ArticlePDF Available

Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy

Authors:
Macroheterocycles
Review
Макрогетероциклы
http://mhc-isuct.ru
Обзор
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 © ISUCT Publishing 207
DOI: 10.6060/mhc224870k
Synthesis Strategy of Tetrapyrrolic Photosensitizers
for Their Practical Application in Photodynamic Therapy
Oskar I. Koifman,a,b Tatyana A. Ageeva,a Natalia S. Kuzmina,c Vasilii F. Otvagin,c
Alexander V. Nyuchev,c Alexey Yu. Fedorov,c Dmitriy V. Belykh,d
Nataliya Sh. Lebedeva,b Elena S. Yurina,b Sergey A. Syrbu,a,b Mikhail O. Koifman,a
Yury A. Gubarev,b Dmitry A. Bunin,e Yulia G. Gorbunova,e,f Alexander G. Martynov,e
Aslan Yu. Tsivadze,e,f Semyon V. Dudkin,g Alexey V. Lyubimtsev,a Larissa A. Maiorova,a,h
Maria B. Kishalova,a Maria V. Petrova,a Vladimir B. Sheinin,b Vladimir S. Tyurin,e
Ilya A. Zamilatskov,e Eduard I. Zenkevich,i Philipp K. Morshnev,a,b Dmitry B. Berezin,a
Eduard A. Drondel,a Andrey V. Kustov,a,b Viktor A. Pogorilyy,k Alexey N. Noev,k,l
Elizaveta A. Eshtukova-Shcheglova,k Ekaterina A. Plotnikova,l Anna D. Plyutinskaya,l
Natalia B. Morozova,l Andrei A. Pankratov,l Mikhail A. Grin,k Olga B. Abramova,m
Ekaterina A. Kozlovtseva,m Valentina V. Drozhzhina,m Elena V. Filonenko,l
Andrey D. Kaprin,l Anastasiya V. Ryabova,n,o Daria V. Pominova,n,o Igor D. Romanishkin,n
Vladimir I.Makarov,n,o Victor B. Loschenov,n,o Kseniya A. Zhdanova,k
Anastasia V. Ivantsova,k Yulia S. Bortnevskaya,k Natal’ya A. Bragina,k Anna B. Solovieva,p
Anastasya S. Kuryanova,p and Petr S. Timashevp,q
aResearch Institute of Macroheterocyclic Compounds, Ivanovo State University of Chemistry and Technology, 153000
Ivanovo, Russia
bG.A. Krestov Institute of Solution Chemistry of the Russian Academy of Sciences, 153045 Ivanovo, Russia
cN.I. Lobachevsky State University of Nizhny Novgorod, 603950 Nizhny Novgorod, Russia
dInstitute of Chemistry of Komi Republic Scientific Centre of the Ural Branch of Russian Academy of Sciences, 167000
Syktyvkar, Russia
eA.N. Frumkin Institute of Physical Chemistry and Electrochemistry, 119991 Moscow, Russia
fN.S. Kurnakov Institute of General and Inorganic Chemistry, Russian Academy of Sciences, 119991 Moscow, Russia
gA.N. Nesmeyanov Institute of Organoelement Compounds of the Russian Academy of Sciences (INEOS RAS), 119334
Moscow, Russia
hInstitute of Pharmacoinformatics, Federal Research Center “Computer Science and Control” of Russian Academy of
Sciences, Moscow, Russia
iBelarussian National Technical University, 220013 Minsk, Belarus
kMIREA-Russian Technological University, 119454 Moscow, Russia
lP. Hertsen Moscow Oncology Research InstituteBranch of the National Medical Research Radiological Centre of the
Ministry of Health of the Russian Federation, 125284 Moscow, Russia
mLaboratory of Experimental Photodynamic Therapy, A.F. Tsyb Medical Radiological Research Center Branch of the
National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249034 Obninsk, Russia
nProkhorov General Physics Institute, Russian Academy of Sciences, 119991 Moscow, Russia
oDepartment of Laser Micro-, Nano- and Biotechnologies, National Research Nuclear University MEPhI, 115409 Moscow,
Russia
pN.N. Semenov Federal Research Center for Chemical Physics of the Russian Academy of Sciences, 119991 Moscow, Russia
qInstitute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
This review presents a wide range of tetrapyrrole photosensitizers used for photodynamic therapy (PDT), antimicro-
bial photodynamic therapy, photoinactivation of pathogens. Methods of synthesis and design of new photosensitizers
with greater selectivity of accumulation in tumor tissue and increased photoinduced antitumor activity are consid-
ered. The issues of studying the properties of new photosensitizers, their photoactivity, the ability to generate singlet
oxygen, and the possibility of using targeted photodynamic therapy in clinical practice are discussed. The review ex-
amines the work on PDT by national and foreign researchers.
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
208 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
Keywords: Photodynamic therapy, antimicrobial photodynamic therapy, tetrapyrrolic macroheterocyclic compounds,
porphyrins, phthalocyanines, chlorins, bacteriochlorins, synthesis, photosensitizers, optical properties, proteins, SARS-
CoV-2, photoinactivation, optochemical diagnostics, self-assembling, nanostructures; Langmuir-Schefer films, extra-
ligation, quantum dots, singlet oxygen generation, amphiphilic polymers, diagnostics, clinical application of PDT.
Стратегия синтеза тетрапиррольных фотосенсибилизаторов
для их практического применения в фотодинамической терапии
О. И. Койфман,a,b T. A. Aгеева,a Н. С. Кузьмина,c В. Ф. Отвагин,c A. В. Нючев,c
A. Ю. Федоров,c Д. В. Белых,d Н. Ш. Лебедева,b Е. С. Юрина,b С. А. Сырбу,a,b
M. O. Koйфман,a Ю. А. Губарев,b Д. А.Бунин,e Ю. Г. Горбунова,e,f A. Г. Мартынов,e
А. Ю. Цивадзе,e,f С. В. Дудкин,g А. В. Любимцев,a Л. А. Майорова,a,h
М. Б. Кишалова,a М. В. Петрова,a В. Б. Шейнин,b В. С. Тюрин,e И. А. Замилацков,e
Э. И. Зенькевич,i Ф. К. Моршнев,a,b Д. Б. Березин,a Э. А. Дрондель,a А. В. Кустов,a,b
В. А. Погорилый,k А. Н. Ноев,k,l E. A. Ештукова-Щеглова,k E. A. Плотникова,l
A. Д. Плютинская,l Н. Б. Морозова,l A. A. Панкратов,l M. A. Грин,k O. Б. Абрамова,m
E. A. Koзловцева,m В. В. Дрожжина,m E. В. Филоненко,n A. Д. Каприн,l
A. В. Рябова,n,o Д. В. Поминова,n,o И. Д. Романишкин,n В. И. Макаров,n,o
В. Б. Лощенов,n,o К. А. Жданова,k А. В. Иванцова,k Ю. С. Бортневская,k
Н. А. Брагина,k A. Б. Соловьева,p A. С. Курьянова,p П. С. Тимашевp,q
Elena V. Guseva,a@ Albina V. Potapova,b and Elena V. Fesikc
aИнститут макрогетероциклических соединений, Ивановский государственный химико-технологический университет,
153000 Иваново, Россия
bИнститут химии растворов им. Г.А. Крестова РАН, 153045 Иваново, Россия
cНижегородский государственный университет им. Н.И. Лобачевского, 603950 Нижний Новгород, Россия
dИнститут химии Коми научного центра Уральского отделения РАН, 167000 Сыктывкар, Россия
eИнститут физической химии и электрохимии им. А.Н. Фрумкина РАН, 119071 Москва, Россия
fИнститут общей и неорганической химии им. Н.С. Курнакова РАН, 119991 Москва, Россия
gИнститут элементоорганических соединений им. А.Н. Несмеянова РАН (ИНЭОС РАН), 119334 Москва, Россия
hИнститут фармакоинформатики Федерального исследовательского центра «И нформатика и управление»
Российской академии наук, Москва, Россия
iБелорусский национальный технический университет, 220013 Минск, Беларусь
kМИРЭА - Российский технологический университет, 119454 Москва, Россия
lМНИОИ им. П.А. Герцена - филиал ФГБУ «НМИЦ радиологии» Минздрава России, 125284 Москва, Россия
mМедицинский радиологический научный центр им. А.Ф. Цыба – филиал ФГБУ «НМИЦ радиологии» Минздрава
России, 249036 Обнинск, Россия
nИнститут общей физики им. А.М. Прохорова РАН, 119991 Москва, Россия
oНациональный исследовательский ядерный университет «МИФИ», 115409 Москва, Россия
pФедеральный исследовательский центр химической физики им. Н.Н. Семенова РАН, 119991 Москва, Россия
qИнститут регенеративной медицины Московского государственного медицинского университета
им. М.М. Сеченова, Москва, Россия
В обзоре представлен широкий спектр тетрапиррольных фотосенсибилизаторов, применяемых для фото-
динамической терапии (ФДТ), антимикробной фотодинамической терапии, фотоинактивации патогенов.
Рассмотрены методы синтеза и дизайн новых фотосенсибилизаторов, обладающих большей селективно-
стью накопления в опухолевой ткани и повышенной фотоиндуцированной противоопухолевой активностью.
Обсуждаются вопросы исследования свойств новых фотосенсибилизаторов, их фотоактивность, способность
генерировать синглетный кислород, возможности применения таргетной фотодинамической терапии в кли-
нической практике. В обзоре рассмотрены работы по ФДТ отечественных и зарубежных исследователей.
Ключевые слова: Фотодинамическая терапия, антимикробная фотодинамическая терапия, тетрапиррольные
макрогетероциклические соединения, порфирины, фталоцианины, хлорины, бактериохлорины, синтез, фото-
сенсибилизаторы, оптические свойства, белки, SARS-CoV-2, фотоинактивация, оптохимическая диагностика,
наноматериалы, самоорганизация, наноструктуры, пленки Ленгмюра-Шеффера, экстракоординация, кванто-
вые точки, генерация синглетного кислорода, амфифильные полимеры, диагностика, клиническое примене-
ние ФДТ.
Macroheterocycles
Review
Макрогетероциклы
http://mhc-isuct.ru
Обзор
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 209
Introduction
The idea of this review was born as a result of a de-
tailed discussion of the problems of photodynamic therapy
at the “PDT Day” section at the XIV International Confer-
ence "Synthesis and Application of Porphyrins and Their
Analogues" (ICPC-14), held at Ivanovo State University of
Chemistry and Technology from 29th June to 4th July,
2022. The unique ability of porphyrins and related com-
pounds to generate reactive oxygen species under the influ-
ence of light necessitates the synthesis of new photosensi-
tizers with the required properties (such as selectivity, viru-
lence, bacteriocide properties, etc.). Such properties are a
crucial part of effective use of photosensitizers in photody-
namic therapy, clinical laboratory diagnostics and photody-
namic inactivation of bacteria and viruses that can be harm-
ful to humans. Therefore, the review focuses on the synthet-
ic aspects of tetrapyrrole macroheterocycles and their modi-
fication for the purposes of targeting photodynamic therapy.
Leading specialists in the photosensitizers’ synthesis and
research, as well as PDT practitioners were involved in
writing this article.
List of Abbreviations:
1O2 singlet oxygen
aPDT antibacterial photodynamic therapy
BC bacteriochlorin (7,8,17,18-tetrahydroporphyrin)
BSA bovine serum albumin
CMPI 2-Chloro-1-methylpyridinium iodide
DAMPs damage-associated molecular patterns
DCC dicyclohexylcarbodiimide
EDAC 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide
FLI fluorescence imaging
FRET fluorescence resonance energy transfer
FRET foerster resonance energy transfer
GFC gel filtration chromatography
GSH glutathione
HCP1 heme carrier protein 1
HDACi histone deacetylase inhibitor
HDACs histone deacetylases
HDL high-density lipoproteins
HLB hydrophilic-lipophilic balance
iBC isobacteriochlorin (2,3,7,8-tetrahydroporphyrin)
LDL low density lipoproteins
MAM metastasis-associated macrophages
MDR multiple drug resistance
MHC macroheterocyclic compounds
MRI magnetic resonance imaging procedure
NIR near infrared
PBS phosphate buffered saline
Pcs phthalocyanines
PDT photodynamic therapy
PL photoluminescence
PSs photosensitizers
QD quantum dots
RBD receptor-binding domain
ROS reactive oxygen species
TAM tumor associated macrophages
TFA trifluoroacetic acid
TKI tyrosine kinases inhibitors
TLC thin-layer chromatography
TNTR the tumor-to-normal tissue ratio
TOPO tri-n-octyl phosphine oxide
1. The Key Role of Tetrapyrrole Macroheterocyclic
Compounds in Photodynamic Therapy
Photodynamic therapy (PDT) is a perspective socially
significant method of treatment successfully applied both
for tumor neoplasms and microbial infections caused by
poly-resistant pathogens.[1-9] This method requires the pres-
ence of three components: irradiation (including NIR radia-
tion), photosensitizer (PSs, phototherapeutic agent) and
molecular oxygen. Photosensitizers, substances selectively
localized in tumor or microbial cells, produce reactive oxy-
gen species (ROS)[4,10-13] when exposed to the red light in
the presence of dioxygen.
