ArticlePDF AvailableLiterature Review

Hypericin-mediated photodynamic therapy for the treatment of cancer: A review

Authors:

Abstract and Figures

Objectives Hypericin is a polycyclic aromatic naphthodianthrone that occurs naturally. It is also an active ingredient in some species of the genus Hypericum. Emerging evidence suggests that hypericin has attracted great attention as a potential anticancer drug and exhibits remarkable antiproliferative effect upon irradiation on various tumour cells. This paper aims to summarise the anticancer effect and molecular mechanisms modulated by hypericin-medicated photodynamic therapy and its potential role in the cancer treatment. Key findings Hypericin-medicated photodynamic therapy could inhibit the proliferation of various tumour cells including bladder, colon, breast, cervical, glioma, leukaemia, hepatic, melanoma, lymphoma and lung cancers. The effect is primarily mediated by p38 mitogen-activated protein kinase (MAPK), JNK, PI3K, CCAAT-enhancer-binding protein homologous protein (CHOP)/TRIB3/Akt/mTOR, TRAIL/TRAIL-receptor, c-Met and Ephrin-Eph, the mitochondria and extrinsic signalling pathways. Furthermore, hypericin-medicated photodynamic therapy in conjunction with chemotherapeutic agents or targeted therapies is more effective in inhibiting the growth of tumour cells. Summary During the past few decades, the anticancer properties of photoactivated hypericin have been extensively investigated. Hypericin-medicated photodynamic therapy can modulate a variety of proteins and genes and exhibit a great potential to be used as a therapeutic agent for various types of cancer.
Content may be subject to copyright.
425
Journal of Pharmacy and Pharmacology, 2021, Vol 73, 425–436
doi:10.1093/jpp/rgaa018
Review
Advance Access publication 24 December 2020
© The Author(s) 2020. Published by Oxford University Press on behalf of Royal Pharmaceutical Society. All rights reserved.
For permissions, please e-mail: journals.permissions@oup.com
Review
Hypericin-mediated photodynamic therapy for
the treatment of cancer: areview
XiaoxvDong1,2,, YawenZeng1, ZhiqinZhang1, JingFu3, LongtaiYou1,
YuanyuanHe2, YangHao2, ZiliGu2, ZhenfengYu2, ChanghaiQu1,
XingbinYin1, JianNi1,4 and LuisJ.Cruz2,*
1School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
2Department of Radiology, Division of Translational Nanobiomaterials and Imaging, Leiden University Medical Center,
Leiden, The Netherlands
3Beijing Institute of Traditional Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical
University, Beijing, China
4Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
*Correspondence: Luis J. Cruz, Department of Radiology, Division of Translational Nanobiomaterials and Imaging, Room
C2-187h, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands. Email: l.j.cruz_ricondo@lumc.nl
Received April 4, 2020; Accepted October 5, 2020.
Abstract
Objectives Hypericin is a polycyclic aromatic naphthodianthrone that occurs naturally. It is also
an active ingredient in some species of the genus Hypericum. Emerging evidence suggests that
hypericin has attracted great attention as a potential anticancer drug and exhibits remarkable
antiproliferative effect upon irradiation on various tumour cells. This paper aims to summarise the
anticancer effect and molecular mechanisms modulated by hypericin-medicated photodynamic
therapy and its potential role in the cancer treatment.
Key findings Hypericin-medicated photodynamic therapy could inhibit the proliferation of various
tumour cells including bladder, colon, breast, cervical, glioma, leukaemia, hepatic, melanoma,
lymphoma and lung cancers. The effect is primarily mediated by p38 mitogen-activated protein kinase
(MAPK), JNK, PI3K, CCAAT-enhancer-binding protein homologous protein (CHOP)/TRIB3/Akt/mTOR,
TRAIL/TRAIL-receptor, c-Met and Ephrin-Eph, the mitochondria and extrinsic signalling pathways.
Furthermore, hypericin-medicated photodynamic therapy in conjunction with chemotherapeutic
agents or targeted therapies is more effective in inhibiting the growth of tumour cells.
Summary During the past few decades, the anticancer properties of photoactivated hypericin have
been extensively investigated. Hypericin-medicated photodynamic therapy can modulate a variety
of proteins and genes and exhibit a great potential to be used as a therapeutic agent for various
types of cancer.
Keywords: Hypericin; photosensitiser; photodynamic therapy; molecular targets
Introduction
Photodynamic therapy (PDT) is an emerging noninvasive therapy
that possesses many advantages over traditional cancer therapies
(surgery, chemotherapy and radiation therapy), such as minimal
invasiveness and limited side effects, and low mutation rate. [1, 2] In
addition, PDT could be used to treat a variety of diseases, including
bacterial infections, psoriasis and age-related macular degener-
ation.[3, 4] PDT consists of three essential elements: photosensitiser,
Downloaded from https://academic.oup.com/jpp/article/73/4/425/6047495 by Universiteit Leiden / LUMC user on 18 October 2021
426 Journal of Pharmacy and Pharmacology, 2021, Vol. 73, No. 4
light and oxygen.[5, 6] The photosensitiser is irradiated under the
appropriate wavelength of light, and the singlet basic energy state
is converted to the excited state due to photon absorption, which
might spontaneously undergo intersystem crossing to form a rela-
tively stable triplet state (Figure1). The triplet state photosensitiser
reacts with molecule oxygen and then generates reactive oxygen
species (ROS), which can direct cell killing, devastate the vascular
system and stimulate immune response. [7] Hence, the photosensitiser
plays a critical role in the PDT process.
Hypericin (4,5,7,4,5,7-hexahydroxy-2,2-
dimethylnaphthodianthrone) (HYP; Figure 2) is a hydroxyl-
ated phenanthroperylenequinone in some species of the genus
Hypericum. The photoactivation behaviour of HYP is induced by
the delocalised π-electron system in its aromatic rings.[8, 9] A lot of
researches have conrmed that HYP selectively accumulates in tu-
mour tissue via diffusion, pinocytosis or endocytosis in PDT of tu-
mours.[1013] Hypericin mainly accumulates in the membranes of the
endoplasmic reticulum (ER), lysosomes, Golgi apparatus and mito-
chondria due to its hydrophobic character.[14, 15] Furthermore, HYP
has a maximum absorption at 590nm and generates high singlet
oxygen. It also has minimal dark toxicity, excellent photosensitivity
and high clearance rate. [16, 17] In all, these advantages make HYP to
be a promising excellent photosensitiser, as well as its photochem-
ical properties in PDT. Hence, the purpose of this review is intended
to summarise the anticancer activity and molecular mechanisms of
HYP-medicated PDT (HYP-PDT) and highlight its potential thera-
peutic value in cancer treatment.
Anticancer Activity
Bladdercancer
De Witte’s group for the rst time reported that HYP-PDT could
induce apoptosis or necrosis in AY-27 urinary bladder carcinoma
in a concentration-dependent manner. This indicated that HYP
might play an important role for the PDT of supercial bladder car-
cinoma.[18] The mechanism of HYP-PDT in AY-27 cells was through
vascular effects rather than mediated by cellular destruction in these
tumours.[19] Moreover, it was shown for the rst time that HYP-
PDT could be used to induce selective AY-27 cells damage without
damaging the detrusor musculature.[20] In another experiment, their
group also evaluated the biodistribution of HYP in AY-27 cells line
and normal bladder wall. The data demonstrated that there was
much more HYP uorescence intensity in tumour cells without
causing damage to the underlying muscle layers. [21] Huygens etal.[22]
showed that HYP combined with hyperoxygenation resulted in a
virtually complete bladder RT-112 cancer cells killing through apop-
tosis. Similarly, peruorocarbons were also applied in increasing
the photodynamic activity of HYP in RT-112 carcinoma spheroids.
Since HYP was specically concentrated in bladder carcinoma cells,
indicating that HYP combined with peruorocarbons could be util-
ised for effective bladder wall photodynamic treatment.[23] Kocanova
etal.[24] studied the signalling pathways leading to the upregulation
of heme oxygenase (HO-1) enzyme in T24 tumour cells treated
with HYP-PDT. This mechanism might involve the stimulation of
HO-1 expression controlled by the PI3K and p38 mitogen-activated
protein kinase (MAPK) pathways. Buytaert etal.[25] investigated the
molecular targets of HYP-induced cell apoptosis in T24 cells. The
results indicated that inhibition of p38 MAPK prevented the autono-
mous regeneration and migration. It has been shown that HYP-PDT
combined with angiogenesis inhibitor (bevacizumab) signicantly
downregulated the expression of vascular endothelial growth factor
(VEGF) proteins in the MGH cells.[26] Moreover, HYP-PDT induced
the upregulation of angiogenic proteins including VEGF, interferon-α
(IFN-α), tumor necrosis growth factor-α (TNF-α) and basic bro-
blast growth factor (bFGF) in MGH xenograft model. Hepatocyte
growth factor (HGF) and Ephrin-A3 (EFNA3) proteins were also
increased by the activation of c-Met and Ephrin-Eph pathways.[27]
Coloncancer
Blank et al.[28] evaluated the wavelength effects of HYP-PDT in a
C26 colon carcinoma model. In-vitro irradiation of HYP-sensitised
cells resulted in a loss of cell viability in a light dose-dependent
manner. In-vivo irradiation of C26-derived tumours, HYP caused
extensive vascular damage and tumour necrosis. It was shown for
the rst time that pre-treatment with proadifen affected the function
Figure 1 Mechanism of photoactivation of photosensitizer.
Figure 2 Chemical structure of hypericin.
Downloaded from https://academic.oup.com/jpp/article/73/4/425/6047495 by Universiteit Leiden / LUMC user on 18 October 2021
Journal of Pharmacy and Pharmacology, 2021, Vol. 73, No. 4 427
of multidrug resistance-associated protein (MRP1) and breast cancer
resistance protein (BCRP), leading to an increase of HYP content
and the improvement of HY-PDT.[29] 5-Lipoxygenase (5-LOX) in-
hibitor (MK-886) and HYP-PDT also induced apoptosis and the
accumulation in the S phase of HT-29 cells, along with the repres-
sion of DNA synthesis.[30] Moreover, the mechanisms associated
with the cell cycle progression of HT-29 cells were accompanied by
increase in cyclin E level and decrease of cyclin A, cdk-2 and pRb
expression. The stimulation of apoptosis was accomplished prefer-
entially via the mitochondrial pathway.[31] Asimilar study showed
that HYP-PDT combined with hyperforin stimulated the onset of
apoptosis in HT-29 cells through inhibiting cell cycle arrest and sup-
pressing expression of matrix metalloproteinase-2 (MMP-2)/-9.[32]
Furthermore, the expression of some pro-angiogenic factors, such as
VEGF-A, PD-ECGF, PDGF-A (V1) and PDGF-A (V2) in HT-29 cells,
was increased.[33, 34] The nonspecic cyclooxygenase (COX) inhibi-
tors or polyunsaturated fatty acids potentiated the cytotoxic effect
of HYP-PDT.[35, 36] Treatment with HYP and the inhibitor of survivin
expression (YM155) caused more severe dissipation of mitochon-
drial membrane potential and induced the activation of caspase-3
and poly(ADP-ribose)polymerase (PARP) proteins in HT-29 cells.[37]
Similarly, a synergistic combination of photoactive HYP and
Manumycin Aexhibited anticancer effects on oxaliplatin-resistant
HT-29-OxR cells. This mechanism might involve the inhibition
of cell viability, colony formation and induction of apoptosis.[38]
Meanwhile, HYP-PDT decreased the volume of the primary tu-
mour and signicantly prolonged the survival of the CT26 tumour-
bearing BALB/c mice by inducing antitumoral immune response.[39]
Lin etal.[40] reported that HYP-PDT caused severe ER stress and led
to the induction of CCAAT-enhancer-binding protein homologous
protein (CHOP), which thereby activated CHOP/TRIB3/Akt/mTOR
pathway and triggered autophagic cell death of HCT116 and HCT8
cells. Their group also found that treatment with HYP-PDT en-
hanced the antitumour activity of oxaliplatin in HCT8 and HCT116
cells. This was mediated by ROS, and its mechanism involved af-
fecting drug efux and GSH-related detoxication.[41] Furthermore,
the inhibition of ABCG2 potentially enhanced the efcacy of HYP-
PDT in 3D spheroid models of HT-29 cells.[42] In another study,
F-actin and dysadherin could be the target proteins for HYP-PDT
in HT-29 cells.[43] Montanha etal.[44] investigated the photodynamic
activity of HYP in Pluronic P123 against HT-29 cells. The data dem-
onstrated that the nanocarrier played an important role in the pene-
tration of HYP through cell membranes, suggesting that HYP was
a promising photosensitiser that might be suitable for the treatment
of colon diseases. Similarly, the phototoxic effects of HYP loaded on
superparamagnetic iron oxide nanoparticles were also studied. The
results indicated that nanoparticle-loaded HYP resulted in a com-
plete stop of HT-29 cells’ proliferation and induction of cell death.[45]
In another study, HYP-PDT exhibited immunomodulatory activities
resulting from an increase of interleukin-8 (IL-8) secretion in SW620
cells and a signicant decrease in IL-8 in the SW480 cells. [46]
Breastcancer
Ferenc et al.[47] found that combination of HYP-PDT and genistein
inhibited the proliferation of MCF-7 and MDA-MB-231 cells and
induced their apoptosis. The mechanisms were related to alterations
in the protein levels of Bcl-2, Bax, Akt and cell cycle perturbations
in G2/M-phase. It has been shown that HYP-PDT combined with
17-dimethylaminoethylamino-17-demethoxygeldanamycin (17-
DMAG) obviously decreased the metabolic activity and viability
of SKBR-3 cells through decreasing the levels of human epidermal
growth factor receptor-2 (HER2), Akt, survivin and P-Erk1/2.[48]
Similarly, photoactivated HYP signicantly decreased the mRNA
and protein expression of HER2 in SKBR-3 and MCF-7 cells.[49]
HY-PDT and MK-886 also synergistically induced the MCF-7 cell
death.[50] Asimilar study showed that combination of HYP-PDT and
carmustine inhibited the reduction of GSH, resulting in an increase in
the sensitivity of MCF-7 cells.[51] Photoactivated HYP increased the
production of ROS in MCF-7 and MDA-MB-231 cells. Additionally,
the superoxid dismutase-2 (SOD-2) inhibitor (methoxyestradiol)
signicantly enhanced the sensitivity of MCF-7 cells to HYP.[52]
In-vitro assays have shown that HYP and hypericinates permeated
the membrane of MCF-7 cells and accumulated in organelles near
the nucleus. Furthermore, the photodynamic studies showed that
HYP inhibited the formation of cellular colonies, indicating a pos-
sible ability to prevent the recurrence of tumours.[53] Gao et al.[54]
demonstrated that single-walled carbon nanohorns (SWNH)-HYP
effectively ablated the 4T1 mouse breast cells upon light irradiations.
