutic levels in the body while avoiding rapid by the kidneys. Micelles and li- posomes incorporating hydrophilic or hydrophobic photoactivators to create or destroy structure upon light irradiation have been reported, Photodynamic therapy (PDT), where small molecules are designed to some of which have successfully utilized in mouse models [39,40]. generate reactive oxygen species (ROSs) upon irradiation, is well repre- These in vivo studies demonstrate long circulation times on the order sented in the literature [13,14]. The entry of several PDT drugs into clin- of 2–4 hours and accumulation in the liver and kidneys, but with little ical trials and FDA approval [15] has led to innovations in light off-target activation or side effects. application devices in the clinic, enabling light to be delivered to any re- In this section, we will discuss past and present efforts in gion in the body with varying levels of invasiveness [1]. “Caging” small photoregulating circulating carrier activity, highlighting unique exam- molecules by blocking their activity until irradiation liberates the bioac- ples and pointing the reader to other references as appropriate (Fig. 2). tive molecule has prompted the elucidation of new neuronal circuits and advancements in epigenetics [16,17]. 2.1. Small-molecule prodrugs Embedding light-responsive nanoparticles or highly conjugated or- ganic molecules in micelles and liposomes has rendered these popular Prodrugs, or caged small molecules, have brought innovation to sev- nanostructures photosensitive, combining their potential for solubiliz- eral areas of drug delivery, from targeted chemotherapeutics to directed ing hydrophobic drugs with the possibility of targeting drug delivery. gene expression. These small molecules can be introduced to cells in Other advances using light to create and/or anisotropically modify culture by inclusion in the media, or by injection into various animal biologic-presenting materials has launched an entirely new field of models. Frequently these designs are not targeted to a specific cell

96 T.L. Rapp and C.A. DeForest Advanced Drug Delivery Reviews 171 (2021) 94–107

Table 1 Common photochemical reactions for

Structure Reaction Citation

Azobenzene [18,19]

Ortho-nitrobenzyl [23]

[22,24]

[25]

[26–30]

Coumarin [20]

[21]

[22]

Ruthenium

[31,32]

(continued on next page)

97 T.L. Rapp and C.A. DeForest Advanced Drug Delivery Reviews 171 (2021) 94–107

Table 1 (continued)

Structure Reaction Citation

[33]

[34]

type, but see uptake in all cells, followed by spatially specific activation or uncaging. Here we will discuss various molecular designs and their applications in recent literature, from caging neurotransmitters to targeted gene expression profiles.

2.1.1. Photocaged small molecules for in vitro discovery Controlled in vitro drug delivery or activation has led to an increased understanding of neuronal systems and intercellular communication. Triggering a physical change in a small molecule to reveal an active form with high spatiotemporal control is one of the most straightfor- ward applications of light-based technologies. For over 200 years, the study of ion channels and neurophysiology has been facilitated by appli- cation of electrodes to cultured neuronal tissue slices. With the advent of light-activated neurotransmitters and optogenetic proteins, light has slowly taken over the field as the most predominant interrogator of these complex networks of cells [41]. The Ellis-Davies research group was one of the early pioneers of this field, using photocaged neu- rotransmitters (e.g., glutamine) for the detailed study of neuronal ion channels and receptors [35]. In this field, two major classifications of photoactivated small fi Fig. 2. Light-responsive small molecules have distinct bene ts in terms of ease of dosing, molecules have emerged, characterized by whether modulation wide circulation, as well as spatiotemporally targeted activation. Light-activated designs rely on the circulatory system for delivery into local tissue, where the active form is occurs reversibly or irreversibly. Reversible photoswitches based on revealed through targeted irradiation. Common strategies exploit prodrugs, dual-action azobenzene or coumarin-diene moieties can control membrane poten- drugs, and reversible photoswitches. tial [42] and the structure of peptides/proteins [18]. Since 1971,

98 T.L. Rapp and C.A. DeForest Advanced Drug Delivery Reviews 171 (2021) 94–107 reversible molecular switches have dramatically altered the study of versatile cages for many different types of biologically active molecules. molecular interactions inside and between cells [43–46]. Recently, the While early work in the design of ruthenium coordination complexes Ellis-Davis group reported the use of the incredibly thermostable focused on caging primary amines [54,61,62], their utilization and tetrafluoroazobenzene (4FAB) for the modulation of voltage-gated ion scope rapidly expanded to include control of more complex biomole- channels in living neurons [47]. By appending two quaternary amines cules. Ruthenium polypyridyl complexes have been employed to cage onto a 4FAB structure, their molecular structure was able to modulate a broad spectrum of biomolecules including nicotine [63], thioethers ion channel flow in cultured brain tissue slices only when in the light- [64], and enzyme inhibitors [65,66]. Small-molecule ruthenium com- activated cis conformation. The same group has also reported the devel- plexation has permitted caging of previously uncageable moieties, par- opment of other molecular photoswitches derived from coumarin [20]. ticularly nitrile and pyridine-based groups [66]. While not as common Irreversible photoresponsive molecules have been primarily used to in neurotransmitters, nitrile and pyridine moieties are common in in- cage bioactive small molecules, dramatically reducing their biological hibitors for metalloenzymes, as they are excellent metal-binding motifs activity through attachment of a large photoresponsive group that bind reversibly to iron and copper-based active sites. Using a metal- (i.e., putting the bioactive molecule in a “cage”). As interest in the based photocaging group to bind the active pyridine or nitrile moiety photocages originated in the field of neuroscience, γ-aminobutyric has been shown to successfully knock down or fully eliminate back- acid (GABA) and glutamate were among the first molecules to be ground activity of these enzyme inhibitors [66]. Inhibitors targeting ca- caged. As the predominant chemical messengers in the brain and thepsins and cytochrome P450 have been successfully caged following a eliciting opposite responses, GABA and glutamate remain the most pop- relatively simple synthetic pathway and photoreleased in vitro [65]. ular photocaged neurotransmitters in modern-day literature. Gluta- Ruthenium complexes as a class of biologically active drug targets mate is an excitatory neurotransmitter that activates a large family of are not new to the scene of targeted drug delivery. The more common glutamate receptors, while GABA is an inhibitory neurotransmitter application of these inorganic complexes is triggered cell apoptosis via that binds ligand-gated chloride channels and G-protein-coupled recep- irreversible DNA binding and generation of ROS [67,68]. As such, ruthe- tors [48]. GABA has been caged by a variety of light-sensitive species, nium photocages can also be designed as a dual-action drug, where all each with their own advantages and disadvantages; oNB- [49,50], photoproducts are bioactive [69]. This dual therapy is purported to (methoxy-)nitroindolino- [51,52], 7-diethylaminocoumarin- [53], and have high efficacy against drug resistant cancers and complex tumor en- ruthenium- [54] based photocages have each been reported. Glutamate vironments. Dual-action Ru(II) complexes have been published from has also been caged with many of the same photosensitive groups several groups, with a focus on the release of enzyme-binding mole- [55–57]. cules as well as light-mediated DNA damage and cell death. Upon re- Photocaging neurotransmitters and other small molecules is not al- lease of the bioactive molecule inside the cell, the Ru(II) core is free to ways an easy task. Caging methodologies are well reported for several migrate to the nucleus and bind DNA, a process which leads to some functional moieties (e.g., amines, alcohols) but not all, and may prove in the dark, and is compounded upon light irradiation and gen- challenging when many such groups are present on a single species. Ad- eration of ROS. Delivery of chemotherapy agent 5-cyanouracil via Ru ditionally, in some cases, bulky caging groups must be employed to polypyridyl complex as a dual-action design has successfully degraded eliminate activity in the dark state. In one example, the Ellis-Davies DNA and slowed HeLa cell growth [64,67]. The Glazer group has studied group used a large dendrimer to fully cloak GABA’s activity before irra- the combination of cytochrome P450 inhibition with Ru(II)-mediated 1 diation [58]. This technique may prove to be necessary in more systems O2 production in vitro [31]. Arora et al. demonstrated this technique than currently described, as researchers further advance their work in 2D and 3D cell culture, targeting surface cathepsin B with a with caged neurotransmitters and other small molecules. In many photoreleased inhibitor, then tracking uptake of the Ru(II) metal center cases, these model compounds have not made it beyond initial in vitro into the cell, leading to photomediated cell death [70]. While examples studies on cells cultured on the benchtop; stability and dark activity in of Ru-biomolecule dual-action therapies abound [71–73], none have an in vivo setting have yet to be determined for many of these constructs. progressed to in vivo trials. While these reports suggest these complexes In vitro discovery extends beyond caged neurotransmitters to other are stable in cell culture medium and serum, effective tumor targeting techniques for monitoring RNA levels and the transcriptome in living has not been reported for these small molecules, whether administered tissue samples. Lovatt et al. demonstrated the use of a sterically caged intraperitoneally or intravenously. Transcriptome In Vivo Analysis tag composed of a poly-U RNA oligomer Conversely, simpler prodrugs may rely on a conformational or mo- for hybridizing messenger RNA (mRNA) poly-A tails, two oNB lecular change to reveal a reactive moiety or drug once inside the cell. photocleavable linkers, two fluorophores to track location and These prodrugs are common in the literature, ranging from activating uncaging, and a biotin tag for pulldown [59]. Upon irradiation with common inorganic cisplatin-like derivatives to revealing a chelating li- single-photon UV or multiphoton red light, the crosslinkers are cleaved gand. Significant work has been done in the Sadler group to generate and blocking strands melt off the construct to reveal the poly-U binding stable Pt(IV)-based prodrugs that are reduced to bioactive Pt(II) upon tag. The tagged mRNA is isolated from irradiated cells (e.g., neurons in irradiation, and/or exchange stabilizing ligands for water, activating cultured brain slices) and analyzed for gene expression. This probe has the Pt-based drug in a similar way to cisplatin. These complexes can been iteratively improved upon, and currently studies are underway be designed to generate ROS in response to low-power red light [74] using it in more complex in vivo environments [60]. and to bind DNA [75]. Other reviews on this extensive area of research have been written in recent years [76–78]. 2.1.2. Caged prodrugs for targeted delivery With innovations in light sources and photoresponsive molecular 2.1.2.2. Small-molecule light-activated theranostic approaches. Several design, an increasing number of small molecules have been caged for photoactivatable small molecules have been reported that can serve on-demand activation in vivo. Interestingly, many prodrugs designed both diagnostic and therapeutic roles in the treatment of disease. for use in vivo have more than one target or purpose. Here, we see the These theranostic species often take advantage of elements at the center advent of dual-action agents where both the caging group and the of the periodic table for either the imaging moiety or the phototoxic caged molecule have biological activity, as well as the expansion of a prodrug. In one example, Chaudhuri et al. recently reported the new field of drug delivery – theranostics – where the same small mole- phototriggered release of a copper-binding ligand that, when chelated cule can both diagnose and treat disease. with endogenous copper, effectively turns on Cu’sapoptoticproperties [79]. Coupling targeted release of this chelator with the sequestration 2.1.2.1. Transition-metal complexes as photocages and dual-action of Cu in cancer cells of the breast, prostate, large intestine, lungs, and prodrugs. Ruthenium complexes have been recently developed as brain could lead to an effective method of shutting down cancer cell

