You are on page 1of 36

pharmaceuticals

Review
Photomodulation Approaches to Overcome
Antimicrobial Resistance
Sofia N. Sarabando 1,2 , Andreia Palmeira 1,3 , Maria Emília Sousa 1,3, * , Maria Amparo F. Faustino 2
and Carlos J. P. Monteiro 2, *

1 Laboratory of Organic and Pharmaceutical Chemistry, Chemical Sciences Department, Faculty of Pharmacy,
University of Porto, 4050-313 Porto, Portugal; sofia.sarabando@ua.pt (S.N.S.); apalmeira@ff.up.pt (A.P.)
2 LAQV-Requimte and Department of Chemistry, University of Aveiro, 3010-193 Aveiro, Portugal;
faustino@ua.pt
3 CIIMAR—Interdisciplinary Centre of Marine and Environmental Research, 4450-208 Porto, Portugal
* Correspondence: esousa@ff.up.pt (M.E.S.); cmonteiro@ua.pt (C.J.P.M.)

Abstract: Photopharmacology is an approach that aims to be an alternative to classical chemotherapy.


Herein, the different classes of photoswitches and photocleavage compounds and their biological
applications are described. Proteolysis targeting chimeras (PROTACs) containing azobenzene moi-
eties (PHOTACs) and photocleavable protecting groups (photocaged PROTACs) are also mentioned.
Furthermore, porphyrins are referenced as successful photoactive compounds in a clinical context,
such as in the photodynamic therapy of tumours as well as preventing antimicrobial resistance,
namely in bacteria. Porphyrins combining photoswitches and photocleavage systems are highlighted,
taking advantage of both photopharmacology and photodynamic action. Finally, porphyrins with an-
tibacterial activity are described, taking advantage of the synergistic effect of photodynamic treatment
and antibiotic therapy to overcome bacterial resistance.

Keywords: photopharmacology; photoswitch; photocaged; porphyrin; antimicrobials

Citation: Sarabando, S.N.; Palmeira,


1. Introduction
A.; Sousa, M.E.; Faustino, M.A.F.;
Monteiro, C.J.P. Photomodulation Photopharmacology is a new pharmacophore modality that is gaining attention as a
Approaches to Overcome promising alternative to classical chemotherapy drugs (Figure 1a) [1–3]. It is characterized
Antimicrobial Resistance. by the combination of photochemistry and pharmacology, and it aims at solving issues
Pharmaceuticals 2023, 16, 682. related to poor drug selectivity, minimizing off-target effects and the emergence of drug
https://doi.org/10.3390/ resistance [4]. With the increased rate of antimicrobial resistance, there are fewer treat-
ph16050682 ments available, so the World Health Organization (WHO) recommends developing new
Academic Editor: Abdelwahab Omri
antimicrobial approaches to overcome this problem [5]. In this way, the use of light on
the inactivation of microorganisms (MO) has been shown to be an outstanding tool, as it
Received: 1 April 2023 is non-evasive and capable of fast and controllable delivery to precise locations, paving
Revised: 24 April 2023 the way for the use of photons for new medical treatment approaches, such as photophar-
Accepted: 30 April 2023 macology, an emerging field that utilizes light to control biological processes with high
Published: 2 May 2023
spatiotemporal precision, offering a promising strategy for overcoming antimicrobial resis-
tance [2,6]. Its principle is the introduction of a photocaged or photoswitchable moiety into
the molecular structure of the bioactive compound, where the light effect can be irreversible
or reversible. Photocleavage molecules are an example of irreversible photoactivation by
Copyright: © 2023 by the authors.
Licensee MDPI, Basel, Switzerland.
using photocleavable protecting groups (PPG) as ‘’cages” [7]. PPG are small moieties that
This article is an open access article
can be released upon irradiation, exposing the bioactive molecule in its active form [7]. On
distributed under the terms and
the other hand, photoswitches are molecules that can be interconverted upon the action of
conditions of the Creative Commons light, in a reversible and fast process, into different structural configurations (cis or trans),
Attribution (CC BY) license (https:// in which one of the configurations is the bioactive form [8]. Light offers the possibility to
creativecommons.org/licenses/by/ control both the pharmacokinetic (PK) and pharmacodynamic (PD) properties of molecules,
4.0/). switching the drug from a low to high affinity with the biological target (cis and trans, in

Pharmaceuticals 2023, 16, 682. https://doi.org/10.3390/ph16050682 https://www.mdpi.com/journal/pharmaceuticals


(PD) properties of molecules, switching the drug from a low to high affinity with the bio-
logical target (cis and trans, in case of photoswitches) and allowing molecular activation
in case of PPG. Therefore, light can improve selectivity, being particularly suitable when
the disease is localized, and consequently reduces adverse effects by adjusting its wave-
Pharmaceuticals 2023, 16, 682 2 of 36
length and intensity [4,9,10]. One of the major challenges in photopharmacology, using
short-wavelength light (ultraviolet (UV), below 350 nm), is the limited penetration into
tissue and off-target toxicity due to theand
case of photoswitches) fact that UV
allowing lightactivation
molecular is toxicinto healthy
case cells (Figure
of PPG. Therefore, light
can improve selectivity, being particularly suitable when
1b,c). At a first glance, utilizing UV light may not appear to be a favorable strategy. Nev- the disease is localized, and con-
sequently reduces adverse effects by adjusting its wavelength and intensity [4,9,10]. One
ertheless, the primaryofadvantage of thisinapproach
the major challenges is the mitigation
photopharmacology, of cytotoxicity
using short-wavelength once the
light (ultraviolet
photoswitches lose their (UV),activity
below 350after the
nm), is thetherapeutic effect
limited penetration into(Figure
tissue and1c). A high-precision
off-target toxicity due to
the fact that UV light is toxic to healthy cells (Figure 1b,c). At a first glance, utilizing UV
photopharmacologicallight approach utilizing long-wavelength light (near infrared, 650–900
may not appear to be a favorable strategy. Nevertheless, the primary advantage of
nm) can effectively overcome this approachthe UV
is the light problem.
mitigation This
of cytotoxicity once approach has lose
the photoswitches the their
added ad-
activity
vantage of deeper tissue after penetration
the therapeutic effect (Figure
without 1c). A high-precision
harming healthy photopharmacological
cells, as it specifically approachtar-
utilizing long-wavelength light (near infrared, 650–900 nm) can effectively overcome the
gets only the desired UV treatment areaThis
light problem. (Figure
approach 1d,e). Photoswitches,
has the added advantage of due to their
deeper reversible
tissue penetration
process, can also minimize without and prevent
harming healthyside
cells, effects, such as
as it specifically generalized
targets cytotoxicity
only the desired and
treatment area
(Figure 1d,e). Photoswitches, due to their reversible process, can also minimize and prevent
antimicrobial resistance (Figure 1e).
side effects, such as generalized cytotoxicity and antimicrobial resistance (Figure 1e).

Figure 1. Comparison ofFigure


the 1.principle behind (a) classical chemotherapy, (b) photopharmacology
Comparison of the principle behind (a) classical chemotherapy, (b) photopharmacology
using UV light—PPG, (c) photopharmacology
using using UV and
UV light—PPG, (c) photopharmacology visible
using UV andlight—photoswitches,
visible light—photoswitches,(d) pho-
(d) pho-
topharmacology using IR light—PPG,
topharmacology and
using IR (e) high-precision
light—PPG, photopharmacological
and (e) high-precision photopharmacological chemotherapy
chemotherapy
using IR-light—photoswitches. Red capsule denotes cytotoxic effects and blue capsule isisan
using IR-light—photoswitches. Red capsule denotes cytotoxic effects and blue capsule an inactive
inactive
form and not cytotoxic.
form and not cytotoxic.
Photodynamic therapy (PDT) is a spatiotemporal therapeutic modality approved by
the Food and Drug Administration (USA-FDA) and European Medicines Agency (EMA)
Photodynamic therapy (PDT) is a spatiotemporal therapeutic modality approved by
to treat skin diseases and other organs where light can reach [1,11–16]. It requires the
the Food and Drug Administration
administration of a (USA-FDA) andwhich
photosensitizer (PS), European Medicines
is a non-toxic drug thatAgency (EMA)
is accumulated in
to treat skin diseases and other organs where light can reach [1,11–16]. It requires the ad-
the target cells and activated by adequate visible light irradiation in presence of dioxygen
(O2 ) to produce reactive oxygen species (ROS) that cause cytotoxic effects at the therapeutic
ministration of a photosensitizer (PS), which is a non-toxic drug that is accumulated in the
target. Briefly, the mechanism of action of PDT is based on PS promotion by light absorption
target cells and activated
fromby the adequate
ground statevisible light
(1 PS) to an irradiation
excited state (1 PS*).in presence
1 PS* can returnof to dioxygen (O2)
the ground state
(1 PS) directly through the emission of light (fluorescence) or heat (internal conversation),
to produce reactive oxygen species (ROS) that cause cytotoxic effects at the therapeutic
or indirectly by intersystem crossing, converting to the excited triplet state (3 PS*). In this
target. Briefly, the mechanism of action
state, 3 PS* can of PDT
interact with O2 by istwo
based on PS
different promotion
mechanisms. The bytypelight absorp-
I mechanism
tion from the groundinvolves
state (electron
1PS) totransfer
an excited
to lead state (1PS*). 1PS*
to the production can
of free return
radicals, to the
which ground
interact with
O2 and produce ROS, such as hydrogen peroxide (H2 O2 ), hydroxyl radicals (HO ), and •
state (1PS) directly through the emission of light (fluorescence) or heat (internal conversa-
superoxide radicals (O2 •− ). The type II mechanism involves energy transfer between the
tion), or indirectly by intersystem crossing, converting to the excited triplet state (3PS*). In
this state, 3PS* can interact with O2 by two different mechanisms. The type I mechanism
involves electron transfer to lead to the production of free radicals, which interact with O2

Pharmaceuticals 2023, 16, 682 3 of 36

PS excited and the ground state dioxygen (3 O2 ), producing singlet oxygen (1 O2 ). As 1 O2


is an uncharged molecule, it can diffuse through plasma and biological membranes and
will trigger a chain of successive oxidations of biological molecules (e.g., DNA, lipids, and
proteins) [17–21]. It is believed that the production of ROS through the type II reaction
is predominant in PDT. This photodynamic approach was also used with great success
on the photoinactivation of microorganisms and demonstrates great advantages when
compared with the conventional antimicrobial agents, namely (i) the non-induction of
resistance mechanisms of the microbial cells and (ii) either resistant and sensitive strains
are responsive to the photodynamic treatment and (iii) can be applied to inactivate a range
of microbial entities (bacteria, fungi, viruses, protozoa, parasites [11,12,22–29], among
others). The versatility of PDT is not limited to clinical applications [23,30]. This technique
is valuable for clinical [31–33] and non-clinical applications, such as removing biofilms
from medical devices and implants [34] and environmental water treatments [35–37].
Herein, different classes of photoswitches and photocleavage compounds and their
biological applications will be described. Furthermore, porphyrins will be referred to as suc-
cessful photoactive compounds for use in antimicrobial photodynamic treatment (aPDT), as
well as promising approaches when combined with photoswitches or photocleavage to take
advantage of both photopharmacology and PDT. Finally, examples combining aPDT and
antibiotic therapy will be reported, highlighting this application to overcome resistance.

2. Different Classes of Photoswitches and Some Examples in Antibacterial Applications


Photoswitches are compounds that can be interconverted into two different structural
configuration forms by using light. These compounds are of great importance in several
fields, including molecular electronics, phytopharmacology, and catalysis [38]. In 1937,
Hartley discovered that trans azobenzene could come from the cis isomer by UV-light
and, conversely, by keeping the molecule in the dark, or irradiating it with blue light,
the thermodynamically more stable trans isomer could be recovered [39]. The trans →
cis photoisomerization of azobenzenes can occur by n → π* excitation (lower energy) at
380–500 nm, or π → π* excitation (higher energy) at 280–380 nm [40].
By coupling a photoswitch moiety with a biologically active compound, it is possible
to turn ON (high-affinity form) and OFF (low-affinity form) this bioactive compound.
In fact, azobenzene-based photoswitches are the most widely used due to their efficacy,
reversibility, and repeatability. However, photoswitchable molecules show incomplete
photoisomerization, insufficient light sensitivity for some applications, and difficulties
in fine-tuning to produce a maximal difference between ON and OFF states of the com-
pounds [41].
Ideal photoswitches are molecules that absorb visible light and have a long half-life
time of the metastable isomers [42]. Although UV-light is typically used, studies have
shown that azoarenes can be activated by visible light [42] and near infrared (NIR) light,
which is required to penetrate tissues [39]. Diarylethenes [4], Schiff base-like [43], oxazolone-
like [44], hydantoin-like [45], pyrrolinone-like [46], and stilbene [47] compounds are also
types of photochromic compounds (Figure 2).
Pharmaceuticals 2023,2023,
Pharmaceuticals 16, 682
16, 682 4 of 364 of 35

(a) (b)
R
R
hv1
visible light
N N hv2
UV-Vis light N R R S S R
N S S R
R open closed
E R Z
Diarylethene's derivatives
Azobenzene

O
(c) (d)
O
O
N N hv1
hv1 O
hv2
hv2 N N

Schiff Base-like Oxazolone-like

(e) (f)

N O

O N O hv1
N hv1 N H
O hv2 N
hv2 N H O
O Pyrrolinone-like
Hydantoin-like

(g)

hv1

hv2

E Z

Stilbene
Figure 2.2.Structures
Figure Structuresof
of some photoswitchesand
some photoswitches and their
their photoisomerization
photoisomerization process.
process. hν is hν
theisphoton
the photon
energy (h—Planck’s
energy (h—Planck’s constant; ν—frequency;ν1ν<1 <ν2ν).2).
constant; ν—frequency;

2.1.
2.1. Azobenzenes
Azobenzenes
Azobenzenes are a class of compounds that present an aromatic molecular structure,
Azobenzenes are a class of compounds that present an aromatic molecular structure,
characterized by the presence of sensitivity to external stimuli -N=N- double bond, with the
characterized by the for
appropriate features presence of sensitivity
incorporation into mosttopharmacophores
external stimuli[48,49].
-N=N-Feringa
doubleetbond,
al. [9] with
the appropriate features for incorporation into most pharmacophores [48,49]. Feringa et
al. [9] investigated the possibility of obtaining pharmacological agents by conjugating
photoswitches with ciprofloxacin (Figure 3) using azobenzenes to treat local diseases,
such as a solid tumor or local inflammation. Structure activity relationship (SAR) studies
indicated that the secondary amine in the piperazine ring of ciprofloxacin can be modified
Pharmaceuticals 2023, 16, 682 5 of 36

ceuticals 2023, 16, 682


investigated the possibility of obtaining pharmacological agents by conjugating photo-
switches with ciprofloxacin (Figure 3) using azobenzenes to treat local diseases, such as a
solid tumor or local inflammation. Structure activity relationship (SAR) studies indicated
that the moiety
azobenzene secondary(Figure
amine in3).theThis
piperazine ring of allowed
approach ciprofloxacin
forcan
thebepreparation
modified without
of an ant
loss of activity; therefore, this position was chosen for conjugation with the azobenzene
with photoswitchable
moiety (Figure 3). Thisactivity (azofloxacin).
approach allowed for theBefore irradiation,
preparation azofloxacin
of an antibiotic occurs in
with pho-
as the toswitchable
trans-isomer (compound
activity (azofloxacin).1), butirradiation,
Before after irradiation
azofloxacinwith
occurslight atas365
in 100% the nm, 6
trans-isomer
azofloxacin (compound
converts to the but after irradiation
1), cis-isomer with light2).
(compound at 365
Thenm,non-total
61% of azofloxacin
conversion o
converts to the cis-isomer (compound 2). The non-total conversion of trans azofloxacin to
azofloxacin to cis azofloxacin might be due to the aggregation between azobenzen
cis azofloxacin might be due to the aggregation between azobenzene and molecules in an
molecules in an
aqueous aqueousnot
environment, environment,
allowing completenotphotoisomerization
allowing complete [9]. photoisomerization

Figure Figure
3. Molecular structure
3. Molecular structureof ciprofloxacin
of ciprofloxacin and and its photoswitchable
its photoswitchable analogue, azofloxacin
analogue, azofloxacin.

