Abstract
Dysregulated cathepsin activity is linked to various human diseases including metabolic disorders, autoimmune conditions, and cancer. Given the overexpression of cathepsin in the tumor microenvironment, cathepsin inhibitors are promising pharmacological agents and drug delivery vehicles for cancer treatment. In this study, we describe the synthesis and photochemical and biological assessment of a dual-action agent based on ruthenium that is conjugated with a cathepsin inhibitor, designed for both photodynamic therapy (PDT) and photochemotherapy (PCT). The ruthenium-cathepsin inhibitor conjugate was synthesized through an oxime click reaction, combining a pan-cathepsin inhibitor based on E64d with the Ru(II) PCT/PDT fragment [Ru(dqpy)(dppn)], where dqpy = 2,6-di(quinoline-2-yl)pyridine and dppn = benzo[i]dipyrido[3,2-a:2′,3′-c]phenazine. Photochemical investigations validated the conjugate’s ability to release a triazole-containing cathepsin inhibitor for PCT and to generate singlet oxygen for PDT upon exposure to green light. Inhibition studies demonstrated the conjugate’s potent and irreversible inactivation of purified and intracellular cysteine cathepsins. Two Ru(II) PCT/PDT agents based on the [Ru(dqpy)(dppn)] moiety were evaluated for photoinduced cytotoxicity in 4T1 murine triple-negative breast cancer cells, L929 fibroblasts, and M0, M1, and M2 macrophages. The cathepsin inhibitor conjugate displayed notable selectivity for inducing cell death under irradiation compared to dark conditions, mitigating toxicity in the dark observed with the triazole control complex [Ru(dqpy)(dppn)(MeTz)]2+ (MeTz = 1-methyl-1H-1,2,4-triazole). Notably, our lead complex is among a limited number of dual PCT/PDT agents activated with green light.
Graphical Abstract

INTRODUCTION
Cancer is the second leading cause of death worldwide, attributed to nearly 10 million global deaths in 2020.1 While recent advancements in screening technology and treatment have resulted in a steady decline in the cancer death rate by 1.5% from 2019 to 2020,2 it is evident that preventative measures and new treatments must continue to progress. While traditional chemotherapy is effective in killing rapidly differentiating cells, it generally cannot discern between cancerous and normal cells, resulting in unwanted side effects. To combat this problem, light activated therapy, such as photodynamic therapy (PDT), has evolved to trigger medicinal effects solely in irradiated areas. In traditional PDT agents, excitation of the photosensitizer with visible light results in the formation of the long-lived 3ππ* state, where upon relaxation back to the singlet ground state (1GS), energy is transferred to ground-state triplet oxygen (3O2) to form singlet oxygen (1O2), a type of reactive oxygen species (ROS). Light activation enables spatiotemporal control of ROS generation to induce oxidative damage in an area of interest, such as tumors, while avoiding toxicity in normal tissue.
PDT agents have evolved since Photofrin was first clinically approved in 1993, and Ru(II)-based complexes have been explored as potential photoactivated therapeutics. The rich photophysical properties,3 ability to intercalate between DNA bases through π-extended ligands,4–6 and improved water solubility and resistance to photobleaching over porphyrins make Ru(II) complexes attractive PDT candidates.7–13 Unfortunately, a large disadvantage of potential Ru(II) PDT agents is that their photochemistry is generally restricted to blue light activation, limiting the treatment to superficial tissues. Thus, ligands with larger π-systems have been investigated coordinated to Ru(II) centers with the goals of increasing absorptivity in the visible range, lower the energy of 3ππ* state to increase the excited state lifetimes, and to red-shift the absorption toward the “PDT window” (from 600–1100 nm). Specifically, Ru(II) photosensitizer TLD-1433 contains an imidazo[4,5-f][1,10]phenanthroline (IP) ligand conjugated to an α-terthienyl group for improved electron delocalization and absorptivity, allowing for green-light activation.14 As a consequence, TLD-1433 has advanced to Phase II clinical trials to treat nonmuscle-invasive bladder cancer, demonstrating the clinical relevance of Ru(II)-based PDT.
An additional light activated approach for cancer treatment is photochemotherapy (PCT). PCT utilizes light activation of a nontoxic, biologically inert precursor, or photocage, to release a biologically active molecule. While there are no examples of clinically approved photocages, spatiotemporal control of therapeutic delivery has great potential to circumvent off-target toxicity of the photolabile molecule.15–20 Both PDT and PCT involve excitation of the Ru(II) complex from the 1GS to the singlet metal-to-ligand charge transfer (1MLCT) state, which undergoes an efficient ultrafast intersystem crossing to the corresponding triplet 3MLCT state. Whereas PDT relies on the presence of a long-lived triplet excited state, 3ππ* or 3MLCT, to produce reactive 1O2, PCT requires access to the dissociative triplet metal-centered (3MC) excited state, which allows for the release of bioactive ligands upon excitation. Ru(II)-based PDT and PCT were thought to be mutually exclusive until the first dual PCT/PDT agent, [Ru(tpy)(Me2dppn)(py)]2+, was reported.21,22 Incorporation of the π-extended ligand 3,6-dimethylbenzo[i]-dipyrido[3,2-a:20,30-c]phenazine (Me2dppn) results in a lowest energy, long-lived dppn-centered 3ππ* state, leading to efficient 1O2 production for PDT, while the introduction of methyl groups generates steric strain and distortion of the octahedral geometry, resulting in a low-lying 3MC state that allows pyridine ligand photodissociation.
We previously reported a Ru(II)-based complex 1 shown in Figure 1, which contains an irreversible cathepsin inhibitor and Medppn ligand for the 1O2 generation and inhibitor delivery, resulting in the important synergism of PCT/PDT dual-action agents.23 Cathepsins are essential proteases responsible for protein catabolism and autophagy. In the acidic tumor microenvironment, cathepsins translocate from lysosomes to the cell surface and extracellular milieu and contribute to degradation of the extracellular matrix (ECM), either directly or indirectly through activation of the proteolytic cascade.24–27 In fact, overexpression of cathepsins has been associated with poor prognosis of various cancers.28,29 Importantly, in vitro studies using cathepsin inhibitors30–37 and in vivo studies involving cathepsin gene knockout mice38–41 have resulted in decreased tumor burden, invasion, and proliferation. Because cathepsins are well-known to reside at the invasive edge of tumors,42 irreversible inhibition of the cathepsin enzyme provides an anticancer effect that can also serve as a delivery method selective to tumor tissue. Following irreversible, covalent inhibition of cathepsins, light activation allows the photolabile inhibitor to be released from the Ru(II) center, where the solvated Ru(II) complex produces 1O2 and induces oxidative damage at the tumor site.23 This study demonstrated the synergism of simultaneous cathepsin inhibition and ROS generation in a 3D pathomimetic model of triple negative human breast cancer, which offered promise for a drug delivery method, albeit limited by blue-light activation.
Figure 1.

Previously reported dual action PCT/PDT agents, cathepsin-based inhibitor 1 (right) reported to show synergistic cell death against breast cancer spheroids and pyridyl photocage 2 (left) reported to show immunogenic cell death in 3D coculture containing macrophages and triple negative breast cancer cells.
Our groups have also reported that dual action PCT/PDT metal complexes such as 2 (Figure 1) show the ability to target MDA-MB-231 triple negative breast cancer cells and tumor-associated macrophages (TAMs).43 Macrophages are myeloid cells responsible for modulating the immune system by phagocytosis, antigen presentation, and secretion of cytokines, chemokines, and growth factors. To mediate the immune system, these cells must be adaptable to the tissue they reside in and to the environmental cues; therefore, macrophages differentiate across a spectrum between pro-inflammatory, immune activating M1 macrophages or anti-inflammatory, immune suppressing M2 macrophages.44–47 Studies have indicated that tumor regression caused by cathepsin inhibitors may be due to depletion of TAMs or repolarization toward M1-like macrophages.30,48–51 While our lead compound 2 showed efficacy against macrophages and promise for metal-based immunotherapy, we predicted it would lack specificity if applied in vivo. Thus, we envisioned implementing a metal-based cathepsin inhibitor to direct the compound not only to tumor cells but also TAMs. Together, we hypothesized that appending a cathepsin inhibitor to a green light activated PCT/PDT agent would aid delivery to the tumor microenvironment and immunogenic response with improved tissue penetration.