In recent three decades, photodynamic therapy has at-
tracted much attention due to its non-invasive features, low
side effects and low systemic toxicity.[14-16] The photosensi-
tizers are activated on exposure to light and become photo-
sensitizers’ triplet, which, react with molecular oxygen to
produce reactive oxygen species (Figure 1).[17,18] The hy-
droxyl radical is another reason which leads to the reaction
between the photosensitizer and molecular oxygen. These
cytotoxic molecules induce a series of biological reactions
that ultimately lead to cell death. The outcomes of PDT
depend on the nature of the cells, as well as the on the prop-
erties and localization of photosensitizer and the illumina-
tion conditions. Its obvious advantage is that cause negligi-
ble damage to the surrounding normal tissues and has little
systemic effects.
Correspondingly, PSs is one of the key factors of
PDT, and its physico-chemical properties (e.g. interaction
with molecular oxygen, efficiency of singlet oxygen gen-
eration, solubility, successful targeting and delivery, etc.)
largely determine the outcome of PDT. In this respect, it
was evidently shown that tetrapyrrolic macrocycles (por-
phyrins, chlorins, bacteriochlorins, phthalocyanines) are
potentially translatable PSs with exceptional properties,
multifunctional uses and excellent biocompatibility for
PDT, thus their special affinity to cancer cells makes them
the ligands par excellence for anticancer drugs.[19-28] One of
the prominent advantages of most porphyrins is that they
have special affinity for cancer cells, which enables them to
selectively remain in cancer cells, providing feasibility for
the research of anti-tumor targeted drugs.[2932]
Figure 1. Schematic diagram of photodynamic therapy.
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
210 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
These tetrapyrrolic macroheterocyclic compounds are
the focus of research because of their stability under a wide
range of environmental conditions, absorption in the visible
and near infra-red ranges of the electromagnetic spectrum
(4001000 nm), many derivatives possess long lived triplet
excited states that sensitize singlet oxygen (1O2) formation
to potentiate phototoxicity as anticancer and antibiotic
agents. These macrocycles can chelate with nearly every
metal in the periodic table which results in variable func-
tional properties such as tuneable excited state lifetimes,
redox potentials, catalytic activities, molar absorption coef-
ficients, and molecular dynamics. Functional groups on the
periphery of structurally rigid porphyrinoids provide topo-
logical diversity, making these pigments well suited for the
engineering of supramolecular materials.[33-36]
Porphyrins and their analogues are organic heterocy-
clic macrocycles with photophysical properties well-suited
for clinical phototherapy and cancer imaging. However,
their wider application in the clinical management of dis-
ease is barred by poor aqueous solubility, bioavailability,
tumour accumulation and skin phototoxicity. These limita-
tions require the search of new photoactive systems devoid
of these disadvantages
Chemists devoted many efforts to improving the effi-
ciency and selectivity of the photosensitizers to prevent the
side-effects of PDT over the last few decades. The perfect
photosensitizers should meet several criteria: (1) pure and
stable molecule, (2) no cytotoxicity in the dark, (3) optimal
absorption, distribution, metabolism and excretion proper-
ties, (4) high molar absorption coefficient in the long wave-
length region (650800 nm) for maximum light penetration
through tissue, (5) high 1O2 quantum yield, (6) tumor selec-
tivity and (7) ease of synthesis and a scalable process.[37-41]
Today, numerous tetrapyrrole macroheterocyclic compounds
that have been investigated as promising photosensitizers
for use in PDT meet several necessary criteria, such as
strong absorbing properties in the range from 650 to 700 nm,
high quantum yield in the triplet state, high photostability
and often minimal toxicity. In the world clinical practice,
many tetrapyrrol photosensitizers have already found their
application for the treatment of various forms of cancer.
As the PDT method was developing, all photosensitiz-
ers used in the clinical practice were grouped into several
generations, mainly based on their spectral and photophysi-
cal properties.[17,23,42] The first generation of PS includes
porphyrin derivatives, for example, hematoporphyrin iso-
lated from blood (Figure 2). Based on this compound, the
first domestic photosensitizer "Photogem"[43] was developed
by Prof. A.F. Mironov, the founder of our scientific team.
The second generation combines natural and synthetic
compounds, including chlorins, bacteriochlorins, phthalo-
cyanines, benzoporphyrins, and others. Finally, the third
generation includes PS with enhanced spectral properties
that absorb in the near-infrared range of the spectrum and
show targeting against tumors of various genesis.
A huge number of reviews have been published on
the chemistry, photochemical characteristics and clinical
applications of PDT photosensitizers.[4461] The number of
publications devoted to tetrapyrrol photosensitizers, which
are at different stages of development and clinical trials, is
growing rapidly.
Figure 2. Classification of the generations of PSs developed so
far.
The history of the development of PDT using
tetrapyrrole photosensitizers dates back almost a hundred
years. The starting date was an observation made in 1924
by A. Policard who revealed accumulation in animal tumors
of endogenous porphyrins able to fluoresce under UV irra-
diation.[62] In 1942, H. Auler and G. Banzer recorded red
fluorescence in the primary tumor and metastases in rats
subjected to subcutaneous and intramuscular administration
of hematoporphyrin.[63]
The modern stage of development of fluorescence di-
agnosis and photodynamic therapy began in the 1960s,
when R. Lipson et al. (the United States) revealed the pos-
sibility to record the fluorescence of porphyrin in tumors of
cancer patients who were administered the hematoporphy-
rin derivative prepared by acetylation and reduction of a
porphyrin mixture enriched in hydrophobic oligomers.[64] It
is commonlythat wide clinical use of photodynamic therapy
of cancer began in 1978, when T. Dougherty et al. reported
the results from application of this method for treatment of
25 patients with 113 primary, recurrent, and metastatic skin
tumors.[65]
In Russia, photodynamic therapy of tumors has been
the subject of experimental research for many years, but it
was not until 1992 that it received development in clinical
practice, when the first dosage form of the domestic photo-
sensitizer Photohem from a group of hematoporphyrin de-
rivatives was created. Successful clinical trials were con-
ducted at Gertsen Moscow Research Oncological Institute
and the State Scientific Center of Laser Medicine. After two
years (in 1994), clinical trials of second generation photo-
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 211
sensitizer Photosens, on the base sulfonated aluminum
phthalocyanine, developed at the SSC Research Institute of
Organic Intermediates and Dyes, (Corresponding Member
of RAS Prof. G.N. Vorozhtsov, Prof. E.A. Luk’yanets)
were started. In 1999, clinical application of Alasens, a 5-
aminolevulinic acid-based drug (GHTs NIOPIK, Corre-
sponding Member of RAS Prof. G.N. Vorozhtsov, Prof.
E.A. Luk’yanets) and in 2002 and 2004, that of drugs pre-
pared from e6 chlorin, Radachlorin [Radafarma, Limited
Liability Company, A.V. Reshetnikov], and Photoditazine
(VETAGRAND, Limited Liability Company, Prof. G.V.
Ponomarev), respectively, were begun. Three decades of
the history of the creation of tetrapyrrol photosensitizers
that have received clinical use in Russia are reflected in
Figure 3. The introduction of new treatment methods in
Russia was facilitated by the development of appropriate
domestic diagnostic and therapeutic equipment. The availa-
ble instrumental base and domestic photosensitizers make
photodynamic therapy not only a highly effective but also
an economically feasible method.[66]
Although a few porphyrin/chlorin based PSs have
been approved for clinical use, PDT is still limited by the
low stability and poor tumor targeting capacity of the large
majority of these macrocycles in vivo. The main obstacles
in effective delivery of PSs are associated with these intrin-
sic properties. At present, two possible approaches to over-
come this problem seem to be of interest in this direction.
One way is the supramolecular organic chemis-
try/photochemistry, a highly interdisciplinary field of sci-
ence covering the chemical, physical, and biological fea-
tures of chemical species held together and organized by
means of intermolecular binding interactions of covalent
and non-covalent nature. The resulting crossover has pro-
vided novel principles and concepts in physico-chemistry
such as molecular recognition, self-organization, regulation,
cooperativity, replication. A significant interest of numer-
ous scientific groups in this direction has been devoted to
the design and investigation of tetrapyrrole compounds that
fold or assemble predictably in order to form multicompo-
nent well-defined arrays which may be perspective in nano-
biomedicine.[72-76]
On the other hand, small molecular PSs (including
tetrapyrrolic macrocycles of various structure and physico-
chemical properties) can be attached to functional organic
or inorganic nanomaterials to enhance the PS stability and
tumor targeted delivery, and, in addition, some functional-
ized nanocarriers themselves can be directly used as
PSs.[51,77,78] To date, nanostructures of different types and
morphology (such as metal nanoparticles, semiconductor
quantum dots, graphene-based nanomaterials, liposomes,
ROS-sensitive nanocarriers and supramolecular nano-
materials)[79-88] have been tested as possible nanoplatforms
for tetrapyrrolic PSs. Various nanomaterial systems have
shown great potentials in improving PDT.[51,78,89]
The application of PDT is not only limited to oncolo-
gy, but is also being explored for the treatment of cardio-
vascular, dermatological, ophthalmic and infectious diseas-
es.[48] In accordance with the achievements of PDT today,
five main areas of application can be distinguished. These
are: (i) the anti-cancer agents for photodynamic therapy
(PDT), (ii) the anti-cancer agents for photoimmunotherapy
(PIT), (iii) the agents for photodynamic therapy (PDT) in
ophthalmology, (iv) the agents for photodynamic therapy
(PDT) in dermatology, and (v) the anti-microbial agents for
photodynamic inactivation (PDI).
How to enhance the selective uptake and uniform dis-
tribution of photosensitizers in tumor tissues is still an ur-
gent conundrum to be solved, especially the specific target-
ing of different cells and tumor tissues, to improve the effi-
cacy and reduce their toxic and side effects on healthy tis-
sues and organs. This may be a promising research direc-
tion, introducing functionally active groups into porphyrin
photosensitizers to improve their targeting and anti-tumor
activity, and then selectively delivering them to tumor tis-
sues with transport carriers such as liposomes, magnetic
nanoparticles and metalorganic frameworks. The ad-
vantages are as follows: photosensitizers bond with bio-
active groups, which can exert synergistic therapeutic
effects and improve the efficacy. Also, combining with a
transport carrier having a bio-recognition function is
conducive to improving the ability of the photosensitiz-
ers to penetrate tissues, enhancing the targeting of tumor
tissues and increasing the drug-level concentration at the
target site.
Further improvement of the photodynamic therapy
technique requires finding new photosensitizers having
higher photoactivity, possessing better tumor-tropic properties,
and excitable in the near-infrared spectral region, as well as
designing highly sensitive and reliable diagnostic and ther-
apeutic equipment. Clinical experience with photodynamic
therapy allows rating this method among the most
promising directions in modern clinical oncology.
= 630 nm
= 676 нм
= 662 nm
= 661-662 nm
= 662 nm
= 747 nm
Photogem[44]
Photosens[67]
Radachlorin[68]
Photoditazine[69]
Photoran E6[70]
Bacteriosens [71]
Figure 3. The Russian tetrapyrrolic photosensitizers that have received clinical use for PDT.
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
212 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
2. Conjugation of photosensitizers with cytotoxic molecules
an emerging trend in combination therapy
One of notorious problems associated with cancer dis-
eases is a lack of the universal treatment protocol for pa-
tients diagnosed with a cancer. Due to genetic uniqueness
of cancer cells, it is almost impossible to propose the treat-
ment, whose outcome is completely predictable. In this
regard, the combined therapy aimed to multiple cellular
targets has many advantages over conventional chemother-
apy.[90] Thus, combining various anticancer drugs it is pos-
sible to reverse drug resistance and fight metastatic can-
cers,[91] what is difficult for singular modalities. Moreover,
combination therapy works in a synergistic manner, and
therefore a lower therapeutic dosage of each individual drug
is required. Among possible drug combinations the utiliza-
tion of photodynamic therapy and chemotherapy together
stands out as an emerging trend.[92,93] The successful out-
come of PDT is highly dependent on a proper irradiation of
a full area where cancer tissues are located. Combined
treatment with a chemotherapeutic drug lowers this risk and
inhibits pathogenic cells survived PDT.[94] Another notable
advantage is the enhanced delivery of chemotherapeutic
drugs after conjugation with porphyrinoid photosenstitizers.
Due to binding with low density lipoproteins (LDL),[95]
heme carrier protein 1 (HCP1)[96] or peripheral benzodiaze-
pine receptor[97] the photosensitizing part can increase con-
centration of the chemotherapeutic counterpart in tumor
cells. Therefore, along with therapeutic properties PS
demonstrates a targeting delivery as well. Thus, such syner-
gistic behavior can lead to reduce of a drug dose and sys-
temic toxicity.[98,99] Another outstanding advantage of com-
bining chemotherapy with PDT is the unique ability of PDT
to induce an immune response to treatment.[100] Since the
result of photodynamic action may be an inflammatory re-
action, the resulting damage-associated molecular patterns
(DAMPs) can be recognized by the corresponding receptors
of neutrophils, which ultimately leads to the activation of
the immune system in response to PDT.