The bacterial activity of azofloxacin was assessed on Escherichia coli (E. coli) CS1562
The bacterial activity of azofloxacin was assessed on Escherichia coli (E. coli) C
and Micrococcus luteus (M. luteus) ATCC 9341, before and after irradiation with light at
and Micrococcus luteus (M.was
365 nm. Ciprofloxacin luteus)
chosen ATCC 9341, before
as the control. and after
The azofloxacin irradiation
analogue provedwithto light
be more efficient than ciprofloxacin in M. luteus
nm. Ciprofloxacin was chosen as the control. The azofloxacin analogue proved to b
(Table 1), specially before irradiation
(trans-azofloxacin, compound 1), which can be modulated by exposure to light, giving
efficient than ciprofloxacin in M. luteus (Table 1), specially before irradiation
rise to cis-azofloxacin (compound 2). While no significant changes were observed on the
azofloxacin, compound
Gram-negative 1),model
bacterial which canthe
E. coli, beantibacterial
modulated by exposure
activity to light,ongiving
of trans-azofloxacin the rise
azofloxacin (compound
Gram-positive bacterial2). While
model no significant
M. luteus was found to changes
be about were
50 timesobserved
greater thanonthethe Gram
native drug ciprofloxacin.
ative bacterial model E. coli, the antibacterial activity of trans-azofloxacin on the
positive bacterial
Table 1. Minimum model M.concentration
inhibitory luteus was(MIC) found
valuestoofbe about 50
ciprofloxacin andtimes greater than the
their derivatives.
drug ciprofloxacin.
MIC (µM)
Before Irradiation After Irradiation
Compound inhibitory concentration (MIC) values of ciprofloxacin and their derivati
Table 1. Minimum Compound 1 Compound 2
E. coli M. luteus E. coli M. luteus
Azofloxacin 0.50 0.250
MIC0.50
(μM) 0.50
Ciprofloxacin Before
0.0125 Irradiation
12.0 0.0125 After12.0Irradiation
Compound
Compound 1 Compound 2
E. coli M. luteus E. coli M. lute
Azofloxacin 0.50 0.250 0.50 0.50
Ciprofloxacin 0.0125 12.0 0.0125 12.0
Pharmaceuticals 2023, 16, 682 6 of 36

The introduction of the azobenzene moiety into an antibacterial drug allows the light
to spatiotemporally control the antibacterial activity and presents plenty of opportunities to
explore new targets in photopharmacology. According to the authors hypothesis, although
on E. coli the results are not promising, it is still an alternative to treatment, even when used
rmaceuticals 2023, 16, 682 in combination with chemotherapy. Clearly, further studies would be needed to confirm 6 of
this hypothesis.
Wegener et al. [50] reported for the first time a red-shifted responsive azobenzene
photoswitch containing the antibiotic trimethoprim as a core (Figure 4). Trimethoprim
interferes
purines with the biosynthesis
and thymidine of folate
triphosphate byimpaired.
are inhibition ofOf dihydrofolate reductase (DHFR),
note, trimethoprim has a higher
which catalyses the reduction of dihydrofolate to the active
lectivity to bacterial DHRF than mammalian DHFR and it is active against cofactor tetrahydrofolate [51,52].
both Gra
Therefore, the biosynthesis of the amino acids glycine and methionine, as well as purines
positive
and and Gram-negative
thymidine triphosphatebacteria to treat
are impaired. urinary
Of note, and respiratory
trimethoprim tract
has a higher infections. Ne
selectivity
ertheless, with the
to bacterial DHRFemergence of bacterial
than mammalian DHFR resistance,
and it is activeit isagainst
important to develop new th
both Gram-positive
apeutic
andentities, such as
Gram-negative photoresponsive
bacteria to treat urinary analogues,
and respiratory a strategy that can
tract infections. be undertaken
Nevertheless,
with the emergence of bacterial resistance, it is important to develop
modifying the structure of trimethoprim without reducing bacterial DHFR affinity. new therapeutic enti- Bas
ties, such as photoresponsive analogues, a strategy that can be undertaken by modifying
on the aforementioned results, the authors prepared a red-shifted analogue (azobenze
the structure of trimethoprim without reducing bacterial DHFR affinity. Based on the
substituted with fluorine
aforementioned or authors
results, the chlorine at ortho-positions),
prepared a red-shifted analoguewhere the photoisomerizati
(azobenzene substi-
with tuted
visiblewithlight was or
fluorine possible,
chlorine while maintaining
at ortho-positions), the same
where antibacterial properties
the photoisomerization with (F
ure 4). These substituents allowed the effective use of lower-energy n  π* excitation
visible light was possible, while maintaining the same antibacterial properties (Figure 4).
These substituents allowed the effective use of lower-energy n → π* excitation to trigger
trigger trans  cis photoisomerization. The photostationary state (PSS) of tetra-fluo
trans → cis photoisomerization. The photostationary state (PSS) of tetra-fluoro-substituted
substituted
azobenzene azobenzene
was found was found
to have to have
an 89:11 ratio ofan 89:11atratio
cis:trans 527 nmof (visible
cis:trans at 527
light). nm (visib
In the
light).case
Inofthe case of tetra-o-chloroazobenzene, irradiation with
tetra-o-chloroazobenzene, irradiation with 652 nm (near-infrared light) produced652 nm (near-infrar
light)photoisomerization
produced photoisomerization to a PSS of
to a PSS of 87:13 proportion 87:13 proportion of cis:trans.
of cis:trans.

FigureFigure
4. Red-shifted responsive
4. Red-shifted responsive trimethoprim derivatives
trimethoprim derivatives and respective
and respective MIC50 values.
MIC50 values.

The effectiveness of antibacterial activity against E. coli was evaluated before and
ter irradiation. Even though compounds 5 and 6, which contain trimethoprim tetra-o-ch
roazobenzene substitution, exhibited less photoisomerization than compounds 3 and
which contain tetra-o-fluoro-substituted azobenzene, they were still of interest. Co
Pharmaceuticals 2023, 16, 682 7 of 36

The effectiveness of antibacterial activity against E. coli was evaluated before and
after irradiation. Even though compounds 5 and 6, which contain trimethoprim tetra-o-
chloroazobenzene substitution, exhibited less photoisomerization than compounds 3 and 4,
which contain tetra-o-fluoro-substituted azobenzene, they were still of interest. Compound
5 displayed no activity with MIC50 > 80 µM, nevertheless it was responsive to irradiation
(652 nm), giving compound 6, which induced bacteriostasis with MIC50 = 10 µM.
However, tetra-o-fluoro-substituted azobenzene is better at inducing bacteriostasis
with an MIC50 = 5 µM. Furthermore, it was important to study the photoswitching re-
versibility to the trans isomer (inactive) found when blue light at 400 nm was used.
Amidohydrolase enzymes are virulence factors found in various pathogenic bacteria
2 responsible for a great number of hospital-acquired infections and deaths. Weston and
7 of 35
co-workers [53] reported inhibitors of these enzymes using azobenzenes as photoswitches
(Figure 5). Amidohydrolase enzymes are homologous to the human histone deacetylases
(HDACs); therefore, with the knowledge of the pharmacophore of HDAC inhibitors [54–56],
the crystal structure of the enzyme active site of the HDAC family [57–59], and knowing
Pseudomonas aeruginosa (P. aeruginosa),
the bacterial enzymes B/A-HDAH PA1409 and deacetylase-like
(histone PA0321 [58]), and PA-HDAHs
amidohydrolase from Bor-
(acetylpolyamine amidohydrolases
detella/Alcaligenesfrom P. aeruginosa,
[59]), APAHs PA3774),
(acetylpolyamine the authorsfrom
amidohydrolases designed and
Pseudomonas
aeruginosa (P. aeruginosa), PA1409 and PA0321 [58]), and PA-HDAHs
conjugated a pharmacophoric moiety with a zinc chelating group (hydroxamic acid), a (acetylpolyamine
amidohydrolases from P. aeruginosa, PA3774), the authors designed and conjugated a phar-
phenyl capping unit,macophoric
and an azobenzene linker.
moiety with a zinc chelating group (hydroxamic acid), a phenyl capping unit,
and an azobenzene linker.

(b)
O
H
N OH
N
H
O
Vorinostat
Figure 5. (a) Structures of trans
Figure and cis
5. Figure azobenzene
5. (a) HDAH
Structures of trans inhibitors
and cis azobenzeneand (b)inhibitors
HDAH structure andof Vorinostat,
(b) structure of
Vorinostat, inhibitor of
inhibitor of histone deacetylation, 2 h half-life.histone deacetylation, 2 h half-life.

With the photoisomerization of the azobenzene compounds 8 activated by a UV-A


With the photoisomerization
lamp at 365 nm, theof the azobenzene
half-maximal compounds
inhibitory concentration 8 activated
values (IC50 ) againstby
theabacterial
UV-A
enzymes B/A-HDAH, PA-HDAH, and HPAHs were assessed using
lamp at 365 nm, the half-maximal inhibitory concentration values (IC50) against the bacte- SAHA (suberoylanilide
hydroxamic acid, also known as vorinostat, Figure 5b) as a positive control (Table 2). It
rial enzymes B/A-HDAH, PA-HDAH,
should be and HPAHs
noted that incomplete were assessed
photoswitching using
is an issue, and SAHA
about >10% of(suberoy-
the former
lanilide hydroxamic acid,
isomeralso known
was present uponas irradiation.
vorinostat, Figure 5b) as a positive control (Table
2). It should be noted that incomplete photoswitching is an issue, and about >10% of the
former isomer was present upon irradiation.

Table 2. IC50 values for trans and cis azobenzenes against bacterial HDACs.

IC50 (μM)
Pharmaceuticals 2023, 16, 682 8 of 36

Table 2. IC50 values for trans and cis azobenzenes against bacterial HDACs.

IC50 (µM)
Compound R B/A-HDAH PA-HDAH APAH PA0321 APAH PA1409
7a (trans) H 0.042 0.0056 0.79 2.0
8a (cis) H 0.062 0.0066 0.40 0.21
7b (trans) t-But 0.37 0.072 2.2 >25
8b (cis) t-But 0.38 0.094 1.8 3.5
7c (trans) OMe 0.059 0.051 2.3 5.1
8c (cis) OMe 0.075 0.090 1.4 0.65
Vorinostat 0.095 0.037 0.50 0.30

In general, compounds 7a and 7c showed better activity than the positive control
against B/A-HDAH. However, treatment with UV-A light was not successful as the cis-
isomer was more effective than the trans-isomer. Compound 8a revealed a better efficacy
than the positive control against the PA-HDAH and APAHs. Here, it was demonstrated
that the efficacy of the cis-isomer obtained after UV-A light treatment is greater than the
trans-isomer. Compound 7b exhibits a greater steric effect due to the presence of the
tert-butyl group, which leads to an unfavourable steric fit. As a consequence, its inhibitory
activity is weaker than that of compounds 7a and 7c.
Initially, the authors proposed that the cis-isomers would induce more unfavourable
steric interactions, reducing inhibitory activity, so the trans-isomer would be the most
active. However, this was not observed, and the results showed that the most active isomer
depends on the inhibitor substituent and the enzyme (the cis-isomers were found to be
better in APAHs).
Gramicidin S is a cyclic peptide known for its antibacterial activity [60]; therefore, Yuan
et al. [61] developed an approach based on an azobenzene moiety containing gramicidin S
to modulate, reversibly, the antibacterial activity (Figure 6).
Peptide 9 was designed by replacing proline and D-phenylalanine amino acid residues
with an azobenzene moiety, maintaining the amphiphilic side chain for biological activity.
Peptide 10a was obtained by removing the leucine residue in peptide 9. Arginine (basic
amino acid residue) and glutamic acid (acidic amino acid residue) were replaced in peptide
10a, to produce peptides 10b and 10c, respectively, aiming to investigate possible differences
in the antibacterial activity due to the positively or negatively charged amino acid residues.
To study the amphiphilicity on the antibacterial activity, basic amino acid residues were
incorporated into the peptide backbone by replacing arginine with leucine and valine
residues (peptides 11a and 11b). When cis-isomers of peptides were irradiated with blue
light (405 nm), trans-isomers are obtained. On the other hand, the trans-enriched PSS
of 9–11 were irradiated by UV-A light of 352 nm for 2 h to convert to the respective cis-
enriched PSS.
The MIC values of gramicidin S and its peptides were determined against Staphylococ-
cus aureus (S. aureus) ATCC 49775 (Table 3).
Peptide 9 only has two intramolecular strong hydrogen bonds linking the upper and
lower strand, showing a distorted β-strand secondary structure, proved in a molecular
modelling study. Gramicidin S has four strong hydrogen bonds, while the cis-isomers of
peptides 10 and 11 have three strong hydrogen bonds, causing a well-defined secondary
structure. The trans-isomers of peptides 10 and 11 lack intramolecular hydrogen bonding
and, therefore, show a weak secondary structure. The cis-isomers of peptides 10a and 10b
proved to be more efficient than trans-isomers, with cis-enriched 10b exhibiting the highest
antibacterial potential in this study. Similar to peptide 10c, peptides 11a and 11b were
inactive, which was attributed to the replacement of hydrophobic residues (valine and
leucine), which may disrupt the amphiphilic nature of peptides, decreasing penetration
through the bacterial membrane. In this study, the use of light was not essential to activate
the ON effect, since the trans-isomer was not more efficient than the cis-isomer. However,
light would be a good strategy to generate an OFF effect and, therefore, reduce side effects.
depends on the inhibitor substituent and the enzyme (the cis-isomers were found to be
better in APAHs).
Gramicidin S is a cyclic peptide known for its antibacterial activity [60]; therefore,
Pharmaceuticals 2023, 16, 682 9 of 36
Yuan et al. [61] developed an approach based on an azobenzene moiety containing gram-
icidin S to modulate, reversibly, the antibacterial activity (Figure 6).

Figure 6. Structure of Gramicidin S and its modified peptides (compounds 9–11) containing
Figure 6. Structure of Gramicidin S and its modified peptides (compounds 9–11) containing azoben-
azobenzene units [61].
zene units [61].
Pharmaceuticals 2023, 16, 682 10 of 36

Table 3. MIC values of Gramicidin S and its mimetics peptides against S. aureus ATCC 49775.

MIC (µg mL−1 )


Compound cis trans
9 64 64
10a 64 256
10b 32 128
10c >256 >256
11a 256 >256
11b >256 >256
Gramicidin S 2

2.2. Diarylethenes
Similar to azobenzenes, diarylethenes have an aromatic molecular structure that
allows perfect incorporation into most pharmacophores. Thus, diarylethenes have been
studied and applied as photoswitch molecules that can be interconverted by modulation
between open and closed forms [62,63]. Li et al. [64] developed an approach of molecular
hybridization by introducing a diarylethene moiety into fluoroquinolone (norfloxacin,
R2 = ethyl and ciprofloxacin, R2 = cyclopropyl) to obtain switchable antibacterial agents
2 (Figure 7). As mentioned before, SAR studies showed that the secondary amine in the 10
piperazine ring of fluoroquinolones can be modified without loss of antibacterial activity,
so this position was chosen for conjugation with a diarylethene moiety.

Figure 7. Diarylethenes based


Figure on fluoroquinolone
7. Diarylethenes antibiotics
based on fluoroquinolone antibiotics[64].
[64].

The antibacterial activity of the diarylethene derivative was assessed before and a
irradiation with UV-C light (254 nm) by performing MIC values on E. coli and S. au
(Table 4).

Table 4. MIC values of diarylethene-based switchable antibacterial agents before and after U
Pharmaceuticals 2023, 16, 682 11 of 36

The antibacterial activity of the diarylethene derivative was assessed before and after
irradiation with UV-C light (254 nm) by performing MIC values on E. coli and S. aureus
(Table 4).

Table 4. MIC values of diarylethene-based switchable antibacterial agents before and after UV-C
(254 nm) light irradiation.