99 T.L. Rapp and C.A. DeForest Advanced Drug Delivery Reviews 171 (2021) 94–107 migration and initializing apoptosis [80]. In another theranostic ap- layers, and theranostics, wherein drug delivery and efficacy can be ob- proach, Li et al. linked a europium-based fluorophore and a ruthenium served in real time [97,98]. The ability to trigger cargo delivery from DNA-binding prodrug to image and trigger release of the Ru DNA- these systems using light further establishes their utility in targeted binding phototoxic core [81]. Imaging with low- (700 nm) and activat- drug delivery. ing with higher-energy light (488 nm) permitted selectivity in this Micelles and liposomes are common drug carriers: their readily ac- approach, with efficacy demonstrated in vivo in a mouse tumor model. cessible syntheses and surface modification provides flexible handles Biswas et al. leveraged coumarin’s native fluorescence to synthesize a for targeting their cellular uptake, and the combination of hydrophobic coumarin-doxorubicin (DOX) chemotherapeutic prodrug. Based on and hydrophilic regions enable efficient loading of chemically distinct progressive structural changes of the coumarin fluorophore in response drugs. Polymer-based micelles have already entered into clinical trials to light and local nitric oxide concentrations, the design facilitated imag- as delivery vehicles for hydrophobic chemotherapeutics including pac- ing cellular uptake, detecting elevated levels of nitric oxide, and subse- litaxel (PTX), DOX, and others [99]. Incorporating light responsiveness quent release DOX upon irradiation [82]. into nanostructures is an effective way to increase targeting to tumors, and is compatible with other targeting methods [e.g., surface 2.1.2.3. Photoregulating gene expression. As therapeutic targets move functionalization, modulating size/geometry, protecting circulating progressively towards large complex biomolecules (e.g., full proteins, drugs by PEG-ylation with poly(ethylene glycol)] [100]. Both organic oligonucleotides), efforts to control efficacy of these therapeutics with photosensitizers and inorganic small molecule and nanoparticle photo- light have increased. Specifically, directing gene function with light sensitizers have been incorporated to endow photoresponsiveness into has proved useful in spatiotemporally regulating gene expression [83]. polymeric nanostructures [101]. Temporary gene knockdown in vivo is often achieved through the use of single-stranded antisense oligonucleotides (ASOs) or double- stranded short interfering RNA (siRNA). ASOs may be modified at 2.2.1. Disrupting nanostructures with organic photosensitizers their backbone to be more stable to deoxyribonucleases present in Organic photosensitizers take many forms and can interact with serum and prolong their half-life; common modifications include ex- light in several ways. To build this functionality into a liposome or mi- changing the phosphodiester linkage for a morpholine-based linker or celle, photosensitizers have been leveraged to break bonds and disrupt thiolating the backbone. ASOs bind to mRNA, and can be designed to the hydrophobic/hydrophilic nature of the polymers (in the case of mi- block any number of translational steps as transcribed RNA is spliced celles) or disrupt intermolecular interactions between hydrophobic and read by the ribosome [84]. polymers (for liposomes). In one interesting example, Poelma et al. de- Caging ASOs can be achieved through stochastic decoration of indi- veloped a donor-acceptor Stenhouse adduct that switched from hydro- vidual nucleotide bases with photocaging groups [85,86] or by enforcing phobic to hydrophilic upon green light irradiation (530–570 nm). In its a secondary structure which blocks hybridization. The latter approach hydrophobic form, the photosensitizer induced micelle formation involves only one photocaging group, compared to multiple cages re- around PTX. When the photosensitizer was converted to a hydrophilic quired for the first design, and can give extended serum stability due molecule through irradiation, the micelle was rendered unstable and to the secondary structure of the oligonucleotide. Both morpholine- the PTX was released [102]. modified (“morpholinos”)andunmodified ASOs have been used Linking hydrophilic and hydrophobic polymers with a cleavable in vivo and ex vivo. The Chen group has long pioneered this method, linker has also shown promise in disrupting micellar structures to re- even using multiple crosslinkers to circularize two different morpholino lease drug cargo. oNB-based photocleavable crosslinkers have been ASOs. In their recent report, the group multiplexed oNB and coumarin used in micelle formulations to break the junction point between hy- photocrosslinkers (photoresponsive molecules used to crosslink two drophobic and hydrophilic polymers [25]. In their material design, Pei parts of a molecule together) to give selective gene silencing with two et al. formed polymer-based micelles around ROS-generating chlorin wavelengths of light [22,24]. The Dmochowski group has also recently e6 (Ce6), attaching doxorubicin to the polymer via an ROS-sensitive published examples improving upon this design, using a ruthenium- thioketal bond [103]. As Ce6 is sensitive to red light (660 nm), localized based photocleavable crosslinker for in vivo gene knockdown [87]and DOX release was demonstrated in tumors in a mouse model. Incorporat- incorporating a non-coding stem to further decrease activity in the ing ROS-sensitive bonds between hydrophobic and hydrophilic poly- dark [88]. mers in micelle structures has also been demonstrated in vivo.Intheir siRNA is more popular than single-stranded ASOs, primarily for its design, Uthaman et al. synthesized a poly(ethylene glycol)-stearamine extended stability and ease of synthesis [89,90]. Caging or deactivating (C18) amphiphilic construct linked with a thioketal linker and loaded siRNA often relies on sterically blocking the binding of the RNA- the resultant micelles with DOX and a photosensitizer pheophorbide A Induced silencing complex (RISC) to the siRNA [91], either through in- [104]. Upon accumulation in tumors, red light (670 nm) was used to cluding azobenzenes to change the structure of the antisense strand generate ROSs and break the micelle structure, releasing the DOX and [92], incorporating photocaging molecules on individual bases [93,94], killing tumor cells. Positive results for mouse studies demonstrated or modifying the 5′ terminal phosphate [95]. Interestingly, serum and the clinical relevance of this technique, while also laying the ground- cellular proteins can also be harnessed to cage siRNA, as shown in recent work for other light-activated nanoparticulate systems usage in vivo. work by Ji et al, who used a vitamin E modification at the 5′ terminus of Photodisrupting liposome structure follows a slightly different the sense strand coupled with an oNB group to give photoactivity. They methodology, with fewer examples of cleaving covalent bonds and found that extensive recruitment of vitamin E-binding proteins helped more examples with larger, conjugated porphyrin molecules that yields block extraneous activity. These recruited proteins and bulky lipid fac- membrane destabilization upon irradiation. This can lead to better drug tors were released upon irradiation with UV light, restoring activity release from endosomes following endocytosis by prompting fusion of and successfully knocking down two genes of interest [96]. the liposome with the endosome membrane, as well as direct liposome structural breakdown. In one example, a malachite green porphyrin was 2.2. Nanoparticle delivery vehicles: micelles, liposomes, and nanoparticles attached to the outside cell membrane of a 1-palmitoyl-2-oleoyl-sn- glycero-3-phosphatidylcholine liposome, which upon UV irradiation Nanoparticle drug delivery systems, including those based on cova- dramatically improved DOX delivery to the cytosol and nucleus [105]. lent polymer networks and micellar structures, have revolutionized the Similarly, a polymer-based system carrying Zn2+ and Ca2+ ions was drug delivery industry. These platforms have led to dramatic improve- disrupted by the incorporation of a fused porphinato-Zn construct in ments in improved and targeted delivery of poorly water-soluble the hydrophobic region that upon blue light irradiation (488 nm) drugs, transcytosis across cellular membranes and tight endothelial underwent a conformational change. This hydrophobic bilayer