Generally, there are many possible ways to connect PS
with a chemotherapeutic molecule. The first way is admin-
istration of two or more drugs independently during one
course of a treatment. To achieve a proper combinational
effect in this case, a dosage has to be carefully chosen.[101]
Another option is the incorporation of drugs in targeted
systems - liposomes or nanoparticles.[91,102] One of the main
problems associated with such approach is unpredictable
properties arising during manufacturing of these delivery
platforms. Thus, the procedures for surface modification
may strongly modify the size, charge, shape, stability, drug
loading, and releasing ability of the nanovectors,[103] what
can cause undesirable side effects. In recent decades,
covalent binding of a photosensitizer and a cytostatic
agent demonstrated exceptional utility and efficiency
(Figure 4).[92,104107] Such platform operates with homoge-
neous, chemically pure and stable compounds, which satis-
fies the requirements for an ideal photosensitizer.[92,108] In
addition, the structure of covalent conjugates can be easily
varied over a wide range, thereby changing their biological
activity. In order to create chemical bonding between active
parts specific linkers are used, which provide sufficient
separation in the space of the active fragments.
Figure 4. Combining chemotherapy with PDT in conjugated system.
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 213
Scheme 1.
Interestingly, in some cases the chemotherapeutic part
can also demonstrate binding to key proteins in cells and as
a consequence improves delivery of PS.[106,109111] Thus,
tyrosine kinases inhibitors (TKI) have been proposed as a
perspective cytostatic vector for the design of conjugates.
One of the widely utilized TKI for construction of conju-
gates is erlotinib, which binds to the epidermal growth fac-
tor receptor (EGFR) known for its overexpression in many
types of tumors.[103] Erlotinib provides targeted binding of
the conjugate with EGFR and inhibits its activity what leads
to a death of cancer cells.
One of the recent examples of photoactive conjugates
designed with utilization of erlotinib is conjugate 1
(Scheme 1).[112] In this work Huang with colleagues sug-
gested a conjugate 1, where the copper(II) porphyrin moiety
plays a dual role both as a photosensitizer and a sensor for
glutathione-dependent apoptotic processes. The decrease of
glutathione (GSH) level is an important benchmark during
apoptotic processes.[113] Interestingly, paramagnetic copper
being bound to NH fragments is capable to coordinate GSH
which leads to fluorescence intensification.[114] However,
when the concentration of glutathione is low, the copper(II)
porphyrin exists in a fluorescence-quenching aggregation
state.[114] This phenomenon was used to visualize apoptotic
cells and monitor the therapeutic effect during PDT.[112]
Copper tetraarylporphyrin and erlotinib were conjugated
with the tetraethyleneglycol linker. The authors showed that
the presence of erlotinib increases the selectivity of con-
jugate accumulation in murine breast cancer cells 4T1
(IC50 (1) = 3.4 μM against IC50 (Cu-porphyrin without er-
lotinib) = 25 μM) with an elevated level of EGFR compared
to the non-tumor human hepatic LO2 line (IC50 (1) = 42.8 μM
against IC50 (Cu-porphyrin without erlotinib) = 31.7 μM)
with a low level of EGFR expression (LED, λ=540 nm).
Furthermore, conjugate 1 could respond to intracellular
glutathione fluctuations. Upon treatment of HepG2 cells
with 1, its fluorescence was restored, which can timely es-
timate the therapeutic efficiency of bimodal chemo- and
photodynamic therapy.
Scheme 2.
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
214 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
The research group under supervision of Prof. A.Yu.
Fedorov is also interested in creation of various multifunc-
tional conjugates with improved pharmacological parame-
ters. Thus, the vandetanib derivative, another inhibitor of
EGFR and VEGFR (vascular endothelial growth factor)
tyrosine kinases, as part of a conjugated system based on
the chlorin e6 zinc complex 2 was used (Scheme 2).[110]
Vanetanib was supposed to play the role of a dual agent of
selective delivery due to preferential binding to VEGFR
and EGFR receptors overexpressed by many types of tu-
mors, as well as cytotoxic action due to inhibition of the
activity of the mentioned tyronis kinases.[115] To increase
water solubility, chlorin derivative was modified by the
introduction of three ammonium groups in the form of
quaternized salts. The conjugate 2 showed a selective ac-
cumulation in A431 cells (EGFR-positive, human epider-
moid carcinoma), which was greater than in non-tumor
CHO cells (EGFR-negative, chinese hamster ovary cells)
and HeLa cells (human epidermoid carcinoma) with a low
level of EGFR expression. Under the action of light (LED,
λ= 615-635 nm), the conjugate 2 inhibited the growth of all
cell lines at low micromolar concentrations, while without
irradiation, the values of the half-inhibition index (IC50)
were approximately an order of magnitude higher. This
indicates that when bound to chlorin-e6, Vandetanib deriva-
tive is unable to inhibit cell growth. It should be noted that
chlorin-e6 without Vandetanib 3 had 2-3 times lower toxici-
ty in all cell lines. In experiments performed on CT-26 tu-
mor-grafted mice, the conjugate 2 also demonstrated selec-
tive accumulation in the tumor.
Then, it was decided to change the type of the linkage
between vandetanib and chlorin-e6 from an ether bond to an
ester bond (Scheme 2). It is known that ester bonds in the
body are easily destroyed by the action of esterase,[116]
therefore, the modified conjugate may be capable of releas-
ing vandetanib, which means that the synergistic effect of
PDT and chemotherapy can be observed. The second
change was to replace the tetramethylammonium groups
with maltose-based carbohydrate moieties. Having hydro-
philic properties, carbohydrates also selectively bind to
their respective receptors (galectins and GLUT transporters)
in tumor cells by the Warbug effect.[117-119] Although there
was almost no selectivity for A431 cells compared to CHO,
the conjugate 4 generally accumulated better in these cells
than the conjugate with ammonium fragments 2, and also
showed less pronounced cytotoxicity in the dark compared
to 2. Under light irradiation (LED, λ = 655−675 nm) in
A431 cells, the conjugate 4 was 2 times more active than in
the CHO cell line and 25 times more active than unconju-
gated chlorin-e6 5. An in vivo study of activity in mice with
grafted A431 tumors showed a moderate selectivity for
tumor-normal cells, as well as slight differences in the ac-
tivity of the conjugate in the light and in the dark. This fact
implies that the conjugate 4 enters the cells and releases
vandetanib by esterase-mediated ester cleavage. Thus, the
obtained results indicate different behavior of vandetanib-
chlorin-e6 conjugates depending on the type of bond be-
tween the key fragments.
Scheme 3.
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 215
Scheme 4.
Metal-containing drugs are broadly known for their
significant antitumor activity.[120] The conjugation of such
metal complexes with PSs can modulate their hydrophilic
properties. Due to the interaction with DNA, metal com-
plexes[121] may serve as a vector to deliver PS to the cell
nuclei. Organotin complexes are an attractive alternative to
the known platinum-containing cytostatics because of low-
ered systemic toxicity.[122] Importantly, such complexes can
be effective in cell lines possessing cisplatin resistance.[123,124]
In 2021, two conjugates of bacteriochlorin derivative and a
tin complex with amino acid ligands (p-aminobenzoic acid
and L-lysine) 5-6 was obtained by Prof. Grin with col-
leagues (Scheme 3).[125]
The dark cytotoxicity of conjugates 5 and 6 in various
cell lines (prostate gland adenocarcinoma (PC-3); breast
adenocarcinoma (MCF-7); lung carcinoma (A549); cervix
adenocarcinoma (Hela) and mouse sarcoma (S-37)) was 3-5
times higher than that of the corresponding organotin com-
plexes. Based on studies in vitro, the authors concluded that
the conjugation of tin-containing cytostatics with bacterio-
chlorin moieties promotes better internalization and accu-
mulation of conjugates in tumor cells and therefore in-
creased cytotoxicity. Among synthesized conjugates, the
molecule 6 demonstrated exceptional photodynamic effi-
ciency (2-3 fold increased IC50 under light) compared to
referenced bacteriochlorin derivative 7 (halogen lamp,
λ≥720 nm). The nature of the intracellular distribution of
the obtained agents showed a diffuse granular distribution
with predominant accumulation in the near nuclear region,
but it was not possible to observe the localization of conju-
gates in the nuclei themselves. Thus, the authors conclude
that the tin complexes act as target fragments for the deliv-
ery of bacteriochlorins into the cells.
Histone deacetylases (HDACs) are directly involved
in carcinogenesis and therefore represent an attractive tar-
gets for anticancer therapy.[90,126] Inhibition of HDACs is
shown to suppress growth of cancer cells, promote cell
death, modulate immune responses, and inhibit formation
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
216 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
of new blood vessels.[127] Mainly approved for hematologi-
cal malignancies, histone deacetylase inhibitor (HDACi)
has been clinically tested for solid tumors and has shown
better results when combined with another chemotherapy
drug.[128] It is also known that, HDAC6 and HDAC8 are
highly expressed in breast cancer cells, therefore their
downregulation may be beneficial.[129] In 2020 Aru with
colleagues reported the 3-hydroxypyridin-2-thione (3-HPT)
substituted silicon phthalocyanine derivative 8 and evaluat-
ed its anti-cancer properties on two different breast cancer
cell lines (non-invasive MCF-7 and invasive MDA-MB-
231 cells) (LED, λ=700 nm). It was shown, that conjugate 8
activated programmed cell death pathways and induced cell
cycle arrest which was accompanied by decreasing levels of
HDAC6 and HDAC8 (Scheme 4).[130]
Among various breast cancers, its triple negative vari-
ant doesn’t respond to classical hormone therapy and al-
ways associated with poor prognosis.[131] In this regard,
promising results obtained after treatment of cell line
MDA-MB-231 (triple negative breast cancer) with 8 can
become a starting point for a novel treatment of such dis-
ease. Thus, the authors demonstrated pronounced inhibition
of MDA-MB-231 cells with the highest late apoptotic cell
population. Inspired by the obtained results, the authors
evaluated how the substitution in α- or β-position of a
phthalocyanine with HDACi inhibitor affects antitumor
behavior of suggested conjugates.[132] Additionally, differ-
ent metals were chosen for complexation with a phthalocy-
anine core. The influence of the central ion on the internal-
ization of conjugates was different for zinc and indium de-
rivatives. While for α-substituted indium conjugates 9b and
10b no differences were found in their cell uptake, β-
substituted Zn-conjugate 10a in demonstrated enhanced
internalization and, therefore, a more pronounced antitumor
effect compared to its α-analogue-conjugate ZnPc 9a (LED
lamp, red light). It was also noted that although all conju-
gates generated ROS in the mitochondrial network, only
indium compounds 9b and 10b directly targeted mitochon-
dria. Treatment of cells with obtained conjugates also led to
cell cycle arrest in different phases of cell cycle.
Camptothecin 11 is a potent antitumor antibiotic
whose action mechanism involves the inhibition of DNA
relaxation by the stabilization of a covalent binary complex
formed between topoisomerase I and DNA.[133] In 2022
Zhang and colleagues proposed the concept of a prodrug 12
based on the camptothecin derivative (Scheme 5).[134] As a
photosensitizing part a BODIPY-based compound was cho-
sen. At the same time camptothecin derivative was attached
to the BODIPY through a labile phenyl benzoate group.
The phenyl benzoate group is prone to cleavage by cell
esterases what was exploited for the release of camptothe-
cin 11. Importantly, camptothecin 12 attached to the BOD-
IPY photosensitizer can’t inhibit tumor proliferation while
its release could lead to pronounced cytotoxicity due to
DNA binding. In addition, the fluorescence of conjugated
camptothecin was dramatically reduced through the intra-
molecular fluorescence resonance energy transfer (FRET)
process from camptothecin to the BODIPY photosensitizer.
Monitoring the fluorescence level, the authors were able to
observe the release of camptothecin 11. Studies of esterase-
mediated release of camptothecin 11 from conjugate 12
proved its liberation under action of exogenous carboxyles-
terase in phosphate-buffered saline and under action of en-
dogenous carboxylesterase overexpressed in cancer cells.
The authors showed a significant decrease in the IC50 val-
ues of the resulting conjugate against HepG2 human hepa-
tocellular carcinoma and HeLa human cervical carcinoma
cells upon irradiation (LED, λ=660 nm) compared with
camptothecin and BODIPY as controls. The combined anti-
tumor effects of the prodrug 12 were also observed in the
mice bearing H22 murine hepatocarcinoma tumors.
Scheme 5.
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 217
Scheme 6.
Combretastatin A-4 13 (CA4) is a strong antimitotic
agent that can bind to the colchicine-binding site and inhibit
tubulin polymerization[135] (Scheme 6). CA-4 in Z-form
exhibits cytotoxicity at the nanomolar level in various hu-
man tumor cell lines, (MCF-7-IC50 = 9 nM) including mul-
ti-drug resistant (MDR) tumor cells.[136] CA4 contributes to
the blockage of tumor vessels, therefore serves as a vascular
disrupting agent. In 2013, Bio with colleagues developed
“photo-unclick chemistry”, a novel chemical tool involving
the cleavage of aminoacrylate linker by singlet oxygen
(1O2). The researchers applied it to visible light-activatable
prodrugs. The proposed delivery system includes a photo-
sensitizer connected by an aminoacrylate linker to an anti-
tumor drug (combretastatin A4) 14[137] (Scheme 6).