MIC (µg mL−1 )


Compound Open-Isomer (before) Closed-Isomer (after)
E. coli S. aureus E. coli S. aureus
12a 32 8 16 8
12b 32 16 2 16
13a 16 32 16 32
13b 16 32 16 32
14a 32 16 8 16
14b 32 16 2 16
15a 32 16 16 16
15b 16 16 4 16
Norfloxacin 0.125 0.125 0.125 0.125
Ciprofloxacin 0.125 0.125 0.125 0.125

All the open-isomers showed a lower antibacterial activity on E. coli (MIC = 16–32 µg
mL−1 ) than closed-isomers. A similar activity against S. aureus (MIC = 8–32 µg mL−1 ) is
shown in both the open and closed forms. However, all of the compounds were less active
than the native drugs, norfloxacin and ciprofloxacin (MIC = 0.125 µg mL−1 ). The activity
decrease can be related to the addition of the switchable moiety. Particularly, compounds
12b and 14b showed a great difference in the antibacterial activity against E. coli before
and after irradiation, with the closed form being 16-fold more efficient than the open form.
The ring-open form in diarylethenes shows a flexible conformation due to free rotation
around the C–C bonds joining the thiophene to the central cyclopentene ring, consequently
adopting various geometries. This flexible conformation was hypothesized not to allow the
formation of stable complexes with DNA gyrase, decreasing the open-isomer antibacterial
activity. Furthermore, the closed-form of compounds 12b and 14b appear to have a rigid
conformation, which allows the formation of stable complexes with the DNA gyrase of
E. coli and, consequently, derivatives were shown to increase the antibacterial activity up to
16-fold. Despite 12b and 14b being less effective than the parent drug ciprofloxacin, they
showed the potential to be used as novel switchable antibacterial agents [64].

2.3. PHOTACs
PROTACs (PROteolysis TArgeting Chimeras) are bifunctional small molecules that
have been extensively studied as a novel method to treat diseases that result from the
aberrant expression of specific disease-associated proteins [65]. PROTACs can degrade
these proteins selectively by ubiquitination. This approach is promising for the treatment of
cancer, neurodegenerative diseases, inflammatory diseases, and viral infections. By the com-
bination of PROTACs with photopharmacology, a new approach arises—PHotoswitchable
proteolysis TArgeting Chimeras (PHOTACs), which are bifunctional small molecules that
target a protein of interest (POI) for ubiquitylation by an E3 ubiquitin ligase complex,
promoting proteasome degradation (Figure 8). This approach allows the change in the
reversibly endogenous protein levels, being helpful for potential treatments of diseases,
such as cancer [66] and eventually Alzheimer’s disease [67]. Several studies [68–70] have
already designed and synthesized PHOTACs by incorporating photoswitches, such as
azobenzenes, into PROTACs.
Pfaff et al. [70] were the first to report a novel approach based on PROTACs, including
o-F4 -azobenzene linkers. AVR-771 is a PROTAC that degrades BRD2 and BRD4 (BET
proteins), so it has (+)-JQ1 as a POI ligand and VHL (Von Hippel-Lindau) as a ligand
2.3. PHOTACs
PROTACs (PROteolysis TArgeting Chimeras) are bifunctional small molecules that
have been extensively studied as a novel method to treat diseases that result from the
aberrant expression of specific disease-associated proteins [65]. PROTACs can degrade
Pharmaceuticals 2023, 16, 682 12 of 36
these proteins selectively by ubiquitination. This approach is promising for the treatment
of cancer, neurodegenerative diseases, inflammatory diseases, and viral infections. By the
combination of PROTACs with photopharmacology, a new approach arises—PHo-
of E3 ubiquitin ligase (Figure 9a). Based on AVR-771, a novel PHOTAC was designed
toswitchable proteolysis TArgeting Chimeras (PHOTACs), which are bifunctional small
(Figure 9b). The biological activity of PHOTACs was tested in Ramos cells and proved to be
molecules that target a protein of interest (POI) for ubiquitylation by an E3 ubiquitin ligase
promising. While the trans-isomer was active in decreasing BRD2 levels, the cis-isomer did
complex, promoting proteasome
not induce protein degradation
degradation, (Figure
hypothesized due8).
toThis approach
the shorter allows
distance the change
between BRD2
in the and
reversibly endogenous protein levels, being helpful for potential treatments
VHL ligands because of the ‘’compact form” of the cis-isomer (Figure 10). Despiteof dis-
the
eases,ARV-771
such as cancer
PROTAC [66]being
and able
eventually Alzheimer’s
to degrade both BRD2disease [67]. Several
and BRD4, studies
degradation [68–70]
of BRD4 was
have already designed
not observed and synthesizedThis
for photoPROTAC. PHOTACs by incorporating
is hypothesized due to thephotoswitches,
reversed amidesuch
bond
as azobenzenes, into PROTACs.
between (+)-JQ-1 and the o-F4 -azobenzene moiety.

FigureFigure
8. Schematic of theof
8. Schematic working model
the working of a PHOTAC.
model TheThe
of a PHOTAC. molecules toggle
molecules between
toggle betweenanan
inactive
inactive
form and an active form upon irradiation. Red pentagon does not fit in E3 ubiquitin ligase complex
form and an active form upon irradiation. Red pentagon does not fit in E3 ubiquitin ligase complex
Pharmaceuticals 2023, 16, 682until irradiation occurs. There, the conformation changes, the ligand fits, and the ubiquitylation
12 of 35
until irradiation occurs. There, the conformation changes, the ligand fits, and the ubiquitylation
process occurs. Adapted with permission from [66]. Copyright, 2020, Elsevier.
process occurs. Adapted with permission from [66]. Copyright, 2020, Elsevier.

(a)
Pfaff et al. [70] were the first to report a novel approach based on PROTACs, includ-
ing o-F4-azobenzene linkers. AVR-771 N is aS PROTAC that degrades BRD2 and BRD4 (BET
HO N
proteins), so it has (+)-JQ1 as a POI N ligand and VHL (Von Hippel-Lindau) as a ligand of
O O
E3 ubiquitin ligase
O
(Figure
O
9a). Based on AVR-771, a novel PHOTAC was designed (Fig-
N
ure 9b).NThe biological N of PHOTACs
activity N was tested in Ramos cells and proved to be
H H
S N O
H
promising. While the trans-isomer was active in decreasing BRD2 levels, the cis-isomer
O
N did not induce protein degradation, hypothesized Cl due to the shorter distance between
BRD2 and VHL ligands because of the ‘’compact (+)-JQ1 form’’ of the cis-isomer (Figure 10). De-
VHL
spite the ARV-771 PROTAC being able to degrade both BRD2 and BRD4, degradation of
BRD4 was not observedAVR-771for photoPROTAC. This is hypothesized due to the reversed am-

ide bond between (+)-JQ-1 and the o-F4-azobenzene moiety.


(b)
F
N N F

F N
HO O O F N
F NH N S
N N F N S NH
H O
N N N 530 nm
O N N
H O
N O N
H F N 415 nm O
S F N
O HO
NH
N Cl
Cl
VHL
(+)-JQ1 cis
trans 17
16 S
N

Figure 9. (a) structure of PROTAC AVR-771 and (b) schematic representation of photoisomerization
Figure 9. Figure 9. (a) structure of PROTAC AVR-771 and (b) schematic representation of pho-
between trans-isomer (active, compound 16) and cis-isomer (inactive, compound 17) [70].
toisomerization between trans-isomer (active, compound 16) and cis-isomer (inactive, compound
17) [70].
Reynders et al. [68] also reported the photopharmacological application to target pro-
tein degradation by incorporating azobenzenes into PROTACs, promoting light-depend-
ent proteolysis. In the dark, the molecules do not have proteolytic activity, but upon irra-
diation with blue-violet light (380–440 nm), they were found to be activated. The azoben-
zene moiety was introduced on the phthalimide-conjugated ligand to bind cereblon E3
ubiquitin ligase complex to allow the affinity change. Based on the previously reported
PROTAC dBET1 (Figure 10a) [71], a POI ligand, (+)-JQ1, was chosen for the design of the
compound 18 series. (+)-JQ1 is an inhibitor of BET proteins BRD2-4. For the design of the
Pharmaceuticals 2023, 16, 682 13 of 36
Pharmaceuticals 2023, 16, 682 13 of 35

Figure 10. Structures of PHOTACs 18 and 19 series.


Figure 10. Structures of PHOTACs 18 and 19 series.
Pharmaceuticals 2023, 16, 682 14 of 36

Reynders et al. [68] also reported the photopharmacological application to target pro-
tein degradation by incorporating azobenzenes into PROTACs, promoting light-dependent
proteolysis. In the dark, the molecules do not have proteolytic activity, but upon irradiation
with blue-violet light (380–440 nm), they were found to be activated. The azobenzene
moiety was introduced on the phthalimide-conjugated ligand to bind cereblon E3 ubiquitin
ligase complex to allow the affinity change. Based on the previously reported PROTAC
dBET1 (Figure 10a) [71], a POI ligand, (+)-JQ1, was chosen for the design of the compound
18 series. (+)-JQ1 is an inhibitor of BET proteins BRD2-4. For the design of the compound
19 series, a synthetic ligand of FK506-binding protein (SLF) was chosen as the POI ligand
based on PROTAC dFKBP-1 (Figure 10b), reported previously. The SLF ligand is a syn-
thetic ligand for FKBP inhibitors. Figure 10 summarizes the small library of PHOTACs
synthesized in this work.
The compound 18 series was evaluated on the viability of RS4;11 lymphoblast leukemic
cells, in which compound 18c (Figure 11c), with a 1,4diaminobuthyl spacer, proved to be
the most effective. Therefore, BET protein (BRD2-4) levels were analysed in the presence of
compound 18c. A decrease in BRD4, BRD3, and BRD2 levels upon compound 18c irradi-
ation (390 nm) was observed, but not in the dark. A light dependent BRD4 degradation
was also demonstrated in breast cancer cell lines (MBMDA231 and MBMDA468) using
compound 18c. Upon irradiation with UV-A light (390 nm), trans-18c was triggered to cis,
obtaining a PSS of >90%. When the cis-isomer was irradiated with green light (500 nm),
a PSS of >70% trans was obtained (Figure 11c). The compound 19 series was tested on
FKBP12 levels; however, a slight activity in the dark was observed.
Jin et al. [69] designed PHOTACs (Figure 12a) to knockdown the BCR-ABL fusion
protein to interact with E3 ubiquitin ligase cereblon (CRBN) using desatinib, a second-
generation tyrosine kinase inhibitor, to target BCR-ABL. The compounds were assessed in
a myelogenous leukaemia cell line K-562 and compound 20c proved to be the most active
in reducing BCR-ABL levels selectively by UV irradiation (Figure 12b). The cis-isomer did
not induce protein degradation.
In general, contrasting with the examples of antibacterial compounds that incorporate
azobenzenes (compounds 1–2 and 7–11), light does not have an activating effect in some
of them. However, in the case of PHOTACs, light triggers a protein destruction reaction.
Nevertheless, it is still possible to reverse the process if it is desired to stop such a reaction.
Although this is a promising strategy, it is still preliminary and has only been tested in
cancer cells.
irradiation (390 nm) was observed, but not in the dark. A light dependent BRD4 degra
tion was also demonstrated in breast cancer cell lines (MBMDA231 and MBMDA468)
ing compound 18c. Upon irradiation with UV-A light (390 nm), trans-18c was triggered
cis, obtaining a PSS of >90%. When the cis-isomer was irradiated with green light (500 n
Pharmaceuticals 2023, 16, 682
a PSS of >70% trans was obtained (Figure 11c). The compound 19 series15was of 36
tested
FKBP12 levels; however, a slight activity in the dark was observed.

Figure 11. Structure


Figure ofofPROTACs
11. Structure (a)bBET1
PROTACs (a) bBET1andand (b) dFKBP-1.
(b) dFKBP-1. (c) Photoswitching
(c) Photoswitching of 18c.
of compound compound
18c.

Jin et al. [69] designed PHOTACs (Figure 12a) to knockdown the BCR-ABL fus
protein to interact with E3 ubiquitin ligase cereblon (CRBN) using desatinib, a seco
generation tyrosine kinase inhibitor, to target BCR-ABL. The compounds were asses
in a myelogenous leukaemia cell line K-562 and compound 20c proved to be the m
Pharmaceuticals 2023, 16, 682 15 of 35

Pharmaceuticals 2023, 16, 682 16 of 36


active in reducing BCR-ABL levels selectively by UV irradiation (Figure 12b). The cis-iso-
mer did not induce protein degradation.

2023, 16, 682 16 of 35

Figure 12. (a) Structures of PHOTACs synthesized and (b) schematic representation of PHOTAC
3. Different Classes
Figureof (a) Structures of PHOTACs
12.Photocleavage and synthesized and (b) schematic representation of PHOTAC 20c
20c (n = 3) and its isomerization [69].Some Examples in Antibacterial
Applications (n = 3) and its isomerization [69].
In general, contrasting with the examples of antibacterial compounds that incorpo-
3. Different
Photocleavage Classes of Photocleavage and Someas Examples in Antibacterial
drugApplications
ratecontrol is another
azobenzenes (compounds category classified
1–2 and light-controlled
7–11), light does not have an activating activity
effect in
[72]. In photocleavage,
some ofthe irradiation
Photocleavage
them. control
However, intriggers
the case ofthe
is another cleavage
category
PHOTACs, oftriggers
a covalent
classified
light bond
as alight-controlled
protein between
drug
destruction re-activ-
ity [72]. In photocleavage,
Nevertheless, it is the
still irradiation
possible to triggers
reverse the the cleavage
the drug and a light-responsive moiety that is responsible for modulating the drug’s ac-
action. process if it of
is a covalent
desired to bond
stop between
such a
the drug and
reaction. a light-responsive
Although this is a moiety strategy,
promising that is responsible
it is still for modulating
preliminary and theonly
has drug’s activity
been
tivity (Figure 13).
(Figure
tested 13).
in cancer cells.

Figure 13. Representation


Figure 13.of photocleavage
Representation control. Adapted
of photocleavage control.from Friedman
Adapted [72]. [72].
from Friedman

Photocleavage reactions need photoremovable protecting groups (PPG). PPG are


small light-responsive units that can be covalently linked to the bioactive molecule. By
light irradiation, PPG releases, irreversibly, the molecule in its active form [7]. Six classes
Pharmaceuticals 2023, 16, 682 17 of 36
Figure 13. Representation of photocleavage control. Adapted from Friedman [72].

Photocleavage reactions need photoremovable protecting groups (PPG). PPG are


Photocleavage reactions need photoremovable protecting groups (PPG). PPG are small
small light-responsive units that can be covalently linked to the bioactive molecule. By
light-responsive units that can be covalently linked to the bioactive molecule. By light
light irradiation,
irradiation, PPG releases,
PPG releases, irreversibly,
irreversibly, the molecule
the molecule in form
in its active its active form
[7]. Six [7]. Six
classes of classes
of PPGs
PPGs havehave already
already been investigated,
been investigated, including o-nitrobenzyl
including o-nitrobenzyl groups, coumarin-4
groups, coumarin-4-ylmethyl
ylmethyl
groups, groups, groups,
arylmethyl arylmethyl groups, arylcarbonylmethyl
arylcarbonylmethyl groups, boron-dipyrromethene
groups, boron-dipyrromethene (BODIPY),
and Ru complexes (Figure 14).
(BODIPY), and Ru complexes (Figure 14).

Figure14.
Figure 14.Different
Different types
types of PPGs.
of PPGs. X unit
X unit means
means leaving
leaving groupsgroups released
released by photocleavage
by photocleavage [7]. [7].

As
Asmentioned
mentionedabove,
above, forfor
light-controlled
light-controlleddrugdrug
delivery, it is important
delivery, to achieve
it is important to achieve
photocleavage groups at long-wavelength light (between 650 nm and 900 nm),
photocleavage groups at long-wavelength light (between 650 nm and 900 nm), such as red such as red
light or near infra-red light (NIR), for clinical use [73]. Therefore, BODIPY [74] and Ru
light or near infra-red light (NIR), for clinical use [73]. Therefore, BODIPY [74] and Ru
complexes [74] have attracted research for having long-wavelength light PPGs and their
complexes [74] have attracted research for having long-wavelength light PPGs and their
photolysis process occurring under visible or NIR light irradiation. However, BODIPY
photolysis have
derivatives process occurring
shown under
cytotoxic visible
effects due toor1 O
NIR light irradiation. However, BODIPY
2 generation, but there are studies
in which after irradiation, this cytotoxicity has been applied to exert a synergistic effect
combined with chemotherapy drugs [75].
The use of PPGs has the greatest interest in some biomedical applications, such as
to treat of skin infections, cancer, neuronal and antibacterial diseases, and other cellular
processes [76].