100 T.L. Rapp and C.A. DeForest Advanced Drug Delivery Reviews 171 (2021) 94–107 disruption successfully released a variety of cargos, including large fluo- 3. Drug-loaded depots: soft biomaterials as drug delivery platforms rescent conjugates [106] and cations (e.g., Zn2+,Ca2+) in vitro and in vivo [107]. Circulating drug delivery vehicles have driven significant innovation Combining different modes of action helps increase the specificity of in improving drug solubility, circulation time, and targeting. In certain targeting these nanostructures to a more localized region. One such de- instances, however, a large, non-circulating depot containing a thera- sign takes advantage of hypoxic environments in tumors and a light- peutic is preferable to oral or intravenous drug administration absorbing conjugated polymer to act as a multifunctional polymersome [125,126]. In the brain, for example, using a drug-loaded polymer mate- construct. The incorporated 2-nitroimidazole group generates ROS rial that is surgically implanted can dramatically increase and prolong under green light irradiation, followed by a change in polarity triggered drug delivery by side-stepping the blood-brain barrier. Larger therapeu- by elevated ROS concentrations and hypoxic conditions. With the loss of tics including full-length proteins can more easily be administered to hydrophobicity upon irradiation, the polymersome is disrupted, releas- local tissue via an injectable or surgically implanted drug depot, as the ing DOX to the intercellular environment [108]. A theranostic liposome material design can protect sensitive cargo. A stationary, implanted has also been developed by Liu et al., combining a gadolinium magnetic drug depot also opens the possibility of more complex release profiles resonance imaging (MRI) sensitizer with a liposome structure linked by and designs that incorporate live cells for real-time responsiveness to oNB groups to both image localization and release DOX once localized biological cues, such as protected implants of islet cells for sensing and identified [109]. The utility of this construct was also demonstrated blood glucose levels. in vivo in a targeted mouse study, establishing the viability of this MRI- For the protection of complex therapeutics and encapsulation of live guided light-mediated therapy in a unique combination of electromag- cells, hydrogels have come to the forefront of these drug depot designs. netic interactions in the body. Hydrogels are made up of 90-99% water, with a 3D structure maintained by a hydrophilic polymer network. These biomaterials can be designed using any number of polymers, with a variety of mesh sizes for con- 2.2.2. Disrupting nanostructures with inorganic photosensitizers trolled cargo release, and a wide range of crosslinking density and stiff- The use of inorganic nanoparticles as photosensitizers is extensively ness to improve the body’s response to their implantation. reported in the literature. Gold nanoparticles, nanoshells, and nanorods Hydrogel biomaterials can be modified for light-triggered drug de- have been used for decades as photothermal sensitizers that generate livery in several ways (Fig. 3). Directly tethering drug cargo (large or local heating upon irradiation with red and near-IR (NIR) light. Such lo- small) to the hydrogel matrix with a photocleavable crosslinker has ef- calized heating has been leveraged to target cancerous tumors and local fectively sequestered and then released drug cargo in several designs. A tissue in several applications separate from drug delivery. Combining hydrogel may also be modified in their polymer backbone with a gold-based nanomaterials with thermosensitive polymers is one widely photocleavable crosslinker for degradation of the structure of the mate- reported method to trigger on-demand drug release from micelles or li- rial and subsequent release of cargo by opening broad pores in the ma- posomes, often by eliciting a phase transition within the lipid mono- or terial. Hydrogels may be rendered fully degradable by irradiation via bi-layer [110,111]. As this field is extensive and beyond the scope of this this method, an outcome that may be desired to eliminate the need to article, the reader is directed to other excellent reviews on the subject retrieve the depot after all the drug has been released. [112–114]. In this section, we will discuss recent examples demonstrating drug Upconverting nanoparticles (UCNPs) have also seen wide use for release from macroscale hydrogels, as well as several examples drug delivery. When coupled with existing photosensitizing technolo- exploiting photoresponsive hydrogels to probe and direct cell fate on gies, UCNPs have the potential to extend light-mediated drug delivery the benchtop. deeper into tissue. Researchers are working to use UCNPs to improve drug delivery in the brain [115] and to drug-resistant tumors [116], in 3.1. Phototriggered drug release from hydrogels addition to many other targets [117]. Taking advantage of their NIR fluo- rescence to image localization as well as deliver drug cargo, UCNPs have Hydrogel depots are most commonly reported as protective drug de- been utilized as theranostic agents in vitro [118,119]. The sensitivity of livery vehicles for large, complex therapeutics, with the appropriately UCNPs to IR light opens the possibility to achieve drug delivery in vivo sized and comparatively inexpensive globular protein bovine serum al- due to the light’s enhanced tissue penetration. UCNPs have been thor- bumin (BSA) frequently serving as a model drug. Delivering large pep- oughly studied and reviewed elsewhere; for a more detailed discussion tides and full-length proteins while maintaining native bioactivity on their uses in vivo and other effective designs, we recommend the remains a challenge in targeted drug delivery. In addition to the chal- reader to several excellent reviews on this topic [117,120,121]. lenges associated with working with inherently fragile species, thera- The use of photoactive inorganic coordination compounds as inte- peutic proteins must be protected from the harsh conditions of the gral parts of micelle drug delivery systems has increased as well, as stomach and blood; direct delivery to local tissue is often desired. more transition metal-based therapeutics enter clinical trials. One de- sign incorporates the cisplatin precursor Pt(IV)(NH3)2(N3)2 into the hy- 3.1.1. Photodegradable hydrogels for drug delivery drophobic polymer, which upon irradiation with sub-500 nm light In some cases, complete degradation of the hydrogel depot repre- ruptured the polymer to release bioactive Pt(II) [122]. The efficacy of sents a desirable trait. In this case, a photodegradable material may be their material design was also demonstrated in vivo in a mouse tumor designed to release cargo by breaking up the polymer matrix, opening model. In a similar vein, Sun et al. published the design of a pore sizes to allow for uninhibited diffusion of proteins from the mate- ruthenium-core micelle, taking advantage of the charged Ru(II) prodrug rial. Generally, attaching photoresponsive moieties to a polymer back- species to form a micelle structure and deliver the small molecule intra- bone as part of the crosslinking network forming the hydrogel can cellularly [123]. The Ru(H2O)2 species is released from the polymer render a hydrogel photodegradable. For example, modifying hyaluronic upon visible light irradiation (530 nm) and is freed to bind DNA and acid (HA) polymers with azobenzene or beta-cyclodextrin (βCD) leads generate ROS [124]. to the formation of photoresponsive crosslinks between polymers: While inorganic photosensitizers may be instrumental in increasing whentheazobenzeneisinthetrans isomeric form it can form a hydro- sensitivity to longer wavelengths of light, concerns over their toxicity, phobic “host-guest” interaction with βCD, forming strong crosslinks. especially for lanthanide-doped UCNPs, continue to persist. However, Upon irradiation, the azobenzene shifts to the cis isomer and can no lon- several biomaterial designs using UCNPs have demonstrated efficacy ger interact with βCD, breaking the crosslinks and opening pores in the in mouse models with limited background toxicity, suggesting they hydrogel. This interaction is reversible, yielding a material that can give may present a viable path forward. varying doses of cargo upon subsequent light treatments. Azobenzene-

101 T.L. Rapp and C.A. DeForest Advanced Drug Delivery Reviews 171 (2021) 94–107

Fig. 3. Different photoactivatable hydrogel designs with engineered light-responsive triggers for the release of bioactive therapeutics.

βCD chemistry has been employed in several formulations. Rosales et al. Taking advantage of reactive handles available as part of a protein’s demonstrated an HA-based hydrogel for the on-demand release of BSA native amino acid side chains is a popular method for covalent with UV light [19]. Subsequent irradiation with visible (405 nm) light crosslinking protein cargo to hydrogels. Primary amines and carboxylic returned a fully crosslinked hydrogel, demonstrating controlled dosing acid groups on various amino acids (e.g., lysine, glutamic acid, aspartic of large protein cargo. Similarly, Wang et al. synthesized a poly(acrylic acid) are reactive through a variety of chemistries and are often acid)-based hydrogel modified with βCD and methoxy-substituted solvent-accessible. In some cases, these residues can be covalently azobenzene for a red-shifted response [127]. Their BSA-loaded hydrogel linked to the hydrogel matrix without significant loss to protein func- responded to red light, releasing the protein cargo in bolus or in dose- tion, demonstrated by Grim et al. using transferrin and transforming dependent manners. growth factor beta-1 (TGF-β1) [131]. The authors used an allyl sulfide Photodegradable hydrogels containing oNB moieties within their that could undergo reversible fragmentation chain-transfer with surface polymeric backbone have also been used to release siRNA. In multiple thiol installed by N-hydroxysuccinimide (NHS) chemistries of several reports using similar material designs, the siRNA is trapped within the protein constructs (e.g., transferrin, ovalbumin, TGF-β1) to pattern in hydrogel until UV irradiation; upon photorelease, the active siRNA is and out of a PEG-based hydrogel. Tethered TGF-β1 stimulated down- able to knock down expression of model proteins (e.g., green fluores- stream signaling, indicating that the patterned protein retained some cent protein, luciferase) in cultured HeLa cells [26] and to direct osteo- biological activity. Rapp et al. used a ruthenium-based photocleavable genesis of human mesenchymal stem cells [27]. crosslinker to simultaneously crosslink an HA-based hydrogel as well Inorganic nanoparticles have also been incorporated into degradable as to tether model enzyme beta-lactamase (bLA) to the matrix [33]. hydrogel matrices for on-demand drug delivery. These material ap- Their Ru-based crosslinker was modified with aldehydes to serve as a proaches again fall into two use categories: taking advantage of thermal crosslinker for hydrazine-modified HA, as well as linking protein cargo responsivity in the hydrogel or coupling common photocleavable (in this case beta-lactamase) via surface lysine residues to the hydrogel crosslinkers with upconverting nanoparticles. Platinum nanoparticles matrix, though they observed significant loss of bLA activity upon re- were incorporated into a supramolecular hydrogel with thermally re- lease. Additionally, work by Sridhar et al. has shown that immobilizing sponsive interactions between PEG sidechains and α-cyclodextrin to proteins in hydrogels can help preserve enzyme activity even under render a NIR-responsive hydrogel [128]. This hydrogel design was in- thermal stress, maintaining activity of otherwise temperature- jectable (due to the supramolecular interactions) and “melted” in re- sensitive proteins for weeks at 60°C [132]. sponse to NIR irradiation, delivering various small-molecule While there are some examples in which random attachment of a fluorophores both in vitro and in a proof-of-concept in vivo trial injecting protein to a hydrogel results in minimal disturbance of activity, the the hydrogel into a tumor. vast majority of these approaches have a severely detrimental effect on protein activity. Using NHS chemistry to stochastically attach reac- tive groups (such as azides or alkynes for subsequent click reactions) 3.1.2. Photocleavable linkers to release tethered cargo onto the surface of a protein can completely eliminate enzyme activity. In cases where complete degradation of the hydrogel material is not This is suggested to be a result of over-modification of a protein to en- desired, cargos can be tethered directly to a stable material through a sure that the reactive group has been successfully installed. To eliminate photocleavable linker. In these instances, covalent attachment of the this challenge, several recent reports have used recombinant protein cargo to the hydrogel material can preserve its activity while minimiz- techniques to site-specifically modify proteins with a chemical or pro- ing untriggered release. Methods to covalently attach molecules have tein handle that can be used to attach a protein of interest (POI) to a hy- drawbacks, most frequently in terms of activity loss accompanying drogel. In one such example, Shadish et al. used a chemoenzymatic chemically unspecified tethering, though recent innovations and alter- transamidation-based strategy, expressing POIs with the recognition se- native modes of site-specifically tethering proteins to hydrogels have quence of a sortase enzyme at the C terminus [29]. When POIs are incu- worked to overcome these disadvantages [129,130]. bated with sortase and a peptide containing an N-terminal triglycine,