Moreover, the remarkable tumour growth inhibition as well as tu-
mour cell death was proved by PDT of SWNH-HYP. In another ex-
periment, the hybrid therapy of HYP-PDT and tamoxifen exhibited
90% cytotoxicity in MDA-MB-231 and MCF-7 cells. The cytotox-
icity was in the form of necrosis and autophagy accompanied by
high levels of lipid peroxidation.[55]
Cervicalcancer
Delaey et al.[56] demonstrated for the rst time that HYP-induced
photocytotoxicity in HeLa cells depends on the cells’ number, as low
cell density cultures were more responsive to PDT than conuent
cell cultures. The inhibition of JNK and p38 MAPK upregulated the
HYP-PDT-induced apoptosis of HeLa cells, which might provide
molecular basis for developing new therapeutic strategies.[57] The
apoptosis induced by HYP-PDT in HeLa cells was mediated by the
mitochondrial pathway, which involved the release of cytochrome c
and the further activation of caspase-3.[58] HYP-PDT and pyridinyl
imidazole might improve the therapeutic effect by preventing the
upregulation of COX-2, which contributed to the growth of HeLa
cells through the release of pro-angiogenic factors, as well as by
sensitising the cells to apoptosis.[59] Similarly, Bianchini etal.[60] re-
ported that the complexation of HYP with apomyoglobin (apoMb)
improved its efcacy as a photosensitiser on HeLa cells. Application
of the HYP-apoMb to HeLa tumour spheroids followed by illumin-
ation obviously decreased the growth rate of the structure. It has
been shown that PDT with ER-associated HYP induced the imme-
diate loss of SERCA2 protein levels, leading to HeLa cells death and
disruption of Ca2+ homeostasis.[61] Moreover, Barras investigated the
photodynamic effect of HYP-loaded lipid nanocapsules on HeLa
cells. The results demonstrated that the nanocapsules held promise
for potential application for cervical cancer due to their extended
circulation and accumulation in tumour-bearing mice.[62]
Nasopharyngealcancer
Du et al.[63] evaluated the expression of IL-6 in two different
nasopharyngeal cancer (NPC) cells after HYP-PDT. The results
showed the transcription of IL-6 was obviously elevated in PDT-
treated poorly differentiated CNE-2 cells but not in well differ-
entiated HK-1 cells. In vivo, the mRNA expression of IL-6 was
also upregulated in CNE-2 cells. However, HYP-PDT did not alter
the expression level of IL-8 in HK-1 and CNE-2 cells.[64] HYP-
PDT induced the phototoxicity in HK1 cells, and led to tumour
shrinkage and necrosis with accompanying lipid peroxidation
in the HK-1 murine tumour model.[65] Moreover, there was
Downloaded from https://academic.oup.com/jpp/article/73/4/425/6047495 by Universiteit Leiden / LUMC user on 18 October 2021
428 Journal of Pharmacy and Pharmacology, 2021, Vol. 73, No. 4
signicant reduction of glutathione S-transferase (GST) activity
in HK-1 and CNE-2 cells as well as in the HK-1 tumour tis-
sues.[66] Photoactivation of HYP elicited an increase in the protein
and mRNA expression of MMP-1 in HK-1 and CNE-2 cells.
Similarly, the mRNA expression of MMP1 was also upregulated
in HYP-PDT-treated HK-l tumours.[67] In addition, HYP-PDT
downregulated the expression of MMP-9 by inhibiting the pro-
duction of granulocyte-macrophage colony-stimulating factor
(GM-CSF), thereby regulating the transcriptional effect of AP1/
NF-kB.[68] HYP-PDT could activate cysteine-type endopeptidase,
which inhibited the growth and induced the apoptosis of CNE-2
cells through the mitochondrial-dependent intrinsic and extrinsic
pathways.[69] Furthermore, HYP-PDT activated the p38 MAPKs
by generating singlet oxygen. The inhibition of p38 MAPKs
pathway with chemical inhibitors could enhance the apoptosis
of HK-1 cells induced by HYP-PDT.[70] HYP-PDT had been re-
ported to exhibit the desirable characteristics of inducing more
apoptosis and lowering serum VEGF levels in the HK-1 tumour
murine model.[71] Olivo etal. conducted a study to improve the
tumour growth by using HYP-PDT and celebrex together. The
data showed that the mRNA level of COX-2 was decreased in
CNE-2 cells in vitro. Meanwhile, the downregulation of COX-2
and hypoxia inducible factor-lα (HIF-lα) genes was observed in
vivo.[72] Additionally, their group reported that HYP-PDT along
with celebrex decreased the protein level of VEGF, indicating that
angiogenesis inhibitors might enhance the therapeutic efcacy of
HYP-PDT in HK-1 cells.[73]
Glioma
PDT can be utilised to regulate the proliferation of glioma cells,
which belong to the most inltrative types of cancers. Miccoli
etal.[74] were the rst to show that photoactivation of HYP affected
the energy metabolism of the SNB-19 cells through suppressing
hexokinase binding to mitochondria. These data demonstrated
that HYP could be a promising phototoxic drug for the treat-
ment of glioma tumours. The intracellular localisation of HYP in
D54Mg cells was investigated by Uzdensky etal.[75] The results
indicated that HYP was concentrated in the perinucleolar cyto-
plasmic area mainly on one side of the nucleus.[75] Furthermore,
HYP-PDT effectively inactivated three glioblastoma cells (U373
MG, LN229 and T98G) after a short incubation and exposure
to low light.[76] Balogová studied the distribution patterns of Bax
and Bak after HYP-PDT treatment in U-87 MG cells. The data
demonstrated that Bak was located predominantly at the plasma
membrane, while Bax was translocated from cytosol to mitochon-
dria.[77] In another experiment, photoactivated HYP exhibited an
inhibitory activity on U-87 MG and U-373 MG cells. However,
the efcacy of the inhibitory effect was involved in key steps of
angiogenesis, such as the cell proliferation, migration and degrad-
ation of MMP.[78] HYP is one of the protein kinase C (PKC) regu-
lators, which are considered as promising drugs that can mediate
glioma cells apoptosis in PDT. HYP-PDT induced the activation
and translocation of PKC along the plasma membrane and par-
tially in the nucleus of U87 MG cells. The prolonged effect of
HYP-PDT resulted in PKC being located mainly in the cytoplasm
and nucleus.[79] In addition, HYP and PKCδ inhibitor (rottlerin)
after light irradiation led to a signicant increase in apoptosis of
U87 MG cells accompanied by the downregulation of intracel-
lular oxidative stress and upregulation of phosphorylated Bcl-2
in the cytoplasm.[80]
Leukaemia
Lavie et al.[81] reported that photoactivation HYP and dimethyl
tetrahydroxyhelianthrone (DTHe) induced apoptosis and necrosis
with nucleolar condensation of chromatin in HL-60 and K-562 cells.
HYP-PDT also inhibited the proliferation of Jurkat cells by induction
of apoptosis. The mechanism might be mediated by the activation
of the TRAIL/TRAIL-receptor system and caspase-8.[82] Moreover,
HYP loaded on superparamagnetic iron oxide nanoparticles
(SPIONs) following laser irradiation induced the death of Jurkat
cells in a dose and illumination time-dependent manner due to the
generation of ROS.[83] Xu etal.[84] investigated the cytotoxic effect
of HYP-PDT on the growth of K562 cells as well as its underlying
mechanism. The results demonstrated that HYP-PDT reduced the
viability and induced mitochondria-dependent apoptosis by regu-
lating the JNK signalling pathway. The overexpression of BCRP
can confer a multidrug-resistant (MDR) phenotype on cancer cells.
Pretreatment with BCRP inhibitor (Ko143) signicantly increased
HYP accumulation and sensitised HL-60 cells to HYP.[85] Treatment
with HYP-PDT was highly effective against adult T-cell leukemia
(ATL) cells through mitochondrial-dependent apoptotic signalling
and suppression of viral transcription.[86]
Livercancer
HYP-PDT resulted in the decrease of cell viability, proliferative ac-
tivity and apoptosis in hepatoblastoma (HUH6 and HepT1) and
HepG2 cells.[87] Barathan etal.[88] also showed that treatment with
HY-PDT induced apoptosis in HepG2 cells by promoting the gen-
eration of ROS, and recruited apoptosis mediators and IL-6. Fadel
et al.[89] found that the binding of anti-hepatocyte-specic antigen
(anti-HAS) to HYP played an important part in increasing the mor-
tality of HepG2 cells compared with free HYP. This suggested that
the conjugation of HYP with anti-HAS might be a potential agent
for liver cancer. Moreover, HYP-loaded on polydopamine-coated
cerium oxide nanorods under light illumination had specic targeting
ability and good biocompatibility. It also specically recognised the
asialo-glycoprotein (ASGP) receptors, which were overexpressed on
the membrane of HepG2 cells.[90]
Melanoma
Melanoma is the main cause of death in skin cancers. Hadjur etal.[91]
for the rst time demonstrated that the activity of total SOD was
increased in G361, M18 and M6 cells, while the glutathione perox-
idase and catalase activities were decreased. Davids etal.[92] showed
that treatment with HYP-PDT induced different cell death modes
in melanoma cells associated with the absence or presence of a pig-
mented phenotype. In addition, UCT Mel-1 pigmented cells died
due to necrosis, whereas UCT Mel-3 nonpigmented cells died via
apoptosis.[92] However, both melanoma cell types exhibited an ini-
tial cytoprotective response to HYP-PDT cytotoxicity by inducing
autophagy.[93] In another study, their group studied that HYP-PDT
signicantly induced UCT Mel-1 cells death after depigmentation
through a likely mechanism involving a necrotic or autophagic mode
of cell death.[94] HYP-PDT was effective in inhibiting the growth of
A375 and UCT Mel-1 cells through participating in the loss of cell
membrane integrity and the externalisation of phosphatidylserine.
In addition, this treatment led to extrinsic and intrinsic caspase-
dependent apoptotic modes of cell death.[95] Under the illumination
effect, HYP in 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC)
liposomes had a higher selectivity index in B16-F10 cells than Hyp
solubilised in ethanol.[96]
Downloaded from https://academic.oup.com/jpp/article/73/4/425/6047495 by Universiteit Leiden / LUMC user on 18 October 2021
Journal of Pharmacy and Pharmacology, 2021, Vol. 73, No. 4 429
Lymphoma
Chen etal.[97] evaluated the PDT efcacy of HYP in a P388 murine
lymphoma model. The results indicated that HYP-PDT was ef-
fective in prolonging the survival time. Moreover, the damage of
tumour vasculature might be the main mechanism of HYP-PDT
action. Treatment with HYP-PDT decreased the viability of U937
cells through altering the levels of apoptosis-related proteins, such
as Bcl-2, Bax and PARP.[98] Asimilar study showed that HYP-PDT
signicantly restrained the proliferation and induced apoptosis of
U937 cells. However, intracellular ATP levels increased markedly.[99]
Compared with the placebo vehicle, HYP combined with visible
light led to a signicant improvement in skin lesions in most patients
with cutaneous T-cell lymphoma.[100]
Squamouscancer
Due to the limitations of surgery and recurrence of the disease, the
clinical treatment of patients with squamous cancer is still poor.