3.1. BODIPY
Kumari and co-workers [77] designed a BODIPY complexed with levofloxacin (BODIPY-
Levo) to deliver fluoroquinolone upon visible blue light (470 nm) irradiation (Figure 15).
treat of skin infections, cancer, neuronal and antibacterial diseases, and other cellular pro-
cesses [76].

3.1. BODIPY
Kumari and co-workers [77] designed a BODIPY complexed with levofloxacin (BOD-
Pharmaceuticals 2023, 16, 682 18 of 36
IPY-Levo) to deliver fluoroquinolone upon visible blue light (470 nm) irradiation (Figure
15).

Figure 15.
Figure 15. Mechanism
Mechanism proposed
proposed for
for generation
generation of
of levofloxacin
levofloxacin after
after photoirradiation
photoirradiation with
with blue
blue light
light
(470 nm).
(470 nm).

The antimicrobial
The antimicrobial activity
activity was
was assessed
assessed against
against E. E. coli
coli and
and S.
S. aureus
aureus bacterial
bacterial strains.
strains.
Before irradiation,
Before irradiation,compound
compound21 21 did
did not
not contribute
contribute to to the
the inhibitory
inhibitory effects
effects towards
towards E. E. coli
coli
and S.
and S. aureus.
aureus. Upon
Upon light
light conditions
conditions (470
(470 nm),
nm), after
after releasing
releasing levoflaxacin
levoflaxacin (5.0
(5.0 µM),
μM), the
the
optical density measurement at 600 nm
optical density measurement at 600 nm (OD600 (OD 600 ) decreased significantly in comparison
in comparison to to
the compound 21 (p-value
the compound 21 (p-value < < 0.001), corroborating the rationale of the design
corroborating the rationale of the design [77]. [77].

3.2.
3.2. o-Nitrobenzyl
o-Nitrobenzyl Derivatives
Derivatives
Shchelik
Shchelik et al.[78]
et al. investigated
[78] investigated twotwo
‘’caged” antibiotics
‘’caged’’ (vancomycin
antibiotics and cephalosporin,
(vancomycin and cephalo-
both of which inhibit bacterial cell wall biosynthesis), where the
sporin, both of which inhibit bacterial cell wall biosynthesis), where the drug drug was planned to be
was planned
photoreleased upon UV
to be photoreleased uponlight
UVexposure (365 nm,
light exposure 5 min),
(365 min), o-nitrobenzyl
nm, 5using as a PPG
using o-nitrobenzyl as group
a PPG
and
grouppolyethylene glycol (PEG)
and polyethylene glycolas(PEG)
a linker
as a(Table
linker5).(Table
PEGylation was an approach
5). PEGylation to allow
was an approach
steric hindrance,
to allow which could
steric hindrance, suppress
which couldvancomycin’s activity and
suppress vancomycin’s prevent
activity theprevent
and bindingtheto
the amino acid residues of the peptide chain during cell wall synthesis.
binding to the amino acid residues of the peptide chain during cell wall synthesis. Ceph- Cephalosporin
PEGylation was expected
alosporin PEGylation was to preventtobinding
expected preventtobinding
penicillin-binding protein or
to penicillin-binding prevent
protein or
transport issues.
prevent transport issues.
The antibacterial activity was evaluated against Bacillus subtilis (B. subtilis) ATCC 6633,
S. aureus ATCC 29,213, MRSA, E. coli ATCC 25922, and P. aeruginosa ATCC 27853, with MIC
values calculated (Table 6).
Compound 22 containing PEG did not show significant activity. However, compound
24 (without PEG) revealed excellent activity against B. subtilis and S. aureus, as well as
vancomycin (compound 23). In the case of the cephalosporin series, contrary to expec-
tations, PEG decreased the antibiotic activity (compound 25). Compound 27 exhibited
better activity than cephalosporin against all the strains except P. aeruginosa, but the activity
was revealed to be more efficient in Gram-negative strains, which could be explained by
the thiadiazole cephalosporin side chain and its zwitterionic properties that allow higher
penetration through the outer membrane (present only on Gram-negative strains). The PEG
linker decreased the antibacterial activity against all the tested strains, both in vancomycin
and cephalosporin series.
Wong and co-workers [79] reported a delivery mechanism targeted to Gram-negative
bacteria based on ciprofloxacin photoreleased by a cell wall-targeted dendrimer nanoconju-
gate (Figure 16a). The delivery system is a dendrimer conjugated with polymyxin B (PMB)
or ethanolamine (EA) that acts as a carrier for ciprofloxacin (inhibitor of DNA gyrase),
using the o-nitrobenzyl as PPG (ONB-cipro). When PMB or EA binds to lipopolysaccharide
(LPS), 80% of ciprofloxacin is photoreleased from ONB-cipro by irradiating UV-A light
(365 nm) for 30 min. The antibacterial activity of the conjugates (29 and 30, Figure 16b) was
evaluated and MIC50 values were 2.0 µM (in E. coli) approximately, which may be due to
an incomplete drug release. The MIC50 value of ciprofloxacin is 0.0125 µM. However, the
Pharmaceuticals 2023, 16, 682 19 of 36

aceuticalsPharmaceuticals
aceuticals 682 2023,
2023, 16, 682
Pharmaceuticals
2023, 2023, 16,
16, 682
682 18 of
of 35
3
2023, 16,
aceuticalsPharmaceuticals
16, 682 2023, 16, 682 18
molecules did not show phototoxicity in human cells and exhibited good selectivity for 18 of 35
E. coli.

Table 5.Table 5. Vancomycin


Table
Vancomycin (22,23,
5. Vancomycin
(22,23, and and andcephalosporin
24)and
(22,23,
24) cephalosporin
and (26, and and
27, 27,
24) and cephalosporin
(26, 28) derivatives.
(26,derivatives.
28) 27, and 28) derivatives.
Table
Table 5.
5. Vancomycin (22,23,
(22,23, and
and 24)
Table 5. Vancomycin
Vancomycin and
and cephalosporin
(22,23,
24) (26,
(26, 27,
27, and
and 24) and cephalosporin
cephalosporin and 28)(26,derivatives.
28) 27, and 28) derivatives.
derivatives.
Vancomycin Derivatives
Vancomycin
Vancomycin Derivatives Derivatives Cephalosporin Derivatives
Cephalosporin
Cephalosporin DerivativesDerivatives
Vancomycin
Vancomycin Derivatives
Vancomycin
DerivativesDerivatives Cephalosporin Derivatives
Cephalosporin
Cephalosporin DerivativesDerivatives

Target
Target Target
Target
Target Target Compounds
mpounds
mpoundsCompounds
mpoundsCompounds

Release
Release Release
Release
Release Release Compounds
mpounds
mpounds Compounds
mpounds Compounds

Control
Control Control
Control
Control Control
mpounds Compounds
Compounds
mpounds
mpoundsCompounds

The The antibacterial


The antibacterial
antibacterial activity wasactivity
activity was evaluated
evaluatedwas against
evaluated
against against
Bacillus
Bacillus Bacillus
subtilis
subtilis (B. subtilis (B.
(B. subtilis) sub
ATCC
The
Table 6. MICThe
antibacterial antibacterial
values activity
(µg was
mL−1 ) of activity
evaluated
vancomycin wascephalosporin
and evaluated
against against
Bacillus subtilis
derivatives. (B. subtilis)
Bacillus subtilis) ATCC
subtilis (B. sub
ATCC
6633, 6633,
6633, S. aureus
aureus S. aureus
ATCC ATCC
29,213, MRSA, 29,213, MRSA,
E. coli ATCC E. 25922,
coli ATCC
and 25922, and P.ATCC
P. aeruginosa aeruginosa A
27853
6633, S.
S. aureus ATCC
6633,
ATCC 29,213,
S. aureus ATCC
29,213, MRSA,
MRSA, E.
E. coli
29,213, ATCC
MRSA,
coli ATCC E. 25922, and
and P.
coli ATCC
25922, P. aeruginosa
25922, and P.ATCC
aeruginosa 27853
aeruginosa
ATCC A
27853
Strainswith with MIC
with MIC values
values values(Table
calculated calculated
6). (Table 6).
MIC (µg − 1
with MIC calculated
with MIC
MIC values values(Table
calculated 6).
calculated
(Table 6). (Table 6).mL )
Vancomycin Series Cephalosporin Series
Table MICTable
Table 6. MIC 6.22(μg
values MICmL
values
−1) of23(μg mL−1
−1) of24
vancomycin vancomycin25and cephalosporin
and cephalosporin derivatives.
26 derivatives.
27
Table 6. values
6.(μgMICmL −1)) of
of vancomycin and
and cephalosporin derivatives.
−1
6. MICTable
values (μg values
mL (μg mL−1) of vancomycin
vancomycin and cephalosporin
cephalosporin derivatives. derivatives.
E. coli ATCC 25922 - - - 8 1–2 1
StrainsATCCStrains
Strains
P. aeruginosa 27853 - - - MIC (μg
MIC (μg
64 mL
mLMIC
−1) (μg mL−1
−1 ) 2–4 −1)
32
Strains Strains MIC (μg mL )MIC
−1 (μg mL −1 )
B. subtilis ATCC 6633 32 0.06–0.125
Vancomycin 0.125
Vancomycin
Series 8
Series 2–4
Cephalosporin 1 Series
Cephalosporin
S. aureus ATCC 6633 >64 Vancomycin
Vancomycin
0.5–1 Series
Vancomycin
Series
0.5 Series
32 Cephalosporin
Cephalosporin
8 4 Series
Cephalosporin
Series
S. aureus ATCC 43300 >64 22
22 2223
1–2 22 23 1 2324
24 64 24 32
25
25 25 16
26
26 26
27
27
22 23 23
24 24 25 25 26 26 27
E. coli ATCC
E. ATCC E.25922
coli ATCC 25922 --- - -- - -- 888-- 1–28 1–2111
E. coli
coli ATCC E.25922
coli ATCC 25922
25922 -- - - 1–2
1–28 1–2
P. aeruginosa
P. P.ATCC
aeruginosa ATCC 27853
27853 --- - -- - - 64 - 64
2–4 2–4
32
P. aeruginosa
aeruginosaP.ATCC
ATCC 27853
aeruginosa ATCC 27853
27853 -- - -- 64
64 - 2–4
64
2–4 32
2–4
32
B. B. subtilis
B. subtilis ATCC
ATCC 6633ATCC 6633 32 32
0.06–0.125 0.06–0.125
0.125 0.125
888 2–48 2–4111
B. subtilis
subtilis ATCC 6633
B. subtilis
6633ATCC 6633 32
32 0.06–0.125
32
0.06–0.125 0.125
0.06–0.125
0.125 0.125 2–4
2–48 2–4
S. aureus ATCC
S. S. aureus
ATCC 6633ATCC 6633 >64 >64
0.5–1 0.5–1
0.5 0.5
32 32
888 84
S. aureus
aureus ATCC 6633
S. aureus
6633ATCC 6633 >64
>64 0.5–1
>64
0.5–1 0.5
0.5–1
0.5 32
0.5
32 32 844
S. aureus ATCC
S. S. aureus
ATCC 43300ATCC 43300 >64 >64
1–2 1–21 64 1 64
32 32
16
S. aureus
aureus ATCC 43300
S. aureus
43300ATCC 43300 >64
>64 1–2
>64
1–2 1–211 64
64 1 32
64
32 16
32
16
CompoundCompound
Compound 22 22 containing
22 containing PEG did notPEG
not did significant
show not show significant activity. com
activity. However, How
22 containing
CompoundCompound PEG
PEG did
22 containing
containing PEG
did not show
did significant
show not show activity.
significant activity. However,
significant activity. com-
However, How
com
saccharide (LPS), 80% of ciprofloxacin is photoreleased from ONB-cipro by irr
UV-A light (365 nm) for 30 min. The antibacterial activity of the conjugates (29
Figure 16b) was evaluated and MIC50 values were 2.0 μM (in E. coli) approximately
may be due to an incomplete drug release. The MIC50 value of ciprofloxacin is 0.0
Pharmaceuticals 2023, 16, 682 However, the molecules did not show phototoxicity in human cells 20and of 36exhibit

selectivity for E. coli.

.
Figure 16. (a) Scheme for light-controlled targeted delivery of photocaged ciprofloxacin carried by
lipopolysaccharide (LPS) adsorbed to the outer membrane of the Gram-negative bacterium (E. coli)
cell wall. Internalized ciprofloxacin inhibits DNA gyrase. (b) Structures of ciprofloxacin, photocaged
ciprofloxacin (28), dendrimers conjugated with ONB-ciprofloxacin and cell wall targeting ligand—EA
(29), or PMB (30). Adapted with permission from [79]. Copyright, 1996, Royal Society of Chemistry.
lipopolysaccharide (LPS) adsorbed to the outer membrane of the Gram-negative bacterium (E. coli)
cell wall. Internalized ciprofloxacin inhibits DNA gyrase. (b) Structures of ciprofloxacin, photo-
caged ciprofloxacin (28), dendrimers conjugated with ONB-ciprofloxacin and cell wall targeting lig-
and—EA (29), or PMB (30). Adapted with permission from [79]. Copyright, 1996, Royal Society of
Pharmaceuticals 2023, 16, 682
Chemistry. 21 of 36

3.3. Photocaged PROTACs


3.3.As in the case
Photocaged of photoswitches, photocaged PROTACs are an approach to photo-
PROTACs
cleavage in which E3 ubiquitin ligase (a linker with a photocleavage moiety) and the POI,
As in the case of photoswitches, photocaged PROTACs are an approach to photocleav-
form
age ainternary
which E3complex to ligase
ubiquitin lead proteasomal
(a linker withdegradation [80] moiety)
a photocleavage (Figure and
17). the
However, pho-
POI, form
tocaged PROTACs are irreversible, unlike photoswitchable PROTACs, which are
a ternary complex to lead proteasomal degradation [80] (Figure 17). However, photocaged reversi-
ble. Several are
PROTACs researchers reported
irreversible, unlike photocaged PROTACs
photoswitchable PROTACs,to induce
which protein degradation
are reversible. Severalby
light.
researchers reported photocaged PROTACs to induce protein degradation by light.

Figure 17. Schematic of the working model of a photocaged PROTAC. The caging moiety attached
Figure 17. Schematic of the working model of a photocaged PROTAC. The caging moiety attached
to either end of the PROTAC is removed upon irradiation. The red geometric figures are in the
to either end of the PROTAC is removed upon irradiation. The red geometric figures are in the
presence of the PPG groups (pink and orange figures) that prevent the structure from binding to the
presence of the PPG groups (pink and orange figures) that prevent the structure from binding to the
E3 ubiquitin ligase and POI. Upon irradiation, the PPG groups leave, allowing the structure to bind
E3 ubiquitin ligase and POI. Upon irradiation, the PPG groups leave, allowing the structure to bind
and, therefore, the ubiquitylation process occurs. Adapted with permission from [80]. Copyright,
and,John
2020, therefore,
Wileythe
andubiquitylation
Sons. process occurs. Adapted with permission from [80]. Copyright,
2020, John Wiley and Sons.
Xue et al. [81], for the first time, designed two photocaged PROTACs based on dBET1
Xue et al. [81], for the first time, designed two photocaged PROTACs based on dBET1
PROTAC
PROTAC[71], [71],which
whichused
used thalidomide
thalidomide asas aa ligand
ligandofofE3
E3ubiquitin
ubiquitinligase
ligasecereblon
cereblon (CRBN)
(CRBN)
and
and (+)-JQ1 as a ligand of BRD4. The 4,5-dimethoxy-2-nitrobenzyl (DMNB) introduced is a is
(+)-JQ1 as a ligand of BRD4. The 4,5-dimethoxy-2-nitrobenzyl (DMNB) introduced
a PPG thatisiscleaved
PPG that cleavedupon
upon irradiation
irradiation (365
(365 nm). nm).
TheThe photocaged
photocaged PROTAC
PROTAC 31 (Figure
31 (Figure 18a)
18a) has
has the PPG group linked to the amide of (+)-JQ1, and the photocaged PROTAC
the PPG group linked to the amide of (+)-JQ1, and the photocaged PROTAC 32 (Figure 18b) 32 (Figure
18b)
hashas
thethe
PPG PPG group
group through
through thethe imide
imide of of thalidomide.
thalidomide. Upon
Upon exposure
exposure to to irradiation,
irradiation,
Pharmaceuticals 2023, 16, 682 21 of 35
photocaged
photocagedPROTAC
PROTAC31 31 generated approximately50%
generated approximately 50%ofofdBET1
dBET1production,
production, whereas
whereas nono
dBET1production
dBET1 productionwas wasobserved
observed with
with photo-caged
photo-cagedPROTACPROTAC32.32.