102 T.L. Rapp and C.A. DeForest Advanced Drug Delivery Reviews 171 (2021) 94–107 the peptide is enzymatically attached to the recognition sequence at the method of directing cell growth, migration, and identity has spread to C terminus of the protein. Using this approach, the authors successfully many other applications. Attempts to eliminate challenges presented modified bLA, epidermal growth factor (EGF), and several fluorescent by slow diffusion rates through hydrogels has led to innovations in proteins with azides, aldehydes, and oNB-based photocleavable immobilizing caged peptides or signals in a hydrogel that can be acti- crosslinkers. The site-specificmodification yielded proteins with native vated instantly upon irradiation. In one such example, Farrukh et al. bioactivity, and photoreleasable growth factors were used to control demonstrated the power of caging a lysine residue on a laminin adhe- signaling with subcellular resolution. Following up on this work, sive peptide which enabled them to direct neuron growth through a hy- Shadish et al. tethered proteins to hydrogel through a photocleavable drogel [140,141]. The del Campo group has shown the broad flexibility protein and demonstrated their spatiotemporal release using 405 nm of photocaged peptide modifications in hydrogels, successfully directing light [133]. cell growth through an oNB-caged alpha5beta1 integrin-binding pep- Site-specificmodification of proteins for incorporation into tide [142]. Caging cyclic RGD peptides similarly directed cell growth hydrogels is becoming the preferred method of attaching protein into fibers, and continues to be an active area of research [23]. Caged cargo to a hydrogel depot. In another example of a reversible protein- RGD peptides have proved very interesting in the study of immune re- hydrogel interaction, Hammer et al. generated a recombinant protein sponse to implanted biomaterials, when a hydrogel bearing islet cells construct taking advantage of the phototriggered, reversible protein was decorated with caged RGD and implanted. Activating the RGD pep- conformational change in light-oxygen-voltage-sensing domain 2 tide on the surface of the hydrogel after a timepoint of 24 hours led to an (LOV2) and the strong binding affinity of Zdark to its dark-state confor- improved immune response and prolonging the lifetime of transplanted mation [134]. POIs were genetically fused to the Zdark domain, demon- islet cells [143]. strating repeated loading and photorelease of model fluorescent proteins from a hydrogel modified with LOV2. 3.3. Light-responsive living hydrogels Combining photodegradable crosslinkers with linkers that cleave under other internal triggers such as local enzyme population or a re- “Living” hydrogels with embedded cells highlight the potential for ducing environment can improve targeting of such therapeutics to a continuous delivery of bioactive compounds while minimizing design highly localized area [134]. Targeting specific tumor or organ microen- complexity. This method of drug delivery also falls under cell therapy vironments may only be possible by rendering pro-drug formulations – using patient-derived or otherwise sourced cells to manufacture and accessible only via multiple inputs. For example, while tumors usually release protein therapeutics. When looking to engineer these living bio- have a lower pH than healthy tissue, so does the stomach and certain or- materials with a photoresponse, the del Campo lab has leveraged photo- ganelles within cells. To avoid drug uptake and off-target effects in these induced protein expression and secretion of proteins from Escherichia areas, an ideal material drug delivery platform may require input from a coli on and within gels. In their first demonstration of such living combination of external and internal triggers. In 2018, Badeau et al. in- hydrogels, the del Campo lab exploited a photoactivatable isopropyl troduced a generalizable strategy to program hydrogel degradation in β-d-1-thiogalactopyranoside (PA-IPTG) applied externally to trigger response to well-defined combinations of external inputs (including protein expression in E. coli immobilized on the surface of a hydrogel light) following Boolean YES/OR/AND logic [30]. Since this initial report, [144]. ClearColi® BL21(DE3) bacteria genetically modified to produce the DeForest group has extended these concepts in using topologically a lipopolysaccharides that do not trigger endotoxic response in human defined molecular crosslinks to control release of small molecules cells were immobilized to the gel surface using electrostatic interactions [135] and full-length proteins [136]fromstablegels. between the negative cell membrane and positive Poly-D-Lysine (PDL) polymer. Upon treatment of the system with PA-IPTG and light, surface 3.2. Directing cell growth In Vitro with light adhesion protein RGD was expressed, leading to mammalian cell adhe- sion and growth. Not only do hydrogels show promise in the safe sequestration and del Campos’ unique approach was expanded up in a subsequent re- delivery of sensitive drug cargo, they can be designed as platforms for port using embedded ClearColi® in a hydrogel to deliver deoxyviolacein encapsulating cells, differentiation of stem cells, and cell-mediated ther- (dVio), a tryptophan derivative with antifungal, antibacterial, and anti- apeutic response. Many types of hydrogels have been used for 3D cell tumor properties [145]. In this case, optogenetic control of enzyme ex- culture, optimizing cell growth conditions on the benchtop, and as sup- pression was incorporated using vioABCE as an upstream phototrigger portive scaffolds for the implantation of healthy cells into the body. of expression, eliminating the need for exogenous PA-IPTG. Sustained, These cell therapies rely on the protective hydrogel material to sustain pulsatile, and spatially resolved expression and release of dVio was a healthy cell population while also providing access to physiological achieved in this system, demonstrating the power of this technique. Fu- cues from the body when implanted. In many cases, researchers direct ture studies should include in vivo studies confirming biocompatibility patient-derived stem cell growth on the benchtop to form organs or of these hydrogels. organoids for future implantation. This benchtop control of cell fate and structure has driven many advancements in the use of light- 4. Perspectives responsive cues, either of signaling proteins attached to the matrix via a photocleavable linker, or in the modulation of material stiffness to Light will always retain its place as one of the most ubiquitous exter- drive cell fate; in these many cases, photochemistry is exploited to de- nal triggers in biology and drug delivery. The excellent and precise con- liver cues to encapsulated cells to alter function. trol it offers clinicians and researchers is unparalleled, and the broad Hydrogels for the study of cellular development on the benchtop range of photosensitizers makes the engineering of light-responsive have used light successfully in many applications to direct cell growth, technologies accessible to labs possessing different skillsets. Recent lit- stem cell differentiation, and cellular release for further study and clas- erature suggests that innovations in modern methods and molecular sification. Hydrogel matrix stiffness can be changed under irradiation, design will soon be able to address most if not all concerns in the directing cell growth towards softer regions. These photomodulated field. Light affords focusability and opportunities for multiplexed deliv- hydrogels have enabled significant additions to our knowledge of ery and response extending well beyond that achieved using other organ growth [137], and especially towards the growth of living tissues triggers. on the benchtop for improved drug efficacy outcomes [138]. Recent re- Because of this incredible flexibility and focusability, researchers views on the subject can be found in the literature [138]. continue to work towards the development of ideal photoresponsive After the initial work by the Anseth lab in directing cell growth by small molecules, nanoparticles, and hydrogels: response to low energy both hydrogel softening and patterning adherent peptides [139], this light, minimal dark activity, and demonstration of improved efficacy