Herein, we report the design, synthesis, and biological evaluation of a Ru(II)-containing photolabile cathepsin inhibitor. While many photoactivated Ru(II) complexes have been designed to display red-shifted absorption windows with high extinction coefficients, they generally feature π-extended ligands that reduce their solubility in aqueous media, organic chromophores that are responsible for its photosensitization properties, or nanoparticles that incorporate many Ru(II) molecules. Moreover, in attempts to shift Ru(II) complexes to the therapeutic window, the PCT mode is sacrificed for PDT capabilities; therefore, only a handful of dual PCT/PDT agents can be activated with green or red light.52–57 In our synthetic design, we utilized an intermediate aldehyde that can be subjected to facile oxime formation via a “click” reaction to efficiently produce conjugates with green light activated dual action PCT/PDT analogs.58 The Ru PCT/PDT-cathepsin inhibitor conjugate prepared by this method efficiently inactivates purified cathepsins B and L both upon irradiation and when kept in the dark. Furthermore, cathepsin inactivation was evident in cells treated with our compound, thereby confirming cellular uptake and the inactivation of intracellular cysteine cathepsins. Photoactivation with green light results in photorelease of the inhibitor conjugate and phototoxicity in breast cancer cells and macrophages by using short intervals of irradiation, which shows promise for further biological applications. Importantly, we demonstrate that conjugation to a cathepsin inhibitor can be used to mitigate the toxicity of Ru PCT/PDT agents in the dark.
RESULTS AND DISCUSSION
Compound Design.
We hypothesized that designing a Ru(II) complex that contains both PDT and PCT modalities in addition to a cathepsin inhibitor would enable specific targeting of the tumor region where these enzymes are overexpressed. In this scenario, the covalent inhibition of cathepsins would allow for localization of the Ru(II) complex at the invasive tumor edge, where cathepsins are known to be found.42 Following localization, the release of the covalently bound inhibitor from the Ru(II) center and ROS production can then be achieved through the irradiation of the complex with visible light, resulting in oxidative damage to the TAMs and tumor cells. We previously reported a Ru PCT/PDT agent conjugated to a cathepsin inhibitor, where the dual action PCT/PDT behavior was crucial for achieving photoactivated cell death. However, our previously reported complexes were photoactivatable only upon blue light irradiation with low tissue penetration depth, thereby limiting their potential biological applications. Here, we have designed a dual PCT/PDT Ru(II)-cathepsin inhibitor that employs green light activation, which penetrates tissue deeper than blue light and has clinically validated applications in PDT.59,60
To ensure our Ru(II) complex could be activated by green light and serve as a PDT agent, ancillary ligands were chosen to move absorption to longer wavelengths, as well as to optimize cellular penetration and 1O2 production. Ru(II) complexes containing benzo[i]dipyrido[3,2-a:2′,3′-c]phenazine (dppn) ligands have been proven to elicit efficient PDT achieved by its lowest-energy, long-lived dppn-centered 3ππ* excited state.61,62 Additionally, inclusion of the more extended π-system in 2,6-di(quinoline-2-yl)pyridine (dqpy) has been shown to result in a red shift of the 1MLCT absorption as compared to the traditional tridentate terpyridine (tpy) ligand.63 Incorporation of these ligands in [Ru(dppn)(dqpy)-(py)](PF6)2 indeed showed phototoxicity using green light (520–525 nm) in a preliminary screen (data not shown), indicating the parent complex serves as a promising PDT agent.
Because in dppn-containing complexes PDT arises from the thermal population of the lowest-lying 3ππ* state from the 3MLCT, this process is generally more favorable than population of the higher-energy dissociative 3MC state required for PCT. To make PCT a feasible process, we appended a less basic and weaker monodentate ligand, 1H-1,2,4-triazole, to serve as the photolabile ligand. To allow for cathepsin inhibition in both the dark and light, the photolabile ligand was designed to contain an epoxysuccinyl group found in commonly known cathepsin inhibitors CA-074, E64d, and NS-134. The epoxysuccinyl warhead enables irreversible inhibition via epoxide opening by the cysteine of the cathepsin. Additionally, while it is known that CA-074 is cell impermeable, its methyl ester analog CA-074Me (Figure 2) behaves as a cell permeable prodrug that it is hydrolyzed to CA-074 by intracellular esterases.64–66 Thus, we sought to conserve the terminal ester of the inhibitor conjugate to allow for cell permeability and the inhibition of intracellular cathepsins.
Figure 2.

Cell permeable cathepsin inhibitors CA-074Me and E64d.
Due to forcing conditions required to complex the weak triazole ligand to the Ru(II) center and opposing lability of the epoxide group, we employed a “click” reaction, where we could separately complex triazole and subsequently attach the epoxysuccinyl warhead. The synthesis of the terminal benzaldehyde intermediate (5) allowed for a facile oxime reaction commonly found in bioconjugation reactions where hydroxylamines can “click” with aldehydes or ketones to generate a stable oxime.67–72 Additionally, the hydroxylamine precursor (8) contained a diethylene chain to aid in water solubility and lengthen the linker between the Ru(II) center and the epoxysuccinyl warhead to allow for covalent binding to the active site under dark conditions. In addition to our Ru(II)-based cathepsin inhibitor, we also synthesized a Ru(II) complex containing an identical core devoid of the inhibitor conjugate [Ru(dqpy)(dppn)(MeTz)]2+ where MeTz = 1-methyl-1H-1,2,4-triazole (6). We hypothesized that 6 would also be phototoxic due to its PDT properties; however, due to the absence of the cathepsin inhibitor, it lacks the drug delivery vector.
Compound Synthesis and Structural Characterization Data.
Because triazole is a weak ligand, forcing conditions such as high temperatures and excess ligand were necessary to complex the triazole ligand to ruthenium. To prevent decomposition of our labile epoxysuccinyl containing ligand, we synthesized our target Ru(II)-based inhibitor in several steps, as depicted in Scheme 1. A terminal benzaldehyde motif was incorporated to perform the oxime reaction. We first attempted to synthesize 4-((1H-1,2,4-triazol-1-yl)methyl)benzaldehyde; however, unwanted side reactions occurred upon complexation to ruthenium, presumably due to activation of the aldehyde carbonyl group. Instead, we used acetal protected 1-(chloromethyl)-4-(diethoxymethyl)benzene and substituted the chloride for 1H-1,2,4-triazole in the presence of catalytic NaI and K2CO3 to give 3.
Scheme 1.

(A) Synthesis of Triazole-Containing Acetal, 3; (B) Synthesis of Ru(II)-Containing Aldehyde, 5, and Control Compound, 6
Before complexing triazole with ruthenium, we first constructed our precursor ruthenium complex. To make [Ru(dppn)(dqpy)(Cl)]PF6 (4), we synthesized [Ru(dppn)-(p-cymene)Cl]Cl according to the literature,73 and subjected it to 2 equiv of dqpy ligand and excess LiCl in DMF at reflux, during which the color changed from orange to purple. Electronic absorption spectroscopy revealed a red-shift of the 1MLCT band from the starting material at 419 to 535 nm for the product,73 indicating successful complexation. The purple solid was isolated via NH4PF6 precipitation and alumina column chromatography. The complex was then subjected to AgBF4 in 1:1 DCM:MeOH to exchange the chloride for a more labile leaving group. The reaction mixture was filtered to remove AgCl, concentrated, and refluxed in 2:1 ethylene glycol:EtOH with excess triazole-containing ligand (3). Isolation by NH4PF6 precipitation and subsequent vapor diffusion using acetonitrile and Et2O serendipitously afforded deprotected aldehyde 5. Hydrolysis of the acetal presumably occurred due to adventitious water in the medium. The control compound 6 was synthesized in a similar fashion containing a methyl group rather than a benzaldehyde motif for conjugation using 1-methyl-1H-1,2,4-triazole as the ligand. Analysis by electronic absorption spectroscopy of both complexation reactions displayed an MLCT shift to 470 nm, demonstrating that the reaction was successful.