Blank et al.[101] evaluated the tumoricidal properties of HYP-PDT
in a highly metastatic adenocarcinoma (DA3Hi) and anaplastic SQ2
tumours in vivo. The data demonstrated that HYP-PDT reduced
the growth of primary tumours and obviously prolonged the sur-
vival of tumour-bearing mice. Additionally, HY-PDT signicantly
upregulated the mRNA levels of inammatory cytokines (IL-1β,
IL-6, GM-CSF and TNF-α). In-vitro testing revealed that HYP-
PDT exhibited an optimal tumoricidal response in SNU-1 cells.
Moreover, in-vivo tumour retention of injected HYP following laser
led to regression of SNU-1 tumour transplants.[102] Moreover, HYP
combined with pulsed laser light resulted in the membrane damage
of the SNU-1 cells. The phototoxic response increased linearly with
HYP dose of 0.1–2µm.[103] Sanovic etal.[104] investigated gene ex-
pression proles of A-431 cells to analyse the molecular background
for skin treatment and apoptosis induced by HYP-PDT. The results
reported that 168 genes were signicantly upregulated and 45 genes
were downregulated. These functions were related to the antioxidant
response, adaptation mechanism and cell damage protection func-
tion against ROS. HYP-PDT also decreased the cell viability of SCC
cells in a concentration-dependent manner via increasing the ROS
production. Moreover, the cell death mode was a necrotic caspase-
independent mechanism.[105] The FaDu cells treated with HYP-PDT
exhibited a large amount of metabolic reduction and excessive apop-
tosis, indicating that HYP might be suitable for clinical application
of head neck squamous cancers.[106] In another study, the mixtures
[HYP and meso-tetra-hydroxyphenyl-chlorin (mTHPC)] decreased
the cell viability and increased the level of ROS in UMB-SCC 745
and 969 cells. Examination of death signalling pathways exhibited
that the mixtures-mediated cell death was apoptotic and necrotic.[107]
Othercancer
Liu was the rst to show that HYP-PDT signicantly decreased
the growth of MiaPaCa-2 and PANC-1 cells in vivo and in vitro,
indicating that HYP and laser therapy might be effective in pancre-
atic cancer.[108] Chen etal.[109,110] reported that HYP-PDT induced vas-
cular injury and apoptosis in a tumour model of radiation-induced
brosarcoma-1 (RIF-1) mice. Their studies also showed that the
Figure 3 Anticancer effect of hypericin.
Downloaded from https://academic.oup.com/jpp/article/73/4/425/6047495 by Universiteit Leiden / LUMC user on 18 October 2021
430 Journal of Pharmacy and Pharmacology, 2021, Vol. 73, No. 4
Table 1 Anticancer activities of HYP-PDT
Cell lines/model Drug Dose Laser Energy Mechanism(s) of action Application Reference
AY-27 cells 0.05–10μm 0.45–3.6 J/cm2Induces apoptosis or necrosis In vitro [18]
30μm 6–48 J/cm2Induces selective the cells damage In vitro [20]
RT-112 cells 5,30,100μm 7.2 J/cm2Induces apoptosis In vitro [22]
T24 cells 125, 150 nm 4 J/cm2Involves the stimulation of HO-1 expression
governed by the p38 MAPK and PI3K
pathways
In vitro [24]
150 nm 4 J/cm2Inhibits p38 MAPK pathway In vitro [25]
Mice, MGH cells 5mg/kg 120 J/cm2Downregulates the VEGF expression In vivo [26]
5mg/kg 120 J/cm2Activates c-Met and Ephrin-Eph signalling
pathways
In vivo [27]
C26 cells 0.5–4μm
5mg/kg
60–120 J/cm2Induces vascular damage and tumour necrosis In vitro and
vivo
[28]
HT-29 cells 0.1μm 3.15 J/cm2Affects the function of MRP1 and BCRP In vitro [29]
0.1μm 4.4 J/cm2Induces S phase arrest and increases apoptosis In vitro [30]
0.04, 0.1μm 4.4 J/cm2The mitochondrial pathway In vitro [31]
30, 50 nm 3.15 J/cm2Suppresses expression of MMP-2 In vitro [32]
50, 500 nm 3.15 J/cm2Increases expression of VEGF-A, PD-ECGF In vitro [33]
50–200 nm 3.15 J/cm2Induces an increase in caspase-3 activation and
subsequent PARP cleavage
In vitro [37]
0–100 nm 4.4 J/cm2Inhibits cell viability and Induces apoptosis In vitro [38]
0–200 nm 1 J/cm2Upregulates the ABCG2 In vitro [42]
Mice, CT26 cells 1mg/ml 14, 60 J/cm2Induces immune response In vivo [39]
HCT116 and
HCT8 cells
0.0625–1μm 7.2 J/cm2Activates CHOP/TRIB3/Akt cascade and
triggered autophagic cell death
In vitro [40]
0–2.5μm 7.2 J/cm2Affects drug efux and GSH detoxication In vitro [41]
MCF-7 cells 0.021μm 4.4 J/cm2Regulates Bcl-2, Bax, Akt In vitro [47]
0.04, 0.1μm 3.15 J/cm2Induces cell death In vitro [50]
2μm 200 mJ/cm2Inhibits the GSH reduction In vitro [51]
0.1–50μm 0.96 J/cm2Inhibits the formation of cellular colonies In vitro [53]
2μm
12.5mg/kg
40 J/cm2Induces necrosis and autophagy In vitro and
vivo
[55]
SKBR-3 cells 0.84–210 nm 4.4 J/cm2Decreases HER2, Akt, P-Erk1/2 and In vitro [48]
50, 250 nm 3.15 J/cm2survivin [49]
Mice, 4T1 cells 10μg/ml
25.6μg/ml
150 J/cm2Inhibits tumour growth In vitro and
vivo
[54]
HeLa cells 125 nm 4 J/cm2Inhibits JNK and p38 MAPK In vitro [57]
80 nM-10μm 4 J/cm2The mitochondrial pathway In vitro [58]
150 nm 4 J/cm2Blocks COX-2 upregulation In vitro [59]
200 nm 4 J/cm2Induces loss of SERCA2 protein levels In vitro [61]
Mice, CNE-2 cells 0.5μm
2mg/kg
0.5 J/cm2Upregulates IL-6 mRNA expression In vitro and
vivo
[63]
0.5–4.0μg/ml 5.67 J/cm2The mitochondria pathway and the extrinsic
pathway
In vitro [69]
2mg/kg 42.4 J/cm2Decreases the COX-2 mRNA expression In vivo [72]
Mice, HK-1 cells 2mg/kg 120 J/cm2 Induces tumour shrinkage and necrosis In vivo [65]
2mg/kg 120 J/cm2Reduces GST activity In vivo [66]
0.125–2μm 0.25 J/cm2Upregulates MMP1 mRNA expression In vitro [67]
0.25, 0.5μm
2mg/kg
0.5 J/cm2Downregulates MMP-9 expression; Induces
apoptosis and lowers VEGF
In vitro and
vivo
[68]
2, 5mg/kg 30 J/cm2levels In vivo [71,73]
SNB-19 cells 0.5, 2.5μg/ml 1.5 J/cm2Inhibits hexokinase bound to mitochondria In vitro [72]
U-87 MG cells 500 nm 4 J/cm2Induces Bax translocated from cytosol to
mitochondria
In vitro [77]
5×10–7 M 5 J/cm2Inhibits angiogenesis In vitro [78]
500 nm 4 J/cm2Activates PKC In vitro [79]
0.5μm 4 J/cm2Induces apoptosis In vitro [80]
HL-60 cells 0–65μm 7.2 J/cm2Induces apoptosis and necrosis In vitro [81]
Jurkat cells 0.25–1μg/ml 5 J/cm2The TRAIL/TRAIL-receptor pathway and
activates the caspase-8
In vitro [82]
0–0.3μg/ml 12 J/cm2Upregulates reactive oxygen species In vitro [83]
K562 cells 0.4μg/ml 90 mJ/cm2Regulates the JNK pathway In vitro [84]
Downloaded from https://academic.oup.com/jpp/article/73/4/425/6047495 by Universiteit Leiden / LUMC user on 18 October 2021
Journal of Pharmacy and Pharmacology, 2021, Vol. 73, No. 4 431
combination of HYP-PDT with hyperthermia or mitomycin C ef-
fectively enhanced tumour response with minimal side effects.[111,112]
HYP might also be an effective tool for treating and detecting
ovarian cancer. HYP-loaded polylactic acid (PLA) nanoparticles
exhibited higher photoactivity on NuTu-19 ovarian cells than free
drugs. In addition, the antitumour activity was improved with the
increasing incubation time and laser dose.[113] The combination of
HYP and illumination resulted in an obvious decrease in the meta-
bolic activity and induced apoptosis of renal cancer cells (A498 and
ACHN) in vitro.[114] In a GH4C1 rat pituitary cell line, treatment
of HYP-PDT in vitro signicantly slowed tumour growth and in-
duced apoptosis in a dose-dependent manner.[115] Penjweini etal.[116]
compared the phototoxicity of HYP on a mixture of human lung
broblast (HLF) cells and A549 lung cancer cells. Compared with
HLF cells, A549 cells were more responsive by polyvinylpyr-
rolidone (PVP)-HYP treatment after 4h PDT. HYP-PDT also in-
duced damage-associated molecular patterns (DAMPs) and surface
integrin-related protein (CD47) dysregulation on the surface of
Lewis cancer cells, leading to the destruction of immune homeo-
stasis.[117] Furthermore, HYP-PDT was effective in inhibiting the
growth of FRO anaplastic thyroid cancer cells and tumours in mice.
The mechanisms were mediated by the intracellular ROS generation
and mitochondrial damage.[118] HYP tetraether liposomes irradiated
with a LED-device were suitable for antivascular targeting of tu-
mours, while HYP encapsulated in hydroxypropyl-β-cyclodextrin
(HPβCD) inclusion complex could be more safely delivered to tu-
mour sites.[119]
Perspectives and Conclusions
PDT is a rapidly developing targeted cancer therapy with less toxic
adverse effect due to its excellent photosensitising and tumoritropic
properties. Hypericin, a powerful candidate photosensitiser, is a
natural naphthodianthrone isolated from the genus Hypericum, es-
pecially Hypericum perforatum L.[120] In recent decades, the photo-
dynamic compound has attracted great attention as a potential
anticancer drug. Many researchers have conrmed its powerful
antitumour effects on different tumour cells in vivo and in vitro
studies, such as bladder, colon, breast, cervical, glioma, leukaemia,
hepatic, melanoma, lymphoma, etc. (Figure3). The photosensitising
effects of HYP are generally described as oxygen-dependent where
singlet oxygen is the main contributor for HYP-PDT.
The underlying antitumour mechanisms of HYP-PDT are associ-
ated with inhibition of various proteins and genes including VEGF,
MMP-2, MMP-9, HER2, Akt, P-Erk1/2, SERCA2, COX-2, GST,
glutathione peroxidase and catalase. However, the levels of MRP-1,
HO-1, VEGF-A, PD-ECGF, ABCG2, Bax, IL-6, IL-1β, MMP-1, PKC,
caspase-3, caspase-8, PARP, GM-CSF and TNF-α are upregulated.
Furthermore, HYP-PDT may affect the p38 MAPK, JNK, PI3K,
CHOP/TRIB3/Akt/mTOR, TRAIL/TRAIL-receptor, c-Met and
Ephrin-Eph, mitochondria and the extrinsic signalling pathways
(Table1; Figure4). Meanwhile, these results strongly establish that
HYP-PDT possesses the ability to inhibit various events related to
cell growth, apoptosis, necrosis, autophagy, angiogenesis, cell cycle
arrest and the formation of cellular colonies, and clearly supports its
great promising in cancer treatment.