Figure
Figure18.
18.(a)
(a)Uncaging
Uncagingreaction
reaction of
of photocaged PROTAC31
photocaged PROTAC 31inindBET
dBETand
and(b)
(b) structure
structure of of photocaged
photocaged
PROTAC
PROTAC32. 32.

Ramos cells were used to assess in vitro BRD4 degradation. BRD4 levels decreased
when photocaged PROTAC 31 was irradiated (365 nm, 3 min), being almost completely
degraded after 4 h. A zebrafish model was used to assess the in vivo activity of photocaged
PROTAC 31, which showed effectiveness.
Pharmaceuticals 2023, 16, 682 22 of 36

Ramos cells were used to assess in vitro BRD4 degradation. BRD4 levels decreased
when photocaged PROTAC 31 was irradiated (365 nm, 3 min), being almost completely
degraded after 4 h. A zebrafish model was used to assess the in vivo activity of photocaged
PROTAC 31, which showed effectiveness.
Naro et al. [82] also designed other two different photocaged PROTACs using two
different PPG groups and ligands recruiting E3 ubiquitin ligases (VHL and CRBN). Pho-
tocaged PROTAC 33 (Figure 19a) contains a VHL ligand, targets ERRα (estrogen related
2 22 of 35
receptor α), and uses a diethylaminocoumarin as a PPG group. Photocaged PROTAC
34 (Figure 19b) has a CRBN ligand and targets BRD4 using a 6-nitropiperonyloxymethyl as
a PPG group.

Figure 19. Chemical structures of (a)structures


Figure 19. Chemical photocaged PROTAC
of (a) photocaged 33 and
PROTAC 33 and(b) photocaged
(b) photocaged PROTAC
PROTAC 34. Upon 34.
irradiation, PPG groups are
Upon irradiation, PPG groups are released [82]. released [82].

MCF-7 cells were used to test the ability of the PPG group of the photocaged PROTAC
MCF-7 cells were used
33 to to test
block ERRα the ability
degradation, of the
and it was PPG
verified that group of theofphotocaged
the degradation ERRα was
blocked before irradiation. In the case of photocaged PROTAC 34, HEK293T cells were
PROTAC 33 to blockused ERRα degradation, and it was verified that the degradation of ERRα
to test the degradation of BRD4. In the absence of light, no degradation was verified.
was blocked before Upon
irradiation.
exposure toIn the(365
UV-A casenm)of photocaged
light, PROTAC
BRD4 was degraded, 34, HEK293T
corroborating cells
the expected
were used to test theresults of the designed
degradation of study.
BRD4. In the absence of light, no degradation was
Jing et al. [67] added a PPG group on pomalidomide based on dBET and dALK
verified. Upon exposure to UV-A (365PROTAC
PROTACs—photocaged nm) light, BRD4
35 (Figure wasphotocaged
20a) and degraded, corroborating
PROTAC 36 (Figure 20b).the
expected results of the designed study. was used as a PPG group that will prevent pomalidomide from
Dimethoxy-2-nitrobenzyl
binding to CRBN E3 ubiquitin ligase. In this way, protein degradation will not occur. This
Jing et al. [67] added a PPG group on pomalidomide based on dBET and dALK
PROTACs—photocaged PROTAC 35 (Figure 20a) and photocaged PROTAC 36 (Figure
20b). Dimethoxy-2-nitrobenzyl was used as a PPG group that will prevent pomalidomide
from binding to CRBN E3 ubiquitin ligase. In this way, protein degradation will not occur.
Pharmaceuticals 2023, 16, 682 23 of 36

armaceuticals 2023, 16, 682 will only occur when there is UV-A irradiation, in which PPG leaves and thus pomalidomide 23 o
can already recruit CRBN ligase for degradation to occur.

Figure 20. Chemical


Figure 20. Chemicalstructures ofphotocaged
structures of photocaged PROTAC
PROTAC 35 (a) 35
and(a) and photocaged
photocaged PROTAC
PROTAC 36 (b). 36 (b).

HEK293FT cells were used to test the effect of photocaged PROTAC 35 to degrade
HEK293FT cells were used to test the effect of photocaged PROTAC 35 to degr
BRD3-4 by UV-A irradiation. SU-DHL-1 cells were used to assess the effect of opto-dALK
BRD3-4 by UV-A
to degrade irradiation.
anaplastic lymphoma SU-DHL-1
kinase (ALK, cells wereprotein)
a fusion used to by assess the effectUpon
UV-A irradiation. of opto-dA
to degrade anaplastic
UV-A irradiation, lymphoma
photocaged PROTAC kinase (ALK, and
35 is uncaged a fusion
leads to protein) by ofUV-A
the induction BRD3-4irradiati
Upon UV-A irradiation,
degradation. photocaged
On the other hand, whenPROTAC
photocaged35 is uncaged
PROTAC and leadswith
36 is irradiated to the induction
UV-A
light, it is also uncaged and promotes the degradation of ALK.
BRD3-4 degradation. On the other hand, when photocaged PROTAC 36 is irradiated w
Kounde and co-workers [83] designed a photocaged PROTAC (Figure 21a) using a
UV-A light, it is also uncaged and promotes the degradation of ALK.
DMNB group binding to VHL E3 ligase-recruiter ligand and (+)-JQ1 as an inhibitor of BET.
Kounde
This approach andwasco-workers [83] designed
based on PROTAC a photocaged
MZ1 [84] (Figure PROTAC
21b). The DMNB moiety(Figure 21a) usin
was used
DMNB group
to block binding to
the recruitment VHLE3E3
of VHL ligase-recruiter
ubiquitin ligase withoutligand and but
irradiation, (+)-JQ1 as an inhibito
it was removed
upon UV-A light (365 nm) irradiation.
BET. This approach was based on PROTAC MZ1 [84] (Figure 21b). The DMNB mo
The ability of photocaged PROTAC 37 to degrade BRD4 was tested in HeLa cells and
was used to block the recruitment of VHL E3 ubiquitin ligase without irradiation, bu
its knockdown in real time was performed using live-cell fluorescence imaging (HEK293
was cells).
removed uponofUV-A
The results lightproved
this study (365 nm)
to be irradiation.
good as BRD4 degradation was achieved.
euticals 2023, 16, 682 2023, 16, 682
Pharmaceuticals 24 of 36 2

Figure Figure
21. Chemical structures
21. Chemical structures of photocaged
of photocaged PROTAC
PROTAC 37 (a)
37 (a) and and PROTAC
PROTAC MZ1 (b). MZ1 (b).
4. Porphyrins Combining Photoswitch Systems and Some Application Examples
The ability of photocaged PROTAC 37 to degrade BRD4 was tested in HeLa cel
Usually, gadolinium complexes are applied to improve magnetic resonance imaging
its knockdown in real
(MRI) structural timeinwas
contrast performed
tissues. using
Light can be used live-cell fluorescence
as a functional MRI contrastimaging
with (H
cells). The results of this
high-spatiotemporal study proved
resolution. to be good
A light-responsive as BRD4
contrast degradation
agent for was achiev
magnetic resonance
imaging based on a Ni(II) porphyrin derivative and a switch moiety has previously been
reported in the literature [85]. This agent can be applied non-invasively to switch magnetic
4. Porphyrins
resonanceCombining
imaging contrastPhotoswitch
ON and OFF with Systems and Some Application
a high-spatiotemporal Examples
resolution by light
stimulus gadolinium
Usually, (Figure 22). complexes are applied to improve magnetic resonance im
Porphyrin derivative 40 was found not to be soluble in water due to the first generation
(MRI) of
structural contrast in tissues. Light can be used as a functional MRI contras
small glycerol dendrimers, so the hydrophobic character of the Ni(II) porphyrin prevails.
high-spatiotemporal
However, compound resolution.
42 (secondAgeneration
light-responsive contrast agent
glycerol dendrimers) preventsforporphyrin
magnetic reso
imagingaggregation
based on andathus increases
Ni(II) water solubility.
porphyrin Non-dendronized
derivative porphyrins
and a switch moiety (compounds
has previousl
38 and 43) form dimers and with increasing concentration become paramagnetic, decreasing
reported in the literature [85]. This agent can be applied non-invasively to switch ma
the efficacy of contrast switching. Second generation dendrimers prevent this phenomenon.
resonance imaging contrast ON and OFF with a high-spatiotemporal resolution b
stimulus (Figure 22).
euticals 2023, 16, 6822023, 16, 682
Pharmaceuticals 25 of 36 2

Figure Figure
22. Structures of of
22. Structures porphyrins irradiated
porphyrins irradiated withwith 505
505 nm nm (contrast
(contrast ON) and
ON) and reversible reversible p
process
(435 nm, contrast
(435 OFF)
nm, contrast [85]
OFF) .
[85].

The trans-isomer was found not to coordinate intramolecularly, being diamagnetic and
Porphyrin derivative
thus is MRI silent (contrast40 was
OFF), foundsince
whereas notthe
tocis-isomer
be soluble in water
coordinates due to the first g
intramolecularly,
1 (R1 Ni(II)
tion ofitsmall glycerol
is paramagnetic anddendrimers, so the
MRI active (contrast ON).hydrophobic
Substitutions oncharacter of the
R with pyridine = H) porp
furnished porphyrins with a PSS of 65% cis-isomer, while 4-methoxyl (R1 = OMe) showed
prevails. However, compound 42 (second generation glycerol dendrimers) preven
a PSS > 95% cis-isomer, allowing the retention of diamagnetic to paramagnetic switching. It
phyrinwasaggregation
concluded thatand thus 47
compound increases water
provided the solubility.
best structures Non-dendronized
in this study. porp
(compounds 38literature,
In the and 43)photoswitchable
form dimers micelles
and with wereincreasing concentration
applied for the become
control of singlet oxy- par
gen (1 O ) generation in PDT, using a zinc(II) complex of 5,10,15,20-tetraphenylporphyrin
netic, decreasing the efficacy of contrast switching. Second generation dendrimers p
2
(ZnTPP) as a photosensitizer and 1,2-bis(5-(4-carbonxyphenyl)-2-methylthien-3-yl)cyclopent-
this phenomenon.
1-ene (BDTE) as a photoswitch (compound 48, Figure 23). Compound 48 was encapsulated
The trans-isomer was found not to coordinate intramolecularly, being diama
and thus is MRI silent (contrast OFF), whereas since the cis-isomer coordinates int
lecularly, it is paramagnetic and MRI active (contrast ON). Substitutions on R1 with
dine (R1 = H) furnished porphyrins with a PSS of 65% cis-isomer, while 4-methoxy
Pharmaceuticals 2023, 16, 682 26 of 36
Pharmaceuticals 2023, 16, 682 26 of 35

in micelles to cause tumour and bacterial cell death or prevent cell damage upon irradia-
tion through 1 O generation [86]. The photoswitching was evaluated in a subcutaneous
irradiation through2 1O2 generation [86]. The photoswitching was evaluated in a subcuta-
tissue model.
neous tissue model.

Figure 23. Singlet-oxygen control upon opening and closing photoswitch


generates 1O2, while the closed is not active in producing 1O2.

5. Porphyrins Combining Photocleavage Systems and Some Ap


Figure
Figure Lin
23.23. et al. [87]
Singlet-oxygen
Singlet-oxygen controlreported
control upon openingfor
upon opening the
andclosing
and closingfirst timeThein
photoswitch.
photoswitch. The 2008,
open
open an(open-48)
form
form antican
(open-48)
generates
generatesO1 O
1 2, while the closed is not active in producing1 1O2.
, while the closed is not active in producing O .
49, Figure 24) based on an o-nitrobenzyl group coupled with a po
2 2

5. 5. PorphyrinsCombining
Combining Photocleavage
Photocleavage Systems
Systemsand Some Application
anticancer prodrug is composed of three parts, such as a porphyr
Porphyrins and Some ApplicationExamples
Examples
Linetetal.
Lin al.[87]
[87]reported
reported for
for the
the first
firsttime
timeinin2008,
2008,anananticancer prodrug
anticancer (compound
prodrug (compound
group), and a parent anticancer drug (Tegafur). First,
49, Figure 24) based on an o-nitrobenzyl group coupled with a porphyrin. The porphyrin the drug i
49, Figure 24) based on an o-nitrobenzyl group coupled with a porphyrin. The porphyrin
with UV-A
anticancer
anticancer prodruglight
prodrug is composed(350of
is composed ofnm). After
three parts,
three parts,
such12
such asmin, it achieved
as a porphyrin, a 50% conv
a PPG (o-nitrobenzyl
a porphyrin, a PPG (o-nitrobenzyl
group), and a parent anticancer drug (Tegafur). First, the drug is released by irradiation
intoUV-A
group),
with tegafur.
and a parent
light 5-Fluorouracil
anticancer
(350 nm). drug
After 12 as an
min,(Tegafur).
it achieved anticancer
First,
a 50% the drug is of
conversion drug
released bywas
compound used a
irradiation
49 into
with UV-A
tegafur. light (350
5-Fluorouracil nm).
as After 12 min,
an anticancer drug it was used as a control, and it was released in 49
achieved a 50% conversion of compound
released in the same way as Tegafur.
into tegafur. 5-Fluorouracil as an anticancer drug was used as a control, and it was
the same way as Tegafur.
released in the same way as Tegafur.

Figure 24. Schematic representation of compound 49 (anticancer prodrug composed by a porphyrin,


a PPG, and a parent anticancer drug, tegafur) and its release upon UV-A irradiation [87].

The authors used MCF-7 human breast adenocarcinoma cells to evaluate the cytotox-
icity of compound 49 in the absence and presence of irradiation. In the absence of UV-A
Figure
Figure
light, tegafur
24. Schematic
24. Schematic
induced
representation
representation
91% of cellofdeath
of compound
compound 49 (anticancer
while the compound
49 (anticancer
prodrug composed
induced onlyby7%
prodrug
a porphyrin,
of cell death
(less PPG,
aaPPG, andWhen
a parent anticancer drug, tegafur)
therefore,and its release upon69% UV-A
and a parent anticancer drug, tegafur) and its release upon UV-A irradiation [87].
cytotoxic). this entity is irradiated and, Tegafur is released, of
cell death is observed. Although there is less cell death, this approach allows for minimiz-
ing the side effects due to the tumour-affinity property of porphyrin and the light-control-
The authors used MCF-7 human breast adenocarcinoma cell
lable system.
icity of compound
Tessaro 49 ainnovel
et al. [88] reported thephotocontrolled
absence andnanoplatform
presencecomprising
of irradiation.
two
photoresponsive
light, tegafur components—a
induced 91% benzochlorin
of cellPSdeath(Figurewhile
25a) forthe
PDTcompound
and a coumarin- indu
Pharmaceuticals 2023, 16, 682 27 of 36

The authors used MCF-7 human breast adenocarcinoma cells to evaluate the cyto-
toxicity of compound 49 in the absence and presence of irradiation. In the absence of
UV-A light, tegafur induced 91% of cell death while the compound induced only 7% of
cell death (less cytotoxic). When this entity is irradiated and, therefore, Tegafur is released,
69% of cell death is observed. Although there is less cell death, this approach allows
for minimizing the side effects due to the tumour-affinity property of porphyrin and the
Pharmaceuticals 2023, 16, 682 light-controllable system. 27 of 3
Tessaro et al. [88] reported a novel photocontrolled nanoplatform comprising two
photoresponsive components—a benzochlorin PS (Figure 25a) for PDT and a coumarin-
photocaged chlorambucil (compound 50, Figure 25b)—into core-shell micelles to amplify
the the
anticancer
anticanceractivity
activity against MCF-7human
against MCF-7 human breast
breast adenocarcinoma
adenocarcinoma cells.
cells. The The author
authors
demonstrated
demonstrated that
thatwith
withsimultaneous irradiation
simultaneous irradiation with
with visible
visible light
light in micelles,
in the the micelles,
ben- benzo
1 O as a PS and compound 50 acts as a prodrug by leaving of PPG
chlorin produces O2 as 2a PS and compound 50 acts as a prodrug by leaving of PPG group
zochlorin produces
1

group25b).
(Figure (Figure 25b).