In order to generate the hydroxylamine portion for the desired oxime reaction, we first attached the hydroxylamine to an ethylene glycol linker. Monocoupling of 2-(((tert-butoxycarbonyl)amino)oxy)acetic acid with 2,2′-(ethylenedioxy)bis(ethylamine) with DCC as a coupling agent afforded 7 (Scheme 2). Subsequent coupling of the terminal amine with 4-nitrophenyl (tert-butoxycarbonyl)-l-leucinate gave the leucine containing Boc-protected hydroxylamine, 8, as shown in Scheme 2. To make the hydroxylamine accessible, the ligand was treated with 1 M HCl to deprotect both Boc groups. The HCl salt intermediate was added in excess to ruthenium-containing aldehyde 5 and was isolated via NH4PF6 precipitation and EtOAc trituration to remove the unreacted ligand (Scheme 2). The successful oxime reaction was validated by ESI/MS analysis displaying the m/z value of 636, and 1H NMR revealed a new proton signal belonging to the oxime proton at 8.23 ppm and the simultaneous disappearance of the aldehyde proton. Additionally, no evidence of imine formation from the terminal amine and aldehyde was found, where the chemical shift of the imine hydrogen would be expected at ~8.1 ppm. This observation is consistent with prior reports indicating that imine formation is fast and reversible, whereas oximes are more thermodynamically stable.70–72
Scheme 2.

Synthesis of Ru(II) PCT/PDT-Cathepsin Inhibitor Conjugate 9
As shown in Scheme 2, the oxime was treated with excess 2-ethyl 3-(4-nitrophenyl) (2S,3S)-oxirane-2,3-dicarboxylate and DIPEA to attach the epoxide-containing activated ester to the leucyl-amine to give final compound 9. This specific epoxide group was chosen in 9 to generate an analog of E64d, a pancathepsin inhibitor with affinity for cathepsins B and L, two prominent cysteine proteases found in the tumor microenvironment.74 High resolution ESI/MS analysis showed the correct m/z value of 706.2241 as well as the expected isotopic distribution (Figure S8). Additionally, 1H NMR analysis depicted a shift of the α-proton of the leucine residue to 4.43 ppm, revealing successful coupling. While chemical and photochemical analysis was carried out with the complexes as PF6− salts, PF6− anions were exchanged for more water-soluble Cl− counteranions in complexes 6 and 9 before biological evaluation. The anion exchange was accomplished by dissolving the PF6− salt of each complex in CH3CN, adding Amberlite IRA-410(Cl) exchange resin, stirring for 10 min, and then filtering. Condensing the filtrate yielded the Cl− salt as verified by the absence of the peak corresponding to the PF6− counteranion in the 19F-NMR spectrum.
Photophysical Properties and Photochemistry.
The ground- and excited-state properties of inhibitor-containing Ru(II) complex 9 and control 6 were investigated, and each complex was evaluated for singlet oxygen production and ligand exchange upon irradiation. The absorption spectra of 6 and 9 in CH3CN are nearly identical in the 350 to 700 nm range and are shown in Figure 3a. Complex 6 exhibits a broad peak at 471 nm (ε = 8,900 M−1 cm−1) and shoulder at 501 nm (ε ~ 8,200 M−1 cm−1) that arise from Ru(II) → dppn and Ru(II)→ dqpy 1MLCT transitions, respectively. These assignments were made from comparison to those of related complexes. For example, [Ru(dqpy)(phen)(CH3CN)]2+ (phen = 1,10-phenantholine) features a broad peak at 469 nm (ε = 7,900 M−1 cm−1), attributed to the Ru(II) → dqpy 1MLCT transition,75 since it is red-shifted relative to the Ru(II) → tpy 1MLCT absorption maximum in [Ru(tpy)-(phen)(CH3CN)]2+ at 455 nm (ε = 11,000 M−1 cm−1).76 The red-shift in the Ru(II) → dqpy 1MLCT maximum in 6 relative to that in [Ru(dqpy)(phen)(CH3CN)]2+ indicates a MeTz serving as a weaker π-acceptor than CH3CN for π-back-bonding, resulting in a lower amount of stabilization of the t2g-type d-orbital set and a smaller energy gap between the metal-centered highest occupied molecular orbitals (HOMOs) and the dqpy(π*) lowest unoccupied orbital (LUMO), since the energy of the latter is expected to remain relatively constant in these complexes. In addition, two lower energy Ru(II) → dqpy 1MLCT bands are observed for [Ru(dqpy)(phen)-(CH3CN)]2+ at 534 nm (ε = 2,300 M−1 cm−1) and 600 nm (ε = 1,100 M−1 cm−1), which appear at 580 nm (ε = 2,600 M−1 cm−1) and 640 nm (ε = 1,200 M−1 cm−1) in 6. The absorption spectrum of 6 also features strong dppn-centered 1ππ* absorption at 408 nm (ε = 10,000 M−1 cm−1), also present in other Ru(II) complexes with dppn ligands.21,61 In addition, 6 features a sharp peak at 373 nm (ε = 24,000 M−1 cm−1) that is not observed in [Ru(dqpy)(phen)(CH3CN)]2+ or [Ru(dqpy)(dppn)(CH3CN)]2+, such that it has been assigned as arising from a transition involving the MeTz ligand.
Figure 3.

(a) Electronic absorption spectra of 6 and 9 in CH3CN and (b) transient absorption spectra of 9 in acetone recorded at various delay times following a 532 nm (fwhm ~8 ns) laser pulse under N2.
The transient absorption spectra of 6 and 9 collected in deareated acetone following 532 nm (fwhm ~8 ns) excitation exhibit a broad positive signal in the 500–600 nm range with a maximum at ~540 nm, shown for 9 in Figure 3b. The signal at 540 nm for each complex can be fitted to a monoexponential function resulting in lifetimes, τ, of 3.8 and 8.5 μs for 6 and 9, respectively. The peak at ~540 nm and its long lifetime are consistent with a 3ππ* excited state localized on the dppn ligand, as previously reported for related complexes with a dppn ligand.21,61
Upon steady-state irradiation under ambient conditions, complexes 6 and 9 produce 1O2 with quantum yields, ΦΔ, of 0.78(4) and 0.8(1), respectively, in methanol (λirr = 460 nm). In addition, under a nitrogen atmosphere to prevent the formation of 1O2, the photolysis of 6 and 9 result in the exchange of triazole ligand for a solvent CH3CN molecule, as shown in Figure 4 for 6. It is evident from Figure 4 that there are isosbestic points at 240, 275, 293, 481, and 503 nm, indicating the clean conversion from the starting material to the solvated product, [Ru(dqpy)(dppn)(CH3CN)]2+. In addition, the maxima of the product are consistent with those previously reported for the related complex [Ru(dqpy)-(phen)(CH3CN)]2+.75 It should be noted that the peak at 373 nm associated with the MeTz ligand decreases in intensity during photolysis, consistent with the exchange of the monodentate ligand with a solvent molecule. The release of MeTz ligand was also confirmed by monitoring photolysis of 6 by 1H NMR spectroscopy (Figure S22). The quantum yields for MeTz ligand exchange upon 500 nm irradiation, Φ500, of 6 and 9 were determined to be 0.025(8) and 0.008(2), respectively, in CH3CN.
Figure 4.

Changes to the electronic absorption spectrum of 6 from 0 min (red) to 210 min (black) upon 670 nm irradiation in CH3CN (inset: expanded view of the visible range).
Complex Stability and Distribution Coefficients.
Before proceeding to the biological evaluation of complexes 6 and 9, preliminary studies were pursued to confirm complex stability and gain insight into its potential ability to penetrate cells. Stock solutions of complexes 6 and 9 in DMSO were diluted in cell growth media (RPMI 1640, containing 10% FBS) at 37 °C and monitored in the dark over the course of 24 h by electronic absorption spectroscopy (Figures S20–S21). While reduction in overall absorbance was noted over time, presumably due to aggregation, no spectral shifts in the MLCT bands indicating release of the triazole ligand were detected.
To further predict how complexes 6 and 9 would behave in cells, the distribution coefficient was determined to estimate their ability to penetrate cellular membranes. A positive distribution coefficient value is generally desired, usually indicating that the compound is lipophilic and can cross the cell membrane. While Ru(II) complexes can show enhanced cellular uptake due to their lipophilic cationic nature, lipophilicity characteristics can vary among complexes, leading to differences in cellular permeability. In our case, ligands dqpy and dppn are highly lipophilic and presented solubility issues during synthesis; thus, it was crucial to offset this lipophilicity to ensure solubility in aqueous cell growth media while maintaining feasible cell permeability. The distribution coefficient of the inhibitor Ru(II) complex 9 and of the control compound 6 were found to be 0.05 ± 0.01 and −0.26 ± 0.02, respectively, using the “Shake-Flask” method. Despite the lipophilic ligands present in both complexes, it is evident that the hydrophilic nature of 6 can be attributed to its overall 2+ charge. While inhibitor complex 9 contains more hydrogenbond donors, it was slightly more lipophilic than 6, presumably due to the presence of the benzyl group or leucine side chain. Nonetheless, lipophilicity is desirable to allow for permeation of the cell membrane. It was also hypothesized that the presence of the ester in 9 would aid in cellular uptake and retention, similar to ester CA-074Me that undergoes cellular uptake, followed by cleavage from intracellular esterases to form the carboxylate product CA-074 that is not cell permeable.64
Cathepsin B and L Inhibition.