The resulting prodrug 14 was activated by far-red light
(diode laser, λ=690 nm) due to the destruction of the ami-
noacrylate linker by singlet oxygen formed upon irradiation
of the dithioporphyrin photosensitizer. Thus, singlet oxygen
acts not only as a cytotoxic agent, but also as a tool for acti-
vating the chemotherapy drug, resulting in synergistic dual
chemo- and photodynamic therapy. In 2020, Bio with col-
leagues improved the above mentioned system by adding a
2,4-dinitrobenzenesulfonate (DNBS) quencher and biotin as
a tumor-targeting ligand to it [138]. Due to the presence of
the DNBS substituting group, zinc phthalocyanine quench-
ing was achieved for 1O2 production and fluorescence emis-
sion. When interacting with glutathione (GSH) in the cell
environment, DNBS group had removed and prodrug had
recovered photoactivity. In cells HepG2 (biotin receptor
positive) conjugate 15a demonstrated elevated uptake than
that for HCT-116 cells (control, human colorectal carcino-
ma), therefore proving its ability to target biotin receptors.
After GSH-OEt pre-treatment the fluorescence intensity
increased. Under conditions of light irradiation (300 W hal-
ogen lamp, λ=610 nm) of HepG2 cells containing the con-
jugate 15a, their growth was inhibited at a nanomolar con-
centration (Hep2G: IC50 light = 48 nM), while in the dark it
was inactive at a concentration of up to 2 μM. At the same
time, the control conjugate without aminoacrylate linker
15b had a much lower activity both with and without irra-
diation, confirming the proposed mechanism of activation
of the conjugate system by singlet oxygen-induced destruc-
tion of the aminoacrylate linker in conjugate 15a. The au-
thors calculated the combination index (CI)[139] for 15a
(CI~0.25), which indicated strong synergism between pho-
tosensitizing and chemotherapeutic parts. Thus, the new
delivery system is favorably distinguished by its dual action
(PDT and chemotherapy) and high controllability.
Our research group decided to take advantage of com-
bretastatin A-4's photoisomerization from a 1000-fold less
active trans-isomer 17 (IC50 = 10-6M) to a clinically active
cis-isomer 13 (IC50 = 10-9M)[140,141] (Scheme 7A). Combin-
ing trans-combretastatin with a porphyrin photosensitizer
using a photoremovable o-nitrobenzyl linker,[142] we as-
sumed that the action of one- or two-photon irradiation
could trigger a cascade of processes: 1) destruction of the o-
nitrobenzyl linker with simultaneous release of trans-
combretastatin; 2) its photoisomerization into toxic cis-
isomer 13; 3) photoactivation of porphyrin and its genera-
tion of singlet oxygen.[143] As a result, we would be able to
control the toxicity of the conjugates 16a-b using light and
enhance the therapeutic effect due to the simultaneous ac-
tion of PDT and chemotherapy. To increase hydrophilicity,
the porphyrin macrocycle was decorated with carbohydrate
fragments (galactose and maltose, respectively). However,
when using various light sources (UV-A, λ=365 nm, UV-B,
λ=311 nm, and UV-C, λ= 254 nm, LED, λ = 450 nm), it
was not possible to observe the desired release of com-
bretastatin A-4. To determine the cause of the intactness of
the conjugates 16a-b, irradiation of the trans-combretastatin-
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
218 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
linker 18 model system without porphyrin was carried out.
It turned out that this system is capable of releasing a thera-
peutic agent, which is immediately not only isomerized to
active cis-combreatstatin, but further cyclized to a phenan-
threne-type derivative 19 (Scheme 7B). TD-DFT calcula-
tions were also performed to identify the best type of bind-
ing between the porphyrin and the o-nitrobenzyl linker. It
has been shown that the excitation of three orbitals (LUMO,
LUMO+1, and LUMO+2) is possible when the conjugates
are irradiated with 365 nm light, in which the maximum
electron density is shifted towards the porphyrin fragment,
which possibly complicates the photocleavage process. At
the same time, if there are no additional functional groups
between the porphyrin fragment and the o-nitrobenzyl link-
er, then on the LUMO + 2 orbital, which at the same time
has an energy higher than for all considered types of bind-
ing, the electron density shifts towards the linker, which
increases the probability of photodegradation. Our result is
consistent with the experimentally photocleaved porphyrin-
o-nitrobenzyl linker-fluorouracil conjugate by Lin,[144] for
which we also calculated the corresponding orbitals. Thus,
further research will focus on an updated design that elimi-
nates linkers between the photosensitizer moiety and the o-
nitrobenzyl group, and the replacement of combretastatin
A-4 with azo analogs that do not undergo cyclization.[145].
Thus, the substantial progress made in the develop-
ment of multifunctional drugs opens broad possibilities for
the creation of efficient antitumor agents. Even though
there are no approved conjugated drugs so far on the mar-
ket, their further investigation will undoubtedly lead to new
treatment options for people suffering from oncological
diseases. Among main beneficials hiding in this concept the
one can point out: 1) broad variability arising from unlim-
ited tools for conjugation including cytostatics, cleavable
linkers, quenchers etc.; 2) predictability of cellular targets
for main active parts of conjugate; 3) possibility to exploit
multiple mechanisms leading to the death of cancer cells,
including MDR-cells; 4) selective delivery to tumors
through decoration with specific oncovectors or targeted
TKI utilization.
Scheme 7.
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 219
3. Formation of Cationic Groups at the Natural
Chlorins Macrocycle Periphery in the Synthesis
of Potential Antitumor and Antibacterial
Photosensitizers
It is known that the introduction of cationic groups to
the periphery of the chlorin PS macrocycle not only in-
creases hydrophilicity, but also promotes interaction with
the cell membrane of gram-negative bacteria.[42,47,92,146-161]
Chlorophyll a derivatives have spectral properties suitable
for use as antitumor and antibacterial PSs and relatively low
toxicity; therefore, they are widely used as a basis for the
synthesis of new PSs for medical purposes.[42,47,92,146-161]
One of the possible directions of chlorophyll a and its de-
rivatives chemical modification is the introduction of cati-
onic substituents to the macrocycle periphery. It should be
noted that a positive charge can arise both in the protona-
tion of nitrogen atoms (as a rule, nitrogen atoms of periph-
eral substituents[47,150,153,162-164]) and in the presence of pH-
independent cationic groups on the macrocycle periph-
ery.[47,146,148153,159,165] The dependence on pH of the charge
presence at the macrocycle periphery can reduce the effec-
tiveness of the PS, so the formation of cationic substituents
with a constant charge is more preferable. This review con-
siders methods for the formation of cationic groups at the
periphery of the macrocycle of chlorophyll a derivatives. It
should be noted that not all cationic chlorins, which will be
discussed below, have been studied as potential PS, howev-
er, the methods used in their synthesis can be applied in the
synthesis of potential PS, and these chlorins themselves are
potential PS. Only cationic derivatives with a quaternary
nitrogen atom have been described for a-series chlorins.
The key step in the formation of a cationic group is the in-
troduction of an amino group substituted to varying degrees
(usually it is tertiary amino group). Quaternization of this
group allows the formation of a charged group and is usual-
ly not difficult. Next, will be considered the main ways of
introducing the previous nitrogen-containing groups and
their subsequent transformation into cationic groups.
The Introduction of Cationic Substituents by Forming an
Ester Bond
To introduce cationic groups by forming an ester bond
the action of an alcohol with a tertiary nitrogen atom on the
activated carboxyl groups of chlorine is used, followed by
quaternization of the nitrogen atom. When chlorin e6 is
treated with 2-(N,N-dimethylamino)ethanol (choline) or 3-
hydroxymethylpyridine, followed by alkylation with methyl
iodide, the corresponding chlorins with three cationic sub-
stituents are obtained. Because when using dicyclohexylcar-
bodiimide (DCC) and 1-ethyl-3-(3-dimethylaminopropyl)car-
bodiimide (EDAC) the products are usually a mixture of
mono-, di-, and tri-substituted chlorin e6, mostly mono- and
di-substituted chlorin e6, probably because the structure of
intermediates is too bulky to esterify all of the three car-
boxyl groups, the carboxyl groups “activation” is carried
out with oxalyl chloride. The corresponding acid chloride is
forms, which makes it possible to carry out the reaction at
all three carboxyl groups of chlorin e6[166] (Scheme 8).
Insertion of Cationic Substituents through the Formation of
an Amide Bond
The formation of an amide bond upon the introduction
of a tertiary amino group to the periphery of the chlorine
macrocycle is carried out both using the usual reactions of
“activated” carboxyl groups and using the reactions of chlo-
rophyll a derivatives associated with the features of their
chemical structure. When, for the introduction of the
N,N(dimethyl)ethylenediamine fragment in reactions with
various 17-carboxy derivatives of chlorophyll a, in most
cases, carbodiimide “activation” (DCC, EDC) is used
(Schemes 9-12). The quaternization of the tertiary nitrogen
atom is carried out by the action of CH3I, if it is necessary,
the exchange of the counterion is carried out on an ion ex-
change resin. So, for the synthesis of the cationic derivative
of pheophorbide a, the DCC-activated carboxyl group of the
substrate is treated with (N,N-dimethyl)ethylenediamine[167]
(Scheme 9).
A similar reaction of pyropheophorbide a[168] (Scheme 10),
pyropheophorbide d[169] (Scheme 11), and phorbin, which is
the active substance of Photochlor PS[170] (Scheme 12), is
carried out with EDC “activation”.
If it is necessary to carry out some additional modifi-
cations of chlorin (for example, the synthesis of a complex
or the reduction of the aldehyde group,[169] Scheme 11),
quatrainization is carried out at the final stage, since it is not
so convenient to work with a compound with a cationic
group due to its increased hydrophilicity and also the possi-
bility of uncontrolled exchange of the counterion and its
participation in side reactions. As will be seen from what
follows, this technique is quite universal and is used regard-
less of the method of introducing a substituent with a ter-
tiary nitrogen atom.
Scheme 8.
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
220 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
Scheme 9.
Scheme 10.
Scheme 11.
Scheme 12.
The interaction of amines with esters to form the cor-
responding amides is a well-known reaction that is possible
for compounds of various classes, including natural chlo-
rins. Amidation of chlorophyll a derivatives ester groups in
most cases, is carried out after the formation chlorin e6 13-
amide derivatives after the exocycle opening (see below).
However, amidation of the phytylpropionate substituent of
pheophytin a by the action of N,N,N,N,N,N-hexapropyl-
penta(aminoethyl)amine (amine oligoamine with five ter-
tiary nitrogen atoms) is possible before exocycle open-
ing[161] (Scheme 13). The reaction is carried out under the
action of a small molar excess of amine in the presence of
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 221
about 10-fold molar excess of TFA. It is known,[161] that the
same amine can open the pheophytin a exocycle with the
formation of the corresponding 13-amide derivative (see
below), but this reaction does not occur under amidation
conditions. The selectivity of amidation can be provided not
only by a small excess of amine, but also by the presence of
TFA. The role of TFA the authors of[161] do not comment
on. Possibly, the presence of a small amount of TPA pro-
vides an "acidic" catalysis of amidation, which, along with
a slight excess of amine, results in the selectivity of the
process. The target compound in this synthesis was the cat-
ionic zinc complex, as in the cases described above, quater-
nization is carried out at the last stage.
A similar reaction of the amide’s formation occurs
upon the action of amines on cyclic lactones formed during
the oxidation of chlorines with a carboxyl group in position
17.[171,172] As a result of the reaction, the lactone exocycle
opens and the corresponding hydroxychlorines are forms.
This reaction can be used to insert the N-methylpyridyl
group[173] (Scheme 14).
It is known that the exocycle of pheophytin a, methyl
pheophorbide a, pheophorbide a and a number of other
similar natural chlorins is relatively easily opened under the
action of nucleophilic agents such as amines. This reaction
can be used to form cationic groups if the opening is per-
formed with an amine containing tertiary nitrogen atoms or
additional amino groups that can be alkylated. The exocycle
opening under the action of (N,N-dimethyl)ethylenediamine
allows the introduction of a tertiary amino group, the
quaternization of which gives a cationic substituent
(Scheme 15). This pathway was implemented for pheophyt-
in a, methylpheophorbide a and a number of its derivatives
with fragments of biomolecules to increase hydrophilici-
ty.[110,118,174-178] Alkylation of the dimethylamino group is
usually carried out with CH3I.[110,174-178] However, other
alkylating agents, such as C2H5Br, are also possible[118]
(Scheme 15).
Scheme 13.
Scheme 14.
Scheme 15.
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
222 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
Scheme 16.
Scheme 17.
Scheme 18.
The use of (N,N-dimethyl)ethylenediamine analogs
with a large number of methylene units between the prima-
ry and tertiary amino groups makes it possible to synthesize
chlorin e6 derivatives with a cationic group remote from the
macrocycle to varying degrees. For example, the reaction
with 1-(N,N-dimethylamino)-3-aminopropane[179] makes it
possible to lengthen the spacer by one methylene group
(Scheme 16).
The exocycle opening with 4-aminomethylpyridine can
be used to introduce the pyridinium fragment[180] (Scheme 16),
but the authors did not perform further alkylation. The cati-
onic group formation is also possible from a primary amino
group. For example, chlorin with a cationic group is synthe-
sized by the action of a large excess of CH3I on the 13-
amide derivative of chlorin e6, obtained by the action of
ethylenediamine on methyl pheophoride a[181] (Scheme 17).
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 223
Scheme 19.
Scheme 20.