Figure 25.25.
Figure Benzochlorin
Benzochlorinused as aaPS
used as PSfor
forPDT
PDT(a)(a)
andand coumarin-photocaged
coumarin-photocaged (compound
(compound 50) releas
50) release
chlorambucil
chlorambucil (b)[88]
(b). [88]..

Results showed chlorambucil used alone induces 25% cell death, while micelles
Results showed chlorambucil used alone induces 25% cell death, while micelles con
containing compound 50 and the sensitizer benzochlorin induce 75% cell death after 20 min
taining compound 50 and the sensitizer benzochlorin induce 75% cell death after 20 min
of blue irradiation (400 nm). This promising multimodal therapy destroys the tumour in
of blue irradiation
two ways—generating(400 1nm). This
O2 and promising
releasing multimodal
chlorambucil duringtherapy destroys
the irradiation the tumour in
procedure,
twoshowing
ways—generating 1 O2 and releasing chlorambucil during the irradiation procedure
a synergistic effect.
showing a synergistic effect.

6. Porphyrins with Antibacterial Activity


Antimicrobial photodynamic treatment (aPDT) requires, besides an adequate PS,
sufficient PS concentration and an adequate light dose to guarantee full microbial inacti
vation. However, achieving these conditions is not always easy. To overcome this prob
Pharmaceuticals 2023, 16, 682 28 of 36

6. Porphyrins with Antibacterial Activity


Antimicrobial photodynamic treatment (aPDT) requires, besides an adequate PS, a
sufficient PS concentration and an adequate light dose to guarantee full microbial inactiva-
tion. However, achieving these conditions is not always easy. To overcome this problem, an
ongoing approach that has been presented by the scientific community is the combination
of PDT and chemotherapy, as such high concentrations of PS are not necessary because
the antibiotic is also used in combination. Thus, the synergistic effect between aPDT and
antibiotic therapy is a promising approach as it is possible to increase aPDT efficiency at
concentrations below the MIC [89,90]. This section will be presented as an overview of
using porphyrins as PS for aPDT and antibiotic therapy combination.
Pharmaceuticals 2023, 16, 682 Xing et al. [91] conjugated vancomycin, a glycopeptide antibiotic, with a porphyrin 28
derivative to generate a divalent dimeric system (Figure 26), which has a rigid structure and
Pharmaceuticals 2023, 16, 682 28
provides a steric hindrance necessary for interactions between vancomycin and the bacterial
strain. The antibacterial activity of compound 51 was tested in B. subtilis (ATCC 33,677), En-
photoinactivation
terococcus activity
faecium (E. faecium) for the
ATCC conjugate
51559, when compared
and Enterococcus to vancomycin
faecalis (E. faecalis) ATCC 51,299,and por
vancomycin-resistant
rin alone.
photoinactivation enterococci
activity forstrains. The results
the conjugate showed
when a strong photoinactivation
compared to vancomycin and po
activity for the conjugate when compared to vancomycin and porphyrin alone.
rin alone.

Figure 26. Chemical structure of the conjugated Van-porphyrin. Van = vancomycin [91].
Figure26.26.
Figure Chemical
Chemical structure
structure of theof the conjugated
conjugated Van-porphyrin.
Van-porphyrin. Van = vancomycin
Van = vancomycin [91]. [91].
Liu et al. [92] also developed an approach based on the bioconjugation of a proto
phyrin
Liu IXet
Liuet al.(PpIX)
[92]
al. asdeveloped
also
[92] alsoa developed
PS with an lipopolysaccharide
approach on the(LPS)
based based
an approach theand an antibacterial
bioconjugation
on of a proto-
bioconjugation pep
of a prot
porphyrin
(YI13WF) IX (PpIX) as a PS with lipopolysaccharide (LPS) and an antibacterial peptide
phyrin IXto(PpIX)
photoinactivate
as a PS withGram-negative bacterial strains
lipopolysaccharide (Figure
(LPS) and
(YI13WF) to photoinactivate Gram-negative bacterial strains (Figure 27).
27).
an antibacterial pe
(YI13WF) to photoinactivate Gram-negative bacterial strains (Figure 27).

Figure27.27.Dimeric
Figure DimericPpIX −peptide conjugate.
PpIX−peptide Reprinted
conjugate. with permission
Reprinted from [92]. Copyright,
with permission from [92].2012,
Copyright,
American Chemical Society.
American Chemical Society.
Figure 27. Dimeric PpIX−peptide conjugate. Reprinted with permission from [92]. Copyright
American Chemical Society.
The MIC values (Table 7) were calculated in E. coli DH5a (ATCC 53868), S. ent
(ATCC 14028), E. coli BL21, and K. pneumoniae (ATCC 700603). The antibacterial pep
The MIC values (Table 7) were calculated in E. coli DH5a (ATCC 53868), S. en
sequence (YI13WF) binds strongly towards lipids at the outer membrane of Gram-n
(ATCC 14028), E. coli BL21, and K. pneumoniae (ATCC 700603). The antibacterial pe
tive bacteria, showing potent MIC values against the strains. However, the MIC valu
sequence (YI13WF) binds strongly towards lipids at the outer membrane of Gram-
Pharmaceuticals 2023, 16, 682 29 of 36

The MIC values (Table 7) were calculated in E. coli DH5a (ATCC 53868), S. enterica
(ATCC 14028), E. coli BL21, and K. pneumoniae (ATCC 700603). The antibacterial peptide
sequence (YI13WF) binds strongly towards lipids at the outer membrane of Gram-negative
bacteria, showing potent MIC values against the strains. However, the MIC values of the
dimeric conjugate (compound 52) demonstrated greater aPDT. The antibacterial activity
of PpIX alone as a control was also evaluated. The results of the conjugates proved to be
significantly better than these components alone.

Table 7. MIC values, expressed in µM, of the conjugate free peptide YI13WF and PpIX.

MIC Values (µM)


E. coli DH5a S. enterica E. coli BL21 K. pneumoniae
maceuticals 2023, 16, 682 PpIX−Peptide Conjugate 2.0 4.0 4.0 8.0
YI13WF 8 ~24 32 32
PpIX >64 >64 >64 >64

Dastgheyb and co-workers [93] tested a 5,10,15,20-tetrakis(4-aminophenyl)porphyrin


TAPP
(TAPP) specifically
combined targets
with antibiotics bacterial
used for membranes,
the treatment while
of bacterial infections, tobramyci
assuming
inhibit
that protein
combining synthesis,
TAPP with and ceftriaxone
antibiotics would and vancomycin
have a complementary effect (Figure 28). impa
TAPP specifically targets bacterial membranes, while tobramycin and chloramphenicol
Results
inhibit indicated
protein that
synthesis, and TAPP and
ceftriaxone with chloramphenicol
vancomycin impair membrane orintegrity.
tobramycin
on S.indicated
Results aureusthatand TAPPE. coli
with and a synergic
chloramphenicol effect
or tobramycin had anonadditive
MRSA effectand
on S. ep
S. aureus and E. coli and a synergic effect on MRSA and S. epidermidis. On the other hand,
hand, vancomycin
vancomycin andonly
and ceftriaxone show ceftriaxone
modest effectsshow only modest
when combined with TAPP.effects when

Figure 28. Chemical structure of 5,10,15,20-tetrakis(4-aminophenyl)porphyrin (TAPP).


Figure 28. Chemical structure of 5,10,15,20-tetrakis(4-aminophenyl)porph
Almeida et al. [94] aimed to allow the efficient treatment of aPDT on multidrug-resistant
bacteria in hospital wastewaters. Therefore, a 5,10,15,20-tetrakis(1-methylpyridinium-4-
Almeida
yl)porphyrin et al.
tetra-iodide [94] aimed
54 (TMPyP, Figure 29)toasallow theused
the PS was efficient treatment
in combination with of
sistant bacteria
chloramphenicol. in hospital
The antibacterial wastewaters.
activity of Therefore,
the conjugate was performed a and
in E. coli 5,10,15,20
it
was proved that the combination effect of the conjugates was more efficient than TMPyP
dinium-4-yl)porphyrin
used alone. The synergic effect could tetra-iodide
be due to the aPDT 54cell
(TMPyP,
membrane andFigure
the cell29)
wallas the
nation with
destabilization, chloramphenicol.
allowing The antibacterial activity of the conj
an easier entry of chloramphenicol.
Branco and co-workers [90] compared the effect of aPDT and aPDT combined with
E. coli and
antibiotics it skin
to treat wasinfected
proved with that theATCC
S. aureus combination
6538. As well effect
as in theof the conjug
previous
thanTMPyP
work, TMPyP used 54,
(compound alone.
FigureThe synergic
29) was effect
used as the PS. Thecould be due
antibacterial to the aP
activity
was performed in vitro and ex vivo (porcine skin), using compound 54 combined with
thewithout
and cell wall destabilization,
antibiotics. allowing
In vitro, a bacterial an~8easier
reduction of log afterentry
180 minof of chloramp
white
Pharmaceuticals 2023, 16, 682 30 of 36

Figure 28. Chemical structure of 5,10,15,20-tetrakis(4-aminophenyl)porphy


light irradiation at an irradiance of 4.0 mW cm−2 was attained using 5.0 µM of 54 without
Almeida
antibiotic. When tested,et the
al.combined
[94] aimed
action ofto
0.5allow the
and 1.0 µg mLefficient
−1 ampicillintreatment
with 5.0 µM of aP
of
sistant bacteria in hospital wastewaters. Therefore, a of5,10,15,20-t
54 increased the efficacy, observing a reduction of ~8 log after 60 min and 30 min white
light irradiation, respectively. When the ampicillin was tested alone, no colony-forming
dinium-4-yl)porphyrin
unit (CFU) reduction was observed,tetra-iodide
even at the highest54tested
(TMPyP, Figure
concentration, 29)
1.0 µg mLas −1 . the P

In ex vivo with
nation experiments, using 50 µM of 54 after
chloramphenicol. The 180 antibacterial
min of irradiation with white light
activity at anconjug
of the
− 2
irradiance of 150 W m , a decrease of ~4 log (99.99% reduction) of bacterial abundance was
E. coli and
observed. it was
However, when proved that
treated the theskin
porcine combination effect
simultaneously with of the
TMPyP conjuga
(50 µM)
and with −
5.0 µg mLused1 ampicillin,
than TMPyP alone.a bacterial reduction of
The synergic ~5.6 log
effect (>99.9995%
could be duereduction)
to the aPD
after 180 min of white light irradiation was observed. Using the antibiotic alone at this
the cell wall
concentration, onlydestabilization,
a CFU reduction of ~1allowing
log was noted.an easier entry of chloramphe

Pharmaceuticals 2023, 16, 682

Iluz et al. [95] searched for a synergistic effect between deuteroporphy


Figure 29. Chemical structure of the cationic porphyrin 54, also known as TMPyP or Tetra-Py+ -Me.
Figure 29. Chemical structure of the cationic porphyrin 54, also known as T
30) and antibiotics usually used to treat infections with S. aureus ATCC 25,92
Iluz et al. [95] searched for a synergistic effect between deuteroporphyrin 55 (Figure 30)
Although gentamicin, vancomycin, rifampin, and fusidic acid treatments w
and antibiotics usually used to treat infections with S. aureus ATCC 25,923 and MRSA.
Branco
combination
Although gentamicin,
andvancomycin,
with co-workers
aPDT, only [90]and
oxacillin
rifampin,
compared
(1 μg acid
fusidic
the
mLtreatments
−1 effect
) proved tooftested
were
aPDT
have and
a syner
in
antibiotics
combination
combination withto treat
with
aPDT, skin
only infected
deuteroporphyrin
oxacillin with
(1 µg mL (4 μM),S. aureus
−1 ) proved
during
to haveATCC
24 h, at6538.
a light
a synergistic As inwell
dose
effect of as
15
combination with deuteroporphyrin (4 µM), during 24 h, at a light dose of 15 J·cm−2 , show-
TMPyP
ing a CFU(compound
reduction of 54, ~5.5Figure 29) was
log (99.9995% used as the
reduction). PS. The
Cultures
ing a CFU reduction of ~5.5 log (99.9995% reduction). Cultures treated with monotherapy
antibact
treated with
(oxacillin
formedorin
(oxacillin orvitro
55 55 alone)
alone) and showed
showed ex
no vivo no log(0%
(porcine
log decrease decrease
skin),(0%
reduction), using
even reduction),
there even
compound
though was an54though
com
increase
increase during 2hInofvitro,
during
out antibiotics. 2h of treatment.
treatment.
a bacterial reduction of ~8 log after 180 min
ation at an irradiance of 4.0 mW cm−2 was attained using 5.0 μM of
When tested, the combined action of 0.5 and 1.0 μg mL−1 ampicil
increased the efficacy, observing a reduction of ~8 log after 60 min
light irradiation, respectively. When the ampicillin was tested alon
unit (CFU) reduction was observed, even at the highest tested conce
In ex vivo experiments, using 50 μM of 54 after 180 min of irradiati
an irradiance of 150 W m−2, a decrease of ~4 log (99.99% reduction) o
was observed. However, when treated the porcine skin simultaneo
μM) and with 5.0 μg mL−1 ampicillin, a bacterial reduction of ~5.6 l
tion) after 180 min of white light irradiation was observed. Using
Figure 30. Chemical structure of
this concentration, the deuteroporphyrin
only aofCFU 55 (DP).
reduction of ~1 log was noted.
Figure 30. Chemical structure the deuteroporphyrin 55 (DP).

7. Concluding Remarks and Future Challenges


The problem of antimicrobial and chemotherapeutic resistance is that
quently in therapeutic failure. With the emergence of new strains of resistan
Pharmaceuticals 2023, 16, 682 31 of 36

7. Concluding Remarks and Future Challenges


The problem of antimicrobial and chemotherapeutic resistance is that is results fre-
quently in therapeutic failure. With the emergence of new strains of resistant bacteria, the
search for effective antibiotics has become a critical global health priority and has motivated
several research groups around the world to find effective alternatives.
Photodynamic treatments, photoswitches, photocaged groups, and recent PHOTACs
and photocaged PROTACs strategies have been explored as promising approaches to tackle
the AMR issue. As it was demonstrated (compounds 3–6 and 12–15), photoswitches have
the advantage of being reversible compared to photocaged compounds. However, the
design of these molecules is not always easy or straightforward because they were related
to a series of azobenzene-based compounds where light would not have the activating
effect but rather result in deactivation, which is very important for deactivation processes.
Despite this, these deactivation approaches will be more relevant in the case of PHOTACs
to stop the degradation protein process if necessary. However, many approaches reported
using UV-A-C irradiation, which is harmful to healthy cells, or with visible blue light, which
has limited tissue penetration. As far as we know, only one example was found in the
literature that reports the use of NIR light in azobenzene-based compounds (compounds
5 and 6). Therefore, it is necessary to improve strategies of incorporation of red-shifted
chromophores to address the tissue penetration problem, even in PHOTACs (not just
restricted to photoswitchable compounds). Apparently, there are not as many studies done
on photocaged compounds as there are on photoswitchable compounds, which could be
due to the fact that the process is not reversible. Photocaged PROTACs do not seem to be
as promising because of they do not allow protein degradation to be stopped if necessary.
Nevertheless, PHOTACs and photocaged PROTACs are reported for cancer treatment, but
it should be important to prove their effectiveness for other diseases, such as antimicrobial
infections, to combat resistant strains. Furthermore, porphyrins and their derivatives (e.g.,
chlorins) have been demonstrating a synergistic effect when used with antibiotic therapy.
However, the most recent study on this topic was published several years ago, so it is critical
to continue researching in this field, specially involving porphyrin derivatives with the
antimicrobial activity in aPDT, since no resistance mechanism seems to be developed by the
biological entities. It is important to note that, to date, examples of porphyrin derivatives
combining photoswitchable or photocaged systems with antimicrobial activity are not
described in the literature and there is still room to develop interesting photoswitchable or
photocaged systems.
Photoswitches and photocleavage compounds have shown promising potential for
use in various clinical applications. However, as with any new therapeutic agent or its
combined action, there are potential safety concerns associated with their use that must be
evaluated [96]. One of the main safety concerns is the potential risk for these compounds
to elicit adverse effects or toxicity. To mitigate these potential safety risks in a clinical
context, it is crucial to conduct comprehensive preclinical studies to evaluate toxicity and
to find the optimal therapeutical conditions in order to carefully control and avoid toxicity.
Additionally, the delivery method must be optimized to ensure that the desired target
compounds are only delivered to the target tissues or organs. [97]. Overall, the successful
translation of externally activated drug delivery systems from preclinical studies to human
clinical trials requires a comprehensive understanding of the complex interactions between
the delivery system and the target biological environment [98]. To address these challenges,
it is important to employ animal models, assess drug dose and administration routes
based on in vitro experiments, and develop effective methods for monitoring the long-
term toxicity and pharmacokinetics of the delivery system in vivo [99]. While external
triggers pose additional challenges, the imaging capabilities of many triggered delivery
systems offer unique opportunities for better monitoring the fate of drugs and drug carriers
in vivo [100].
Our aim for this review is to offer new perspectives on the use of photopharmacol-
ogy, not only for bacterial inactivation to mitigate antibiotic resistance but also for cancer
Pharmaceuticals 2023, 16, 682 32 of 36

treatment, motivating the medical and scientific communities to devote greater attention
and resources to these issues. This, in turn, will enhance the comprehension of the prob-
lem, enabling the identification of potential solutions that could lead to improved public
health outcomes.