To evaluate the ability of the inhibitor conjugate of compound 9 to inactivate cysteine cathepsins and serve as a delivery vector to TAMs and tumor cells, we measured its inhibitory properties using a purified enzyme assay. We chose to study inhibition of cathepsins B (CTSB) and L (CTSL) specifically for their relevance in cancer.40,77–79 Fluorogenic substrates (100 μM), Z-Arg-Arg-AMC (CTSB) or Z-Phe-Arg-AMC (CTSL), and varying concentrations of complex 9 (0.05 to 0.30 μM final) or 6 (0.5 to 25 μM final) were treated with green light (520–525 nm) for 10 min or left in the dark. Activated CTSB or CTSL was added, and the activity was immediately monitored by following the formation of the hydrolyzed fluorescent 7-amino-4-methylcoumarin (AMC) product. Data were fit to a two-step model for irreversible inhibition (Figure S12), which provided the quotient kinact/Ki representing the second-order rate constant for enzyme inactivation, that includes covalent bond formation via epoxide opening. Compound 9 successfully inhibited CTSB and CTSL under light and dark conditions with kinact/Ki values within error (0.8 to 3 × 104 M−1 s−1) (Table 1, Figure S14). Furthermore, we found that control complex 6 was unable to inhibit CTSB or CTSL in the dark or upon irradiation at concentrations as high as 25 μM (Figure S15). This result shows that the epoxysuccinyl warhead is crucial for irreversible cathepsin inhibition, rather than the cysteine active site thiolate binding to the open coordination site of the metal following triazole photorelease, which has been reported as a mechanism of cysteine protease inhibition by Ru(II) complexes.80,81
Table 1.
Second Order Rate Constants, kinact/Ki (M−1 s−1), for CTSB and CTSL Inactivation
| CTSBa | CTSLb | |||
|---|---|---|---|---|
| Compound | Dark | Light | Dark | Light |
| 6 | <10 | <10 | <10 | <10 |
| 9 | (3 ± 2) × 104 | (9.4 ± 0.3) × 103 | (8 ± 2) × 103 | (9 ± 2) × 103 |
Conditions: The CTSB stock was diluted to 16 nM (4 nM final concentration) in assay buffer (0.4 M acetate, pH 5.5, 5 mM EDTA, 0.01% Triton X-100) containing 8 mM DTT. Inhibitors were prepared as <1% DMSO stock solutions in assay buffer to achieve final concentrations ranging from 0.30 to 0.05 μM for 9 and 25 to 0.5 μM for 6. Complexes 6 and 9 were irradiated with green (520–25 nm) light (tirr = 10 min) or left in the dark. The substrate Z-Arg-Arg-AMC in assay buffer (200 μM, 50 μL) was diluted to achieve a final concentration of 100 μM. CA-074 was used as a positive control for cathepsin B inhibition. Fluorescent readings (λex = 360 nm, λem = 430 nm, 5 flashes/well) were immediately taken every 30 s for 15 min at room temperature.
CSTL assay was carried out analogously, however in 0.4 M acetate buffer, 4 mM EDTA, pH 5.5, 0.01% Triton X-100 with substrate Z-Phe-Arg-AMC. Data was fit to a two-step model for irreversible inhibition using DynaFit, which provided the quotient kinact/Ki.
Confirmed inhibition of isolated CTSB and CTSL supported the idea that 9 should be able to inhibit cathepsin secreted into the extracellular milieu, which is commonly found in the tumor microenvironment. To further investigate the ability of 9 to inhibit intracellular cathepsins, we treated cancer cells with 6 or 9 and then examined cathepsin inhibition in cell lysates. Murine breast cancer 4T1 cells were chosen to investigate biological properties of 6 and 9 to ensure data collected would be informative for future experiments using syngeneic mice models to measure immunogenic effects. 4T1 cells were treated with vehicle or 1 μM known, permeable cathepsin inhibitor E64d as a positive control, 6, or 9. After 4 h incubation, cell growth media was replaced with fresh media to remove impermeable compounds. The cells were then irradiated or left in the dark for 10 min, and then incubated for 1 h. Next, the media was aspirated, and the cells were washed with PBS and lysed. Lysates were diluted and activated with assay buffer, and fluorogenic substrate was added. Again, cathepsin inhibition was monitored by a progression curve related to hydrolysis, and formation of fluorescent AMC. It is important to note that Z-Arg-Arg-AMC is a selective substrate for CTSB, whereas Z-Phe-Arg-AMC lacks high selectivity toward CTSL; thus, inhibition of Z-Phe-Arg-AMC hydrolysis shows more general cathepsin inhibition. Collectively, these studies demonstrated that complex 9 can indeed inactivate intracellular cathepsins both under visible light irradiation and in the dark at concentrations as low as 1 μM (Table 2, Figures S18–S19). Consistent with data in Table 1, complex 6 showed no significant inhibition except under light conditions with substrate Z-Arg-Arg-AMC, presumably due to ROS generation in cells, which either directly or indirectly led to partial loss of cathepsin B activity. Effective inhibition of intracellular cathepsins also supports the hypothesis that complex 9 can penetrate the cells and behave as a delivery vector to cancer and stromal cells in the tumor microenvironment where cysteine cathepsins are overexpressed.
Table 2.
Percent Activity of Cysteine Cathepsins in 4T1 Cells Treated with E64d, 6 or 9 Using Substrates Z-Arg-Arg-AMC or Z-Phe-Arg-AMCa
| Substrate Z-Arg-Arg-AMC | Substrate Z-Phe-Arg-AMC | |||
|---|---|---|---|---|
| Compound | Light | Dark | Light | Dark |
| E64d | <5 | <5 | 21 ± 4 | 18 ± 4 |
| 6 | 53 ± 8 | 118 ± 4 | 110 ± 16 | 94 ± 8 |
| 9 | <5 | <5 | 6 ± 2 | 6 ± 2 |
Conditions: 4T1 cells were treated with vehicle or 1 μM E64d, 6, or 9, incubated for 4 h, irradiated with green (520–25 nm) light (tirr = 10 min) or left in the dark, and then incubated for an additional hour before lysing. Pretreated cell lysates were diluted in assay buffer (0.4 M acetate, pH 5.5, 5 mM EDTA, 0.01% Triton X-100) containing 8 mM DTT. Following activation of lysates for 10 min at 37 °C, lysates were diluted 2× with 200 μM substrate Z-Arg-Arg-AMC or Z-Phe-Arg-AMC, to give final concentration 100 μM. Fluorescent readings (λex = 360 nm, λem = 430 nm, 5 flashes/well) were immediately taken every 30 s for 5 min at 37 °C. Percent activities were determined relative to the vehicle control.
Cellular Studies.