Cell lines/model Drug Dose Laser Energy Mechanism(s) of action Application Reference
ATL cells 0–200ng/ml 11.28 J/cm2The mitochondrial pathway and suppresses the
viral transcription
In vitro [86]
HUH6 and
HepT1 cells
0–12.5μm 1000 Lux Decreases the cell viability and Induces apoptosis In vitro [87]
HepG2 cells 0–1μg/ml 21 J/cm2Facilitates cytotoxic ROS and recruits IL-6 In vitro [88]
0.025–0.5μm 15.48 J/cm2Recognises ASGP-R In vitro [90]
G361, M18 and
M6 cells
5×10–8 M 12 J/cm2Decreases glutathione peroxidase and catalase
activities
In vitro [91]
UCT Mel-1 and
UCT Mel-3 cells
0.2–10μm 1 J/cm2Induces necrosis or apoptosis In vitro [92]
0.2–10μm 1 J/cm2Induces autophagy In vitro [93]
A375 cells 3μm 1 J/cm2The extrinsic and intrinsic caspase-dependent
apoptotic pathway
In vitro [95]
Mice, P388 cells 1, 5, 20mg/kg 120 J/cm2Induces tumour vasculature damage In vivo [97]
SQ2 cells 10mg/kg 7.2 J/cm2Upregulates the mRNA expression levels of
IL-1β, IL-6, TNF-α and GM-CSF
In vivo [101]
SNU-1 cells 0.1–2μm 3 J/cm2Induces the membrane damage In vitro [103]
A-431 cells 200ng/ml 0.6 J/cm2Induces apoptosis In vitro [104]
human SCC cells 3μm 1 J/cm2The necrotic caspase-independent pathway In vitro [105]
FaDu cells 5–50μm 50 000 Lux Reduces metabolism and induces apoptosis In vitro [106]
UMB-SCC 745
and 969 cells
2.5μg/ml 1.92 J/cm2Induces apoptosis and necrosis In vitro [107]
RIF-1 mouse
tumour
5mg/kg 120 J/cm2Induces vascular damage and apoptosis In vivo [109,110]
A498 and ACHN
cells
50μm 50 000 Lux Decreases in metabolic activity and causes
apoptosis
In vitro [114]
Rats, GH4C1 cells 1mg/kg 130 J/cm2Inhibits cellular growth In vivo [115]
Mice, Lewis cells 0.25μm 1.85 J/cm2Induces dysregulation of DAMPs In vitro and
vivo
[117]
FRO cells 1–20μg/ml 46.8 J/cm2Induces mitochondrial damage In vivo [118]
SK-OV-3 cells 0.2–1μm 2.1–12.4 J/cm2Induces antivascular targeting of the tumor In vitro [119]
Table 1 Continued
Downloaded from https://academic.oup.com/jpp/article/73/4/425/6047495 by Universiteit Leiden / LUMC user on 18 October 2021
432 Journal of Pharmacy and Pharmacology, 2021, Vol. 73, No. 4
Nevertheless, hypericin has a very hydrophobic behav-
iour and easily forms aggregates in water, resulting in the loss
of its spectroscopic and tumour-selective properties as well as
low bioavailability.[121] To overcome these limitations, previous
studies have shown that HYP-PDT in conjunction with tar-
geted therapies, such as nanoencapsulation, superparamagnetic
iron oxide nanoparticles, single-walled carbon nanohorns, lipid
nanocapsules, cerium oxide nanorods, PLA nanoparticles, lipo-
somes and HPβCD inclusion complex, is a more potential ap-
proach in cancer treatment.
Moreover, previous researches have demonstrated that HYP-
PDT combined with various chemotherapeutic agents, such as
peruorocarbons bevacizumab, MK-886, hyperforin, diclofenac,
urbiprofen, ibuprofen, YM155, 17-DMAG, carmustine,
methoxyestradiol, tamoxifen, pyridinyl imidazole, apoMb, celebrex,
rottlerin, DTHe and Ko143, is more effective in killing cancer cells.
Hence, it is essential to investigate the activity of HYP-PDT on
the metabolism of other agents in the future. What’s more, further
studies are still needed to improve its oral bioavailability and en-
hance its efciency before HYP-PDT can be safely used in clinical
application for the treatment ofcancer.
Acknowledgements
This work was nancially supported by the China Scholarship Council
(201906557012) and Beijing Natural Science Foundation (7194289,
7202121, 7202123).
Conflict of Interest
The authors have declared that there is no conict of interest.
References
1. AgostinisP, BergK, CengelKA etal. Photodynamic therapy of cancer: an
update. CA Cancer J Clin 2011; 61: 250–81. doi:10.3322/caac.20114
2. YanoS, Hirohara S, ObataM et al. Current states and future views in
photodynamic therapy. J Photochem Photobiol C Photochem Rev 2011;
12: 46–67. doi:10.1016/j.jphotochemrev.2011.06.001
3. ChoiYM, AdelzadehL, WuJJ etal. Photodynamic therapy for psoriasis. J
Dermatolog Treat 2015; 26: 202–7. doi:10.3109/09546634.2014.927816
4. LiJ, Qin M, LiuC et al. Antimicrobial photodynamic therapy against
multidrug-resistant Acinetobacter baumannii clinical isolates mediated by
aloe-emodin: an in vitro study. Photodiagnosis Photodyn Ther 2020; 29:
101632. doi:10.1016/j.pdpdt.2019.101632
5. Kwiatkowski S, KnapB, PrzystupskiD et al. Photodynamic therapy –
mechanisms, photosensitizers and combinations. Biomed Pharmacother
2018; 106: 1098–107. doi:10.1016/j.biopha.2018.07.049
6. Jamali Z, KhoobiM, Hejazi SM etal. Evaluation of targeted curcumin
(CUR) loaded PLGA nanoparticles for in vitro photodynamic therapy on
human glioblastoma cell line. Photodiagnosis Photodyn Ther 2018; 23:
190–201. doi:10.1016/j.pdpdt.2018.06.026
7. WenX, LiY, HamblinMR. Photodynamic therapy in dermatology beyond
non-melanoma cancer: an update. Photodiagnosis Photodyn Ther 2017;
19: 140–52. doi:10.1016/j.pdpdt.2017.06.010
8. Do MH, Kim SY. Hypericin, a naphthodianthrone derivative, prevents
methylglyoxal-induced human endothelial cell dysfunction. Biomol Ther
2017; 25: 158–64. doi:10.4062/biomolther.2016.034
Figure 4 Schematic illustration depicting the process of cell death by apoptosis and cell cycle arrest.
Downloaded from https://academic.oup.com/jpp/article/73/4/425/6047495 by Universiteit Leiden / LUMC user on 18 October 2021
Journal of Pharmacy and Pharmacology, 2021, Vol. 73, No. 4 433
9. AgostinisP, VantieghemA, Merlevede W etal. Hypericin in cancer treat-
ment: more light on the way. Int J Biochem Cell Biol 2002; 34: 221–41.
doi:10.1016/S1357-2725(01)00126-1
10. GonçalvesJLdaS, BernalC, Imasato H etal. Hypericin cytotoxicity in
tumor and non-tumor cell lines: a chemometric study. Photodiagnosis
Photodyn Ther 2017; 20: 86–90. doi:10.1016/j.pdpdt.2017.08.005
11. OckerL, AdamusA, HempingL etal. Hypericin and its radio iodinated
derivatives – Anovel combined approach for the treatment of pediatric
alveolar rhabdomyosarcoma cells in vitro. Photodiagnosis Photodyn Ther
2020; 29: 101588. doi:10.1016/j.pdpdt.2019.101588
12. SattlerS, SchaeferU, Schneider W etal. Binding, uptake, and transport
of hypericin by Caco-2 cell monolayers. J Pharm Sci 1997; 86: 1120–6.
doi:10.1021/js970004a
13. SiboniG, WeitmanH, FreemanD etal. The correlation between hydro-
philicity of hypericins and helianthrone: internalization mechanisms,
subcellular distribution and photodynamic action in colon carcinoma
cells. Photochem Photobiol Sci 2002; 1: 483–91. doi:10.1039/b202884k
14. GalanouMC, Theodossiou TA, TsiourvasD etal. Interactive transport,
subcellular relocation and enhanced phototoxicity of hypericin encapsu-
lated in guanidinylated liposomes via molecular recognition. Photochem
Photobiol 2008; 84: 1073–1083. doi:10.1111/j.1751-1097.2008.00392.x
15. MikešJ, HýžďalováM, KočíL etal. Lower sensitivity of FHC fetal colon
epithelial cells to photodynamic therapy compared to HT-29 colon adeno-
carcinoma cells despite higher intracellular accumulation of hypericin.
Photochem Photobiol Sci 2011; 10: 626–32. doi:10.1039/c0pp00359j
16. KariotiA, BiliaAR. Hypericins as potential leads for new therapeutics. Int
J Mol Sci 2010; 11: 562–94. doi:10.3390/ijms11020562
17. Krammer B, Verwanger T. Molecular response to hypericin-
induced photodamage. Curr Med Chem 2012; 19: 793–8.
doi:10.2174/092986712799034842
18. Kamuhabwa AR, Agostinis PM, D’Hallewin MA et al.
Cellular photodestruction induced by hypericin in AY-27 rat
bladder carcinoma cells. Photochem Photobiol 2001; 74: 126.
doi:10.1562/0031-8655(2001)074<0126:CPIBHI>2.0.CO;2
19. ZupkoI, KamuhabwaAR, D’HallewinMA etal. In vivo photodynamic
activity of hypericin in transitional cell carcinoma bladder tumors. Int J
Oncol 2001; 18: 1099–105. doi:10.3892/ijo.18.5.1099
20. KamuhabwaAAR, RoskamsT, D’HallewinMA etal. Whole bladder wall
photodynamic therapy of transitional cell carcinoma rat bladder tumors
using intravesically administered hypericin. Int J Cancer 2003; 107: 460–
7. doi:10.1002/ijc.11396
21. KamuhabwaAAR, CosseratGerardinI, DidelonJ etal. Biodistribution of
hypericin in orthotopic transitional cell carcinoma bladder tumors: impli-
cation for whole bladder wall photodynamic therapy. Int J Cancer 2002;
97: 253–60. doi:10.1002/ijc.1594
22. HuygensA, KamuhabwaAR, VanLaethemA etal. Enhancing the photo-
dynamic effect of hypericin in tumour spheroids by fractionated light
delivery in combination with hyperoxygenation. Int J Oncol 2005; 26:
1691–7. doi:10.3892/ijo.26.6.1691
23. KamuhabwaAR, Huygens A, Roskams T et al. Enhancing the photo-
dynamic effect of hypericin in human bladder transitional cell carcinoma
spheroids by the use of the oxygen carrier, peruorodecalin. Int J Oncol
2006; 28: 775–80. doi:10.3892/ijo.28.3.775
24. KocanovaS, BuytaertE, MatrouleJY etal. Induction of heme-oxygenase
1 requires the p38MAPK and PI3K pathways and suppresses apoptotic
cell death following hypericin-mediated photodynamic therapy. Apoptosis
2007; 12: 731–41. doi:10.1007/s10495-006-0016-x
25. BuytaertE, MatrouleJY, DurinckS etal. Molecular effectors and modu-
lators of hypericin-mediated cell death in bladder cancer cells. Oncogene
2008; 27: 1916–29. doi:10.1038/sj.onc.1210825
26. BhuvaneswariR, Thong PSP, Gan-YapYY etal. Evaluation of hypericin-
mediated photodynamic therapy in combination with angiogenesis in-
hibitor bevacizumab using in vivo uorescence confocal endomicroscopy.
J Biomed Opt 2010; 15: 011114.
27. BhuvaneswariR, GanYY, LuckySS etal. Molecular proling of angio-
genesis in hypericin mediated photodynamic therapy. Mol Cancer 2008;
7: 1–14. doi:10.1186/1476-4598-7-56
28. Blank M, Kostenich G, Lavie G et al. Wavelength-dependent
properties of photodynamic therapy using hypericin in vitro
and in an animal model. Photochem Photobiol 2002; 76: 335.
doi:10.1562/0031-8655(2002)076<0335:wdpopt>2.0.co;2
29. JendželovskýR, MikešJ, SoučekK etal. Drug efux transporters, MRP1
and BCRP, affect the outcome of hypericin-mediated photodynamic
therapy in HT-29 adenocarcinoma cells. Photochem Photobiol Sci 2009;
8: 1716–23. doi:10.1039/b9pp00086k
30. KlebanJ, SzilárdiováB, MikešJ et al. Pre-treatment of HT-29 cells with
5-LOX inhibitor (MK-886) induces changes in cell cycle and increases
apoptosis after photodynamic therapy with hypericin. J Photochem
Photobiol B Biol 2006; 84: 79–88. doi:10.1016/j.jphotobiol.2006.02.003
31. KlebanJ, MikešJ, HorváthV etal. Mechanisms involved in the cell cycle
and apoptosis of HT-29 cells pre-treated with MK-886 prior to photo-
dynamic therapy with hypericin. J Photochem Photobiol B Biol 2008; 93:
108–18. doi:10.1016/j.jphotobiol.2008.07.007
32. Šemeláková M, Mikeš J, Jendželovský R et al. The pro-apoptotic and
anti-invasive effects of hypericin-mediated photodynamic therapy are
enhanced by hyperforin or aristoforin in HT-29 colon adenocarcinoma
cells. J Photochem Photobiol B Biol 2012; 117: 115–25. doi:10.1016/j.
jphotobiol.2012.09.003
33. Šemeláková M, Jendželovský R, Fedoročko P. Drug membrane trans-
porters and CYP3A4 are affected by hypericin, hyperforin or aristoforin
in colon adenocarcinoma cells. Biomed Pharmacother 2016; 81: 38–47.
doi:10.1016/j.biopha.2016.03.045
34. Majerník M, JendželovskýR, Babinčák M et al. Novel insights into
the effect of hyperforin and photodynamic therapy with hypericin
on chosen angiogenic factors in colorectal micro-tumors created
on chorioallantoic membrane. Int J Mol Sci 2019; 20. doi:10.3390/
ijms20123004
35. KlebanJ, Mikeš J, SzilárdiováB et al. Modulation of hypericin photo-
dynamic therapy by pretreatment with 12 various inhibitors of arachi-
donic acid metabolism in colon adenocarcinoma HT-29 cells. Photochem
Photobiol 2007; 83: 1174–85. doi:10.1111/j.1751-1097.2007.00127.x
36. Kello M, MikešJ, JendželovskýR et al. PUFAs enhance oxidative stress
and apoptosis in tumour cells exposed to hypericin-mediated PDT.