103 T.L. Rapp and C.A. DeForest Advanced Drug Delivery Reviews 171 (2021) 94–107 in vivo. Many new reports in the literature outline a pathway to low- [8] P. Rai, S. Mallidi, X. Zheng, R. Rahmanzadeh, Y. Mir, S. Elrington, A. Khurshid, T. Hasan, Development and applications of photo-triggered theranostic agents, Adv. energy light responsive biomaterials and photoactivatable prodrugs, Drug Deliv. Rev. 62 (2010) 1094–1124, https://doi.org/10.1016/j.addr.2010.09. some demonstrated in vivo using small animal models. Despite these 002. encouraging results, additional characterization is required to elucidat- [9] Y. Qiao, J. Wan, L. Zhou, W. Ma, Y. Yang, W. Luo, Z. Yu, H. Wang, Stimuli-responsive ing the true stability and dark activity of many of these constructs prior nanotherapeutics for precision drug delivery and cancer therapy, Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 11 (2019), e1527. https://doi.org/10.1002/wnan. to clinical translation. 1527. With the advent of advanced photosensitizers that respond to lower [10] C.D. Jones, J.W. Steed, Gels with sense: supramolecular materials that respond to – energy light, it is increasingly possible to photochemically trigger multi- heat, light and sound, Chem. Soc. Rev. 45 (2016) 6546 6596, https://doi.org/10. 1039/c6cs00435k. ple events orthogonally within a single material design. As a critical step [11] M. Karimi, P. Sahandi Zangabad, S. Baghaee-Ravari, M. Ghazadeh, H. Mirshekari, towards capturing and capitalizing on biology’s complexity, the capabil- M.R. Hamblin, Smart nanostructures for cargo delivery: uncaging and activating ity to trigger two or more activating events with one biomaterial or drug by light, J. Am. Chem. Soc. 139 (2017) 4584–4610, https://doi.org/10.1021/jacs. 6b08313. cocktail represents a powerful innovation. There are several potential [12] E.R. Ruskowitz, C.A. DeForest, Photoresponsive biomaterials for targeted drug de- application spaces for such multiplexed biomaterials, including sequen- livery and 4D cell culture, Nat. Rev. Mater. 3 (2018), 17087. https://doi.org/10. tial release of proteins to direct an immune response [146]and 1038/natrevmats.2017.87. [13] X. Li, N. Kwon, T. Guo, Z. Liu, J. Yoon, Innovative strategies for hypoxic-tumor pho- supporting the regeneration of blood vessels by sequential release of todynamic therapy, Angew. Chemie - Int. Ed. 57 (2018) 11522–11531, https://doi. growth factors [147]. Bringing thermally stable low energy-responsive org/10.1002/anie.201805138. photosensitizers into biomaterials and prodrug designs is currently [14] S. Kwiatkowski, B. Knap, D. Przystupski, J. Saczko, E. Kędzierska, K. Knap-Czop, J. ń – inhibited by synthetic challenges, through researchers are actively ex- Kotli ska, O. Michel, K. Kotowski, J. Kulbacka, Photodynamic therapy mecha- nisms, photosensitizers and combinations, Biomed. Pharmacother. 106 (2018) ploring several promising avenues researchers to bring this technology 1098–1107, https://doi.org/10.1016/j.biopha.2018.07.049. forward. [15] R. Baskaran, J. Lee, S.-G. Yang, Clinical development of photodynamic agents and The last great hurdle for many of these innovative designs is the therapeutic applications, Biomater. Res. 22 (2018) 25, https://doi.org/10.1186/ s40824-018-0140-z. journey from 2D and 3D cell culture into animal models. Some of the [16] G.C.R. Ellis-Davies, Useful caged compounds for cell physiology, Acc. Chem. Res. 53 technologies mentioned here have been demonstrated with varying (2020) 1593–1604, https://doi.org/10.1021/acs.accounts.0c00292. levels of success in mouse models, suggesting their efficacy in complex [17] G.C.R. Ellis-Davies, Caged compounds: photorelease technology for control of cellu- lar chemistry and physiology, Nat. Methods. 4 (2007) 619–628, https://doi.org/10. tissues as well as their response to light through skin, muscle, and fat. To 1038/nmeth1072. continue bolstering this field of photomediated drug delivery, the most [18] D. Hoersch, Let there be light: how to use photoswitchable cross-linker to repro- successful approaches that will be developed in coming years must gram proteins, Biochem. Soc. Trans. 45 (2017) 831–837, https://doi.org/10.1042/ demonstrate design efficacy in vivo. BST20160386. [19] A.M. Rosales, C.B. Rodell, M.H. Chen, M.G. Morrow, K.S. Anseth, J.A. Burdick, Revers- Photomediated approaches continues to dominate the drug delivery ible control of network properties in azobenzene-containing hyaluronic acid-based literature, with exciting new molecular and material designs reported hydrogels, Bioconjug. Chem. 29 (2018) 905–913, https://doi.org/10.1021/acs. each year. With demonstrated application in probing fundamental bio- bioconjchem.7b00802. [20] M.T. Richers, D. Du Tran, J. Wachtveitl, G.C.R. Ellis-Davies, Coumarin-diene logical understanding to near-instantaneously triggering therapeutic photoswitches for rapid and efficient isomerization with visible light, Chem. release from biomaterials, possibilities for in vitro applications are end- Commun. 54 (2018) 4983–4986, https://doi.org/10.1039/c8cc01091a. less and ever expanding. With the concerted push to utilize higher [21] L. Fan, R.W. Lewis, G.P. Hess, B. Ganem, A new synthesis of caged GABA compounds – fl for studying GABAA receptors, Bioorg. Med. Chem. Lett. 19 (2009) 3932 3933, wavelengths of light, photocontrolled drug delivery will soon ourish https://doi.org/10.1016/J.BMCL.2009.03.065. in vivo, bringing materials into the clinic and broadening the horizons [22] S. Yamazoe, Q. Liu, L.E. McQuade, A. Deiters, J.K. Chen, Sequential gene silencing of scientific discovery. using wavelength-selective caged morpholino oligonucleotides, Angew. Chemie Int. Ed. 53 (2014) 10114–10118, https://doi.org/10.1002/anie.201405355. [23] M.J. Salierno, L. García-Fernandez, N. Carabelos, K. Kiefer, A.J. García, A. del Campo, Phototriggered fibril-like environments arbitrate cell escapes and migration from Acknowledgements endothelial monolayers, Biomaterials. 82 (2016) 113–123, https://doi.org/10. 1016/j.biomaterials.2015.12.001. [24] S. Yamazoe, I.A. Shestopalov, E. Provost, S.D. Leach, J.K. Chen, Cyclic caged This work was supported by a Faculty Early Career Development morpholinos: conformationally gated probes of embryonic gene function, CAREER Award (DMR 1652141, C.A.D.) and a grant (DMR 1807398, Angew. Chemie Int. Ed. 51 (2012) 6908–6911, https://doi.org/10.1002/anie. C.A.D.) from the National Science Foundation, a Maximizing Investiga- 201201690. [25] C. Zhao, S. Shuai, S. Zhou, Y. Liu, W. Huo, H. Zhu, Z. Rao, Y. Li, K. Zhou, W. Ge, J. Hao, tors’ Research Award (R35GM138036, C.A.D.) from the National Insti- Synthesis and characterization of photo-responsive flower-like copolymer micelles tutes of Health, and a Postdoctoral Fellowship from the Washington with o-nitrobenzyl as the junction point, Mater. Lett. 261 (2020), 127151. https:// Research Foundation (T.L.R.). doi.org/10.1016/j.matlet.2019.127151. [26] C.T. Huynh, M.K. Nguyen, G.Y. Tonga, L. Longé, V.M. Rotello, E. Alsberg, Photocleavable hydrogels for light-triggered siRNA release, Adv. Healthc. Mater. References 5(2016)305–310, https://doi.org/10.1002/adhm.201500778. [27] C.T. Huynh, Z. Zheng, M.K. Nguyen, A. McMillan, G. Yesilbag Tonga, V.M. Rotello, E. [1] S.H. Yun, S.J.J. Kwok, Light in diagnosis, therapy and surgery, Nat. Biomed. Eng. 1 Alsberg, Cytocompatible catalyst-free photodegradable hydrogels for light- (2017), 0008. https://doi.org/10.1038/s41551-016-0008. mediated RNA release to induce hMSC osteogenesis, ACS Biomater. Sci. Eng. 3 [2] G. Stochel, A. Wanat, E. Kuliś, Z. Stasicka, Light and metal complexes in medicine, (2017) 2011–2023, https://doi.org/10.1021/acsbiomaterials.6b00796. Coord. Chem. Rev. 171 (1998) 203–220, https://doi.org/10.1016/s0010-8545(98) [28] C.K. Arakawa, B.A. Badeau, Y. Zheng, C.A. DeForest, Multicellular vascularized 90033-9. engineered tissues through user-programmable biomaterial photodegradation, [3] C. Ash, M. Dubec, K. Donne, T. Bashford, Effect of wavelength and beam width on Adv. Mater. 29 (2017), 1703156. https://doi.org/10.1002/adma.201703156. penetration in light-tissue interaction using computational methods, Lasers Med. [29] J.A. Shadish, G.M. Benuska, C.A. DeForest, Bioactive site-specifically modified pro- Sci. 32 (2017) https://doi.org/10.1007/s10103-017-2317-4. teins for 4D patterning of gel biomaterials, Nat. Mater. 18 (2019) 1005–1014, [4] T.L. Rapp, C.A. DeForest, Visible light-responsive dynamic biomaterials: going https://doi.org/10.1038/s41563-019-0367-7. deeper and triggering more, Adv. Healthc. Mater. 9 (2020), 1901553. https://doi. [30] B.A. Badeau, M.P. Comerford, C.K. Arakawa, J.A. Shadish, C.A. DeForest, Engineered org/10.1002/adhm.201901553. modular biomaterial logic gates for environmentally triggered therapeutic deliv- [5] P. Avci, A. Gupta, M. Sadasivam, D. Vecchio, Z. Pam, N. Pam, M.R. Hamblin, Low- ery, Nat. Chem. 10 (2018) 251–258, https://doi.org/10.1038/nchem.2917. level laser (light) therapy (LLLT) in skin: stimulating, healing, restoring, Semin. [31] A. Zamora, C.A. Denning, D.K. Heidary, E. Wachter, L.A. Nease, J. Ruiz, E.C. Glazer, Cutan. Med. Surg. 32 (2013) 41–52. https://www.ncbi.nlm.nih.gov/pmc/articles/ Ruthenium-containing P450 inhibitors for dual enzyme inhibition and DNA dam- PMC4126803/ (accessed September 28, 2020). age, Dalt. Trans. 46 (2017) 2165–2173, https://doi.org/10.1039/C6DT04405K. [6] A.W. Sainter, T.A. King, M.R. Dickinson, Effect of target biological tissue and choice [32] R.N. Garner, J.C. Gallucci, K.R. Dunbar, C. Turro, [Ru(bpy) 2 (5-cyanouracil) 2 ] 2+ of light source on penetration depth and resolution in optical coherence tomogra- as a potential light-activated dual-action therapeutic agent, Inorg. Chem. 50 (2011) phy, J. Biomed. Opt. 9 (2004) 193–199, https://doi.org/10.1117/1.1628243. 9213–9215, https://doi.org/10.1021/ic201615u. [7] E.R. Ruskowitz, C.A. DeForest, Proteome-wide analysis of cellular response to ultra- [33] T.L. Rapp, C.B. Highley, B.C. Manor, J.A. Burdick, I.J. Dmochowski, Ruthenium- violet light for biomaterial synthesis and modification, ACS Biomater. Sci. Eng. 5 crosslinked hydrogels with rapid, visible-light degradation, Chem. - A Eur. J. 24 (2019) 2111–2116, https://doi.org/10.1021/acsbiomaterials.9b00177. (2018) 2328–2333, https://doi.org/10.1002/chem.201704580.