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
224 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
The interaction of purpurin 18 and its analogs with
amines giving cycloimide derivatives not only significantly
improves the spectral characteristics of chlorin, but also
introduces an additional functional group.[182] This reaction
can be used both for the directly insertion of a substituent
with a tertiary nitrogen fragment[173] and for the formation
of a reaction center, which can then be used to synthesize a
derivative with a tertiary nitrogen fragment. The purpurin
methyl ester 18 cycloimide derivative is obtained from pur-
purin 18 by the action of 4-aminomethylpyridine in three
stages performed in one flask”, subsequent alkylation of
the pyridine substituent with CH3I gives the target cationic
derivative[173] (Scheme 18). Starting from purpurin methyl
ester 18, a similar derivative can be obtained in one step
by refluxing in toluene with 4-aminomethylpyridine[180]
(Scheme 18). In addition to the direct introduction of a sub-
stituent with a tertiary nitrogen atom, the formation of cy-
cloimide can be used to introduce an additional reaction
center. The action of hydrazine on bacteriopurpurin methyl
ester 18 gives a cycloimide derivative with a reactive amino
group, acylation of which with pyridinecarboxylic acids
leads to derivatives with a pyridinium fragment[183-185]
(Scheme 19). Quaternization of pyridinium fragments by
the action of CH3I gives the corresponding cationic de-
rivatives.
Chlorins with a seven-membered exocycle closed by
an imide bond can be synthesized by the action of CMPI on
13-amd-15-carboxy derivatives of chlorin e6, provided that
the amide group at position 13 is secondary or primary.[186]
In the case when there is a dimethylaminomethyl group at
the amide nitrogen atom, cyclization results in the for-
mation of chlorin, the action of which with CH3I gives the
corresponding cationic derivative[186] (Scheme 20).
Formation of C-N(amine) Bonds in the Synthesis of Cationic
Chlorins
The formation of C-N(amine) in a number of cases is used
to introduce substituents with a tertiary nitrogen atom, the
subsequent quaternization of which gives a cationic substit-
uent. Epoxidation of the vinyl group of chlorin with meta-
chloroperoxybenzoic acid followed by treatment of the in
situ formed polyamine epoxide gives chlorin with a poly-
amine fragment in position 3, a precursor of the pentacation
substituent[161] (Scheme 21). Quaternization of tertiary ni-
trogen atoms in the molecule by the action of CH3I is car-
ried out at the last stage.
Cationic derivatives with a pyridinium group were
synthesized by the action of heterocyclic compounds with a
pyridinium fragment, I2 and AgPF6, on vinylchlorins; the
cationic group is immediately formed in the course of
chemical oxidative pyridylation[152,187] (Scheme 22). Using
the reaction involving pyridine as an example, it was
shown[187] that more accessible silver salts, for example,
halides, can be used instead of AgPF6.
A similar reaction can also be carried out in a series of
bactriochlorophyll a derivatives after the formation of a
vinyl group in position 3[187] (Scheme 23).
In the absence of a vinyl group, the insertion of a cati-
onic substituent during chemical oxidative pyridylation
occurs in meso-positions[188] (Scheme 24). As a result, a
mixture of regioisomers with comparable isolated yields is
formed, which significantly complicates the use of this re-
action for the synthesis of cationic PSs.
A similar reaction can also be carried out in a series of
bactriochlorophyll a derivatives after the formation of a
vinyl group in position 3[187] (Scheme 23).
Scheme 21.
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 225
Scheme 22.
Scheme 23.
Scheme 24.
Scheme 25.
Scheme 26.
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
226 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
In the absence of a vinyl group, the insertion of a cati-
onic substituent during chemical oxidative pyridylation
occurs in meso-positions[188] (Scheme 24). As a result, a
mixture of regioisomers with comparable isolated yields is
formed, which significantly complicates the use of this re-
action for the synthesis of cationic PSs.
C-C and C=C Bonds Formation in the Synthesis
of Cationic Chlorins
The formation of a carbon-carbon bond is considered
to be the most preferred when introducing a substituent
with a tertiary nitrogen atom. In the chemical modification
of chlorins in this direction, both some "classical" reactions
(variants of the Mannich reaction, other condensations) and
relatively recently discovered cross-coupling reactions are
used. Under the action of Eschenmoser's reagent on zinc
porphyrinates obtained from methyl pheophorbide a, me-
thyl pyropheophorbide a, and chlorin e6 trimethyl ester,
stereoselective aminomethylation of the vinyl group oc-
curs[189-193] (Scheme 25, methyl pyropheophorbide a as an
example). As a result of the reaction, trans-isomers are
formed, subsequent demetallation with trifluoroacetic acid
and alkylation of the dimethylaminomethyl group of the
obtained derivatives with CH3I gives the corresponding
metal-free monocationic chlorins.
It was shown[194] that, in contrast to Eschenmoser's re-
agent, the action of bis(N,N-dimethylamino)methane on
metal-free 13-amide derivatives of chlorin e6 by refluxing
in acetic acid results in the nonstereoselective insertion of
two dimethylaminomethyl substituents into the vinyl group
(Scheme 26).
A similar reaction occurs at room temperature with the
participation of zinc[195] and nickel[196]) complexes of a-
series vinyl-chlorins.
The vinyl group aminomethylation reaction[194-196] is
used to synthesize dicationic chlorins containing additional
hydrophilic and hydrophobic fragments[197] (Scheme 27), as
well as dicationic derivatives of chlorin e6 with membrano-
tropic substituents (myristic acid fragments[198] (Scheme 28)
and phytol[176,177] (Scheme 29). In addition to aminomethyl-
ation, the introduction of a cationic group can be carried out
using palladium-catalyzed cross-coupling reaction. This
reaction was used to insert a pyridine moiety at position 20
of mesomethylpyropheophorbide a, which was then quaternized
by the action of iodo triethylene oxide[199-201] (Scheme 30).
Condensation of the methylpyropheophorbide d alde-
hyde group with methyl groups of the corresponding heter-
ocycles is used to introduce a heterocyclic fragment with a
pyridyl nitrogen atom. The heterocyclic fragment of the
substituted vinyl group formed as a result of condensation
is quaternized by the action of CH3I[202] (Scheme 31).
Scheme 27.
Scheme 28.
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 227
Scheme 29.
Scheme 30.
Scheme 31.
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
228 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
Combination of Reaction Centers of Various Chlorophyll a
Derivatives in the Synthesis of Cationic Chlorins
It often becomes necessary to synthesize chlorins with
different natures, amounts, and positions of cationic sub-
stituents to reveal structure-activity relationships. Combina-
tions of modifications using reactions of various reaction
centers allow not only to increase the number of introduced
cationic groups, but also to synthesize chlorins with differ-
ent arrangement of cationic groups on the periphery of the
macrocycle, which is widely used in the synthesis of cation-
ic PSs based on chlorophyll a derivatives. The chlorin e6
diamide derivative with ten cationic groups was synthe-
sized by combining the amidation of the pheophytin a
phytylpropionate substituent with the action of
N,N,N,N,N,N-hexapropyl-penta(aminoethyl)amine leads to
the opening of the exocycle of the 17-amide derivative con-
taining a fragment of the same amine[161] (Scheme 32). As
in all previous cases, the cationic groups are formed by the
action of CH3I in the last step.
Opening of the cyclic lactone in combination with the
formation of cycloimide makes it possible to synthesize the
purpurin 18 cycloimide derivative with two cationic groups[173]
(Scheme 33).
Scheme 32.
Scheme 33.
Scheme 34.
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 229
Scheme 35.
Reaction conditions: i: CH3NH2-H2O/THF, r.t., 3 h; ii: [(CH3)2N=СH2]+I-, CH2Cl2, r.t., 12 h; iii: H2NCH2CH2N(CH3)2, CH2Cl2, r.t.; iv:
CH2(N(CH3)2)2, AcOH-THF, boiling for 30 min; v:[92] CH3I, CH2Cl2, r.t.
Scheme 36.
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
230 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
Scheme 37.
The combination of exocycle opening with ami-
nomethylation of the vinyl group by the action of bis(N,N-
dimethylamino)methane makes it possible to synthesize
tricationic chlorin with a membranotropic phytol frag-
ment[176] (Scheme 34). The same reactions were used for the
synthesis of tricationic chlorin with 4-arylaminoquinazoline
moiety[110] (Scheme 35).
The combination of different variants of the vinyl group
aminomethylation with the exocycle opening makes it pos-
sible to synthesize chlorins with a different number of cati-
onic groups and their arrangement in the macrocycle[178]
(Scheme 36).
Alkylation of amino groups in chlorin e6 di- and triamide
derivatives allows the formation of two and three cationic
groups, respectively[181] (Scheme 37).
Thus, chlorophyll a and its derivatives, having a num-
ber of convenient reaction centers in the molecule, are a
good basis for the synthesis of cationic photosensitizers for
medical purposes.
4. Water-Soluble Cationic Porphyrins
for Photoinactivation of Pathogens
The ability of porphyrins and related compounds to
generate reactive oxygen species under the action of light
necessitates the synthesis of new photosensitizers with the
required properties (selectivity, virucidality, bactericidality,
etc.) for their effective use in photodynamic therapy, clini-
cal laboratory diagnostics, pharmacopoeia,[203] antimicrobi-
al water and air purification,[204] photodynamic inactivation
of bacteria and viruses dangerous to humans and plants.[205]
Photodynamic inactivation of bacteria and viruses is
considered the most promising treatment for drug-resistant
bacteria[206] and zoonotic viruses.[207] The method of pho-
toinactivation is based on the reactions of biomolecules,
biosubstrates, and pathogens with ROS, which are generat-
ed with the participation of PS. It should be noted that, in
addition to tetrapyrrole macroheterocyclic compounds, oth-
er classes of compounds are also considered as PS for pho-
toinactivation of pathogens.[208] For example, the well-
known cationic methylene blue or anionic rose bengal also
show an antibacterial photoelectric effect, but their effec-
tiveness is much lower than that of porphyrin photosensi-
tizers. To date, it is reliably known that cationic tetrapyrrole
MHCs more effectively damage pathogens than anionic and
neutral PSs. The positive charges of PS promote the elec-
trostatic binding of porphyrin to negatively charged sites on
the outer membrane of bacteria and induce damage that
enhances the penetration of the photosensitizer into the
cell.[209,210]
The structure of the outer shell of the pathogen is no
less significant. In general, gram-positive bacteria show a
higher susceptibility to photoinactivation than gram-
negative ones. This is probably the result of differences in
the structure of the cell membranes of these two microbial
groups. gram-positive and gram-negative bacteria have a
cytoplasmic membrane. The membrane of gram-positive
bacteria is more permeable; peptidoglycan structure is po-
rous, filled with teichoic and teichuronic acids. The main
structural difference between gram-positive and gram-
negative bacteria is the thickness of the peptidoglycan and
the presence of an outer membrane. The outer membrane is
present only in gram-negative species. It is a very effective
barrier to the penetration of exogenous substances. This
membrane is very dense, the porins present in it ensure the
passage of only small hydrophilic substances.[211,212] In ad-
dition, due to the presence of lipopolysaccharides, the outer
membrane has a strong negative charge and a high surface
potential.[204]
In order to damage a bacterium, PS must reach the cy-
toplasmic membrane after passing through the outer mem-
brane.[213] Differences in the structure of the cell membrane
of gram-negative and gram-positive bacteria determine the
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 231
possibility of photoinactivation of the latter with the help of
not only cationic, but also anionic and neutral PS, while
gram-negative bacteria are more resistant to photoinactivation.
Photoinactivation of viruses by MHC is a less ex-
plored topic compared to photoinactivation of bacteria, but
as with bacteria, singlet oxygen generated (type II photoox-
idation) and radicals (type I photooxidation)[214, 215] play a
key role in virus inactivation. Potential targets for photo-
sensitizer binding are the protein capsid, nucleic acids, and
virion envelopes.
It is a priori clear that the intracellular localiza-
tion/binding site of a cationic PS is highly dependent on the
structure and intramolecular charge distribution of the por-
phyrin compound and is an important factor in photody-
namic antimicrobial chemotherapy. Therefore, the purpose
of this work was to summarize the literature data and our
own data obtained recently and reflecting the interaction of
cationic porphyrins with biosubstrates in the context of
photoinactivation of the corresponding pathogens.
Very interesting data were obtained in a study[ 217] that
evaluated the efficiency of photoinactivation of bacterio-
phage T4 using tetra-, tri-, and dicationic porphyrins. The
efficiency of photoinactivation of this virus depends on the
charge of the MHC: the greater the charge, the greater the
virucidality of the MHC. The authors attribute the revealed
pattern to the fact that the T4 phage capsid in aqueous solu-
tions with a pH of about 7 is negatively charged; therefore,
tetracationic porphyrins interact electrostatically with the
surface of the virus, and partially oxidize it upon photoirra-
diation. Tricationic porphyrins are also able to interact with
the bacteriophage capsid, in contrast to dicationic MHCs.
The significance of the positive charge of MHC for pho-
toinactivation can also be traced on the example of trisubsti-
tuted porphyrins: 5-(4-methoxycarbonylphenyl)-10,15,20-
tris(N-methylpyridinium-4-yl)porphyrin tri-iodide and 5-(4-
carbonylphenyl)-10,15,20-tris(N-methylpyridinium-4-yl)por-
phyrin tri-iodide. These porphyrins differ only in the nature
of the substituent in one of the phenyl rings. The carboxyl
group of 5-(4-carbonylphenyl)-10,15,20-tris(N-methylpyri-
dinium-4-yl)porphyrin tri-iodide dissociates -CO2H -CO2-+
+ H+[218] in an aqueous medium, unlike -CO2CH3 group,
which is part of 5-(4-methoxycarbonylphenyl)-10,15,20-
tris(N-methylpyridinium-4-yl)porphyrin tri-iodide and is
incapable of dissociation. As a result, the total formal
charge of a porphyrin with a carboxyl group and virucidal
activity are lower than those of a porphyrin with a -CO2CH3
group.