Photochem Photobiol Sci 2010; 9: 1244–51. doi:10.1039/c0pp00085j
37. GyurászováK, MikešJ, HalaburkováA etal. YM155, a small molecule in-
hibitor of survivin expression, sensitizes cancer cells to hypericin-mediated
photodynamic therapy. Photochem Photobiol Sci 2016; 15: 812–21.
doi:10.1039/c5pp00438a
38. Macejová M, Sačková V, Hradická P et al. Combination of photo-
active hypericin and Manumycin Aexerts multiple anticancer effects on
oxaliplatin-resistant colorectal cells. Toxicol Vitr 2020; 66. doi:10.1016/j.
tiv.2020.104860
39. SanovicR, VerwangerT, HartlA et al. Low dose hypericin-PDT induces
complete tumor regression in BALB/c mice bearing CT26 colon car-
cinoma. Photodiagnosis Photodyn Ther 2011; 8: 291–6. doi:10.1016/j.
pdpdt.2011.04.003
40. LinS, YangL, ShiH etal. Endoplasmic reticulum-targeting photosensitizer
Hypericin confers chemo-sensitization towards oxaliplatin through
inducing pro-death autophagy. Int J Biochem Cell Biol 2017; 87: 54–68.
doi:10.1016/j.biocel.2017.04.001
41. Lin S, Lei K, Du W et al. Enhancement of oxaliplatin sensitivity in
human colorectal cancer by hypericin mediated photodynamic therapy
via ROS-related mechanism. Int J Biochem Cell Biol 2016; 71: 24–34.
doi:10.1016/j.biocel.2015.12.003
42. KhotMI, PerrySL, MaiseyT etal. Inhibiting ABCG2 could potentially en-
hance the efcacy of hypericin-mediated photodynamic therapy in spher-
oidal cell models of colorectal cancer. Photodiagnosis Photodyn Ther
2018; 23: 221–9. doi:10.1016/j.pdpdt.2018.06.027
43. SüloğluAK, SelmanoğluG, AkayMT. Alterations in dysadherin expression
and F-actin reorganization: a possible mechanism of hypericin-mediated
photodynamic therapy in colon adenocarcinoma cells. Cytotechnology
2015; 67: 311–30.
44. MontanhaMC, SilvaLL, PangoniFBB etal. Response surface method op-
timization of a novel Hypericin formulation in P123 micelles for colorectal
Downloaded from https://academic.oup.com/jpp/article/73/4/425/6047495 by Universiteit Leiden / LUMC user on 18 October 2021
434 Journal of Pharmacy and Pharmacology, 2021, Vol. 73, No. 4
cancer and antimicrobial photodynamic therapy. J Photochem Photobiol
B Biol 2017; 170: 247–55. doi:10.1016/j.jphotobiol.2017.04.008
45. MühleisenL, AlevM, UnterwegerH etal. Analysis of hypericin-mediated
effects and implications for targeted photodynamic therapy. Int J Mol Sci
2017; 18. doi:10.3390/ijms18071388
46. Kaleta-Richter M, Aebisher D, Jaworska D et al. The inuence of
hypericin-mediated photodynamic therapy on interleukin-8 and
-10 secretion in colon cancer cells. Integr Cancer Ther 2020; 19.
doi:10.1177/1534735420918931
47. FerencP, SolárP, KlebanJ et al. Down-regulation of Bcl-2 and Akt in-
duced by combination of photoactivated hypericin and genistein in human
breast cancer cells. J Photochem Photobiol B Biol 2010; 98: 25–34.
doi:10.1016/j.jphotobiol.2009.10.004
48. SolárP, ChytilováM, SolárováZ etal. Photodynamic therapy with hy-
pericin improved by targeting HSP90 associated proteins. Pharmaceuticals
2011; 4: 1488–502. doi:10.3390/ph4111488
49. Solár P, Ferenc P, Koval’ J et al. Photoactivated hypericin induces
downregulation of HER2 gene expression. Radiat Res 2011; 175: 51–6.
doi:10.1667/rr2276.1
50. KuchárováB, Mikeš J, JendželovskýR etal. Potentiation of hypericin-
mediated photodynamic therapy cytotoxicity by MK-886: focus on ABC
transporters, GDF-15 and redox status. Photodiagnosis Photodyn Ther
2015; 12: 490–503. doi:10.1016/j.pdpdt.2015.04.008
51. Theodossiou TA, Olsen CE, Jonsson M et al. The diverse roles of
glutathione-associated cell resistance against hypericin photodynamic
therapy. Redox Biol 2017; 12: 191–7. doi:10.1016/j.redox.2017.02.018
52. KimákováP, SolárP, FeckováB etal. Photoactivated hypericin increases
the expression of SOD-2 and makes MCF-7 cells resistant to photo-
dynamic therapy. Biomed Pharmacother 2017; 85: 749–55. doi:10.1016/j.
biopha.2016.11.093
53. de Andrade GP, Manieri TM, Nunes EA et al. Comparative in vitro
study of photodynamic activity of hypericin and hypericinates in MCF-7
cells. J Photochem Photobiol B Biol 2017; 175: 89–98. doi:10.1016/j.
jphotobiol.2017.08.025
54. GaoC, JianJ, LinZ etal. Hypericin-loaded carbon nanohorn hybrid for
combined photodynamic and photothermal therapy in vivo. Langmuir
2019; 35: 8228–37. doi:10.1021/acs.langmuir.9b00624
55. Theodossiou TA, Ali M, Grigalavicius M et al. Simultaneous defeat of
MCF7 and MDA-MB-231 resistances by a hypericin PDT–tamoxifen hy-
brid therapy. npj Breast Cancer 2019; 5. doi:10.1038/s41523-019-0108-8
56. DelaeyEM, VandenbogaerdeAL, AgostinisP etal. Conuence dependent
resistance to photo-activated hypericin in HeLa cells. Int J Oncol 1999;
14: 759–63. doi:10.3892/ijo.14.4.759
57. AssefaZ, Vantieghem A, Declercq W et al. The activation of the c-Jun
N-terminal kinase and p38 mitogen-activated protein kinase signaling
pathways protects HeLa cells from apoptosis following photodynamic
therapy with hypericin. J Biol Chem 1999; 274: 8788–96. doi:10.1074/
jbc.274.13.8788
58. AgostinisP, AssefaZ, VantieghemA et al. Apoptotic and anti-apoptotic
signaling pathways induced by photodynamic therapy with hypericin. Adv
Enzyme Regul 2000; 40: 157–82. doi:10.1016/S0065-2571(99)00021-7
59. HendrickxN, VolantiC, MoensU etal. Up-regulation of Cyclooxygenase-2
and apoptosis resistance by p38 MAPK in hypericin-mediated photo-
dynamic therapy of human cancer cells. J Biol Chem 2003; 278: 52231–9.
doi:10.1074/jbc.M307591200
60. Bianchini P, Cozzolino M, Oneto M et al. Hypericin-apomyoglobin:
an enhanced photosensitizer complex for the treatment of tumor
cells. Biomacromolecules 2019; 20: 2024–33. doi:10.1021/acs.
biomac.9b00222
61. BuytaertE, Callewaert G, Hendrickx N et al. Role of endoplasmic re-
ticulum depletion and multidomain proapoptotic BAX and BAK proteins
in shaping cell death after hypericin-mediated photodynamic therapy.
FASEB J 2006; 20: 756–8. doi:10.1096/fj.05-4305fje
62. Barras A, Boussekey L, Courtade E et al. Hypericin-loaded lipid
nanocapsules for photodynamic cancer therapy in vitro. Nanoscale 2013;
5: 10562–72. doi:10.1039/c3nr02724d
63. DuH, BayBH, MahendranR etal. Hypericin-mediated photodynamic
therapy elicits differential interleukin-6 response in nasopharyngeal
cancer. Cancer Lett 2006; 235: 202–8. doi:10.1016/j.canlet.2005.04.013
64. Du H, Bay BH, Mahendran R et al. Endogenous expression of inter-
leukin-8 and interleukin-10 in nasopharyngeal carcinoma cells and
the effect of photodynamic therapy. Int J Mol Med 2002; 10: 73–6.
doi:10.3892/ijmm.10.1.73
65. Du HY, Olivo M, Tan BKH et al. Hypericin-mediated photodynamic
therapy induces lipid peroxidation and necrosis in nasopharyngeal cancer.
Int J Oncol 2003; 23: 1401–5. doi:10.3892/ijo.23.5.140
66. Du HY, Olivo M, TanBKH et al. Photoactivation of hypericin down-
regulates glutathione S-transferase activity in nasopharyngeal cancer cells.
Cancer Lett 2004; 207: 175–81. doi:10.1016/j.canlet.2003.12.024
67. Du H, Olivo M, Mahendran R et al. Modulation of matrix
metalloproteinase-1 in nasopharyngeal cancer cells by photoactivation of
hypericin. Int J Oncol 2004; 24: 657–62. doi:10.3892/ijo.24.3.657
68. DuHY, OlivoM, Mahendran R et al. Hypericin photoactivation trig-
gers down-regulation of matrix metalloproteinase-9 expression in well-
differentiated human nasopharyngeal cancer cells. Cell Mol Life Sci 2007;
64: 979–88. doi:10.1007/s00018-007-7030-1
69. WangX, Guo Y, YangS et al. Cellular and molecular mechanisms of
photodynamic hypericin therapy for nasopharyngeal carcinoma cells. J
Pharmacol Exp Ther 2010; 334: 847–53. doi:10.1124/jpet.110.168856
70. Chan PS, Koon HK, Wu ZG et al. Role of p38 mapks in hy-
pericin photodynamic therapy-induced apoptosis of nasopharyn-
geal carcinoma cells. Photochem Photobiol 2009; 85: 1207–17.
doi:10.1111/j.1751-1097.2009.00572.x
71. Thong PSP, Watt F, Ren MQ et al. Hypericin-photodynamic therapy
(PDT) using an alternative treatment regime suitable for multi-fraction
PDT. J Photochem Photobiol B Biol 2006; 82: 1–8. doi:10.1016/j.
jphotobiol.2005.08.002
72. Yee KKL, Soo KC, Olivo M. Anti-angiogenic effects of hypericin-
photodynamic therapy in combination with Celebrex in the treatment of
human nasopharyngeal carcinoma. Int J Mol Med 2005; 16: 993–1002.
doi:10.3892/ijmm.16.6.993
73. BhuvaneswariR, GanYYY, YeeKKL etal. Effect of hypericin-mediated
photodynamic therapy on the expression of vascular endothelial growth
factor in human nasopharyngeal carcinoma. Int J Mol Med 2007; 20:
421–8. doi:10.3892/ijmm.20.4.421
74. Miccoli L, Beurdeley-Thomas A, De Pinieux G et al. Light-induced
photoactivation of hypericin affects the energy metabolism of human
glioma cells by inhibiting hexokinase bound to mitochondria. Cancer Res
1998; 58: 5777–86.