Author Contributions: Conceptualization, C.J.P.M., M.E.S., A.P. and M.A.F.F.; writing—original draft
preparation, S.N.S., C.J.P.M., M.E.S., A.P. and M.A.F.F.; writing—review and editing, C.J.P.M., M.E.S.,
A.P. and M.A.F.F. All authors have read and agreed to the published version of the manuscript.
Funding: This work received support from PT national funds (FCT/MCTES, Fundação para a
Ciência e a Tecnologia and Ministério da Ciência, Tecnologia e Ensino Superior) through the
projects UIDB/50006/2020 and UIDP/50006/2020 (LAQV-REQUIMTE) and UIDB/04423/2020,
UIDP/04423/2020 (Group of Marine Natural Products and Medicinal Chemistry CIIMAR).
Data Availability Statement: Data sharing not applicable.
Acknowledgments: The authors thank the University of Aveiro and FCT/MCT for the financial
support provided to LAQV-REQUIMTE (UIDB/50006/2020 and UIDP/50006/2020), Group of Ma-
rine Natural Products and Medicinal Chemistry CIIMAR (UIDB/04423/2020, UIDP/04423/2020),
through national funds (OE), and where applicable, co-financed by the FEDER-Operational The-
matic Program for Competitiveness and Internationalization-COMPETE 2020, within the PT2020
Partnership Agreement.
Conflicts of Interest: The authors declare no conflict of interest. The funders had no role in the design
of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or
in the decision to publish the results.

References
1. Reessing, F.; Szymanski, W. Beyond Photodynamic Therapy: Light-Activated Cancer Chemotherapy. Curr. Med. Chem. 2017, 24,
4905–4950. [CrossRef]
2. Jia, S.; Sletten, E.M. Spatiotemporal Control of Biology: Synthetic Photochemistry Toolbox with Far-Red and Near-Infrared Light.
ACS Chem. Biol. 2022, 17, 3255–3269. [CrossRef] [PubMed]
3. Broichhagen, J.; Frank, J.A.; Trauner, D. A Roadmap to Success in Photopharmacology. Acc. Chem. Res. 2015, 48, 1947–1960.
[CrossRef] [PubMed]
4. Velema, W.A.; Szymanski, W.; Feringa, B.L. Photopharmacology: Beyond Proof of Principle. J. Am. Chem. Soc. 2014, 136,
2178–2191. [CrossRef] [PubMed]
5. World Health Organization: Antimicrobial Resistance. Available online: https://www.who.int/news-room/fact-sheets/detail/
antimicrobial-resistance (accessed on 21 March 2023).
6. Weinstain, R.; Slanina, T.; Kand, D.; Klan, P. Visible-to-NIR-Light Activated Release: From Small Molecules to Nanomaterials.
Chem. Rev. 2020, 120, 13135–13272. [CrossRef]
7. Liu, J.Z.; Kang, W.R.; Wang, W.P. Photocleavage-based Photoresponsive Drug Deliver. Photochem. Photobiol. 2022, 98, 288–302.
[CrossRef]
8. Hull, K.; Morstein, J.; Trauner, D. In Vivo Photopharmacology. Chem. Rev. 2018, 118, 10710–10747. [CrossRef]
9. Velema, W.A.; Hansen, M.J.; Lerch, M.M.; Driessen, A.J.M.; Szymanski, W.; Feringa, B.L. Ciprofloxacin-Photoswitch Conjugates:
A Facile Strategy for Photopharmacology. Bioconjugate Chem. 2015, 26, 2592–2597. [CrossRef]
10. Mosinger, J.; Lang, K.; Kubát, P. Photoactivatable Nanostructured Surfaces for Biomedical Applications. In Light-Responsive
Nanostructured Systems for Applications in Nanomedicine; Sortino, S., Ed.; Springer International Publishing: Cham, Switzerland,
2016; pp. 135–168. [CrossRef]
11. Mesquita, M.Q.; Dias, C.J.; Neves, M.G.P.M.S.; Almeida, A.; Faustino, M.A.F. The Role of Photoactive Materials Based on
Tetrapyrrolic Macrocycles in Antimicrobial Photodynamic Therapy. In Handbook of Porphyrin Science; World Scientific: Singapore,
2022; Volume 46, pp. 201–277.
12. Q Mesquita, M.; J Dias, C.; P M S Neves, M.G.; Almeida, A.; F Faustino, M.A. Revisiting Current Photoactive Materials for
Antimicrobial Photodynamic Therapy. Molecules 2018, 23, 2424. [CrossRef]
13. Henderson, B.W. Photodynamic Therapy: Basic Principles and Clinical Applications; CRC Press: Boca Raton, FL, USA, 1992.
14. De Silva, P.; Saad, M.A.; Thomsen, H.C.; Bano, S.; Ashraf, S.; Hasan, T. Photodynamic therapy, priming and optical imaging:
Potential co-conspirators in treatment design and optimization—A Thomas Dougherty Award for Excellence in PDT paper.
J. Porphyr. Phthalocyanines 2020, 24, 1320–1360. [CrossRef]
15. Santos, L.L.; Oliveira, J.; Monteiro, E.; Santos, J.; Sarmento, C. Treatment of Head and Neck Cancer with Photodynamic Therapy
with Redaporfin: A Clinical Case Report. Case Rep. Oncol. 2018, 11, 769–776. [CrossRef] [PubMed]
Pharmaceuticals 2023, 16, 682 33 of 36

16. Van Straten, D.; Mashayekhi, V.; de Bruijn, H.S.; Oliveira, S.; Robinson, D.J. Oncologic Photodynamic Therapy: Basic Principles,
Current Clinical Status and Future Directions. Cancers 2017, 9, 19. [CrossRef]
17. Kwiatkowski, S.; Knap, B.; Przystupski, D.; Saczko, J.; K˛edzierska, E.; Knap-Czop, K.; Kotlińska, J.; Michel, O.; Kotowski, K.;
Kulbacka, J. Photodynamic therapy—Mechanisms, photosensitizers and combinations. Biomed. Pharmacother. 2018, 106, 1098–1107.
[CrossRef]
18. Plaetzer, K.; Krammer, B.; Berlanda, J.; Berr, F.; Kiesslich, T. Photophysics and photochemistry of photodynamic therapy:
Fundamental aspects. Lasers Med. Sci. 2009, 24, 259–268. [CrossRef]
19. Rkein, A.M.; Ozog, D.M. Photodynamic Therapy. Dermatol. Clin. 2014, 32, 415–425. [CrossRef] [PubMed]
20. Kim, M.; Jung, H.Y.; Park, H.J. Topical PDT in the Treatment of Benign Skin Diseases: Principles and New Applications. Int.
J. Mol. Sci. 2015, 16, 23259–23278. [CrossRef] [PubMed]
21. Castano, A.P.; Demidova, T.N.; Hamblin, M.R. Mechanisms in photodynamic therapy: Part one—Photosensitizers, photochemistry
and cellular localization. Photodiagn. Photodyn. Ther. 2004, 1, 279–293. [CrossRef]
22. Almeida, A.; Faustino, M.A.F.; Neves, M.G.P.M.S. Antimicrobial Photodynamic Therapy in the Control of COVID-19. Antibiotics
2020, 9, 320. [CrossRef]
23. Alves, E.; Faustino, M.A.F.; Neves, M.G.P.M.S.; Cunha, Â.; Nadais, H.; Almeida, A. Potential applications of porphyrins in
photodynamic inactivation beyond the medical scope. J. Photochem. Photobiol. C Photochem. Rev. 2015, 22, 34–57. [CrossRef]
24. Youf, R.; Müller, M.; Balasini, A.; Thétiot, F.; Müller, M.; Hascoët, A.; Jonas, U.; Schönherr, H.; Lemercier, G.; Montier, T.;
et al. Antimicrobial Photodynamic Therapy: Latest Developments with a Focus on Combinatory Strategies. Pharmaceutics 2021,
13, 1995. [CrossRef]
25. Pucelik, B.; Dabrowski,
˛ J.M. Chapter Three—Photodynamic inactivation (PDI) as a promising alternative to current pharma-
ceuticals for the treatment of resistant microorganisms. In Advances in Inorganic Chemistry; van Eldik, R., Hubbard, C.D., Eds.;
Academic Press: Cambridge, MA, USA, 2022; Volume 79, pp. 65–108.
26. Varzandeh, M.; Mohammadinejad, R.; Esmaeilzadeh-Salestani, K.; Dehshahri, A.; Zarrabi, A.; Aghaei-Afshar, A. Photodynamic
therapy for leishmaniasis: Recent advances and future trends. Photodiagn. Photodyn. Ther. 2021, 36, 102609. [CrossRef] [PubMed]
27. Sadraeian, M.; da Cruz, E.F.; Boyle, R.W.; Bahou, C.; Chudasama, V.; Janini, L.M.R.; Diaz, R.S.; Guimarães, F.E.G. Photoinduced
Photosensitizer–Antibody Conjugates Kill HIV Env-Expressing Cells, Also Inactivating HIV. ACS Omega 2021, 6, 16524–16534.
[CrossRef] [PubMed]
28. Wu, X.; Hu, Y. Photodynamic Therapy for the Treatment of Fungal Infections. Infect. Drug Resist. 2022, 15, 3251–3266. [CrossRef]
29. Barbosa, A.F.S.; Santos, I.P.; Santos, G.M.P.; Bastos, T.M.; Rocha, V.P.C.; Meira, C.S.; Soares, M.B.P.; Pitta, I.R.; Pinheiro, A.L.B.
Anti–Trypanosoma cruzi effect of the photodynamic antiparasitic chemotherapy using phenothiazine derivatives as photosensi-
tizers. Lasers Med. Sci. 2020, 35, 79–85. [CrossRef] [PubMed]
30. Yoo, S.W.; Oh, G.; Ahn, J.C.; Chung, E. Non-Oncologic Applications of Nanomedicine-Based Phototherapy. Biomedicines 2021,
9, 113. [CrossRef]
31. Niculescu, A.-G.; Grumezescu, A.M. Photodynamic Therapy—An Up-to-Date Review. Appl. Sci. 2021, 11, 3626. [CrossRef]
32. Oyama, J.; Fernandes Herculano Ramos-Milaré, Á.C.; Lopes Lera-Nonose, D.S.S.; Nesi-Reis, V.; Galhardo Demarchi, I.; Alessi
Aristides, S.M.; Juarez Vieira Teixeira, J.; Gomes Verzignassi Silveira, T.; Campana Lonardoni, M.V. Photodynamic therapy in
wound healing in vivo: A systematic review. Photodiagn. Photodyn. Ther. 2020, 30, 101682. [CrossRef]
33. Diogo, P.; F. Faustino, M.A.; PMS Neves, M.G.; Palma, P.J.; P. Baptista, I.; Gonçalves, T.; Santos, J.M. An Insight into Advanced
Approaches for Photosensitizer Optimization in Endodontics—A Critical Review. J. Funct. Biomater. 2019, 10, 44. [CrossRef]
34. Li, X.; Sun, L.; Zhang, P.; Wang, Y. Novel Approaches to Combat Medical Device-Associated BioFilms. Coatings 2021, 11, 294.
[CrossRef]
35. Almeida, A. Photodynamic Therapy in the Inactivation of Microorganisms. Antibiotics 2020, 9, 138. [CrossRef]
36. Bartolomeu, M.; Neves, M.G.P.M.S.; Faustino, M.A.F.; Almeida, A. Wastewater chemical contaminants: Remediation by advanced
oxidation processes. Photochem. Photobiol. Sci. 2018, 17, 1573–1598. [CrossRef] [PubMed]
37. Bartolomeu, M.; Oliveira, C.; Pereira, C.; Neves, M.G.P.M.S.; Faustino, M.A.F.; Almeida, A. Antimicrobial Photodynamic
Approach in the Inactivation of Viruses in Wastewater: Influence of Alternative Adjuvants. Antibiotics 2021, 10, 767. [CrossRef]
[PubMed]
38. Toro, P.M.; Jara, D.H.; Klahn, A.H.; Villaman, D.; Fuentealba, M.; Vega, A.; Pizarro, N. Spectroscopic Study of theE/ZPhotoisomerization
of a New Cyrhetrenyl Acylhydrazone: A Potential Photoswitch and Photosensitizer(dagger). Photochem. Photobiol. 2021, 97, 61–70.
[CrossRef] [PubMed]
39. Dong, M.X.; Babalhavaeji, A.; Samanta, S.; Beharry, A.A.; Woolley, G.A. Red-Shifting Azobenzene Photoswitches for in Vivo Use.
Acc. Chem. Res. 2015, 48, 2662–2670. [CrossRef] [PubMed]
40. Ciminelli, C.; Granucci, G.; Persico, M. The photoisomerization mechanism of azobenzene: A semiclassical simulation of
nonadiabatic dynamics. Chem. A Eur. J. 2004, 10, 2327–2341. [CrossRef] [PubMed]
41. Gutzeit, V.A.; Acosta-Ruiz, A.; Munguba, H.; Hafner, S.; Landra-Willm, A.; Mathes, B.; Mony, J.; Yarotski, D.; Borjesson, K.;
Liston, C.; et al. A fine-tuned azobenzene for enhanced photopharmacology in vivo. Cell Chem. Biol. 2021, 28, 1648–1663.e16.
[CrossRef]
Pharmaceuticals 2023, 16, 682 34 of 36