Results for 5-(4-methoxycarbonylphenyl)-10,15,20-
tris(N-methylpyridinium-4-yl)porphyrin triiodide and 5-
(pentafluorophenyl)-10,15,20-tris(N-methylpyridinium-
4-yl)porphyrin triiodide demonstrate that the presence of a
lipophilic aryl group in one of the meso positions of the
tetrapyrrole macrocycle contributes to phage inactiva-
tion.[219] This conclusion is consistent with the results of the
study of photoinactivation of hepatitis A virus and bacteri-
ophage MS2 with PS differing in the length of the side al-
kyl chain (tetrakis(N-[n-butyl]-4-pyridiniumyl) porphyrin,
tetrakis(N-[n-hexadecyl]-4-pyridinium tetrayl)porphyrin,
tetrakis(N-methyl-4-pyridiniumyl)porphyrin, tetrakis(N-[n-
octyl]-4-pyridiniumyl) porphyrin).[203] At the same concen-
tration of PS and identical conditions for complete photoin-
activation of the hepatitis A virus using tetrakis(N-[n-
butyl]-4-pyridiniumyl) porphyrin, irradiation was required
for 30 min, while when using tetrakis(N-[n-octyl]-4-
pyridiniumyl)porphyrin took 1 min to achieve the same
effect.[203]
The introduction of lipophilic fragments has a positive
effect on the bactericidal activity of PS. Photodynamic ef-
fect of meso-substituted cationic porphyrins, 5-[4-
(trimethylammonium)phenyl]-10,15,20-tris(2,4,6-trimethoxyp-
henyl)porphyrin iodide, 5,10-di(4-methylphenyl)- 15,20-
di(4-trimethylammoniumphenyl)porphyrin iodide and 5-(4-
trifluorophenyl)-10,15,20-tris(4-trimethylammoniumphenyl)-
porphyrin iodide have been studied in vitro in gram-
negative Escherichia coli bacteria.[220] It turned out that
after the washing step, only 5-(4-trifluorophenyl)-10,15,20-
tris(4-trimethylammoniumphenyl)porphyrin iodide had bacte-
ricidal activity.
The authors,[221] while studying the antibacterial prop-
erties of meso-substituted tetracationic pyridylporphyrins
quaternized with hydroxyalkyl and haloalkyl residues
against gram-negative and gram-positive microorganisms,
hypothesized that the main factor of photoinactivation is the
presence of positively charged groups in the PS molecule,
which have electrostatic interaction with negative groups on
the surface of bacterial cell membrane. The final binding of
PS to cells is regulated by the lipophilicity of functional
groups, which contributes to the strong association of PS
with the cytoplasmic membrane.
Figure 5. The structure of cell membrane.[216]
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
232 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
When designing a PS, it is also necessary to take into
account the fact that the introduction of a lipophilic substit-
uent can lead to a shift in aggregation equilibria in porphy-
rin solutions towards self-association, which will adversely
affect antibacterial activity. It is this phenomenon that was
discovered by the authors[222] when studying the photophys-
ical properties and antibacterial activity of a number of deriva-
tives of 5,10,15,20-tetrakis-(4-N-methylpyridyl)porphine, in
which one N-methyl group is replaced by a hydrocarbon
chain in the range from C6 to C22. It turned out that the
antibacterial photoactivity of porphyrins increased with an
increase in the length of the N-alkyl chain to C14, which
led to complete inhibition of the growth of E. coli even at
doses of 0.8 μM. A further increase in the size of the sub-
stituent to C18 and C22 was accompanied by a decrease in
the photosensitizing efficiency.
It is known that an asymmetric charge distribution in
the peripheral position of a porphyrin leads to an increase in
the amphiphilic nature of MHC, which can contribute to a
better accumulation in cells during PDT,[223] as well as inac-
tivation of bacteria. For example, 5-(4-trifluorophenyl)-
10,15,20-tris(4-trimethylammoniumphenyl)porphyrin io-
dide,[224] which has a lipophilic trifluoromethyl group in its
composition, exhibits the ability to bind with high affinity
to E. coli (gram-negative) and provides effective photoinac-
tivation. At the same time, 5,10-di(4-methylphenyl)-15,20-
di(4-trimethylammoniumphenyl)porphyrin iodide is able to
cause photodamage of E. coli in a lesser extent, and after
washing out the PS, it does not inhibit the growth of bacte-
ria at all. This fact suggests that the dicationic porphyrin
does not enter the cell.
The inactivation of gram-positive and gram-negative
bacteria with 5,10,15-tris(1-methylpyridinium-4-yl)-20-
(pentafluorophenyl)porphyrin tri-iodide is reported in the
work,[225] which also emphasizes the importance of the lip-
ophilic substituent in the PS molecule for photoinactivation
of gram-negative bacteria.
Quite interesting data were obtained by the authors[209]
in the study of photoinactivation of gram-negative bacteria
Vibrio anguillarum and E. coli by porphyrins. Tetra(4-
sulfonatophenyl)porphine, tetra(4N-methyl-pyridyl)porphine
tetraiodide and the tetra(4-N,N,N,-trimethyl-anilinium)por-
phine were evaluated as PS. All three porphyrins show a
similar pattern of subcellular distribution, localizing mainly
in protoplasts. However, only cationic porphyrins have pho-
toinactivating ability.
It is known that the mechanism of photooxidation with
the participation of PS can proceed in two types: type I,
with the participation of radical forms, and type II, with the
participation of singlet oxygen. As the data show,[226] pho-
toinactivation of gram-negative bacteria of the genus Sal-
monella is carried out to a greater extent due to the genera-
tion of superoxide anion and hydroxyl radical upon irradia-
tion of tetracationic porphyrin containing [Ru(bpy)2Cl]+ -
peripheral groups. The results obtained are credible, since
the formation of radical forms was confirmed by the EPR
method.
As noted above, the cell membrane of gram-negative
bacteria is characterized by low permeability for large mol-
ecules. Nevertheless, the authors[227] succeeded in obtaining
an effective PS for the gram-negative bacterium E. coli
based on a conjugate of tricationic porphyrin with cy-
clodextrin, which, judging by the information presented by
the authors, penetrates into the bacterial cell. The work[228]
emphasizing the significance of the influence of the charged
group position and the bridging heteroatom in porphyrin
conjugates with cyclodextrin on the efficiency of E. coli
photoinactivation is quite indicative. The authors of this
publication found that the replacement of thiopyridine by
methoxypyridine units in the conjugates leads to a signifi-
cant decrease in the antibacterial effect.
Figure 6. Atomic force microscopy for a mycobacterium in the initial state (a), after light exposure (b), after treatment with tetracationic
ZnTMPyP (c), after treatment with tetracationic ZnTMPyP and light exposure (d).
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 233
Figure 7. Effect of photosensitization by the exogenous porphyrins
on the ultrastructure of C. albicans cells. TEM images of C. albicans
cells treated with 3.5 μM TMPyP illuminated with 12 J·cm-2
(TMPyP+Light). C. albicans cells illuminated with 12 J·cm-2 without
an exogenous porphyrin (Light Control) served as control.[234]
Establishing the localization and accumulation sites of
PS in virions and most bacterial cells is a complex and un-
resolved issue in most cases. For bacterial cells, it is ex-
tremely difficult to analyze individual structural elements of
the cell, for example, by flow cytometry methods, where
the bacterial cells themselves are often at the limit of the
resolution of the method[216]. Obtaining information about
the role or behavior of individual cell structures in the pro-
cess of photoinactivation of bacteria requires the use of
complex methods. In some cases, it is possible to establish
the localization of a PS by the presence of its own fluores-
cence[229]. Recently, atomic force microscopy[230-232] or
transmission electron microscopy[323] has been used to
prove the localization of PS in the bacterial cell wall. Figure
6 shows, as an example, atomic force microscopy data for a
mycobacterium in the initial state (a), after light exposure
(b), after treatment with tetracationic zinc(II) 5,10,15,20-
tetra-[4-N-methylpyridyl]porphine (ZnTMPyP) (c), after
treatment with tetracationic ZnTMPyP and light exposure
(d), as well as TEM results for C. albicans with 5,10,15,20-
tetra-[4-N-methylpyridyl]porphine (TMPyP) porphyrin.[230]
TEM makes it possible to detect ultrastructural chang-
es in the cell, so in the presented photo (Figure 7), after
treatment with PS and light, disorganization of the cyto-
plasm is visible, there is a lack of a nucleus and the appear-
ance of white spots, a massive accumulation of wall-like
material in the cytoplasm and damage of cells with a rup-
ture of the membrane, which reflects the leakage of intra-
cellular components.[234]
However, few such data have been obtained so far,
which does not allow us to analyze the dependence of the
PS structure and its predominant localization in the patho-
gen. Nevertheless, the conducted mini-review of the litera-
ture data allows us to conclude that for the effective interac-
tion of meso-substituted porphyrins with pathogens and
their subsequent inactivation, three cationic groups in the
photosensitizer composition are sufficient, and the fourth
peripheral substituent must have a lipophilic character and
can provide the required selectivity. Based on this conclu-
sion, we attempted to design a cationic porphyrin capable to
binding with the SARS-CoV-2 spike protein in the recep-
tor-binding domain (RBD).[235,236] The ultimate goal was to
obtain a porphyrin capable of causing photodestruction of
the RBD S-protein, which would make it impossible for the
SARS-CoV-2 virion to bind to the surface enzymes of hu-
man cells. As a result of theoretical studies,[235-239] the some
porphyrins were designed, which, according to the results
of molecular docking with the S-protein, are localized be-
tween the RBDs of three S-protein polypeptide chains. Us-
ing the C-H-activation method,[240] a targeted synthesis of
porphyrins was carried out: 5-[4′-(1″,3″-benzothiazol-2″-
yl)phenyl]-10,15,20-tris(N-methylpyridinium-3′-yl)porphyrin
triiodide (S-Por), 5- [4′-(1″,3″-benzoxazol-2″-yl)phenyl]-
10,15,20-tris(N-methylpyridinium-3′-yl)porphyrin triiodide
(O-Por), 5-[4-(N-Methyl-1″,3″-benzimidazol-2″-yl)phenyl]-
10,15,20-tris(N-methylpyridinium-3′-yl)-porphyrin triio-
dide (N-Por). For the compounds obtained, the quantum
yields of singlet oxygen were determined, which amounted
to 0.61-0.86.[235,241] In monohetaryl substituted porphyrins,
the replacement of a heteroatom has almost no effect on
both the quantum yield of fluorescence (0.170.21) and sin-
glet oxygen, and the lifetime (8.39.8 ms) in the triplet state.
To confirm the results of molecular docking, a spectral
study of S-protein binding to the synthesized monoheteryl
substituted porphyrins was performed. Quantitative charac-
teristics reflecting the affinity[242,243] of the S-protein to he-
taryl porphyrins were obtained by spectrophotometric titra-
tion of the S-protein with asymmetrically monosubstituted
hetaryl porphyrins (Table 1).
Table 1. The affinity parameters of S-protein with hetaryl porphy-
rins according to fluorescent titration data.
System
Affinity
The number of
binding sites
S-protein·N-por
5.94·105
1.18·105
3.6
4.7
S-protein·O-por
1.2·106
2.4
S-protein·S-por
7.96·105
2.6
S-protein·ACE2·N-por
1.56·105
1.25·105
2.2
2.4
Figure 8. Structure of the ACE2-RBD (6vw1) complex with N-
por according to X-Ray data. Yellow ACE2, blue RBD. Orange
indicates the RBD region responsible for binding to the N-por.[244]
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
234 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
The results of competitive titration[243] of the S-
protein·ACE2 complex with N-por (Figure 8) clearly demon-
strated that N-por leads to the destruction of the S-protein
complex with ACE2. The competitive influence of N-por and
ACE2 for binding to the S-protein confirms that the binding of
porphyrin occurs precisely in the RBD site of the S-protein.
Determination and evaluation of the bactericidal activ-
ity of porphyrins was carried out on the surface of a dense
nutrient medium (Columbian Agar with sheep's blood) con-
taminated with museum strains of microorganisms -
S.aureus ATCC 29213 and S.epidermidis ATCC 14990. It
was found that the synthesized compounds upon photoacti-
vation exhibit antibacterial activity against staphylococci
(Figure 9, Table 2). Thus, the conducted microbiological
study showed that with photoirradiation for 15', monoheter-
yl substituted porphyrins containing benzothiazole and N-
methylbezimidazole residues provide complete lysis of
S.aureus and S.epidermidis strains. In the case of a hetaryl
substituted porphyrin with a benzooxazole residue, com-
plete lysis was observed in the S.aureus strain and incom-
plete lysis was observed in the S.epidermidis test strain.
Increasing the exposure time to 30' ensured complete lysis
in the S.epidermidis strain.
This review of literature data showed that tri- and tet-
ra-cationic porphyrins are effective photosensitizers; reac-
tive oxygen species generated by them initiate oxidative
destructive reactions in biomolecules and biostructures of
pathogens. Variation of the lipophilic nature of the PS
makes it possible to adjust the bactericidal and virucidal
activity of MHC. To increase the efficiency of photoinacti-
vation of pathogens, further studies are needed to study the
influence of the nature of photosensitizers, their interaction
with cells and subcellular structures, biomolecules, and the
assessment of photoinduced damage.