Following verification of intra- and extracellular cathepsin inhibition, complexes 6 and 9 were evaluated against bone-marrow-derived BALB/c M0, M1, and M2 macrophages, 4T1 cells, and murine L929 fibroblasts in 2D culture. The cells were treated with 6 or 9 (0.1 to 18 μM) and incubated for 4 h, after which time the cell growth media was replaced with fresh media and the cells were irradiated with green light (520–525 nm) for 10 min. Cells under dark conditions were treated similarly in the absence of irradiation, however, with a larger range of concentrations (0.1 to 50 μM). After 72 h, cell viability was assessed by the MTT assay. As anticipated, both 6 and 9 showed strong phototoxicity due to their ability to produce ROS (Table 3). While 6 and 9 did show undesired phototoxicity in L929 fibroblasts, phototoxicity in normal tissue should be avoided if the complexes were irradiated only in cancerous tissue. Moreover, in the same cell line, 6 and 9 displayed EC50 values within error under light conditions, consistent with 1O2 generation through energy transfer (PDT mechanism) rather than cathepsin inhibition, driving cell death. Fortunately, appending the inhibitor conjugate in complex 9 ameliorated the cellular toxicity found with 6 in the dark. Compound 9 showed excellent photoselectivity with PI values as high as >19 with no evidence of toxicity at concentrations up to 50 μM across all cell lines in the dark; higher concentrations of 9 were not evaluated due to low solubility. In contrast, 6 showed EC50 values ranging from 19 to 36 μM in the dark in all cell lines tested. Additionally, an ~5-fold higher potency was observed for 9 in macrophages vs tumor 4T1 cells, consistent with our previously reported compound 2, which was also more potent in macrophages as compared to TNBC cells.43 Importantly, Blum et al. reported a cathepsin inhibitor that was able to kill macrophage cells in vitro and reduce tumor cell mass in vivo; however, no effect was elicited in 4T1 cells grown in 2D.30 This finding suggests that while the complex may not be potent against tumor cells, TAM cell death can induce an immune response and subsequent cell death of neighboring tumor cells when further applied in a 3D coculture or in vivo.
Table 3.
EC50 Values (μM) and Phototherapeutic Indexes (PI) of 6 and 9 in Murine Macrophage (M0, M1, M2), Triple Negative Breast Cancer (4T1), and Fibroblast (L929) Cell Linesa
| Compound 6 | Compound 9 | |||||
|---|---|---|---|---|---|---|
| Cell line | Light EC50 | Dark EC50 | PI | Light EC50 | Dark EC50 | PI |
| M0 | 2.0 ± 0.6 | 20 ± 1 | 10 | 2.7 ± 0.6 | >50 | >19 |
| M1 | 2.3 ± 0.7 | 19 ± 1 | 8 | 2.9 ± 0.9 | >50 | >17 |
| M2 | 2.0 ± 0.3 | 19 ± 3 | 10 | 2.4 ± 0.3 | >50 | >21 |
| 4T1 | 14 ± 1 | 30 ± 5 | 2 | 15 ± 1 | >50 | >3 |
| L929 | 2.6 ± 0.9 | 36 ± 9 | 14 | 3 ± 1 | >50 | >17 |
Cells treated with 6 or 9 for 4 h, treatments were replaced with vehicle, and cells were irradiated with green (520–25 nm) light (tirr = 10 min) or left in the dark. Cell viability was determined by MTT assay 72 h after irradiation. Data are the average of three independent experiments using quadruplicate wells, and errors shown are standard deviations. PI = phototherapeutic index = ratio dark EC50/light EC50.
While attachment of the cathepsin inhibitor conjugate did not make 9 more potent than 6, 2D cell culture does not accurately mimic the tumor microenvironment, especially phenotypes where cysteine proteases are translocated to the cell surface and act along the invasive edge of tumors. Future studies will investigate the biological effect of complex 9 in 3D coculture experiments containing both macrophages and tumor cells. This heterogeneous coculture provides a more accurate depiction of the tumor microenvironment, including interactions with the extracellular matrix (ECM), cell polarity, and cell-to-cell contacts. Our previous cathepsin inhibitor 1 (Figure 1) was found to be more toxic in 3D MDA-MB-231 spheroids versus cells grown in 2D, presumably due to greater translocation of cathepsins to the cell surface.23 Thus, complex 9 has the potential to show additional anticancer effects that are not presented in 2D culture. Meanwhile employing green light irradiation is expected to broaden the scope of compounds such as 9 in anticancer applications as compared to previous analogs activated with blue light. Finally, our previously reported dual PCT/PDT agent [Ru(tpy)-(Me2dppn)(py)]2+ 2 was able to target macrophages and yield an immunogenic response without a method of drug delivery.43 Therefore inclusion of cathepsin inhibitor to deliver our Ru(II) PCT/PDT agent to TAMs and tumor cells has great potential to show an enhanced immunotherapeutic effect. Activity-based probes activated by cathepsins have shown that cathepsins are highly expressed in cancerous tissues82–84 and cathepsin inhibition provides a useful pharmacological effect for inhibiting metastasis and for enhancing drug delivery.85–88
CONCLUSIONS
We report the design, synthesis, and biological evaluation of novel Ru(II) PCT/PDT agents. Triazole-containing compounds 6 and 9 were characterized to show photoactivated ligand dissociation and photosensitization of 1O2 for dual action PCT/PDT applications with green light. Importantly, we demonstrate that Ru PCT/PDT complex 5 containing an aldehyde can be used in an oxime “click” reaction for conjugation to drug delivery vectors to prepare compounds such as cathepsin conjugate 9. Complex 9 is able to inactivate intra- and extracellular cathepsins in the dark and light for delivery vector applications to macrophages and tumor cells. Additionally, cell viability studies indicated that PDT is the driving force for cell death where complexes 6 and 9 show similar efficacy in the light. Fortunately, cathepsin inhibitor conjugation was able to reduce the toxicity of the Ru PCT/PDT agent in the dark for 9 vs the control complex 6. Importantly previous PCT/PDT agents displayed immunogenic effects in 3D coculture containing macrophages and tumor cells. Thus, conjugate 9 has great potential for metalloimmunotherapy applications. Studies are now under-way to evaluate 9 in 3D coculture and in vivo models of breast cancer.
Supplementary Material
ACKNOWLEDGMENTS
We gratefully acknowledge the National Science Foundation (CHE 2102508), National Instutites of Health (T32GM142519), and Wayne State University (Grants Boost and Rumle Fellowship to M.D.) for support of this research. NMR resources are supported by the National Institutes of Health (S10OD028488), and High Resolution Mass Spectrometry Resources are supported by R01 GM098285-07S1.
Footnotes
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.inorgchem.4c01008.
Detailed experimental procedures of synthesis of compounds 6 and 9, corresponding 1H NMR spectral data; biological assays (cell viability MTT assay for 6 and 9; cathepsin inhibition assay in purified enzymes and in 4T1 cell lysates), stability studies of 6 and 9, photophysical properties of complexes 6 and 9. (PDF)
Complete contact information is available at: https://pubs.acs.org/10.1021/acs.inorgchem.4c01008
The authors declare no competing financial interest.
Contributor Information
Madeline Denison, Department of Chemistry, Wayne State University, Detroit, Michigan 48202, United States.
Santana P. Garcia, Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210, United States
Alexander Ullrich, Department of Oncology, Wayne State University, Detroit, Michigan 48201, United States.
Izabela Podgorski, Department of Pharmacology, School of Medicine, Wayne State University, Detroit, Michigan 48201, United States; Karmanos Cancer Institute, Detroit, Michigan 48201, United States.
Heather Gibson, Department of Oncology, Wayne State University, Detroit, Michigan 48201, United States; Karmanos Cancer Institute, Detroit, Michigan 48201, United States.
Claudia Turro, Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210, United States.
Jeremy J. Kodanko, Department of Chemistry, Wayne State University, Detroit, Michigan 48202, United States; Karmanos Cancer Institute, Detroit, Michigan 48201, United States
REFERENCES
- (1).Cancer. https://www.who.int/news-room/fact-sheets/detail/cancer (accessed 2024-01-29).