42. Mukadum, F.; Nguyen, Q.; Adrion, D.M.; Appleby, G.; Chen, R.; Dang, H.; Chang, R.; Garnett, R.; Lopez, S.A. Efficient Discovery
of Visible Light-Activated Azoarene Photoswitches with Long Half-Lives Using Active Search. J. Chem. Inf. Model. 2021, 61,
5524–5534. [CrossRef]
43. Blanco-Lomas, M.; Martinez-Lopez, D.; Campos, P.J.; Sampedro, D. Tuning of the properties of rhodopsin-based molecular
switches. Tetrahedron Lett. 2014, 55, 3361–3364. [CrossRef]
44. Blanco-Lomas, M.; Funes-Ardoiz, I.; Campos, P.J.; Sampedro, D. Oxazolone-Based Photoswitches: Synthesis and Properties. Eur.
J. Org. Chem. 2013, 2013, 6611–6618. [CrossRef]
45. Martinez-Lopez, D.; Yu, M.L.; Garcia-Iriepa, C.; Campos, P.J.; Frutos, L.M.; Golen, J.A.; Rasapalli, S.; Sampedro, D. Hydantoin-
Based Molecular Photoswitches. J. Org. Chem. 2015, 80, 3929–3939. [CrossRef]
46. Carcia-Iriepa, C.; Ernst, H.A.; Liang, Y.; Unterreiner, A.N.; Frutos, L.M.; Sampedro, D. Study of Model Systems for Bilirubin
and Bilin Chromophores: Determination and Modification of Thermal and Photochemical Properties. J. Org. Chem. 2016, 81,
6292–6302. [CrossRef]
47. Waldeck, D.H. Photoisomerization dynamics of stilbenes. Chem. Rev. 1991, 91, 415–436. [CrossRef]
48. Di Martino, M.; Sessa, L.; Di Matteo, M.; Panunzi, B.; Piotto, S.; Concilio, S. Azobenzene as Antimicrobial Molecules. Molecules
2022, 27, 5643. [CrossRef] [PubMed]
49. Jerca, F.A.; Jerca, V.V.; Hoogenboom, R. Advances and opportunities in the exciting world of azobenzenes. Nat. Rev. Chem. 2022,
6, 51–69. [CrossRef]
50. Wegener, M.; Hansen, M.J.; Driessen, A.J.M.; Szymanski, W.; Feringa, B. Photocontrol of Antibacterial Activity: Shifting from UV
to Red Light Activation. J. Am. Chem. Soc. 2017, 139, 17979–17986. [CrossRef]
51. Burchall, J.J. Mechanism of Action of Trimethoprim-Sulfamethoxazole—II. J. Infect. Dis. 1973, 128, S437–S441. [CrossRef]
52. Hitchings, G.H. Mechanism of Action of Trimethoprim-Sulfamethoxazole—I. J. Infect. Dis. 1973, 128, S433–S436. [CrossRef]
[PubMed]
53. Weston, C.E.; Kramer, A.; Colin, F.; Yildiz, O.; Baud, M.G.J.; Meyer-Almes, F.J.; Fuchter, M.J. Toward Photopharmacological
Antimicrobial Chemotherapy Using Photoswitchable Amidohydrolase Inhibitors. ACS Infect. Dis. 2017, 3, 152–161. [CrossRef]
[PubMed]
54. Jung, M.; Hoffmann, K.; Brosch, G.; Loidl, P. Analogues of trichostatin A and trapoxin B as histone deacetylase inhibitors. Bioorg.
Med. Chem. Lett. 1997, 7, 1655–1658. [CrossRef]
55. Miller, T.A.; Witter, D.J.; Belvedere, S. Histone deacetylase inhibitors. J. Med. Chem. 2003, 46, 5097–5116. [CrossRef]
56. Mai, A.; Massa, S.; Rotili, D.; Cerbara, I.; Valente, S.; Pezzi, R.; Simeoni, S.; Ragno, R. Histone deacetylation in epigenetics: An
attractive target for anticancer therapy. Med. Res. Rev. 2005, 25, 261–309. [CrossRef] [PubMed]
57. Vannini, A.; Volpari, C.; Filocamo, G.; Casavola, E.C.; Brunetti, M.; Renzoni, D.; Chakravarty, P.; Paolini, C.; De Francesco, R.;
Gallinari, P.; et al. Crystal structure of a eukaryotic zinc-dependent histone deacetylase, human HDAC8, complexed with a
hydroxamic acid inhibitor. Proc. Natl. Acad. Sci. USA 2004, 101, 15064–15069. [CrossRef] [PubMed]
58. Kramer, A.; Herzer, J.; Overhage, J.; Meyer-Almes, F.J. Substrate specificity and function of acetylpolyamine amidohydrolases
from Pseudomonas aeruginosa. BMC Biochem. 2016, 17, 4. [CrossRef]
59. Hildmann, C.; Ninkovic, M.; Dietrich, R.; Wegener, D.; Riester, D.; Zimmermann, T.; Birch, O.M.; Bernegger, C.; Loidl, P.;
Schwienhorst, A. A new amidohydrolase from Bordetella or Alcaligenes strain FB188 with similarities to histone deacetylases.
J. Bacteriol. 2004, 186, 2328–2339. [CrossRef]
60. Yao, H.; Wynendaele, E.; Xu, X.L.; Kosgei, A.; De Spiegeleer, B. Circular dichroism in functional quality evaluation of medicines.
J. Pharm. Biomed. Anal. 2018, 147, 50–64. [CrossRef] [PubMed]
61. Yeoh, Y.Q.; Yu, J.X.; Polyak, S.W.; Horsley, J.R.; Abell, A.D. Photopharmacological Control of Cyclic Antimicrobial Peptides.
Chembiochem 2018, 19, 2591–2597. [CrossRef] [PubMed]
62. Irie, M.; Fulcaminato, T.; Matsuda, K.; Kobatake, S. Photochromism of Diarylethene Molecules and Crystals: Memories, Switches,
and Actuators. Chem. Rev. 2014, 114, 12174–12277. [CrossRef] [PubMed]
63. Matsuda, K.; Irie, M. Diarylethene as a photoswitching unit. J. Photochem. Photobiol. C Photochem. Rev. 2004, 5, 169–182. [CrossRef]
64. Huang, Y.-Y.; Wintner, A.; Seed, P.C.; Brauns, T.; Gelfand, J.A.; Hamblin, M.R. Antimicrobial photodynamic therapy mediated by
methylene blue and potassium iodide to treat urinary tract infection in a female rat model. Sci. Rep. 2018, 8, 7257. [CrossRef]
65. Zhao, L.; Zhao, J.; Zhong, K.H.; Tong, A.P.; Jia, D. Targeted protein degradation: Mechanisms, strategies and application. Signal
Transduct. Target. Ther. 2022, 7, 113. [CrossRef]
66. Wang, Z.W.; Liu, Y.; Zhu, X.Q. PhotoPROTACs: A Novel Biotechnology for Cancer Treatment. Trends Cell Biol. 2020, 30, 749–751.
[CrossRef] [PubMed]
67. Liu, J.; Chen, H.; Ma, L.N.; He, Z.X.; Wang, D.; Liu, Y.; Lin, Q.; Zhang, T.H.; Gray, N.; Kaniskan, H.U.; et al. Light-induced control
of protein destruction by opto-PROTAC. Sci. Adv. 2020, 6, eaay5154. [CrossRef]
68. Reynders, M.; Matsuura, B.S.; Berouti, M.; Simoneschi, D.; Marzio, A.; Pagano, M.; Trauner, D. PHOTACs enable optical control
of protein degradation. Sci. Adv. 2020, 6, eaay5064. [CrossRef] [PubMed]
69. Jin, Y.H.; Lu, M.C.; Wang, Y.; Shan, W.X.; Wang, X.Y.; You, Q.D.; Jiang, Z.Y. Azo-PROTAC: Novel Light-Controlled Small-Molecule
Tool for Protein Knockdown. J. Med. Chem. 2020, 63, 4644–4654. [CrossRef] [PubMed]
70. Pfaff, P.; Samarasinghe, K.T.G.; Crews, C.; Carreira, E. Reversible Spatiotemporal Control of Induced Protein Degradation by
Bistable PhotoPROTACs. ACS Cent. Sci. 2019, 5, 1682–1690. [CrossRef]
Pharmaceuticals 2023, 16, 682 35 of 36

71. Winter, G.E.; Buckley, D.L.; Paulk, J.; Roberts, J.M.; Souza, A.; Dhe-Paganon, S.; Bradner, J.E. Phthalimide conjugation as a strategy
for in vivo target protein degradation. Science 2015, 348, 1376–1381. [CrossRef]
72. Sharma, M.; Friedman, S.H. The Issue of Tissue: Approaches and Challenges to the Light Control of Drug Activity. ChemPhotoChem
2021, 5, 611–618. [CrossRef]
73. Yang, Y.M.; Mu, J.; Xing, B.G. Photoactivated drug delivery and bioimaging. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2017,
9, e1408. [CrossRef] [PubMed]
74. Peterson, J.A.; Wijesooriya, C.; Gehrmann, E.J.; Mahoney, K.M.; Goswami, P.P.; Albright, T.R.; Syed, A.; Dutton, A.S.; Smith, E.A.;
Winter, A.H. Family of BODIPY Photocages Cleaved by Single Photons of Visible/Near-Infrared Light. J. Am. Chem. Soc. 2018,
140, 7343–7346. [CrossRef]
75. Liu, M.; Meng, J.Q.; Bao, W.E.; Liu, S.Y.; Wei, W.; Ma, G.H.; Tian, Z.Y. Single-Chromophore-Based Therapeutic Agent Enables
Green-Light-Triggered Chemotherapy and Simultaneous Photodynamic Therapy to Cancer Cells. ACS Appl. Bio Mater. 2019, 2,
3068–3076. [CrossRef]
76. Silva, J.M.; Silva, E.; Reis, R.L. Light-triggered release of photocaged therapeutics—Where are we now? J. Control. Release 2019,
298, 154–176. [CrossRef]
77. Kumari, P.; Kulkarni, A.; Sharma, A.K.; Chakrapani, H. Visible-Light Controlled Release of a Fluoroquinolone Antibiotic for
Antimicrobial Photopharmacology. ACS Omega 2018, 3, 2155–2160. [CrossRef]
78. Shchelik, I.S.; Tomio, A.; Gademann, K. Design, Synthesis, and Biological Evaluation of Light-Activated Antibiotics. ACS Infect.
Dis. 2021, 7, 681–692. [CrossRef]
79. Wong, P.T.; Tang, S.Z.; Mukherjee, J.; Tang, K.; Gam, K.; Isham, D.; Murat, C.; Sun, R.; Baker, J.R.; Choi, S.K. Light-controlled
active release of photocaged ciprofloxacin for lipopolysaccharide-targeted drug delivery using dendrimer conjugates. Chem.
Commun. 2016, 52, 10357–10360. [CrossRef] [PubMed]
80. Verma, S.; Manna, D. Controlling PROTACs with Light. ChemMedChem 2020, 15, 1258–1261. [CrossRef]
81. Xue, G.; Wang, K.; Zhou, D.L.; Zhong, H.B.; Pan, Z.Y. Light-Induced Protein Degradation with Photocaged PROTACs. J. Am.
Chem. Soc. 2019, 141, 18370–18374. [CrossRef]
82. Naro, Y.; Darrah, K.; Deiters, A. Optical Control of Small Molecule-Induced Protein Degradation. J. Am. Chem. Soc. 2020, 142,
2193–2197. [CrossRef]
83. Kounde, C.S.; Shchepinova, M.M.; Saunders, C.N.; Muelbaier, M.; Rackham, M.D.; Harling, J.D.; Tate, E.W. A caged E3 ligase
ligand for PROTAC-mediated protein degradation with light. Chem. Commun. 2020, 56, 5532–5535. [CrossRef]
84. Zengerle, M.; Chan, K.H.; Ciulli, A. Selective Small Molecule Induced Degradation of the BET Bromodomain Protein BRD4. ACS
Chem. Biol. 2015, 10, 1770–1777. [CrossRef] [PubMed]
85. Dommaschk, M.; Grobner, J.; Wellm, V.; Hovener, J.B.; Riedel, C.; Herges, R. Dendronised Ni(II) porphyrins as photoswitchable
contrast agents for MRI. Phys. Chem. Chem. Phys. 2019, 21, 24296–24299. [CrossRef] [PubMed]
86. Zhai, Y.; Busscher, H.J.; Liu, Y.; Zhang, Z.K.; van Kooten, T.G.; Su, L.Z.; Zhang, Y.M.; Liu, J.J.; Liu, J.F.; An, Y.L.; et al. Photoswitch-
able Micelles for the Control of Singlet-Oxygen Generation in Photodynamic Therapies. Biomacromolecules 2018, 19, 2023–2033.
[CrossRef] [PubMed]
87. Lin, W.Y.; Peng, D.W.; Wang, B.; Long, L.; Guo, C.; Yuan, J. A model for light-triggered porphyrin anticancer prodrugs based on
an o-nitrobenzyl photolabile group. Eur. J. Org. Chem. 2008, 2008, 793–796. [CrossRef]
88. Tessaro, A.L.; Fraix, A.; Failla, M.; Cardile, V.; Graziano, A.C.E.; Estevao, B.M.; Rescifina, A.; Sortino, S. Light-Controlled
Simultaneous “On Demand” Release of Cytotoxic Combinations for Bimodal Killing of Cancer Cells. Chem. A Eur. J. 2018, 24,
7664–7670. [CrossRef]
89. Feng, Y.F.; Tonon, C.C.; Ashraf, S.; Hasan, T. Photodynamic and antibiotic therapy in combination against bacterial infections:
Efficacy, determinants, mechanisms, and future perspectives. Adv. Drug Deliv. Rev. 2021, 177, 113941. [CrossRef]
90. Branco, T.M.; Valerio, N.C.; Jesus, V.I.R.; Dias, C.J.; Neves, M.; Faustino, M.A.F.; Almeida, A. Single and combined effects of
photodynamic therapy and antibiotics to inactivate Staphylococcus aureus on skin. Photodiagn. Photodyn. Ther. 2018, 21, 285–293.
[CrossRef]
91. Xing, B.G.; Jiang, T.T.; Bi, W.G.; Yang, Y.M.; Li, L.H.; Ma, M.L.; Chang, C.K.; Xu, B.; Yeow, E.K.L. Multifunctional divalent
vancomycin: The fluorescent imaging and photodynamic antimicrobial properties for drug resistant bacteria. Chem. Commun.
2011, 47, 1601–1603. [CrossRef] [PubMed]
92. Liu, F.; Ni, A.S.Y.; Lim, Y.; Mohanram, H.; Bhattacharjya, S.; Xing, B.G. Lipopolysaccharide Neutralizing Peptide-Porphyrin
Conjugates for Effective Photoinactivation and Intracellular Imaging of Gram-Negative Bacteria Strains. Bioconjugate Chem. 2012,
23, 1639–1647. [CrossRef]
93. Dastgheyb, S.S.; Eckmann, D.M.; Composto, R.J.; Hickok, N.J. Photo-activated porphyrin in combination with antibiotics:
Therapies against Staphylococci. J. Photochem. Photobiol. B Biol. 2013, 129, 27–35. [CrossRef]
94. Almeida, J.; Tome, J.P.C.; Neves, M.; Tome, A.C.; Cavaleiro, J.A.S.; Cunha, A.; Costa, L.; Faustino, M.A.F.; Almeida, A. Photody-
namic inactivation of multidrug-resistant bacteria in hospital wastewaters: Influence of residual antibiotics. Photochem. Photobiol.
Sci. 2014, 13, 626–633. [CrossRef]
95. Iluz, N.; Maor, Y.; Keller, N.; Malik, Z. The synergistic effect of PDT and oxacillin on clinical isolates of Staphylococcus aureus.
Lasers Surg. Med. 2018, 50, 535–551. [CrossRef]
Pharmaceuticals 2023, 16, 682 36 of 36

96. Thangudu, S.; Kaur, N.; Korupalli, C.; Sharma, V.; Kalluru, P.; Vankayala, R. Recent advances in near infrared light responsive
multi-functional nanostructures for phototheranostic applications. Biomater. Sci. 2021, 9, 5432–5443. [CrossRef]
97. Wang, X.; Xuan, Z.; Zhu, X.; Sun, H.; Li, J.; Xie, Z. Near-infrared photoresponsive drug delivery nanosystems for cancer
photo-chemotherapy. J. Nanobiotechnol. 2020, 18, 108. [CrossRef]
98. Vargason, A.M.; Anselmo, A.C.; Mitragotri, S. The evolution of commercial drug delivery technologies. Nat. Biomed. Eng. 2021, 5,
951–967. [CrossRef] [PubMed]
99. Lu, C.; Di, L. In vitro and in vivo methods to assess pharmacokinetic drug– drug interactions in drug discovery and development.
Biopharm. Drug Dispos. 2020, 41, 3–31. [CrossRef] [PubMed]
100. Said, S.S.; Campbell, S.; Hoare, T. Externally Addressable Smart Drug Delivery Vehicles: Current Technologies and Future
Directions. Chem. Mater. 2019, 31, 4971–4989. [CrossRef]

Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.

You might also like