104 T.L. Rapp and C.A. DeForest Advanced Drug Delivery Reviews 171 (2021) 94–107

[34] T.L. Rapp, Y. Wang, M.A. Delessio, M.R. Gau, I.J. Dmochowski, Designing photolabile [61] L. Zayat, M. Salierno, R. Etchenique, Ruthenium(II) bipyridyl complexes as ruthenium polypyridyl crosslinkers for hydrogel formation and multiplexed, photolabile caging groups for amines, Inorg. Chem. 45 (2006) 1728–1731, visible-light degradation, RSC Adv. 9 (2019) 4942–4947, https://doi.org/10.1039/ https://doi.org/10.1021/ic0512983. C8RA09764J. [62] L. Zayat, O. Filevich, L.M. Baraldo, R. Etchenique, Ruthenium polypyridyl [35] P. Paoletti, G.C.R.R. Ellis-Davies, A. Mourot, Optical Control of Neuronal Ion Chan- phototriggers: from beginnings to perspectives, Philos. Trans. A. Math. Phys. Eng. nels and Receptors, Nature Publishing Group, 2019https://doi.org/10.1038/ Sci. 371 (2013), 201203. https://doi.org/10.1098/rsta.2012.0330. s41583-019-0197-2. [63] R.O. Filevich, Marcello Salierno, Etchenique, A caged nicotine with nanosecond [36] G.C.R. Ellis-Davies, Two-photon uncaging of glutamate, Front. Synaptic Neurosci. range kinetics and visible light sensitivity, J. Inorg. Biochem. 104 (2010) 10 (2019) 48, https://doi.org/10.3389/fnsyn.2018.00048. 1248–1251, https://doi.org/10.1016/J.JINORGBIO.2010.08.003. [37] J.K. White, R.H. Schmehl, C. Turro, An overview of photosubstitution reactions of Ru [64] R.N. Garner, L.E. Joyce, C. Turro, Effect of electronic structure on the photoinduced (II) imine complexes and their application in photobiology and photodynamic ligand exchange of Ru(II) polypyridine complexes, Inorg. Chem. 50 (2011) therapy, Inorganica Chim. Acta. 454 (2017) 7–20, https://doi.org/10.1016/j.ica. 4384–4391, https://doi.org/10.1021/ic102482c. 2016.06.007. [65] M. Huisman, J.K. White, V.G. Lewalski, I. Podgorski, C. Turro, J.J. Kodanko, Caging [38] J. Tian, B. Huang, M.H. Nawaz, W. Zhang, Recent advances of multi-dimensional the uncageable: using metal complex release for photochemical control over irre- porphyrin-based functional materials in photodynamic therapy, Coord. Chem. versible inhibition, Chem. Commun. 52 (2016) 12590–12593, https://doi.org/10. Rev. 420 (2020), 213410. https://doi.org/10.1016/j.ccr.2020.213410. 1039/C6CC07083C. [39] Y. Tao, H.F. Chan, B. Shi, M. Li, K.W. Leong, Light: a magical tool for controlled drug [66] A. Li, C. Turro, J.J. Kodanko, Ru(II) polypyridyl complexes as photocages for bioac- delivery, Adv. Funct. Mater. (2020) https://doi.org/10.1002/adfm.202005029. tive compounds containing nitriles and aromatic heterocycles, Chem. Commun. [40] H.P. Lee, A.K. Gaharwar, Light-responsive inorganic biomaterials for biomedical ap- 54 (2018) 1280–1290, https://doi.org/10.1039/C7CC09000E. plications, Adv. Sci. 7 (2020), 2000863. https://doi.org/10.1002/advs.202000863. [67] M.A. Sgambellone, A. David, R.N. Garner, K.R. Dunbar, C. Turro, Cellular toxicity in- [41] M. Scanziani, M. Häusser, Electrophysiology in the age of light, Nature. 461 (2009) duced by the photorelease of a caged bioactive molecule: design of a potential – 930 939, https://doi.org/10.1038/nature08540. dual-action Ru ( II ) complex, J. Am. Chem. Soc. 135 (2013) 11274–11282, [42] E. Bartels, N.H. Wassermann, B.F. Erlanger, Photochromic activators of the acetyl- https://doi.org/10.1021/ja4045604. – choline receptor, Proc. Natl. Acad. Sci. U. S. A. 68 (1971) 1820 1823, https://doi. [68] L. Zeng, P. Gupta, Y. Chen, E. Wang, L. Ji, H. Chao, Z.S. Chen, The development of an- org/10.1073/pnas.68.8.1820. ticancer ruthenium(II) complexes: from single molecule compounds to ń [43] W. Szyma ski, J.M. Beierle, H.A.V. Kistemaker, W.A. Velema, B.L. Feringa, Reversible nanomaterials, Chem. Soc. Rev. 46 (2017) 5771–5804, https://doi.org/10.1039/ photocontrol of biological systems by the incorporation of molecular c7cs00195a. – photoswitches, Chem. Rev. 113 (2013) 6114 6178, https://doi.org/10.1021/ [69] A.M. Palmer, B. Peña, R.B. Sears, O. Chen, M. El Ojaimi, R.P. Thummel, K.R. Dunbar, cr300179f. C. Turro, Cytotoxicity of cyclometallated ruthenium complexes: the role of ligand [44] C. Brieke, F. Rohrbach, A. Gottschalk, G. Mayer, A. Heckel, Light-controlled tools, exchange on the activity, Philos. Trans. A. Math. Phys. Eng. Sci. 371 (2013), – Angew. Chemie - Int. Ed. 51 (2012) 8446 8476, https://doi.org/10.1002/anie. 20120135. https://doi.org/10.1098/rsta.2012.0135. 201202134. [70] K. Arora, M. Herroon, M.H. Al-Afyouni, N.P. Toupin, T.N. Rohrabaugh, L.M. Loftus, I. [45] D. Bléger, S. Hecht, Visible-light-activated molecular switches, Angew. Chemie Int. Podgorski, C. Turro, J.J. Kodanko, Catch and release photosensitizers: combining – Ed. 54 (2015) 11338 11349, https://doi.org/10.1002/anie.201500628. dual-action ruthenium complexes with protease inactivation for targeting invasive [46] M. Kathan, S. Hecht, Photoswitchable molecules as key ingredients to drive sys- cancers, J. Am. Chem. Soc. 140 (2018) 14367–14380, https://doi.org/10.1021/jacs. tems away from the global thermodynamic minimum, Chem. Soc. Rev. 46 8b08853. – (2017) 5536 5550, https://doi.org/10.1039/c7cs00112f. [71] L.M. Loftus, J.K. White, B.A. Albani, L. Kohler, J.J. Kodanko, R.P. Thummel, K.R. [47] S. Passlick, E.R. Thapaliya, Z. Chen, M.T. Richers, G.C.R. Ellis-Davies, Optical probing Dunbar, C. Turro, New Ru (II) complex for dual activity: photoinduced ligand re- of acetylcholine receptors on neurons in the medial habenula with a novel caged lease and 1O2 production, Chem. - A Eur. J. 22 (2016) 3704–3708, https://doi. – nicotine drug analogue, J. Physiol. 596 (2018) 5307 5318, https://doi.org/10. org/10.1002/chem.201504800. 1113/JP276615. [72] B.A. Albani, B. Peña, K.R. Dunbar, C. Turro, New cyclometallated Ru(II) complex for [48] M.N. Stanton-Humphreys, R.D.T. Taylor, C. McDougall, M.L. Hart, C.T.A. Brown, N.J. potential application in photochemotherapy? Photochem. Photobiol. Sci. 13 Emptage, S.J. Conway, Wavelength-orthogonal photolysis of neurotransmitters (2014) 272–280, https://doi.org/10.1039/c3pp50327e. in vitro, Chem. Commun. 48 (2012) 657–659, https://doi.org/10.1039/c1cc15135e. [73] L.N. Lameijer, S.L. Hopkins, T.G. Brevé, S.H.C. Askes, S. Bonnet, D- versus L-glucose [49] K.R. Gee, R. Wieboldt, G.P. Hess, Synthesis and photochemistry of a new conjugation: mitochondrial targeting of a light-activated dual-mode-of-action photolabile derivative of GABA. Neurotransmitter release and receptor activation ruthenium-based anticancer prodrug, Chem. - A Eur. J. 22 (2016) 18484–18491, in the microsecond time region, J. Am. Chem. Soc. 116 (1994) 8336–8367, https://doi.org/10.1002/chem.201603066. https://doi.org/10.1021/ja00097a054. [74] Z. Wang, N. Wang, S.C. Cheng, K. Xu, Z. Deng, S. Chen, Z. Xu, K. Xie, M.K. Tse, P. Shi, [50] P. Molnár, J.V. Nadler, γ-Aminobutyrate, α-carboxy-2-nitrobenzyl ester selectively H. Hirao, C.C. Ko, G. Zhu, Phorbiplatin, a highly potent Pt(IV) antitumor prodrug blocks inhibitory synaptic transmission in rat dentate gyrus, Eur. J. Pharmacol. 391 that can be controllably activated by red light, Chem. 5 (2019) 3151–3165, (2000) 255–262, https://doi.org/10.1016/S0014-2999(00)00106-0. https://doi.org/10.1016/j.chempr.2019.08.021. [51] M. Canepari, L. Nelson, G. Papageorgiou, J.E.T. Corrie, D. Ogden, Photochemical and [75] H. Shi, C. Imberti, P.J. Sadler, Diazido platinum(IV) complexes for photoactivated pharmacological evaluation of 7-nitroindolinyl-and 4-methoxy-7-nitroindolinyl- anticancer chemotherapy, Inorg. Chem. Front. 6 (2019) 1623–1638, https://doi. amino acids as novel, fast caged neurotransmitters, J. Neurosci. Methods. 112 org/10.1039/c9qi00288j. (2001) 29–42, https://doi.org/10.1016/S0165-0270(01)00451-4. [76] K.L. Ciesienski, K.J. Franz, Keys for unlocking photolabile metal-containing cages, [52] M. Matsuzaki, T. Hayama, H. Kasai, G.C.R. Ellis-Davies, Two-photon uncaging of i 3- Angew. Chemie Int. Ed. 50 (2011) 814–824, https://doi.org/10.1002/anie. aminobutyric acid in intact brain tissue, Nat. Chem. Biol. 6 (2010) 255–257, https:// 201002542. doi.org/10.1038/nchembio.321. [53] L. Fan, R.W. Lewis, G.P. Hess, B. Ganem, A new synthesis of caged GABA compounds [77] H. Shi, G.J. Clarkson, P.J. Sadler, Dual action photosensitive platinum(II) anticancer for studying GABAA receptors, Bioorganic Med. Chem. Lett. 19 (2009) 3932–3933, prodrugs with photoreleasable azide ligands, Inorganica Chim. Acta. 489 (2019) – https://doi.org/10.1016/j.bmcl.2009.03.065. 230 235, https://doi.org/10.1016/j.ica.2019.02.016. [54] L. Zayat, M.G. Noval, J. Campi, C.I. Calero, D.J. Calvo, R. Etchenique, A new inorganic [78] M. Imran, W. Ayub, I.S. Butler, Zia-ur Rehman, Photoactivated platinum-based an- – photolabile protecting group for highly efficient visible light GABA uncaging, ticancer drugs, Coord. Chem. Rev. 376 (2018) 405 429, https://doi.org/10.1016/j. ChemBioChem. 8 (2007) 2035–2038, https://doi.org/10.1002/cbic.200700354. ccr.2018.08.009. [55] M. Matsuzaki, G.C.R. Ellis-Davies, T. Nemoto, Y. Miyashita, M. Iino, H. Kasai, Den- [79] A. Chaudhuri, R. Mengji, Y. Venkatesh, A. Jana, N.D. Pradeep Singh, An improved fi dritic spine geometry is critical for AMPA receptor expression in hippocampal tumor-speci c therapeutic strategy by the spatio-temporally controlled in situ for- CA1 pyramidal neurons, Nat. Neurosci. 4 (2001) 1086–1092, https://doi.org/10. mation of a Cu(II) complex, leading to prompt cell apoptosis via photoactivation of – 1038/nn736. a prodrug, Chem. Commun. 56 (2020) 4559 4562, https://doi.org/10.1039/ [56] J.P. Olson, H.B. Kwon, K.T. Takasaki, C.Q. Chiu, M.J. Higley, B.L. Sabatini, G.C.R.R. Ellis- d0cc00667j. Davies, Optically selective two-photon uncaging of glutamate at 900 nm, J. Am. [80] C. Santini, M. Pellei, V. Gandin, M. Porchia, F. Tisato, C. Marzano, Advances in copper Chem. Soc. 135 (2013) 5954–5957, https://doi.org/10.1021/ja4019379. complexes as anticancer agents, Chem. Rev. 114 (2014) 815–862, https://doi.org/ [57] E. Fino, R. Araya, D.S. Peterka, M. Salierno, R. Etchenique, R. Yuste, RuBi-glutamate: 10.1021/cr400135x. two-photon and visible-light photoactivation of neurons and dendritic spines, [81] H. Li, C. Xie, R. Lan, S. Zha, C.F. Chan, W.Y. Wong, K.L. Ho, B.D. Chan, Y. Luo, J.X. Front. Neural Circuits. 3 (2009) 2, https://doi.org/10.3389/neuro.04.002.2009. Zhang, G.L. Law, W.C.S. Tai, J.C.G. Bünzli, K.L. Wong, A smart europium- [58] M.T. Richers, J.M. Amatrudo, J.P. Olson, G.C.R. Ellis-Davies, Cloaked caged com- ruthenium complex as anticancer prodrug: controllable drug release and real- pounds: chemical probes for two-photon optoneurobiology, Angew. Chemie Int. time monitoring under different light excitations, J. Med. Chem. 60 (2017) Ed. 56 (2017) 193–197, https://doi.org/10.1002/anie.201609269. 8923–8932, https://doi.org/10.1021/acs.jmedchem.7b01162. [59] D. Lovatt, B.K. Ruble, J. Lee, H. Dueck, T.K. Kim, S. Fisher, C. Francis, J.M. Spaethling, [82] S. Biswas, Y. Rajesh, S. Barman, M. Bera, A. Paul, M. Mandal, N.D. Pradeep Singh, A J.A. Wolf, M.S. Grady, A.V. Ulyanova, S.B. Yeldell, J.C. Griepenburg, P.T. Buckley, J. dual-analyte probe: hypoxia activated nitric oxide detection with phototriggered Kim, J.-Y. Sul, I.J. Dmochowski, J. Eberwine, Transcriptome in vivo analysis (TIVA) drug release ability, Chem. Commun. 54 (2018) 7940–7943, https://doi.org/10. of spatially defined single cells in live tissue, Nat. Methods. 11 (2014) 190–196, 1039/c8cc01854e. https://doi.org/10.1038/nmeth.2804. [83] L. Kowalik, J.K. Chen, Illuminating developmental biology through photochemistry, [60] S.B. Yeldell, L. Yang, J. Lee, J.H. Eberwine, I.J. Dmochowski, Oligonucleotide probe for Nat. Chem. Biol. 13 (2017) 587–598, https://doi.org/10.1038/nchembio.2369. transcriptome in vivo analysis (TIVA) of single neurond with minimal background, [84] C.F. Bennett, Therapeutic antisense oligonucleotides are coming of age, Annu. Rev. ACS Chem. Biol. (2020) https://doi.org/10.1021/acschembio.0c00499. Med. 70 (2019) 307–321, https://doi.org/10.1146/annurev-med-041217-010829.