- (2).Siegel RL; Miller KD; Wagle NS; Jemal A Cancer Statistics, 2023. CA Cancer J. Clin 2023, 73 (1), 17–48. [DOI] [PubMed] [Google Scholar]
- (3).Juris A; Balzani V; Barigelletti F; Campagna S; Belser P; von Zelewsky A Ru(II) Polypyridine Complexes Photophysics, Photochemistry, Eletrochemistry, and Chemiluminescence. Coord. Chem. Rev 1988, 84, 85–277. [Google Scholar]
- (4).Friedman AE; Chambron JC; Sauvage JP; Turro NJ; Barton JK A Molecular Light Switch for DNA: Ru(Bpy)2(Dppz)-2+. J. Am. Chem. Soc 1990, 112 (12), 4960–4962. [Google Scholar]
- (5).Kumar CV; Barton JK; Turro NJ Photophysics of Ruthenium Complexes Bound to Double Helical DNA. J. Am. Chem. Soc 1985, 107 (19), 5518–5523. [Google Scholar]
- (6).Greguric I; Aldrich-Wright JR; Collins JGA 1H NMR Study of the Binding of Δ-[Ru(Phen)2 DPQ]2+ to the Hexanucleotide d(GTCGAC)2. Evidence for Intercalation from the Minor Groove. J. Am. Chem. Soc 1997, 119 (15), 3621–3622. [Google Scholar]
- (7).Fong J; Kasimova K; Arenas Y; Kaspler P; Lazic S; Mandel A; Lilge L A Novel Class of Ruthenium-Based Photosensitizers Effectively Kills in Vitro Cancer Cells and in Vivo Tumors. Photochem. Photobiol. Sci 2015, 14 (11), 2014–2023. [DOI] [PubMed] [Google Scholar]
- (8).Kaspler P; Lazic S; Forward S; Arenas Y; Mandel A; Lilge L A Ruthenium(Ii) Based Photosensitizer and Transferrin Complexes Enhance Photo-Physical Properties, Cell Uptake, and Photodynamic Therapy Safety and Efficacy. Photochem. Photobiol. Sci 2016, 15 (4), 481–495. [DOI] [PubMed] [Google Scholar]
- (9).Gandosio A; Purkait K; Gasser G Recent Approaches towards the Development of Ru(II) Polypyridyl Complexes for Anticancer Photodynamic Therapy. Chimia 2021, 75 (10), 845. [DOI] [PubMed] [Google Scholar]
- (10).Martínez-Alonso M; Gasser G Ruthenium Polypyridyl Complex-Containing Bioconjugates. Coord. Chem. Rev 2021, 434, 213736. [Google Scholar]
- (11).Wu Y; Li S; Chen Y; He W; Guo Z Recent Advances in Noble Metal Complex Based Photodynamic Therapy. Chem. Sci 2022, 13 (18), 5085–5106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (12).Silva MJSA; Vinck R; Wang Y; Saubaméa B; Tharaud M; Dominguez-Jurado E; Karges J; Gois PMP; Gasser G Towards Selective Delivery of a Ruthenium(II) Polypyridyl Complex-Containing Bombesin Conjugate into Cancer Cells. ChemBioChem. 2023, 24 (4), e202200647. [DOI] [PubMed] [Google Scholar]
- (13).Raza A; Archer SA; Fairbanks SD; Smitten KL; Botchway SW; Thomas JA; MacNeil S; Haycock JW A Dinuclear Ruthenium(II) Complex Excited by Near-Infrared Light through Two-Photon Absorption Induces Phototoxicity Deep within Hypoxic Regions of Melanoma Cancer Spheroids. J. Am. Chem. Soc 2020, 142 (10), 4639–4647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (14).Monro S; Colón KL; Yin H; Roque J; Konda P; Gujar S; Thummel RP; Lilge L; Cameron CG; McFarland SA Transition Metal Complexes and Photodynamic Therapy from a Tumor-Centered Approach: Challenges, Opportunities, and Highlights from the Development of TLD1433. Chem. Rev 2019, 119 (2), 797–828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (15).Cuello-Garibo J-A; Meijer MS; Bonnet S To Cage or to Be Caged? The Cytotoxic Species in Ruthenium-Based Photoactivated Chemotherapy Is Not Always the Metal. Chem. Commun 2017, 53 (50), 6768–6771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (16).Bonnet S Why Develop Photoactivated Chemotherapy? Dalton Trans. 2018, 47 (31), 10330–10343. [DOI] [PubMed] [Google Scholar]
- (17).Farrer NJ; Salassa L; Sadler PJ Photoactivated Chemotherapy (PACT): The Potential of Excited-State d-Block Metals in Medicine. Dalton Trans. 2009, No. 48, 10690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (18).Busemann A; Flaspohler I; Zhou X-Q; Schmidt C; Goetzfried SK; van Rixel VHS; Ott I; Siegler MA; Bonnet S Ruthenium-Based PACT Agents Based on Bisquinoline Chelates: Synthesis, Photochemistry, and Cytotoxicity. J. Biol. Inorg. Chem 2021, 26 (6), 667–674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (19).Mari C; Pierroz V; Leonidova A; Ferrari S; Gasser G Towards Selective Light-Activated RuII-Based Prodrug Candidates. Eur. J. Inorg. Chem 2015, 2015 (23), 3879–3891. [Google Scholar]
- (20).Bonnet S Ruthenium-Based Photoactivated Chemotherapy. J. Am. Chem. Soc 2023, 145 (43), 23397–23415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (21).Knoll JD; Albani BA; Turro C Excited State Investigation of a New Ru(II) Complex for Dual Reactivity with Low Energy Light. Chem. Commun 2015, 51 (42), 8777–8780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (22).Knoll JD; Albani BA; Durr CB; Turro C Unusually Efficient Pyridine Photodissociation from Ru(II) Complexes with Sterically Bulky Bidentate Ancillary Ligands. J. Phys. Chem. A 2014, 118 (45), 10603–10610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (23).Arora K; Herroon M; Al-Afyouni MH; Toupin NP; Rohrabaugh TN; Loftus LM; Podgorski I; Turro C; Kodanko JJ Catch and Release Photosensitizers: Combining Dual-Action Ruthenium Complexes with Protease Inactivation for Targeting Invasive Cancers. J. Am. Chem. Soc 2018, 140 (43), 14367–14380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (24).Brix K; Dunkhorst A; Mayer K; Jordans S Cysteine Cathepsins: Cellular Roadmap to Different Functions. Biochimie 2008, 90 (2), 194–207. [DOI] [PubMed] [Google Scholar]
- (25).Fonović M; Turk B Cysteine Cathepsins and Extracellular Matrix Degradation. Biochim. Biophys. Acta, Gen. Subj 2014, 1840 (8), 2560–2570. [DOI] [PubMed] [Google Scholar]
- (26).Mason SD; Joyce JA Proteolytic Networks in Cancer. Trends Cell Biol. 2011, 21 (4), 228–237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (27).Mohamed MM; Sloane BF Multifunctional Enzymes in Cancer. Nat. Rev. Cancer 2006, 6 (10), 764–775. [DOI] [PubMed] [Google Scholar]
- (28).Yadati T; Houben T; Bitorina A; Shiri-Sverdlov R The Ins and Outs of Cathepsins: Physiological Function and Role in Disease Management. Cells 2020, 9 (7), 1679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (29).Rudzińska M; Parodi A; Soond SM; Vinarov AZ; Korolev DO; Morozov AO; Daglioglu C; Tutar Y; Zamyatnin AA The Role of Cysteine Cathepsins in Cancer Progression and Drug Resistance. Int. J. Mol. Sci 2019, 20 (14), 3602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (30).Salpeter SJ; Pozniak Y; Merquiol E; Ben-Nun Y; Geiger T; Blum G A Novel Cysteine Cathepsin Inhibitor Yields Macrophage Cell Death and Mammary Tumor Regression. Oncogene 2015, 34 (50), 6066–6078. [DOI] [PubMed] [Google Scholar]
- (31).Bell-McGuinn KM; Garfall AL; Bogyo M; Hanahan D; Joyce JA Inhibition of Cysteine Cathepsin Protease Activity Enhances Chemotherapy Regimens by Decreasing Tumor Growth and Invasiveness in a Mouse Model of Multistage Cancer. Cancer Res. 2007, 67 (15), 7378–7385. [DOI] [PubMed] [Google Scholar]