Por-
phyrin
Exposure time, min
Control in dark
15'
30'
60'
S-por
О-por
N-por
Figure 9. The results of the bactericidal effect of porphyrins in the dark and upon activation by light on the S. aureus ATCC 29213 test
culture.
Table 2. Evaluation of lytic activity on a five-point scale (“-” no lytic activity; “+” low activity; “++” formation of a lysis zone with a
large number of colonies of secondary growth of the bacterium; “+++” lysis zone with single colonies of secondary growth ; "++++"
transparent zone of lysis without secondary growth colonies).
Porphyrin
S-por
O-por
N-por
Exposure time
15'
30'
60'
15'
30'
60'
15'
30'
60'
S.aureus
++++
++++
++++
+++
++++
++++
++++
++++
++++
S.epidermidis
++++
++++
++++
++++
++++
++++
++++
++++
++++
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 235
5. Approaches toward Cationic Photosensitizers
Based on Phthalocyanines
One of the most attractive photosensitizers in PDT are
phthalocyanines (Pcs) and their metal complexes.[10,98, 245248]
The singlet oxygen mediated process is predominant for
Pcs that possess strong absorbance in the visible region
between 600 nm and 700 nm (referred to as the Q-bands)
resulting from the HOMO LUMO (π→π*)
excitations.[249,250] This part of spectrum favours the optimal
penetration of the light through a tissue. Moreover, Pcs,
compared with porphyrins, have low absorbance at wave-
lengths between 400 nm and 500 nm, where daylight inten-
sity is highest. The presence of a heavy atom in the phthal-
ocyanine leads to the enhancement of singlet oxygen gener-
ation due to an increase in spin-orbit coupling owing to so
called “heavy atom effect”, and consequently, the transition
of the photosensitizer from the excited singlet state to the
excited triplet state. Outstanding photophysical properties
of phthalocyanines are tailorable through variation of the
central cation, axial ligands, nature of substituents and their
position in the Pc aromatic ring.[98,251253] Introduction of
cationic substituents into phthalocyanine’s rings may pro-
vide better solubility in water as well as decrease their ten-
dency to aggregation [254,255]. In addition, many cationic Pcs
show a higher photodynamic efficiency than anionic or
uncharged hydrophilic Pcs [255] due to better binding to
DNA molecules.
Cationic function in Pc can be formed via different
ways, such as: quaternization of the nitrogen atom in ali-
phatic and aromatic substituents,[256258] quaternization of pyri-
do(pyrazo)porphyrazines [255] and exocyclic metalation,[259]
introducing of pnictogenes(V) in the cavity of phthalocya-
nine ring,[260264] or via meso-N-protonation of phthalocya-
nines bearing alkoxy-groups in non-peripheral positions[265]
(Figure 10).
The synthesis and photophysical properties of some
cationic Pcs bearing quaternized nitrogen-containing groups
have been summarized in two comprehensive reviews.[266,267]
This chapter of the review is an attempt to illustrate novel
examples of Pcs with quaternized nitrogen-containing
groups and other pathways to others types of cationic Pcs.
N-Centered Cationic Charges
Intriguing examples of cationic phthalocyanines with
four and eight extremely bulky 2,6-di(N-methylpyridine-3-
yl)phenoxy substituents were reported by P. Zimcik group
(Figure 11).[257] These Pcs exist as monomers in water and
phosphate buffered saline (PBS) and exhibit moderate fluo-
rescence quantum yields (0.17 and 0.16) both in DMF and
PBS that may be used for bioimaging. Also, high values of
phototherapeutic indices were obtained. The ratio of dark
toxicity to light toxicity (TC50/EC50) for HeLa cell line was
found to be 1409 and 5691 for tetra- and octa-substituted
complexes respectively.
Introduction of imidazolium units into Pcs’s macro-
heterocycles was accomplished by Lioret et al. through SN2
reaction of 4,5-dichlorophthalonitrile and imidazole in the
presence of K2CO3 at 50 °C yielding 75 % of the diimidaz-
olyl derivative. The further template cyclotetramerization of
the resulting phthalonitrile with Zn(OAc)2 lead to octa-
imidazolyl Pc derivative (Figure 12).[258] After quaterni-
zation with methyliodide, the target zinc octa(N-
methylimidazolium)phthalocyaninate was obtained in al-
most quantitively yield. For latter complex photocytotoxici-
ty studies were carried out on murine melanoma B16F10
cancer cells and IC50 = 5.379 mM was determined.
Figure 10. Approaches toward cationic phthalocyanines and tet-
ra(pyrido/pyrazino)porphyrazines.
Figure 11. Synthesis of quaternized zinc complexes with tetra-[257] and octa-substituted[268] [2,6-di(3-pyridyl)phenoxy]phthalocyanines.
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
236 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
Figure 12. Synthesis of zinc octa(N-methylimidazolium) phthalocyaninate.[258]
Figure 13. Robust route toward cationic phthalocyanines through reductive amination.[256]
Recently Bunin et al. developed robust route toward
cationic phthalocyanines through reductive amination.[256]
In the synthesis of cationic Pcs reductive amination was
used for the first time. Thus, reaction of 3- and 4-
(formylphenoxy)phthalonitriles with diethylamine and so-
dium tris-acetoxyborohydride in THF resulted in corre-
sponding diethylamino-derivatives of phthalonitriles isolat-
ed in high yields (Figure 13). New Zn(II), Mg(II) and metal-
free phthalocyanines containing four or eight cationic
groups in non-peripheral (-) or peripheral (-) positions
were synthesized starting from the aminated phthalonitrile
derivatives followed by quaternization with CH3I. All novel
cationic Pcs exhibited strong absorption in the photothera-
peutical range from 670 to 700 nm and high quantum yields
of singlet oxygen generation in DMSO where all phthalo-
cyanines existed in monomeric forms. Also, cationic com-
plexes showed moderate solubility in water with high ten-
dency to aggregation, however -substituted Zn(II) and
Mg(II) complexes existed in water in predominantly mon-
omeric states. Further monomerization of tetra--
substituted Zn(II) complex in aqueous solution was
achieved by interaction either with non-ionic surfactant
Tween-80 or bovine serum albumin (BSA). The advantage
of these results was not limited to new convenient synthetic
approach to the cationic phthalocyanines but it also consist-
ed in preparation of new promising phototherapeutic agents
with good photophysical and photochemical properties.
Cationic Charges Centered on Other Atoms
Schneider et al. report the first example of exocyclic
metalation of tetra(pyrido)porphyrazine (Figure 14).[259] In this
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 237
intriguing work, zinc complex with tetra(pyrido)porphyrazine
(as a mixture of regioisomers) was coupled with transplatine
fragments via reaction with fivefold excess of tetrakis-
[trans-Pt(NH3)2Cl]NO3, derived in situ from transplatine
(tetrakis-(trans-Pt(NH3)2Cl2)) and AgNO3 in DMF. Mass-
spectrometry in combination with 195Pt-NMR spectrum for
platinated compound indicates the formation of the tet-
raplatinated derivative as a sole product. This exo-
platinated Pc demonstrate red shift of Q-band in UV-Vis
spectra up to 682-692 nm in comparison with 663-674 nm
for its non-platinated precursor. Interesting, platinated com-
plex of demonstrated low absolute quantum yield of fluo-
rescence (0.01 in DMF) expectedly due to large percentage
of inter system crossing (ISC) that provides «heavy atom
effect» of Pt-atoms, but no generation of singlet oxygen
was also indicated (by direct method in which luminescence
of 1O2 was registered at 1270 nm in DMF-d7). However, in
phototoxicity experiments with HeLa cell line this complex
exhibited IC50 value at 1.6 M at the light and 12 M in the
dark. These results could be rationalized by production of
other forms of reactive oxygen species (ROS) rather than
singlet oxygen. Notably, for zinc complex of tet-
ra(pyrido)porphyrazine light and dark toxicities IC50 were
28 and 34 M respectively. Difference in dark toxicity of
precursor and platinated complex was explained by DNA-
binding ability of transplatine fragments, which was shown
in this work; after irradiation difference in toxicity was ex-
plained by «double-punch» effect (photoxicity in combina-
tion with DNA-binding ability) provided by platinated
complex. Thus, authors proposed further investigation of this
novel exocyclically platinated tetra(pyrido)porphyrazine as a
photosensitizer.
Also, positive charges can be obtained by introduction
of quaternized phosphonium moieties (Figure 15).[269271]
For example, such complexes were synthesized with the
intention to be used for enhanced mitochondria penetration
owing to four triphenylphosphonium units, they demon-
strated both photodynamic and sonodynamic efficacy against
MCF-7 and HeLa cell lines (IC50 = 12.7 45.0 M).
Figure 14. Synthesis of phthalocyaninate modified by four transplatine units.[259]
Figure 15. Synthesis of phthalocyanines with trisphenylphosphonium moieties.[269271]
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
238 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
Figure 16. Reversible nucleophilic addition to phosphorus(V)
phthalocyaninate (Nu- = OH- or MeO).[264]
Recently Kolomeychuk et al. performed detailed in-
vestigation of phthalocyaninate where the cationic phospho-
rus(V) centre was placed into the cavity of the
macrocycle[264] in order to compare its photophysical prop-
erties with corresponding porphyrin derivatives.[272,273] Dur-
ing this study the authors discovered the unprecedent
switching of aromaticity (Figure 16),[264] which could be
"turned off" by reversible aromatic nucleophilic addition of
OH- or OMe- to the pyrrole C-atom affording the nonflu-
orescent adduct. Aromaticity and fluorescence could be
restored by acidic treatment of the adduct. Structure of the
adduct was confirmed by single crystal X-ray analysis.
Kobayashi et al. demonstrated the prospective of cati-
onic phthalocyanines with endocyclic pnictogene atoms.
They synthesized non-peripherally substituted octa(2,6-
dimethylthiophenoxy) phthalocyaninates of P(V), As(V)
and Sb(V) and thoroughly investigated their optical proper-
ties (Figure 17).[260262] Absorption maxima of these com-
plexes (Q-bands) were found at 1018, 1032 and 1056 nm,
respectively, whereas the analogous zinc complex had a Q-
band located at 790 nm (in CH2Cl2). Such strong shifts of
Q-bans into near-IR region for P(V), As(V) and Sb(V)
complexes occurred due to decrease in HOMOLUMO gap
that was confirmed by DFT calculations. In addition, the
insertion of electron-donating S-atoms in non-peripheral
positions also contributed to the narrowing of the HOMO
LUMO gap.
Figure 17. Complexes of P(V), As(V) and Sb(V) with non-peripherally 2,6-dimethylthiophenoxy-substituted phthalocyanine.[261]
(a)
(b)
(c)
Figure 18. (a) Zinc(II) complex with crown-substituted oxanthrenocyanine ZnOc; (b) reversible formation of bifurcate proton bonds
upon acidic treatment; (c) UV-vis spectra of ZnOc and its protonated forms in CHCl3.[265] Adapted with permission from ref. [265] Copy-
right 2016, American Chemical Society.
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 239
Apart from strong bathochromic shifts of Q-bands
non-peripheral substitution provides phthalocyanines with
enhanced basicity of their meso-nitrogen atoms.[274,275] Pro-
tonation has profound effect on optical properties of phthal-
ocyanines resulting in strong bathochromic shifts of their
Q-bands and quenching of fluorescence.[276] For example,
Safonova et al. developed approach toward tetra-15-crown-
5-annelated zinc oxanthrenocyaninate ZnOc (Figure 18a,b)
and demonstrated consecutive formation of four proto-
nated forms having absorption maxima at 798, 867, 974
and 1028 nm respectively whereas non-protonated form
had maximum of absorbance at 732 nm (Figure 18c).[265]
Notably, this protonation is reversable and neutral form can
be recovered under addition of amines. Such proton-
susceptible compound can serve as a molecular switcher for
optoelectronic materials, sensors as well as switchable pho-
tosensitizers.
To conclude, cationization of phthalocyanines is an ef-
fective way to complexes, that are soluble in water and ex-
ist in aqueous solutions predominantly as monomeric spe-
cies. Also, some cationic phthalocyanines and their analogs
have absorbance beyond 1000 nm due to complexation with
pnictogens or formation of protonated species. These prop-
erties of cationic Pcs in conjunction with excellent photo-
dynamic efficiency may be useful in photomedicine and
also, cationic Pcs might be suitable NIR dyes, molecular
switchers and sensors.[277]
6. Effective Near-Infrared Photosensitizers for
PDT and aPDT Based
on meso-Tetrakis(3-pyridyl)bacteriochlorin
The reduced derivatives of porphyrins such as, chlo-
rins and bacteriochlorins, are used as photosensitizers (PSs)
for photodynamic therapy (PDT) of cancer due to their
high-intensity optical absorption and luminescence in the
red and near-IR range.[46,148,278-281] Thus, well-known photo-
sensitizers based on hydrogenated porphyrin derivatives
such as Foscan® (Temoporfin, meso-tetrakis(3-hydroxyp-
henyl)porphyrin),[282,283] Visudyne® (Verteporfin),[46,284] and
Tookad[285, 286] have been designed (Scheme 38).
A porphine molecule has a multiple-contour conjugat-
ed system; its interior chromophore contains 18 π electrons.