105 T.L. Rapp and C.A. DeForest Advanced Drug Delivery Reviews 171 (2021) 94–107

[85] L. Kröck, A. Heckel, Photoinduced transcription by using temporarily mismatched [110] C. Zhan, W. Wang, J.B. McAlvin, S. Guo, B.P. Timko, C. Santamaria, D.S. Kohane, caged oligonucleotides, Angew. Chemie Int. Ed. 44 (2005) 471–473, https://doi. Phototriggered local anesthesia, Nano Lett. 16 (2016) 177–181, https://doi.org/ org/10.1002/anie.200461779. 10.1021/acs.nanolett.5b03440. [86] D.D. Young, H. Lusic, M.O. Lively, J.A. Yoder, A. Deiters, Gene silencing in mamma- [111] C. Zhan, W. Wang, C. Santamaria, B. Wang, A. Rwei, B.P. Timko, D.S. Kohane, Ultra- lian cells with light-activated antisense agents, ChemBioChem. 9 (2008) sensitive phototriggered local anesthesia, Nano Lett. 17 (2017) 660–665, https:// 2937–2940, https://doi.org/10.1002/cbic.200800627. doi.org/10.1021/acs.nanolett.6b03588. [87] J.C. Griepenburg, T.L. Rapp, P.J. Carroll, J. Eberwine, I.J. Dmochowski, Ruthenium- [112] Y. Liu, P. Bhattarai, Z. Dai, X. Chen, Photothermal therapy and photoacoustic imag- caged antisense morpholinos for regulating gene expression in zebrafish embryos, ing: via nanotheranostics in fighting cancer, Chem. Soc. Rev. 48 (2019) 2053–2108, Chem. Sci. 6 (2015) 2342–2346, https://doi.org/10.1039/C4SC03990D. https://doi.org/10.1039/c8cs00618k. [88] L. Yang, H.-B. Kim, J.-Y. Sul, S.B. Yeldell, J.H. Eberwine, I.J. Dmochowski, Efficient [113] R.S. Riley, E.S. Day, Gold nanoparticle-mediated photothermal therapy: applica- synthesis of light-triggered circular antisense oligonucleotides targeting cellular tions and opportunities for multimodal cancer treatment, Wiley Interdiscip. Rev. protein expression, ChemBioChem. 19 (2018) 1250–1254, https://doi.org/10. Nanomed. Nanobiotechnol. 9 (2017) https://doi.org/10.1002/wnan.1449. 1002/cbic.201800012. [114] L. Zou, H. Wang, B. He, L. Zeng, T. Tan, H. Cao, X. He, Z. Zhang, S. Guo, Y. Li, Current [89] S.M. Elbashir, J. Harborth, W. Lendeckel, A. Yalcin, K. Weber, T. Tuschl, Duplexes of approaches of photothermal therapy in treating cancer metastasis with 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells, Na- nanotherapeutics, Theranostics. 6 (2016) 762–772, https://doi.org/10.7150/thno. ture. 411 (2001) 494–498, https://doi.org/10.1038/35078107. 14988. [90] D.A. Braasch, S. Jensen, Y. Liu, K. Kaur, K. Arar, M.A. White, D.R. Corey, RNA interfer- [115] L. Fu, R. Chung, B. Shi, Upconversion nanoparticle-based strategy for crossing the ence in mammalian cells by chemically-modified RNA, Biochemistry. 42 (2003) blood-brain barrier to treat the central nervous system disease, Methods Mol 7967–7975, https://doi.org/10.1021/bi0343774. Biol. (2019) 263–282, https://doi.org/10.1007/978-1-4939-9769-5_17Humana [91] J.P. Casey, R.A. Blidner, W.T. Monroe, Caged siRNAs for spatiotemporal control of Press Inc. gene silencing, Mol. Pharm. 6 (2009) 669–685, https://doi.org/10.1021/ [116] C. Yao, P. Wang, X. Li, X. Hu, J. Hou, L. Wang, F. Zhang, Near-infrared-triggered mp900082q. azobenzene-liposome/upconversion nanoparticle hybrid vesicles for remotely [92] M.L. Hammill, C. Isaacs-Trépanier, J.-P. Desaulniers, siRNAzos: a new class of controlled drug delivery to overcome cancer multidrug resistance, Adv. Mater. – azobenzene-containing siRNAs that can photochemically regulate gene expres- 28 (2016) 9341 9348, https://doi.org/10.1002/adma.201503799. sion, ChemistrySelect 2 (2017) 9810–9814, https://doi.org/10.1002/slct. [117] G. Jalani, V. Tam, F. Vetrone, M. Cerruti, Seeing, targeting and delivering with – 201702322. upconverting nanoparticles, J. Am. Chem. Soc. 140 (2018) 10923 10931, https:// [93] W.T. Monroe, M.M. McQuain, M.S. Chang, J.S. Alexander, F.R. Haselton, Targeting doi.org/10.1021/jacs.8b03977. expression with light using caged DNA, J. Biol. Chem. 274 (1999) 20895–20900, [118] Z. Zhang, M.K.G. Jayakumar, S. Shikha, Y. Zhang, X. Zheng, Y. Zhang, Modularly as- https://doi.org/10.1074/jbc.274.30.20895. sembled upconversion nanoparticles for orthogonally controlled cell imaging and drug delivery, ACS Appl. Mater. Interfaces. 12 (2020) 12549–12556, https://doi. [94] H. Ando, T. Furuta, R.Y. Tsien, H. Okamoto, Photo-mediated gene activation using caged RNA/DNA in zebrafish embryos, Nat. Genet. 28 (2001) 317–325, https:// org/10.1021/acsami.0c00672. doi.org/10.1038/ng583. [119] G. Jalani, R. Naccache, D.H. Rosenzweig, L. Haglund, F. Vetrone, M. Cerruti, Photocleavable hydrogel-coated upconverting nanoparticles: a multifunctional [95] Q.N. Nguyen, R.V. Chavli, J.T. Marques, P.G. Conrad, D. Wang, W. He, B.E. Belisle, A. theranostic platform for NIR imaging and on-demand macromolecular delivery, Zhang, L.M. Pastor, F.R. Witney, M. Morris, F. Heitz, G. Divita, B.R.G. Williams, G.K. J. Am. Chem. Soc. 138 (2016) 1078–1083, https://doi.org/10.1021/jacs.5b12357. McMaster, Light controllable siRNAs regulate gene suppression and phenotypes [120] Q. Liu, C. Zhan, D.S. Kohane, Phototriggered drug delivery using inorganic in cells, Biochim. Biophys. Acta - Biomembr. 1758 (2006) 394–403, https://doi. nanomaterials, Bioconjug. Chem. 28 (2017) 98–104, https://doi.org/10.1021/acs. org/10.1016/j.bbamem.2006.01.003. bioconjchem.6b00448. [96] Y. Ji, J. Yang, L. Wu, L. Yu, X. Tang, Photochemical regulation of gene expression [121] A. Bagheri, H. Arandiyan, C. Boyer, M. Lim, Lanthanide-doped upconversion nano- using caged siRNAs with single terminal Vitamin E modification, Angew. Chemie particles: emerging intelligent light-activated drug delivery systems, Adv. Sci. 3 Int. Ed. 55 (2016) 2152–2156, https://doi.org/10.1002/anie.201510921. (2016), 1500437. https://doi.org/10.1002/advs.201500437. [97] O.C. Farokhzad, R. Langer, Impact of nanotechnology on drug delivery, ACS Nano. 3 [122] D. Zhou, J. Guo, G.B. Kim, J. Li, X. Chen, J. Yang, Y. Huang, Simultaneously photo- (2009) 16–20, https://doi.org/10.1021/nn900002m. cleavable and activatable prodrug-backboned block copolymer micelles for precise [98] D. Mishra, J.R. Hubenak, A.B. Mathur, Nanoparticle systems as tools to improve anticancer drug delivery, Adv. Healthc. Mater. 5 (2016) 2493–2499, https://doi. drug delivery and therapeutic efficacy, J. Biomed. Mater. Res. Part A. 101 (2013) org/10.1002/adhm.201600470. 3646–3660, https://doi.org/10.1002/jbm.a.34642. [123] W. Sun, R. Thiramanas, L.D. Slep, X. Zeng, V. Mailänder, S. Wu, Photoactivation of [99] Q. Zhou, L. Zhang, T.H. Yang, H. Wu, Stimuli-responsive polymeric micelles for drug anticancer Ru complexes in deep tissue: how deep can we go? Chem. - A Eur. J. – delivery and cancer therapy, Int. J. Nanomedicine. 13 (2018) 2921 2942, https:// 23 (2017) 10832–10837, https://doi.org/10.1002/chem.201701224. doi.org/10.2147/IJN.S158696. [124] W. Sun, Y. Wen, R. Thiramanas, M. Chen, J. Han, N. Gong, M. Wagner, S. Jiang, M.S. [100] G. Shim, S. Ko, D. Kim, Q.V. Le, G.T. Park, J. Lee, T. Kwon, H.G. Choi, Y.B. Kim, Y.K. Oh, Meijer, S. Bonnet, H.-J. Butt, V. Mailänder, X.-J. Liang, S. Wu, Red-light-controlled Light-switchable systems for remotely controlled drug delivery, J. Control. Release. release of drug-ru complex conjugates from metallopolymer micelles for photo- – 267 (2017) 67 79, https://doi.org/10.1016/j.jconrel.2017.09.009. therapy in hypoxic tumor environments, Adv. Funct. Mater. 28 (2018), 1804227. [101] Y. Li, Y. Zhang, W. Wang, Phototriggered targeting of nanocarriers for drug deliv- https://doi.org/10.1002/adfm.201804227. – ery, Nano Res. 11 (2018) 5424 5438, https://doi.org/10.1007/s12274-018-2132-7. [125] T.R. Hoare, D.S. Kohane, Hydrogels in drug delivery: progress and challenges, Poly- [102] S.O. Poelma, S.S. Oh, S. Helmy, A.S. Knight, G.L. Burnett, H.T. Soh, C.J. Hawker, J. Read mer (Guildf). 49 (2008) 1993–2007, https://doi.org/10.1016/j.polymer.2008.01. De Alaniz, Controlled drug release to cancer cells from modular one-photon visible 027. – light-responsive micellar system, Chem. Commun. 52 (2016) 10525 10528, [126] R. Dimatteo, N.J. Darling, T. Segura, In situ forming injectable hydrogels for drug de- https://doi.org/10.1039/c6cc04127b. livery and wound repair, Adv. Drug Deliv. Rev. 127 (2018) 167–184, https://doi. [103] P. Pei, C. Sun, W. Tao, J. Li, X. Yang, J. Wang, ROS-sensitive thioketal-linked org/10.1016/j.addr.2018.03.007. polyphosphoester-doxorubicin conjugate for precise phototriggered locoregional [127] D. Wang, M. Wagner, H.-J. Butt, S. Wu, Supramolecular hydrogels constructed by chemotherapy, Biomaterials. 188 (2019) 74–82, https://doi.org/10.1016/j. red-light-responsive host-guest interactions for photo-controlled protein release biomaterials.2018.10.010. in deep tissue, Soft Matter. 11 (2015) 7656–7662, https://doi.org/10.1039/ [104] S. Uthaman, S. Pillarisetti, A.P. Mathew, Y. Kim, W.K. Bae, K.M. Huh, I.K. Park, Long c5sm01888a. circulating photoactivable nanomicelles with tumor localized activation and ROS [128] X. Wang, C. Wang, Q. Zhang, Y. Cheng, Near infrared light-responsive and inject- triggered self-accelerating drug release for enhanced locoregional chemo- able supramolecular hydrogels for on-demand drug delivery, Chem. Commun. 52 photodynamic therapy, Biomaterials. 232 (2020), 119702. https://doi.org/10. (2016) 978–981, https://doi.org/10.1039/C5CC08391E. 1016/j.biomaterials.2019.119702. [129] J.A. Shadish, C.A. DeForest, Site-selective protein modification: from functionalized [105] K. Hayashi, M. Watanabe, T. Iwasaki, M. Shudou, R.M. Uda, Endosomal escape by proteins to functional biomaterials, Matter. 2 (2020) 50–77, https://doi.org/10. photo-activated fusion of liposomes containing a malachite green derivative: a 1016/j.matt.2019.11.011. novel class of photoresponsive liposomes for drug delivery vehicles, Photochem. [130] J.C. Grim, I.A. Marozas, K.S. Anseth, Thiol-ene and photo-cleavage chemistry for Photobiol. Sci. 18 (2019) 1471–1478, https://doi.org/10.1039/c8pp00495a. controlled presentation of biomolecules in hydrogels, J. Control. Release. 219 [106] N.P. Kamat, G.P. Robbins, J. Rawson, M.J. Therien, I.J. Dmochowski, D.A. Hammer, A (2015) 95–106, https://doi.org/10.1016/j.jconrel.2015.08.040. generalized system for photoresponsive membrane rupture in polymersomes, [131] J.C. Grim, T.E. Brown, B.A. Aguado, D.A. Chapnick, A.L. Viert, X. Liu, K.S. Anseth, A re- Adv. Funct. Mater. 20 (2010) 2588–2596, https://doi.org/10.1002/adfm. versible and repeatable thiol-ene bioconjugation for dynamic patterning of signal- 201000659. ing proteins in hydrogels, ACS Cent. Sci. 4 (2018) 909–916, https://doi.org/10. [107] J.C. Griepenburg, N. Sood, K.B. Vargo, D. Williams, J. Rawson, M.J. Therien, D.A. 1021/acscentsci.8b00325. Hammer, I.J. Dmochowski, Caging metal ions with visible light-responsive [132] B.V. Sridhar, J.R. Janczy, Ø. Hatlevik, G. Wolfson, K.S. Anseth, M.W. Tibbitt, Thermal nanopolymersomes, Langmuir. 31 (2015) 799–807, https://doi.org/10.1021/ stabilization of biologics with photoresponsive hydrogels, Biomacromolecules. 19 la5036689. (2018) 740–747, https://doi.org/10.1021/acs.biomac.7b01507. [108] C. Qian, J. Yu, Y. Chen, Q. Hu, X. Xiao, W. Sun, C. Wang, P. Feng, Q.-D. Shen, Z. Gu, [133] J.A. Shadish, A.C. Strange, C.A. DeForest, Genetically encoded photocleavable linkers Light-activated hypoxia-responsive nanocarriers for enhanced anticancer therapy, for patterned protein release from biomaterials, J. Am. Chem. Soc. 141 (2019) Adv. Mater. 28 (2016) 3313–3320, https://doi.org/10.1002/adma.201505869. 15619–15625, https://doi.org/10.1021/jacs.9b07239. [109] C. Liu, K.K. Ewert, N. Wang, Y. Li, C.R. Safinya, W. Qiao, A multifunctional lipid that [134] J.A. Hammer, A. Ruta, J.L. West, Using tools from optogenetics to create forms contrast-agent liposomes with dual-control release capabilities for precise light-responsive biomaterials: LOVTRAP-PEG Hydrogels for dynamic peptide MRI-guided drug delivery, Biomaterials. 221 (2019), 119412. https://doi.org/10. immobilization, Ann. Biomed. Eng. (2019) 1–10, https://doi.org/10.1007/s10439- 1016/j.biomaterials.2019.119412. 019-02407-w.