75. UzdenskyAB, MaLW, IaniV etal. Intracellular localisation of hypericin
in human glioblastoma and carcinoma cell lines. Lasers Med Sci 2001; 16:
276–83. doi:10.1007/PL00011364
76. RitzR, WeinHT, DietzK etal. Photodynamic therapy of malignant glioma
with hypericin: comprehensive in vitro study in human glioblastoma cell
lines. Int J Oncol 2007; 30: 659–67. doi:10.3892/ijo.30.3.659
77. Balogová LL, Maslanakova M, Dzurová L et al. Bcl-2 proapoptotic
proteins distribution in U-87 MG glioma cells before and after hy-
pericin photodynamic action. Gen Physiol Biophys 2013; 32: 179–87.
doi:10.4149/gpb_2013021
78. StupákováV, VarinskáL, MiroššayA etal. Photodynamic effect of hy-
pericin in primary cultures of human umbilical endothelial cells and
glioma cell lines. Phyther Res 2009; 23: 827–32. doi:10.1002/ptr.2681
79. DzurováL, Petrovajova D, NadovaZ et al. The role of anti-apoptotic
protein kinase Cα in response to hypericin photodynamic therapy in
U-87 MG cells. Photodiagnosis Photodyn Ther 2014; 11: 213–26.
doi:10.1016/j.pdpdt.2014.02.010
80. MisuthM, HorvathD, MiskovskyP etal. Synergism between PKCδ regu-
lators hypericin and rottlerin enhances apoptosis in U87 MG glioma cells
after light stimulation. Photodiagnosis Photodyn Ther 2017; 18: 267–74.
doi:10.1016/j.pdpdt.2017.03.018
81. LavieG, KaplinskyC, TorenA etal. A photodynamic pathway to apop-
tosis and necrosis induced by dimethyl tetrahydroxyhelianthrone and
Downloaded from https://academic.oup.com/jpp/article/73/4/425/6047495 by Universiteit Leiden / LUMC user on 18 October 2021
Journal of Pharmacy and Pharmacology, 2021, Vol. 73, No. 4 435
hypericin in leukaemic cells: possible relevance to photodynamic therapy.
Br J Cancer 1999; 79: 423–32.
82. SchemppCM, Simon-Haarhaus B, TermeerCC et al. Hypericin photo-
induced apoptosis involves the tumor necrosis factor-related apoptosis-
inducing ligand (TRAIL) and activation of caspase-8[J]. FEBS letters
2001; 493: 26–30.
83. UnterwegerH, SubatzusD, TietzeR etal. Hypericin-bearing magnetic iron
oxide nanoparticles for selective drug delivery in photodynamic therapy.
Int J Nanomedicine 2015; 10: 6985–96. doi:10.2147/IJN.S92336
84. Xu Y, Wang D, Zhuang Z et al. Hypericin-mediated photodynamic
therapy induces apoptosis in K562 human leukemia cells through JNK
pathway modulation. Mol Med Rep 2015; 12: 6475–82. doi:10.3892/
mmr.2015.4258
85. Jendželovský R, Jendželovská Z, Kuchárová B et al. Breast cancer re-
sistance protein is the enemy of hypericin accumulation and toxicity of
hypericin-mediated photodynamic therapy. Biomed Pharmacother 2019;
109: 2173–81. doi:10.1016/j.biopha.2018.11.084
86. XuL, ZhangX, ChengW etal. Hypericin-photodynamic therapy inhibits
the growth of adult T-cell leukemia cells through induction of apoptosis
and suppression of viral transcription. Retrovirology 2019; 16: 1–13.
doi:10.1186/s12977-019-0467-0
87. SeitzG, KrauseR, FuchsJ etal. In vitro photodynamic therapy in pediatric
epithelial liver tumors promoted by hypericin. Oncol Rep 2014: 1277–82.
doi:10.3892/or_00000141
88. BarathanM, MariappanV, ShankarEM etal. Hypericin-photodynamic
therapy leads to interleukin-6 secretion by HepG2 cells and their apop-
tosis via recruitment of BH3 interacting-domain death agonist and
caspases. Cell Death Dis 2013; 4: 1–10. doi:10.1038/cddis.2013.219
89. FadelM, Kassab K, Youssef T. Photodynamic efcacy of hypericin tar-
geted by two delivery techniques to hepatocellular carcinoma cells. Lasers
Med Sci 2010; 25: 675–83. doi:10.1007/s10103-010-0787-8
90. WangY, ZhangY, Jin M et al. A hypericin delivery system based on
polydopamine coated cerium oxide nanorods for targeted photodynamic
therapy. Polymers (Basel) 2019; 11. doi:10.3390/polym11061025
91. Hadjur C, Richard MJ, Parat MO et al. Photodynamic effects of hy-
pericin on lipid peroxidation and antioxidant status in melanoma cells.
Photochem Photobiol 1996; 64: 375–81. doi:10.1111/j.1751-1097.1996.
tb02474.x
92. DavidsLM, Kleemann B, Kacerovská D et al. Hypericin phototoxicity
induces different modes of cell death in melanoma and human skin
cells. J Photochem Photobiol B Biol 2008; 91: 67–76. doi:10.1016/j.
jphotobiol.2008.01.011
93. DavidsLM, KleemannB, CooperS etal. Melanomas display increased
cytoprotection to hypericin-mediated cytotoxicity through the induc-
tion of autophagy. Cell Biol Int 2009; 33: 1065–72. doi:10.1016/j.
cellbi.2009.06.026
94. Sharma KV, Davids LM. Depigmentation in melanomas increases
the efcacy of hypericin-mediated photodynamic-induced cell death.
Photodiagnosis Photodyn Ther 2012; 9: 156–63. doi:10.1016/j.
pdpdt.2011.09.003
95. Kleemann B, Loos B, Scriba TJ et al. St John’s Wort (Hypericum
perforatum L.) photomedicine: Hypericin-photodynamic therapy induces
metastatic melanoma cell death. PLoS One 2014; 9. doi:10.1371/journal.
pone.0103762
96. De Morais FAP, Gonçalves RS, Vilsinski BH et al. Hypericin photo-
dynamic activity in DPPC liposomes-part II: stability and application
in melanoma B16-F10 cancer cells. Photochem Photobiol Sci 2020; 19:
620–30. doi:10.1039/c9pp00284g
97. ChenB, ZupkoI, deWittePA. Photodynamic therapy with hypericin in
a mouse P388 tumor model: vascular effects determine the efcacy. Int J
Oncol 2001; 18: 737–42. doi:10.3892/ijo.18.4.737
98. PabaV, QuartoM, VarrialeL etal. Photo-activation of hypericin with low
doses of light promotes apparent photo-resistance in human histiocytic
lymphoma U937 cells. J Photochem Photobiol B Biol 2001; 60: 87–96.
doi:10.1016/S1011-1344(01)00125-7
99. NakajimaN, KawashimaN. A basic study on hypericin-PDT in vitro.
Photodiagnosis Photodyn Ther 2012; 9: 196–203. doi:10.1016/j.
pdpdt.2012.01.008
100. RookAH, WoodGS, DuvicM etal. A phase II placebo-controlled study
of photodynamic therapy with topical hypericin and visible light irradi-
ation in the treatment of cutaneous T-cell lymphoma and psoriasis. J Am
Acad Dermatol 2010; 63: 984–90. doi:10.1016/j.jaad.2010.02.039
101. BlankM, LavieG, MandelM et al. Effects of photodynamic therapy
with hypericin in mice bearing highly invasive solid tumors. Oncol Res
2000; 12: 409–18. doi:10.3727/096504001108747864
102. HeadCS, LuuQ, SercarzJ etal. Photodynamic therapy and tumor im-
aging of hypericin-treated squamous cell carcinoma. World J Surg Oncol
2006; 4: 1–10. doi:10.1186/1477-7819-4-87
103. BublikM, HeadC, BenharashP etal. Hypericin and pulsed laser therapy
of squamous cell cancer in vitro. Photomed Laser Surg 2006; 24: 341–7.
doi:10.1089/pho.2006.24.341
104. SanovicR, KrammerB, GrumboeckS etal. Time-resolved gene expres-
sion proling of human squamous cell carcinoma cells during the apop-
tosis process induced by photodynamic treatment with hypericin. Int J
Oncol 2009; 35: 921–39. doi:10.3892/ijo_00000407
105. SharmaKV, DavidsLM. Hypericin-PDT-induced rapid necrotic death in
human squamous cell carcinoma cultures after multiple treatment. Cell
Biol Int 2012; 36: 1261–6. doi:10.1042/cbi20120108
106. LaffersW, BusseAC, MahrtJ etal. Photosensitizing effects of hypericin
on head neck squamous cell carcinoma in vitro. Eur Arch Oto-Rhino-
Laryngology 2015; 272: 711–8. doi:10.1007/s00405-014-2984-8
107. Gyenge EB, Lüscher D, FornyP et al. Photodynamic mechanisms in-
duced by a combination of hypericin and a chlorin based-photosensitizer
in head and neck squamous cell carcinoma cells. Photochem Photobiol
2013; 89: 150–62. doi:10.1111/j.1751-1097.2012.01217.x
108. LiuCD, KwanD, SaxtonRE etal. Hypericin and photodynamic therapy
decreases human pancreatic cancer in vitro and in vivo. J Surg Res 2000;
93: 137–43. doi:10.1006/jsre.2000.5949
109. ChenB, RoskamsT, XuY etal. Photodynamic therapy with hypericin
induces vascular damage and apoptosis in the RIF-1 mouse tumor
model. Int J Cancer 2002; 98: 284–90. doi:10.1002/ijc.10175
110. ChenB, RoskamsT, deWittePAM. Antivascular tumor eradication by
hypericin-mediated photodynamic therapy. Photochem Photobiol 2002;
76: 509. doi:10.1562/0031-8655(2002)076<0509:atebhm>2.0.co;2
111. ChenB, RoskamsT, deWitte PAM. Enhancing the antitumoral effect
of hypericin-mediated photodynamic therapy by hyperthermia. Lasers
Surg Med 2002; 31: 158–63. doi:10.1002/lsm.10089
112. Chen B, Ahmed B, Landuyt W et al. Potentiation of photodynamic
therapy with hypericin by mitomycin c in the radiation-induced bro-
sarcoma–1 mouse tumor model. Photochem Photobiol 2003; 78: 278.
doi:10.1562/0031-8655(2003)078<0278:poptwh>2.0.co;2
113. Zeisser-Labouèbe M, Lange N, Gurny R et al. Hypericin-loaded
nanoparticles for the photodynamic treatment of ovarian cancer. Int J
Pharm 2006; 326: 174–81. doi:10.1016/j.ijpharm.2006.07.012
114. Wessels JT, Busse AC, Rave-Fränk M et al. Photosensitizing
and radiosensitizing effects of hypericin on human renal car-
cinoma cells in vitro. Photochem Photobiol 2008; 84: 228–35.
doi:10.1111/j.1751-1097.2007.00225.x
115. Cole CD, Liu JK, Sheng X et al. Hypericin-mediated photo-
dynamic therapy of pituitary tumors: preclinical study in a GH4C1
rat tumor model. J Neurooncol 20 08; 87: 255–61. doi:10.1007/
s11060-007-9514-0
116. Penjweini R, Loew HG, Breit P et al. Optimizing the antitumor se-
lectivity of PVP-Hypericin re A549 cancer cells and HLF normal cells
through pulsed blue light. Photodiagnosis Photodyn Ther 2013; 10:
591–99. doi:10.1016/j.pdpdt.2013.06.005
117. ZhengY, YinG, LeV et al. Photodynamic-therapy activates immune
response by disrupting immunity homeostasis of tumor cells, which
generates vaccine for cancer therapy. Int J Biol Sci 2016; 12: 120–32.
doi:10.7150/ijbs.12852
Downloaded from https://academic.oup.com/jpp/article/73/4/425/6047495 by Universiteit Leiden / LUMC user on 18 October 2021
436 Journal of Pharmacy and Pharmacology, 2021, Vol. 73, No. 4
118. Kim H, Kim SW, Seok KH et al. Hypericin-assisted photodynamic
therapy against anaplastic thyroid cancer. Photodiagnosis Photodyn
Ther 2018; 24: 15–21. doi:10.1016/j.pdpdt.2018.08.008
119. Plenagl N, Duse L, Seitz BS et al. Photodynamic therapy–hypericin
tetraether liposome conjugates and their antitumor and antiangiogenic
activity. Drug Deliv 2019; 26: 23–33. doi:10.1080/10717544.2018.153
1954
120. Liang C, HaoF, YaoX etal. Hypericin maintians PDX1 expression
via the Erk pathway and protects islet β-cells against glucotoxicity
and lipotoxicity. Int J Biol Sci 2019; 15: 1472–87. doi:10.7150/
ijbs.33817
121. VanDePutteM, RoskamsT, BormansG etal. The impact of aggrega-
tion on the biodistribution of hypericin. Int J Oncol 2006; 28: 655–60.
doi:10.3892/ijo.28.3.655
Downloaded from https://academic.oup.com/jpp/article/73/4/425/6047495 by Universiteit Leiden / LUMC user on 18 October 2021
... Hypericin is known as a natural photosensitizing agent used in photodynamic therapy. In addition to its anticancer effect, it is characterized by fungicidal, bactericidal and antiviral properties [9][10][11][12][13][14][15][16][17][18][19][20][21]. HY's optical properties configure its absorption of the visible light spectrum from 500 to 620 nm, with a maximum absorption at 595 nm, showing light emission of the order of 603 nm and is characterized by an intense red fluorescence [22]. ...