- (32).Withana NP; Blum G; Sameni M; Slaney C; Anbalagan A; Olive MB; Bidwell BN; Edgington L; Wang L; Moin K; Sloane BF; Anderson RL; Bogyo MS; Parker BS Cathepsin B Inhibition Limits Bone Metastasis in Breast Cancer. Cancer Res. 2012, 72 (5), 1199–1209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (33).Elie BT; Gocheva V; Shree T; Dalrymple SA; Holsinger LJ; Joyce JA Identification and Pre-Clinical Testing of a Reversible Cathepsin Protease Inhibitor Reveals Anti-Tumor Efficacy in a Pancreatic Cancer Model. Biochimie 2010, 92 (11), 1618–1624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (34).Shree T; Olson OC; Elie BT; Kester JC; Garfall AL; Simpson K; Bell-McGuinn KM; Zabor EC; Brogi E; Joyce JA Macrophages and Cathepsin Proteases Blunt Chemotherapeutic Response in Breast Cancer. Genes Dev. 2011, 25 (23), 2465–2479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (35).Premzl A; Zavasnik-Bergant V; Turk V; Kos J Intracellular and Extracellular Cathepsin B Facilitate Invasion of MCF-10A neoT Cells through Reconstituted Extracellular Matrix in Vitro. Exp. Cell Res 2003, 283 (2), 206–214. [DOI] [PubMed] [Google Scholar]
- (36).Sever N; Filipic M; Brzin J; Lah TT Effect of Cysteine Proteinase Inhibitors on Murine B16 Melanoma Cell Invasion in Vitro. Biol. Chem 2002, 383 (5), 839–842. [DOI] [PubMed] [Google Scholar]
- (37).Matarrese P; Ascione B; Ciarlo L; Vona R; Leonetti C; Scarsella M; Mileo AM; Catricalà C; Paggi MG; Malorni W Cathepsin B Inhibition Interferes with Metastatic Potential of Human Melanoma: An in Vitro and in Vivo Study. Mol. Cancer 2010, 9 (1), 207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (38).Sevenich L; Schurigt U; Sachse K; Gajda M; Werner F; Müller S; Vasiljeva O; Schwinde A; Klemm N; Deussing J; Peters C; Reinheckel T Synergistic Antitumor Effects of Combined Cathepsin B and Cathepsin Z Deficiencies on Breast Cancer Progression and Metastasis in Mice. Proc. Natl. Acad. Sci. U.S.A 2010, 107 (6), 2497–2502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (39).Vasiljeva O; Korovin M; Gajda M; Brodoefel H; Bojic L; Krüger A; Schurigt U; Sevenich L; Turk B; Peters C; Reinheckel T Reduced Tumour Cell Proliferation and Delayed Development of High-Grade Mammary Carcinomas in Cathepsin B-Deficient Mice. Oncogene 2008, 27 (30), 4191–4199. [DOI] [PubMed] [Google Scholar]
- (40).Gocheva V; Zeng W; Ke D; Klimstra D; Reinheckel T; Peters C; Hanahan D; Joyce JA Distinct Roles for Cysteine Cathepsin Genes in Multistage Tumorigenesis. Genes Dev. 2006, 20 (5), 543–556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (41).Ruffell B; Affara NI; Cottone L; Junankar S; Johansson M; DeNardo DG; Korets L; Reinheckel T; Sloane BF; Bogyo M; Coussens LM Cathepsin C Is a Tissue-Specific Regulator of Squamous Carcinogenesis. Genes Dev. 2013, 27 (19), 2086–2098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (42).Rothberg JM; Bailey KM; Wojtkowiak JW; Ben-Nun Y; Bogyo M; Weber E; Moin K; Blum G; Mattingly RR; Gillies RJ; Sloane BF Acid-Mediated Tumor Proteolysis: Contribution of Cysteine Cathepsins. Neoplasia 2013, 15 (10), 1125–IN9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (43).Toupin N; Herroon MK; Thummel RP; Turro C; Podgorski I; Gibson H; Kodanko JJ Metalloimmunotherapy with Rhodium and Ruthenium Complexes: Targeting Tumor Associated Macrophages. Chem. - Eur. J 2022, 28 (24). DOI: 10.1002/chem.202104430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (44).Lendeckel U; Venz S; Wolke C Macrophages: Shapes and Functions. ChemTexts 2022, 8 (2), 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (45).Hirayama D; Iida T; Nakase H The Phagocytic Function of Macrophage-Enforcing Innate Immunity and Tissue Homeostasis. Int. J. Mol. Sci 2018, 19 (1), 92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (46).Shapouri Moghaddam A; Mohammadian S; Vazini H; Taghadosi M; Esmaeili S; Mardani F; Seifi B; Mohammadi A; Afshari JT; Sahebkar A Macrophage Plasticity, Polarization, and Function in Health and Disease. J. Cell. Physiol 2018, 233 (9), 6425–6440. [DOI] [PubMed] [Google Scholar]
- (47).Ricketts TD; Prieto-Dominguez N; Gowda PS; Ubil E Mechanisms of Macrophage Plasticity in the Tumor Environment: Manipulating Activation State to Improve Outcomes. Front. Immunol 2021, 12, 642285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (48).Yang M; Liu J; Shao J; Qin Y; Ji Q; Zhang X; Du J Cathepsin S-Mediated Autophagic Flux in Tumor-Associated Macrophages Accelerate Tumor Development by Promoting M2 Polarization. Mol. Cancer 2014, 13 (1), 43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (49).Alam S; Liu Q; Liu S; Liu Y; Zhang Y; Yang X; Liu G; Fan K; Ma J Up-Regulated Cathepsin C Induces Macrophage M1 Polarization through FAK-Triggered P38 MAPK/NF-κB Pathway. Exp. Cell Res 2019, 382 (2), 111472. [DOI] [PubMed] [Google Scholar]
- (50).Dykes SS; Fasanya HO; Siemann DW Cathepsin L Secretion by Host and Neoplastic Cells Potentiates Invasion. Oncotarget 2019, 10 (53), 5560–5568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (51).Oelschlaegel D; Weiss Sadan T; Salpeter S; Krug S; Blum G; Schmitz W; Schulze A; Michl P Cathepsin Inhibition Modulates Metabolism and Polarization of Tumor-Associated Macrophages. Cancers 2020, 12 (9), 2579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (52).He G; He M; Wang R; Li X; Hu H; Wang D; Wang Z; Lu Y; Xu N; Du J; Fan J; Peng X; Sun W A Near-Infrared Light-Activated Photocage Based on a Ruthenium Complex for Cancer Phototherapy. Angew. Chem., Int. Ed 2023, 62 (24), e202218768. [DOI] [PubMed] [Google Scholar]
- (53).He M; Chen F; Shao D; Weis P; Wei Z; Sun W Photoresponsive Metallopolymer Nanoparticles for Cancer Theranostics. Biomaterials 2021, 275, 120915. [DOI] [PubMed] [Google Scholar]
- (54).Zhang B; Xu Z; Zhou W; Liu Z; Zhao J; Gou S A Light-Controlled Multi-Step Drug Release Nanosystem Targeting Tumor Hypoxia for Synergistic Cancer Therapy. Chem. Sci 2021, 12 (35), 11810–11820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (55).Toupin NP; Steinke SJ; Herroon MK; Podgorski I; Turro C; Kodanko JJ Unlocking the Potential of Ru(II) Dual action Compounds with the Power of the Heavy atom Effect †. Photochem. Photobiol 2022, 98 (2), 378–388. [DOI] [PubMed] [Google Scholar]
- (56).Lameijer LN; Ernst D; Hopkins SL; Meijer MS; Askes SHC; Le Dévédec SE; Bonnet S A Red-Light-Activated Ruthenium-Caged NAMPT Inhibitor Remains Phototoxic in Hypoxic Cancer Cells. Angew. Chem., Int. Ed 2017, 56 (38), 11549–11553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (57).Zhang L; Wang P; Zhou X-Q; Bretin L; Zeng X; Husiev Y; Polanco EA; Zhao G; Wijaya LS; Biver T; Le Dévédec SE; Sun W; Bonnet S Cyclic Ruthenium-Peptide Conjugates as Integrin-Targeting Phototherapeutic Prodrugs for the Treatment of Brain Tumors. J. Am. Chem. Soc 2023, 145 (27), 14963–14980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (58).Karges J; Heinemann F; Maschietto F; Patra M; Blacque O; Ciofini I; Spingler B; Gasser G A Ru(II) Polypyridyl Complex Bearing Aldehyde Functions as a Versatile Synthetic Precursor for Long-Wavelength Absorbing Photodynamic Therapy Photosensitizers. Bioorg. Med. Chem 2019, 27 (12), 2666–2675. [DOI] [PubMed] [Google Scholar]
- (59).Ash C; Dubec M; Donne K; Bashford T Effect of Wavelength and Beam Width on Penetration in Light-Tissue Interaction Using Computational Methods. Lasers Med. Sci 2017, 32 (8), 1909–1918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (60).Fritsch C; Stege H; Saalmann G; Goerz G; Ruzicka T; Krutmann J Green Light Is Effective and Less Painful than Red Light in Photodynamic Therapy of Facial Solar Keratoses. Photoderm. Photoimm. Photomed 1997, 13 (5–6), 181–185. [DOI] [PubMed] [Google Scholar]