106 T.L. Rapp and C.A. DeForest Advanced Drug Delivery Reviews 171 (2021) 94–107

[135] E.R. Ruskowitz, M.P. Comerford, B.A. Badeau, C.A. DeForest, Logical stimuli- [142] R.V. Nair, A. Farrukh, A. del Campo, A photoactivatable α 5 β 1 -specificintegrinli- triggered delivery of small molecules from hydrogel biomaterials, Biomater. Sci. gand, ChemBioChem. 19 (2018) 1280–1287, https://doi.org/10.1002/cbic. 7(2019)542–546, https://doi.org/10.1039/c8bm01304g. 201800180. [136] P.M. Gawade, J.A. Shadish, B.A. Badeau, C.A. DeForest, Logic-based delivery of site- [143] T.T. Lee, J.R. García, J.I. Paez, A. Singh, E.A. Phelps, S. Weis, Z. Shafiq, A. Shekaran, A. specifically modified proteins from environmentally responsive hydrogel biomate- Del Campo, A.J. García, Light-triggered in vivo activation of adhesive peptides reg- rials, Adv. Mater. 31 (2019), 1902462. https://doi.org/10.1002/adma.201902462. ulates cell adhesion, inflammation and vascularization of biomaterials, Nat. Mater. [137] E.A. Hushka, F.M. Yavitt, T.E. Brown, P.J. Dempsey, K.S. Anseth, Relaxation of extra- 14 (2015) 352–360, https://doi.org/10.1038/nmat4157. cellular matrix forces directs crypt formation and architecture in intestinal [144] S. Sankaran, S. Zhao, C. Muth, J. Paez, A. del Campo, Toward light-regulated living organoids, Adv. Healthc. Mater. 9 (2020), 1901214. https://doi.org/10.1002/ biomaterials, Adv. Sci. 5 (2018) 1800383, https://doi.org/10.1002/advs.201800383. adhm.201901214. [145] S. Sankaran, J. Becker, C. Wittmann, A. del Campo, Optoregulated drug release from [138] L.C. Bahlmann, A. Fokina, M.S. Shoichet, Dynamic bioengineered hydrogels as scaf- an engineered living material: self-replenishing drug depots for long-term, light- folds for advanced stem cell and organoid culture, MRS Commun. 7 (2017) regulated delivery, Small. 15 (2019), 1804717. https://doi.org/10.1002/smll. 472–486, https://doi.org/10.1557/mrc.2017.72. 201804717. [139] C.A. DeForest, K.S. Anseth, Cytocompatible click-based hydrogels with dynamically [146] K.L. Spiller, S. Nassiri, C.E. Witherel, R.R. Anfang, J. Ng, K.R. Nakazawa, T. Yu, G. tunable properties through orthogonal photoconjugation and photocleavage reac- Vunjak-Novakovic, Sequential delivery of immunomodulatory cytokines to facili- tions, Nat. Chem. 3 (2011) 925–931, https://doi.org/10.1038/nchem.1174. tate the M1-to-M2 transition of macrophages and enhance vascularization of [140] A. Farrukh, J.I. Paez, A. del Campo, 4D biomaterials for light-guided angiogenesis, bone scaffolds, Biomaterials. 37 (2015) 194–207, https://doi.org/10.1016/j. Adv. Funct. Mater. 29 (2019), 1807734. https://doi.org/10.1002/adfm.201807734. biomaterials.2014.10.017. [141] A. Farrukh, W. Fan, S. Zhao, M. Salierno, J.I. Paez, A. del Campo, Photoactivatable ad- [147] K.J. Rambhia, P.X. Ma, Controlled drug release for tissue engineering, J. Control. Re- hesive ligands for light-guided neuronal growth, ChemBioChem. 19 (2018) lease. 219 (2015) 119–128, https://doi.org/10.1016/j.jconrel.2015.08.049. 1271–1279, https://doi.org/10.1002/cbic.201800118.

107