... At the same time, immediately after irradiation, JNK1 and p38 MAPK are activated, while ERK are inhibited [1]. Clinical trials have shown that the use of HY-PDT has potential in the treatment of different neoplasmatic diseases, including recurrent mesothelioma and basal cell, and squamous cell carcinoma as well as psoriasis, warts and skin cancer malignant glioma, pituitary adenoma and cutaneous T-cell lymphoma [20,[25][26][27][28]. ...
... A new, undeniably interesting direction may be the use of antimicrobial PDT (aPDT) with HY, which is confirmed by research and the associated published results using HY-PDT f.e. in onychomycosis [33,34]. It has been observed that HY inhibits the proliferation of various tumor cells, including lung cancers, bladder, colon, glioma, breast, cervical, leukemia, hepatic, melanoma and lymphoma [20,21]. Its use in inflammatory skin diseases, with its immunomodulating effect, may allow this form of therapy in chronic inflammatory skin diseases that cannot be treated conventionally, such as psoriasis, atopic dermatitis, or skin infection [7,28,32,35]. ...
Article
Full-text available
Determination of the hypericin–photodynamic (HY–PDT) effect on the secretion of cytokines secreted by the skin cells, may be the basis for using the immunomodulatory effect of photodynamic action in the treatment of inflammatory skin diseases. The study aimed to evaluate the cytotoxic and immunomodulatory effects of hypericin (HY) in photodynamic therapy (PDT) performed in vitro on cultures of selected skin cell lines. The study used two human cell lines, primary dermal fibroblast (HDFa) and primary epidermal keratinocytes (HEKa). The MTT test was used to define the metabolic activity of treated cells. Cell supernatants subjected to sublethal PDT were assessed to determine the interleukins: IL-2, IL-8, IL-10, IL-11, IL-19, IL-22, and metalloproteinase 1 (MMP-1). The results confirm the destructive effect of HY–PDT and the immunomodulatory effects of sublethal doses on the selected skin cells, depending on the concentration of HY and the light doses. No statistically significant differences were noted in IL-2 and IL-10 concentration after HY–PDT for HEKa and HDFa lines. After using HY–PDT, the concentration of IL-8, MMP-1, IL-22, and IL-11 significantly decreased in the HEKa line. Moreover, the concentration of IL-19 and MMP-1 significantly decreased in the HDFa line. The concentration of IL-11 in the HDFa line after using only the HY, without the light, increased but decreased after HY–PDT. Our experiment confirmed that HY–PDT has not only a cytotoxic effect but, used in sublethal doses, also presents immunomodulatory properties. These may be an advantage of HY–PDT when used in the treatment of persistent skin inflammation, connected with the release of pro-inflammatory cytokines resistant to conventional treatment methods.
... In the last few decades, researchers have attempted to determine the usage of ICD in fundamental and clinical studies for cancer treatment [18][19][20][21][22][23][24][25][26][27]. Photodynamic therapy (PDT) along with several immunogenic dyes can be used as an ICD inducer [28][29][30][31][32][33][34][35][36]. These are produced by cell death during PDT, with PDT-treated cells breaking down into antigens that are easily taken up by antigenpresenting cells. ...
... Notably, certain tumors, such as mammary carcinoma 4T1, melanoma B16F10, and colon adenocarcinoma CT26, exhibit increased immunogenic potential for immunogenic PDT [73][74][75][76][77][78][79]. ICD-PDT is used for the treatment of highly challenging and life-threatening tumors, such as glioma, which are usually resistant to traditional therapies [30,80]. Further research should concentrate on discovering novel ICD inducers that have clinical potential. ...
Article
Full-text available
The development of cancer immunotherapy approaches gives hope to millions of patients for a better clinical outcome after tumor treatment. Investigating the fundamental mechanisms of antitumor immunity activation is an urgent task. The induction of immunogenic cell death is told by many authors, although the process is elegant and simple. Different stimuli can be brought to ICD, but photodynamic exposure has proven to be an effective inducer of programmed death on par with radiotherapy. The link between triggering a photodynamic response on cancer cells and triggering immunogenic cell death has been poorly described in experimental work. The question of which molecular cascades are activated after PDT irradiation and how they lead to the release of DAMPs is intriguing. Much is known about ROS generation and endoplasmic reticulum stress, but rarely about the Golgi apparatus. Photosensitizers of different natures can lead to different effects, including completely nonimmunogenic ones. In this review, we describe the cascades that link the induction of cell death by photodynamic exposure and the immunogenic pattern of DAMPs release. We discuss those photosensitizers that have shown potential as ICD inducers and talk about the different pathways of programmed death that occur during PDT exposure.
... For example, Hypercin is reported to accumulate and generate ROS in ER, and photosensitize cancer cells. 43 Redaporfin targets the endoplasmic reticulum and Golgi apparatus and shows its PDT effects though its direct antineoplastic action and indirect immunedependent destruction of malignant lesions. 44 Another important cellular component, lipid droplets (LDs) are closely associated with ER. ...
Article
Full-text available
Currently, effective therapeutic modalities for pan-creatic ductal adenocarcinoma (PDAC) are quite limited, leading to gloomy prognosis and ∼6-months median patient survival. Recent advances showed the promise of photodynamic therapy (PDT) for PDAC patients. Next generation photosensitizers (PS) are based on "organelle-targeted-PDT" and provide new paradigm in the field of precision medicines to address the current challenge for treating PDAC. In this investigation, we have constructed a novel PS, named as NbB, for precise and simultaneous targeting of endoplasmic reticulum (ER) and lipid droplets (LDs) in PDAC, based on the fact that malignant PDAC cells are heavily relying on ER for hormone synthesis. Our live cell imaging and fluorescence recovery after photobleaching (FRAP) experiments revealed that NbB is quickly targeted to ER and subsequently to LDs and shows simultaneous dual fluorescence color due to polar and nonpolar milieu of ER and LDs. Interestingly, the same molecule generates triplet state and singlet oxygen efficiently and causes robust ER stress and cellular lipid peroxidation, leading to apoptosis in two different PDAC cells in the presence of light. Together, we present, for the first time, a potential next generation precision medicine for ER-LD organelle specific imaging and PDT of pancreatic cancer.
... Hypericin is an important phytochemical derived from the genus Hypericum, especially from H. perforatum. It is a naphthodianthrone molecule with the chemical name 4,5,7,4′,5′,7′-hexahydroxy-2,2′dimethylnaphthodianthrone, and molecular weight of 504 g/ mol (Dong et al. 2021). Hypericin is isolated and extracted from the minute-coloured glands located on the aerial parts of the plant body, especially in flowers, young stems and leaves (Fig. 1). ...
Article
Full-text available
Hypericin, a polycyclic naphthodianthrone and active plant pigment with the molecular formula C30H16O8, is a crucial phytochemical extracted from the dark-colored glands present on the aerial parts of the genus Hypericum. It is biosynthesized through the polyketide pathway by plant-specific type III polyketide synthases (PKSs). In addition to hypericin, the genus Hypericum is rich in various classes of phytochemicals. Alongside other bioactive compounds like hyperforin and flavonoids, hypericin exhibits antidepressant activity. Recently, hypericin has gained increased importance in the research due to its unique properties. Its photodynamic nature makes it an effective natural photosensitizer, extending its use in investigating skin disorders. Moreover, hypericin demonstrates antiviral and antitumoral properties. Despite its effectiveness in treating cancers and neurological disorders, hypericin production faces challenges due to its site-specific nature. Conventional methods struggle to meet the growing demand for hypericin. Biotechnological approaches, including plant tissue culture and bioreactor-based large-scale production, offer promising solutions to address this demand. This review focuses on various plant tissue culture techniques, such as cell and organ culture, and elucidates their biosynthetic pathways. It also discusses hypericin production using elicitation strategies involving biotic and abiotic components, as well as genetic engineering approaches to enhance hypericin yields. Bioreactor-scale production presents significant potential for sustainable hypericin production. Further advancements in understanding and engineering biosynthetic pathways hold promise for unlocking new avenues in hypericin production.
... A significant correlation exists between BC and mitochondria, particularly in the realms of cellular energy metabolism, oxidative stress, and the regulation of apoptosis. In BC, cells exhibit a heightened demand for energy, and mitochondria, serving as the cellular energy center, generate adenosine triphosphate (ATP) through the process of oxidative phosphorylation to meet the rapid proliferation and growth requirements [35,36]. However, oxidative stress may lead to mitochondrial dysfunction, causing damage to mitochondrial DNA and consequently impacting normal cellular metabolism. ...
Article
Full-text available
Bladder cancer (BC) is the fourth and tenth most common malignancy in men and women worldwide, respectively. The complexity of the molecular biological mechanism behind BC is a major contributor to the lack of effective treatment management of the disease. The development and genesis of BC are influenced by mitochondrial retrograde control and mitochondria-nuclear cross-talk. However, the role of mitochondrial-related genes in BC remains unclear. In this study, we analyzed TCGA datasets and identified 752 DE-MRGs in BC samples, including 313 down-regulated MRGs and 439 up-regulated MRGs. Then, the results of machine-learning screened four critical diagnostic genes, including GLRX2, NMT1, PPP2R2B and TRAF3IP3. Moreover, we analyzed their prognostic value and confirmed that only PPP2R2B was associated with clinical prognosis of BC patients and Cox regression assays validated that PPP2R2B expression was a distinct predictor of overall survival in BC patients. Them, we performed RT-PCR and found that PPP2R2B expression was distinctly decreased in BC specimens and cell lines. Functional experiments revealed that overexpression of PPP2R2B distinctly suppressed the proliferation, migration and invasion of BC cells via Wnt signaling pathway. In summary, these research findings offer potential molecular markers for the diagnosis and prognosis of BC, with the discovery of PPP2R2B particularly holding significant biological and clinical significance. This study provides valuable clues for future in-depth investigations into the molecular mechanisms of BC, as well as the development of new diagnostic markers and therapeutic targets.
Chapter
Full-text available
With the emergence of several strains of antibiotic-resistant bacteria, there has been a demand for new therapeutic alternatives to combat the growth of microbial pathogens. PACT (Photodynamic Antimicrobial Chemotherapy) is a therapeutic modality, inspired by the treatment of tumors, that aims to kill microorganisms by photodamage, by employing the combination of a photosensitizer and light irradiation, usually of the wavelength of visible light. Thus, PACT can be applied in different contexts, with Dentistry as one of the greatest applications, but PACT can be used in the treatment of skin infections and even disinfection of surfaces in hospital and industrial environments. This paper addresses historical and technical aspects of PACT, in terms of mechanism of action, sensitive microorganisms and future perspectives. This document is the result of the final work of the discipline "Special Topics in Pharmacology II", prepared by graduate students in Natural Products, Synthetics, Bioactives, Cellular and Molecular Biology.
Article
Colorectal cancer (CRC) is significantly contributed to global cancer mortality rates. Treating CRC is particularly challenging due to metastasis and drug resistance. There is a pressing need for new treatment strategies against metastatic CRC. Photodynamic therapy (PDT) offers a well-established, minimally invasive treatment option for cancer with limited side effects. Hypericin (HYP), a potent photosensitizer for PDT, has been documented to induce cytotoxicity and apoptosis in various types of cancers. However, there are few reports on the inhibitory effects of HYP-mediated PDT on the metastatic ability of CRC cells. Here, we evaluate the inhibitory effects of HYP-mediated PDT against metastatic CRC cells and define its underlying mechanisms. Wound-healing and Transwell assays show that HYP-mediated PDT suppresses migration and invasion of CRC cells. F-actin visualization assays indicate HYP-mediated PDT decreases F-actin formation in CRC cells. TEM assays reveal HYP-mediated PDT disrupts pseudopodia formation of CRC cells. Mechanistically, immunofluorescence and western blotting results show that HYP-mediated PDT upregulates E-cadherin and downregulates N-cadherin and Vimentin. HYP-mediated PDT also suppresses key EMT regulators, including Snail, MMP9, ZEB1 and α-SMA. Additionally, the expressions of RhoA and ROCK1 are downregulated by HYP-mediated PDT. Together, these findings suggest that HYP-mediated PDT inhibits the migration and invasion of HCT116 and SW620 cells by modulating EMT and RhoA–ROCK1 signaling pathway. Thus, HYP-mediated PDT presents a potential therapeutic option for CRC.
Chapter
Fungal endophytes are present in every species of living plant throughout the earth forming a balanced symbiotic relationship with the plants. They produce diverse range of bioactive compounds that protect the host plant from biotic and abiotic stress contributing to the plant’s ecological success as well as their own. These bioactive compounds can be beneficial to not only the plants but also to human and the environment. A multitude of secondary metabolites produced by fungal endophytes showed antimicrobial activity against many plant and human pathogens including the antibiotic-resistant pathogens. They also produce antiparasitic compounds which can be used to eliminate deadly diseases including malaria. Many bioactive compounds produced by fungal endophytes also showed anticancer activity by disrupting the cell cycle, inducing apoptosis, and disrupting important pathways and gene expression. Furthermore, they also play a huge role in soil remediation. In this chapter, we discuss in detail fungal endophytes and their symbiotic relationship with their plant host. We also discuss the various bioactive compounds they produce and the different biological activities of these bioactive compounds.