- (61).Sun Y; Joyce LE; Dickson NM; Turro C Efficient DNA Photocleavage by [Ru(Bpy)2(Dppn)]2+ with Visible Light. Chem. Commun 2010, 46 (14), 2426. [DOI] [PubMed] [Google Scholar]
- (62).Albani BA; Peña B; Leed NA; de Paula NABG; Pavani C; Baptista MS; Dunbar KR; Turro C Marked Improvement in Photoinduced Cell Death by a New Tris-Heteroleptic Complex with Dual Action: Singlet Oxygen Sensitization and Ligand Dissociation. J. Am. Chem. Soc 2014, 136 (49), 17095–17101. [DOI] [PubMed] [Google Scholar]
- (63).Gupta S; Vandevord JM; Loftus LM; Toupin N; Al-Afyouni MH; Rohrabaugh TN; Turro C; Kodanko JJ Ru(II)-Based Acetylacetonate Complexes Induce Apoptosis Selectively in Cancer Cells. Inorg. Chem 2021, 60 (24), 18964–18974. [DOI] [PubMed] [Google Scholar]
- (64).Buttle DJ; Murata M; Knight CG; Barrett AJ CA074 Methyl Ester: A Proinhibitor for Intracellular Cathepsin B. Arch. Biochem. Biophys 1992, 299 (2), 377–380. [DOI] [PubMed] [Google Scholar]
- (65).Yoon MC; Christy MP; Phan VV; Gerwick WH; Hook G; O’Donoghue AJ; Hook V Molecular Features of CA-074 pH-Dependent Inhibition of Cathepsin B. Biochemistry 2022, 61 (4), 228–238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (66).Montaser M; Lalmanach G; Mach L CA-074, but Not Its Methyl Ester CA-074Me, Is a Selective Inhibitor of Cathepsin B within Living Cells. Biol. Chem 2002, 383 (7–8), 1305–1308. [DOI] [PubMed] [Google Scholar]
- (67).Wong DYQ; Yeo CHF; Ang WH Immuno-Chemotherapeutic Platinum(IV) Prodrugs of Cisplatin as Multimodal Anticancer Agents. Angew. Chem., Int. Ed 2014, 53 (26), 6752–6756. [DOI] [PubMed] [Google Scholar]
- (68).Ulrich S; Boturyn D; Marra A; Renaudet O; Dumy P Oxime Ligation: A Chemoselective Click-Type Reaction for Accessing Multifunctional Biomolecular Constructs. Chem. - Eur. J 2014, 20 (1), 34–41. [DOI] [PubMed] [Google Scholar]
- (69).Collins J; Xiao Z; Müllner M; Connal LA The Emergence of Oxime Click Chemistry and Its Utility in Polymer Science. Polym. Chem 2016, 7 (23), 3812–3826. [Google Scholar]
- (70).Hering A; Braga Emidio N; Muttenthaler M Expanding the Versatility and Scope of the Oxime Ligation: Rapid Bioconjugation to Disulfide-Rich Peptides. Chem. Commun 2022, 58 (65), 9100–9103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (71).Kölmel DK; Kool ET Oximes and Hydrazones in Bioconjugation: Mechanism and Catalysis. Chem. Rev 2017, 117 (15), 10358–10376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (72).Dirksen A; Dawson PE Rapid Oxime and Hydrazone Ligations with Aromatic Aldehydes for Biomolecular Labeling. Bioconjugate Chem. 2008, 19 (12), 2543–2548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (73).Zhou Q; Lei W; Chen Y; Li C; Hou Y; Zhang B; Wang X Ruthenium(II)-Arene Complexes with Strong Fluorescence: Insight into the Underlying Mechanism. Chem. - Eur. J 2012, 18 (28), 8617–8621. [DOI] [PubMed] [Google Scholar]
- (74).Tamai M; Matsumoto K; Omura S; Koyama I; Ozawa Y; Hanada K In Vitro and in Vivo Inhibition of Cysteine Proteinases by EST, a New Analog of E-64. J. Pharmacobio-Dyn 1986, 9 (8), 672–677. [DOI] [PubMed] [Google Scholar]
- (75).Al-Afyouni MH; Rohrabaugh TN; Al-Afyouni KF; Turro C New Ru(II) Photocages Operative with near-IR Light: New Platform for Drug Delivery in the PDT Window. Chem. Sci 2018, 9 (32), 6711–6720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (76).Loftus LM; Al-Afyouni KF; Rohrabaugh TN; Gallucci JC; Moore CE; Rack JJ; Turro C Unexpected Role of Ru(II) Orbital and Spin Contribution on Photoinduced Ligand Exchange: New Mechanism To Access the Photodynamic Therapy Window. J. Phys. Chem. C 2019, 123 (16), 10291–10299. [Google Scholar]
- (77).Jakoš T; Pišlar A; Jewett A; Kos J Cysteine Cathepsins in Tumor-Associated Immune Cells. Front. Immunol 2019, 10, 2037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (78).Gondi CS; Rao JS Cathepsin B as a Cancer Target. Expert Opin. Ther. Targets 2013, 17 (3), 281–291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (79).Sudhan DR; Siemann DW Cathepsin L Targeting in Cancer Treatment. Pharmacol. Ther 2015, 155, 105–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (80).Karges J; Cohen SM Rhenium(V) Complexes as Cysteine-Targeting Coordinate Covalent Warheads. J. Med. Chem 2023, 66 (4), 3088–3105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (81).Mosi R; Baird IR; Cox J; Anastassov V; Cameron B; Skerlj RT; Fricker SP Rhenium Inhibitors of Cathepsin B (ReO(SYS)X (Where Y = S, Py; X = Cl, Br, SPhOMe- p)): Synthesis and Mechanism of Inhibition. J. Med. Chem 2006, 49 (17), 5262–5272. [DOI] [PubMed] [Google Scholar]
- (82).Ofori LO; Withana NP; Prestwood TR; Verdoes M; Brady JJ; Winslow MM; Sorger J; Bogyo M Design of Protease Activated Optical Contrast Agents That Exploit a Latent Lysosomotropic Effect for Use in Fluorescence-Guided Surgery. ACS Chem. Biol 2015, 10 (9), 1977–1988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (83).Kennedy GT; Holt DE; Azari FS; Bernstein E; Nadeem B; Chang A; Sullivan NT; Segil A; Desphande C; Bensen E; Santini JT; Kucharczuk JC; Delikatny EJ; Bogyo M; Egan AJM; Bradley CW; Eruslanov E; Lickliter JD; Wright G; Singhal S A Cathepsin-Targeted Quenched Activity-Based Probe Facilitates Enhanced Detection of Human Tumors during Resection. Clin. Cancer Res 2022, 28 (17), 3729–3741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (84).Suurs FV; Qiu S-Q; Yim JJ; Schröder CP; Timmer-Bosscha H; Bensen ES; Santini JT; De Vries EGE; Bogyo M; Van Dam GM Fluorescent Image-Guided Surgery in Breast Cancer by Intravenous Application of a Quenched Fluorescence Activity-Based Probe for Cysteine Cathepsins in a Syngeneic Mouse Model. EJNMMI Res. 2020, 10 (1), 111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (85).Karnthaler Benbakka C; Koblmüller B; Mathuber M; Holste K; Berger W; Heffeter P; Kowol CR; Keppler BK Synthesis, Characterization and in Vitro Studies of a Cathepsin B Cleavable Prodrug of the VEGFR Inhibitor Sunitinib. Chem. Biodiversity 2019, 16 (1), e1800520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (86).Van Dalen FJ; Verdoes M Inhibitory Prodrug Mechanism for Cysteine Cathepsin-Targeted Self-Controlled Drug Release. J. Enzyme Inhibit. Med. Chem 2022, 37 (1), 2566–2573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (87).Dubowchik GM; Firestone RA; Padilla L; Willner D; Hofstead SJ; Mosure K; Knipe JO; Lasch SJ; Trail PA Cathepsin B-Labile Dipeptide Linkers for Lysosomal Release of Doxorubicin from Internalizing Immunoconjugates: Model Studies of Enzymatic Drug Release and Antigen-Specific In Vitro Anticancer Activity. Bioconjugate Chem. 2002, 13 (4), 855–869. [DOI] [PubMed] [Google Scholar]
- (88).Zhang X; Tang K; Wang H; Liu Y; Bao B; Fang Y; Zhang X; Lu W Design, Synthesis, and Biological Evaluation of New Cathepsin B-Sensitive Camptothecin Nanoparticles Equipped with a Novel Multifuctional Linker. Bioconjugate Chem. 2016, 27 (5), 1267–1275. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