Article
Full-text available
The aim of this study was to measure the secretion of interleukin (IL)-8 and -10 during an elicited immune response following sublethal doses of hypericin-mediated photodynamic therapy (HY-PDT) in experimental models of residual colon cancer cells in vitro. Investigations were performed on the cancer cell lines SW480 and SW620. Each cell line was exposed to 3 different concentrations of the photosensitizer HY and various doses of irradiation. The cell metabolic activity using an MTT assay was performed and then the measurement of IL-8 and IL-10 secretion was achieved using the Bio-Plex ProTMAssay. There was a statistically significant amplification of IL-8 secretion during HY-PDT in the SW620 cell line (at 1 J/cm2: P = .01, 5 J/cm2: P = .002, and 10 J/cm2: P = .025) and a statistically significant decrease in IL-8 during HY-PDT in the SW480 cell line (at 1 J/cm2: P = .05, 5 J/cm2: P = .035, and 10 J/cm2: P = .035). No statistically significant differences in IL-10 concentration were found following HY-PDT in the SW480 (at 1 J/cm2: P > .4, 5 J/cm2: P = .1, and 10 J/cm2: P = .075) or in the SW620 cell line (at 1 J/cm2: P > .4, 5 J/cm2: P > .4, and 10 J/cm2: P > .4). HY-PDT can both eliminate and control a primary tumor via cytotoxic effects, and at sublethal doses, it can affect IL release by colon cancer cells. In this experiment, this influence depended on the level of tumor cell metastatic activity.
Article
Full-text available
Photodynamic therapy with hypericin (HY-PDT) and hyperforin (HP) could be treatment modalities for colorectal cancer (CRC), but evidence of their effect on angiogenic factors in CRC is missing. Convenient experimental model utilization is essential for angiogenesis research. Therefore, not only 2D cell models, but also 3D cell models and micro-tumors were used and compared. The micro-tumor extent and interconnection with the chorioallantoic membrane (CAM) was determined by histological analyses. The presence of proliferating cells and HY penetration into the tumor mass were detected by fluorescence microscopy. The metabolic activity status was assessed by an colorimetric assay for assessing cell metabolic activity (MTT assay) and HY accumulation was determined by flow cytometry. Pro-angiogenic factor expression was determined by Western blot and quantitative real-time polymerase chain reaction (RT-qPCR). We confirmed the cytotoxic effect of HY-PDT and HP and showed that their effect is influenced by structural characteristics of the experimental model. We have pioneered a method for analyzing the effect of HP and cellular targeted HY-PDT on pro-angiogenic factor expression in CRC micro-tumors. Despite the inhibitory effect of HY-PDT and HP on CRC, the increased expression of some pro-angiogenic factors was observed. We also showed that CRC experimental micro-tumors created on quail CAM could be utilized for analyses of gene and protein expression.
Article
Full-text available
A decrease in islet β-cell mass is closely associated with the development and progression of diabetes. Therefore, protection against β-cell loss is an essential measure to prevent and treat diabetes. In this study, we investigated the protective effects of non-photoactivated hypericin, a natural compound, on β-cells both in vitro and in vivo. In vitro, hypericin greatly improved INS-1 cell viability under high-glucose and high-fatty-acid conditions by inhibiting glucotoxicity- and lipotoxicity-induced apoptosis and nitric oxide (NO) production. Then, we further demonstrated that hypericin elicited its protective effects against glucotoxicity and lipotoxicity in INS-1 cells by attenuating the reduction in pancreatic duodenal homeobox-1 (PDX1) expression and Erk activity. In vivo, prophylactic or therapeutic use of hypericin inhibited islet β-cell apoptosis and enhanced the anti-oxidative ability of pancreatic tissue in high-fat/high-sucrose (HFHS)-fed mice, thus alleviating β-cell loss and maintaining or improving β-cell mass and islet size. More importantly, hypericin treatment decreased fasting blood glucose, improved glucose intolerance and insulin intolerance, and alleviated hyperinsulinaemia in HFHS-fed mice. Therefore, hypericin showed preventive and therapeutic effects against HFHS-induced onset of type II diabetes in mice. Hypericin possesses great potential for development as an anti-diabetes drug in the future.
Article
Full-text available
Photodynamic therapy (PDT) as a non-aggressive therapy with fewer side effects has unique advantages over traditional treatments. However, PDT still has certain limitations in clinical applications, mainly because most photosensitizers utilized in PDT are hydrophobic compounds, which will self-aggregate in the aqueous phase and cause undesirable effects. In order to resolve this, we utilized the self-polymerization of dopamine molecules under alkaline conditions to coat cerium oxide nanorods (CeONR) with a dense polydopamine (PDA) film. Thereafter, thiolated galactose (Gal-SH) and hypericin (Hyp) were modified and loaded onto the surface to construct CeONR@PDA-Gal/Hyp, respectively, which can be used for targeted photodynamic therapy of human hepatoma HepG2 cells. CeONR@PDA-Gal/Hyp was characterized by transmission electron microscope (TEM), Zeta potential, Ultraviolet-visible (UV-Vis), and fluorescence spectroscopy, respectively. This hypericin delivery system possesses good biocompatibility and specific targeting ability, where the galactose units on the surface of CeONR@PDA-Gal/Hyp can specifically recognize the asialo-glycoprotein receptors (ASGP-R), which overexpress on HepG2 cell membrane. Furthermore, Hyp will detach from the surface of CeONR@PDA-Gal/Hyp after the nanorods enter cancer cells, and shows excellent PDT effect under the irradiation of light with a wavelength of 590 nm. Our work exemplifies a novel targeted delivery of hydrophobic photosensitizers for cancer treatment.
Article
The use of natural products as chemotherapeutic agents and tools for manipulation of apoptosis represent an attractive therapeutic concept. In this study, we investigated the anticancer activities of a combination of two natural compounds with different origin, hypericin (plant product) in its photoactive state and Manumycin A (yeast product) and explored the underlying mechanisms of their pro-apoptotic action using an oxaliplatin-resistant variant of human colon adenocarcinoma cell line HT-29-OxR as the experimental model. CCK-8 assay was performed to evaluate the cytotoxicity of the drugs. CalcuSyn software was used to identify the type of interaction between the two agents. BrdU incorporation assay and colony forming assay were performed to study the short- and long-term proliferation of cells. To evaluate the ability of the drug combination to induce apoptosis, PARP p85 fragment was detected using the ELISA method. Changes in apoptosis-related proteins were examined by immunoassays. Our results showed that a synergistic combination of photoactive hypericin and Manumycin A decreased viability, inhibited both short- and long-term cell proliferation, decreased levels of IAPs proteins (cIAP1, cIAP2, XIAP and survivin), induced an apoptotic PARP cleavage associated with decline in procaspase-3 level, promoted phagocytosis of cancer cells, and restored chemosensitivity to oxaliplatin.
Article
Hypericin (Hyp) is considered a promising photosensitizer for Photodynamic Therapy (PDT) due its high hydrophobicity, affinity for cell membranes, low toxicity and high photooxidation activity. In this study, Hyp photophysical properties and photodynamic activity against melanoma B16-F10 cells were optimized using DPPC liposome (1,2-dipalmitoyl-sn-glycero-3-phosphocholine) as a drug delivery system. This nanoparticle is used as cell membranes biomimetic model and solubilizes hydrophobic drugs. Hyp oxygen singlet lifetime (τ) in DPPC was approximately 2 fold larger than in P-123 micelles (PluronicTM surfactants), reflecting a more hydrophobic environment provided by the DPPC liposome. On the other hand, singlet oxygen quantum yield (ΦΔ1O2) in DPPC and in P-123 were similar; Hyp molecules were preserved as monomers. Hyp/DPPC liposome aqueous dispersion was stable for fluorescence emission and liposome diameter for at least 5 days at 30 oC. However, the liposomes collapsed after lyophilization/rehydration process, which was resolved by adding the lyoprotectant Trehalose to liposome the dispersion prior to lyophilization. Cell viability of the Hyp/DPPC formulation was assessed against the healthy HaCat cells and high-metastatic melanoma B16-F10 cells. Hyp incorporated in the DPPC carrier presented higher selectivity index than Hyp sample previously solubilized in ethanol under illumination effect. Moreover, the IC50 was lower for Hyp in DPPC than for Hyp pre-solubilized in ethanol. These results enable the formulation Hyp/DPPC for future biomedical applications in PDT treatment
Article
Background and aim: Antimicrobial photodynamic therapy (aPDT) has received considerable attention as an emerging and promising approach for treating superficial infections. The aim of this study was to investigate aPDT mediated by aloe emodin (AE), a natural compound isolated from Aloe vera and Rheum palmatum, against multidrug-resistant (MDR) Acinetobacter baumannii clinical isolates in vitro. Methods: The photodynamic inactivation (PDI) efficacies of AE on three MDR A. baumannii isolates were assessed by colony forming units (CFU) assay. The aPDT effects mediated by AE on the genomic DNA, membrane integrity, and cellular structure of MDR A. baumannii were also investigated. Results: AE showed no obvious dark toxicity, but inactivated the MDR A. baumannii isolates in an AE concentration and light energy dose-dependent manner. Agarose gel electrophoresis and LIVE/DEAD BacLight Bacterial Viability kit assay indicated that the genomic DNA and membrane integrity of MDR A. baumannii were damaged after AE-mediated aPDT treatment. Transmission electron microscopy (TEM) images demonstrated that AE-mediated aPDT could induce rupture of bacterial cell wall and membrane, and condensation of ribosomes in the cytoplasm. Conclusions: The results obtained in this study suggested that AE could serve as a potential antibacterial photosensitizer in the treatment of superficial infections caused by MDR A. baumannii.
Article
Background: Alveolar rhabdomyosarcoma (RMA) is a highly malignant soft tissue tumor in children with poor prognosis and failure of established therapies in advanced stages. Therefore, novel treatment options are required. Photodynamic therapy (PDT) has been found useful for the treatment of different tumor entities and might represent such a novel treatment option. A major limitation of PDT remains the restriction to superficial tumor cell layers as illumination with light is essential for the generation of reactive oxygen species. Current research focusses on the development of modified Hypericin (HYP)-based photosensitizers, as well as combining PDT and targeted internal radiotherapy with 131I, to generate an additive anti-tumor effect. Methods: A standardized protocol for in vitro Hypericin-PDT was established in RMA cells. The anti-tumor properties of this photosensitizer were analyzed on molecular and metabolic levels. Changes in cell morphology were visualized using bright field-, fluorescence- and scanning-electron microscopy. Iodinated Hypericin derivatives with both radioactive and non-radioactive isotopes 131I/127I were employed to establish a targeted radionuclide therapy and investigate the potential of a combined treatment with PDT. Results: In vitro photodynamic treatment with Hypericin showed a strong anti-tumor efficiency with favorable cellular uptake and compromised cancer cells on metabolic and molecular levels. Iodination of the photosensitizer did not impair the photosensitizer´s properties. Targeted radiotherapy with 131I-HYP led to distinct reductions of tumor viability. A simultaneously performed PDT leads to a reduction of cell viability that begins earlier in time. However, an additive enhancement of the cell viability was not observed in the selected dose range. Conclusion: In this in vitro study, we got a first insight of a possible potential of Hypericin for the treatment of pediatric soft tissue sarcoma. By coupling with radioiodine, we developed a novel approach for a combined anti-tumor treatment. The in vitro experiments lay the foundation for further in vivo experiments, which are needed to study the effects of a sequential administration of 131I-HYP and HYP.
Article
Photodynamic therapy (PDT) of hypericin is hampered by poor water solubility and photostability. Incorporation of photosensitizers into nanocarriers has been designed to solve these issues. Herein, SWNH-Hyps nanohybrids were first fabricated by loading hypericin (Hyp) on the surface of single-walled carbon nanohorns (SWNHs) through ππ interaction and exhibited high solubility and stability in aqueous water. SWNH-Hyps could be utilized for a single platform for cancer therapy because it could simultaneously generate enough reactive oxygen species and hyperthermia using light irradiation. What’s more, the SWNHs not only improved water solubility, photostability and therapy effect of Hyp but also protected it from light degradation. SWNH-Hyps could effectively ablate 4T1 cells by photodynamic/photothermal synergistic therapy upon 590 and 808 nm light irradiation compared to PDT. Furthermore, remarkable tumor cell death as well as tumor growth inhibition were proved via photothermal therapy (PTT) and PDT of SWNH-Hyps under 590 and 808 nm light irradiation, which demonstrated synergistic anticancer ability of SWNH-Hyps was better than that of free Hyp in vivo. Such a simple and facile adsorption method improved water solubility of Hyp and then enhanced its therapy effect, which displays that SWNHs are hopefully used in medicine in the future.