Two peripheral Сβ=Сβʹ bonds are quasi-isolated, and their
hydrogenation does not disrupt the main macrocyclic aro-
matic π-conjugated system (Scheme 39).[287290]
Reduction of the double bond in one of the pyrrolyc
moieties of porphine gives chlorin (2,3-dihydroporphyrin);
reduction of double bonds in two opposite pyrrole moieties
give rise to bacteriochlorin (7,8,17,18-tetrahydroporphyrin,
ВС), while hydrogenation of two adjacent pyrrole moieties
yields isobacteriochlorin (2,3,7,8-tetrahydroporphyrin, iBC)
(Scheme 40).[291]
Scheme 38.
Scheme 39.
Scheme 40.
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
240 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
Scheme 41.
Scheme 42.
Scheme 43.
The main data on preparation of synthetic chlorins and
bacteriochlorins have been summarized in reviews.[292-294]
The simplest and most available method for preparation
reduced porphyrin derivatives is Whitlock reduction (reduc-
tion by diimide generated in situ from p-toluenesulfonyl
hydrazide, TsNHNH2) (Scheme 41).[295]
Solubility in bodily fluids, and water in particular, is
one of the key requirements posed on PSs, along with the
high quantum yield generation of singlet oxygen and tropic-
ity to tumor cells [14,296-299]. Therefore, meso-tetrakis(3-
pyridyl)bacteriochlorin is a promising platform for design-
ing novel photosensitizers: on the one hand, this compound
is characterized by strong light absorption in the long-
wavelength region (λmax = 747 nm; ε ~ 112,000 mol1 L cm1;
chloroform [300]), while on the other hand, quaternary am-
monium salts can be produced by quaternization of pyridyl
nitrogen atoms upon exposure to various alkylating agents,
giving rise to water-soluble species.
The initial meso-tetra(3-pyridyl)bacteriochlorin
(BC 1) was synthesized using the conventional method: by
Whitlock reduction of meso-tetrakis(3-pyridyl)porphyrin
(Scheme 41, R = 3-Py), and its two water-soluble salts
(tetraiodide (BC 2) and tetratosylate (BC 3)) were immedi-
ately produced by boiling of BC 1 with excess amount of
methyl iodide or methyl-p-toluenesulfonate, respectively
(Scheme 42).[301]
Later on, it was shown that other water-soluble qua-
ternary salts can be produced by boiling BC 1 with excess
amount of 1,4-dibromobutane (BC 4) or ethyl chloroacetate
(ВС 5), respectively, in nitromethane under an inert atmos-
phere (Scheme 42).[300] Aqueous solutions of tetracationic
salts are stable under dark conditions for up to 6 months.[302]
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 241
The presence of bromine atoms in the alkyl chain of
ВС 4 made it possible to conduct further quaternization and
increase the number of cationic sites to n = 8. Boiling BC 4
with excess amount of dry pyridine or dimethylaminoethanol
(DMAE) in methanol under an inert atmosphere for 4.5 h
yielded octabromide (BC 6) and octabromide (ВС 7), re-
spectively (Scheme 43).[300]
Hermann et al.[303] found that tetratosylate ВС 3 gen-
erates singlet oxygen via type I photosensitization. During
type I photosensitization reactions, an electron is detached
from the substrate by a photoexcited photosensitizer mole-
cule, which is accompanied by formation of hydroxyl radi-
cals, superoxide anion radicals, and hydrogen perox-
ide;[304,305] in turn, these species can easily penetrate biolog-
ical membranes, are highly reactive in oxidation processes,
and can further generate radical particles, thus ensuring an
oxidation cascade.
Tetratosylate ВС 3 exhibited high photodynamic ac-
tivity (> 86% dead cells) with respect to bile duct cancer
(BDC, GBC) cell lines in in vitro and in vivo experiments;
furthermore, it possesses low systemic toxicity and derma-
tological toxicity.[306] Later, in vivo experiments showed
that BC 3 is rapidly absorbed by human choroidal melano-
ma cells and has low dark toxicity after 24 h incubation.[307]
The studies of pharmacokinetics and biodistribution of ВС
3 in vivo (in live mice) and ex vivo (in resected organs) in
Balb/c colon-26 carcinoma tumor-bearing mice showed that
ВС 3 is predominantly accumulated in the tumor compared
to most normal tissues except for the kidneys: the tumor-to-
normal tissue ratio (TNTR) for the liver, colon, muscle, and
spleen tissues ranged from 8 to 50.[307]
Physicochemical properties, biodistribution in animal
tissues and photoinduced antitumor efficacy of ВС 4 and
ВС 6 were studied using epidermoid carcinoma of the lar-
ynx (Hep2) cell culture and the Lewis lung carcinoma
(LLC) animal tumor model. In vitro studies showed that
photosensitizers remained stable for a long time and exhib-
ited a high photoinduced activity with respect to НЕр2 cell
culture (IC50 varied from 0.34±0.07 to 0.71±0.09 μМ)[308].
These compounds were shown to be rapidly accumulated in
tumor tissue in mice (the tumor-to-normal tissue ratio being
2.33.3), exhibit a high photoinduced activity against LLC
tumor cells for BC 4 and BC 6 in the in vitro (IC50 is
0.34±0.03 and 0.37±0.08 μМ, respectively) and in vivo sys-
tem (inhibition of tumor growth and response rate are
100%).[308]
Along with being used as conventional photosensitiz-
ers for PDT, BC 1 and its water-soluble derivatives exhibit
antibacterial activity against various pathogens. Thus, ВС 3
was used against Gram-positive S.aureus strains (MSSA
and MRSA) and Gram-negative E.coli and P. aeruginosa
strains.[309,310] BC 3 was found to strongly bind to the cell
wall of both Gram-positive and Gram-negative bacteria and
exhibit no inhibitory activity against S.aureus in the dark,
while irradiation causes almost complete inactivation of the
pathogen.[309] Incubation of the S. aureus MRSA strain in
phosphate-buffered saline (PBS) in the presence of ВС 3
for 30 min followed by irradiation causes almost complete
death of the pathogen.[310]
Along with quaternary ammonium salts, lyophilisate
for solution for infusion based on BC 1 and containing a
non-ionogenic surfactant, Bacteriosens drug, can be used as
a water-soluble photosensitizer.[311] This lyophilisate is sta-
ble during storage under dry conditions for 6 months. Bac-
teriosens exhibits a moderate in vitro hemolytic activity,
does not possess dark cytotoxicity against cultured human
and mouse cancer cells, and exhibits high specific activity
when exposed to light.[312,313]
Loschenov et al. described the application of ВС 1 as
a photobactericidal coating for a hydroxyapatite implant
and demonstrated that this coating was resistant to washout
of photosensitizer particles over time.[314]
The Intrachlorin composition based on meso-
tetrakis(3-pyridyl)bacteriochlorin and being a mixture of
water-soluble meso-tetra(3-pyridyl)bacteriochlorin and me-
so-tetra(3-pyridyl)chlorin (88 : 12, wt. %) was designed.[315,316]
The mixture was prepared by Whitlock reduction of meso-
tetrakis(3-pyridyl)porphyrin, and pyridyl nitrogen atoms
were subsequently quaternized using methyliodide.[316] The
resulting composition is characterized by stability, high-
intensity long-wavelength optical absorption, and high
quantum yields of singlet oxygen generation (~
0.45±5).[317] The Intrachlorin composition was active when
used at a dose of 515 mg per 1 g of tumor against model
Lewis tumors, spontaneous pituitary cancer, and breast can-
cer.[318] Rzhevskii et al. reported using the Intrachlorin
composition for photodynamic therapy of C26 colon carci-
noma subcutaneously grafted to Balb/c mice.[319] The recur-
rence-free efficacy during 6 months was 70 %. A systemic
toxicity study of the Intrachlorin composition in Wistar
mice was conducted.[319] The 10 mg/kg dose of the In-
trachlorin composition was found to be safe when adminis-
tered daily during 7 days, both intravenously and subcuta-
neously; the dose > 20 mg/kg repeatedly administered in-
travenously and the dose > 50 mg/kg repeatedly adminis-
tered subcutaneously were found to induce toxic effects.[319]
7. Spectral Properties of Photosensitizers Based
on Tetra(pyridin-3-yl)porphine and Its Reduced
Forms in Solutions and Thin Films
There are numerous tetrapyrrolic compounds that are
used as PSs in PDT, whereas porphyrins and phthalocya-
nines are the most frequently used PSs in antibacterial pho-
todynamic therapy (aPDT). Cationic porphyrins like TMPyP
(5,10,15,20-tetrakis(1-methyl-4-pyridinium)porphyrin tetra(p-
toluenesulfonate)) have fourfold positive charge. Collins et
al. used TMPyP against Pseudomonas aeruginosa biofilms
both wild and mutant strains.[320,321] It was postulated that
this effect might be due to the large molecular structure of
TMPyP or strong electrostatic interaction between the four-
fold positive charge of cationic porphyrin and negative
charge of extracellular polymeric substance molecules.[32 2]
Nowadays cationic antimicrobial peptides or cell pen-
etrating peptides are conjugated to porphyrins to improve
their efficiency. These conjugated porphyrins show a great
cell inactivation during aPDT. Recent studies strongly up-
hold the hypothesis that aPDT can be a satisfactory alterna-
tive since there is a substantial difference in the mode of
action of PSs than that of antibiotics. The key benefits of
aPDT can be outlined as follows: a broad spectrum of ac-
tion compared to antibiotics since PS can act on diverse
organisms such as bacteria, protozoa, fungi; bactericidal
effects independent of antibiotic resistance pattern; more
limited adverse effect profile and damage to the host tissue;
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
242 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
no resistance following multiple sessions of therapy. The
synthetic photosensitizers of the porphyrin series,[323,324]
conjugates of porphyrins with amino acids[325] and saccha-
rides[326,327] are the most studied. A special place in this
range of compounds is occupied by reduced forms of por-
phyrins - chlorins and bacteriochlorins, which can compete
with existing photosensitizers used in clinical practice.
During the last decade, nanotechnology has had a
great impact on PDT. Most of these studies have used na-
noparticles to improve the efficacy of classical PDT while a
few of them have been done on the antimicrobial as-
pects.[328] The results with nanoparticles were more satisfac-
tory than with the PS alone. Furthermore, PS bound to a
nanoparticle penetrates through the membrane better than
free PS. Nanoparticles themselves can act as PS.[329] To date,
low-dimensional porphyrin-based nanostructures with
promising properties have been obtained in three-
dimensional (3D) environments, on solid surfaces using
two-dimensional (2D) templates, exploiting covalent bond-
ing[330,331] or various non-covalent intermolecular interac-
tions.[332,333] Self-assembly is a key player in materials
nanoarchitectonics.[334336] The nanostructures formed in
this way arise from molecules carrying side groups that
promote the process. Supramolecular polymers were creat-
ed using diverse self-assembly strategies.[337,338] Formation
of different kinds of supramolecular assemblies from some
compounds at the water surface,[339] during electrochemical
deposition on electrodes[340] and in metal-organic frame-
works[341] was reported.
Previously, we demonstrated the possibility of supra-
molecular design at the air-water interface by controlled
self-assembly of porphyrins into 2D and 3D nanoaggre-
gates.[342349] The concept of nanostructuring of organic
compounds at the air-water interface and a model of a float-
ing layer, the structural units of which can be both individ-
ual molecules (Langmuir's approach, special case) and their
major nanoaggregates (so-called M-nanoaggregates, gen-
eral case), were presented.[342,350] The formation of the por-
phyrin nanoparticles from magnesium porphine, a function-
al element of chlorophyll, was reported.[342] The formation
of nanoparticles of biological and bioactive compounds is a
non-trivial, but very important and urgent task. Recently,
we have established the possibility of formation of nanopar-
ticles by a hydrophobic derivative of vitamin B12 (porphy-
rin-type compound) whose properties differ from the prop-
erties of the parent molecules. They were formed at the
water-air interface. This was important for pharmacology
because this derivative is a neuroprotective agent.[351] Mi-
cro- and nanocapsulation technologies occupy an important
place in the development of means for targeted delivery of
hydrophilic and hydrophobic drugs and modulation of their
biological properties.[352-354] It was found that vitamin B12
itself can also form nanosized ensembles in delivery sys-
tems.[354]
However, nanostructures of photosensitizers based on
3-pyridylporphine, which is parent compound of such im-
portant photosensitizers as chlorin and bacteriochlorin, have
not yet been obtained.
The aim of this work is to synthesize and characterize
the 5,10,15,20-tetra(pyridine-3-yl)chlorin (T3PyCh) and
5,10,15,20-tetra(pyridine-3-yl) bacteriochlorin (T3PyBCh),
which are the products of 5,10,15,20-tetra(pyridine-3-
yl)porphine (T3PyP)[355] reduction according to the proce-
dure of Whitlock
[295] (Figure 19). The possibility of form-
ing T3PyP nanostructures by self-assembly at the water-air
interface and in thin films on solid supports was studied.
Figure 19. Structural formulae of 5,10,15,20-tetra(pyridine-3-
yl)porphine (T3PyP); 5,10,15,20-tetra(pyridine-3-yl)chlorin
(T3PyCh); and 5,10,15,20-tetra(pyridine-3-yl) bacteriochlorin
(T3PyBCh).
T3PyP in pyridine was heated at 100-105 °C in the presence of
potassium carbonate with p-toluenesulfonylhydrazide (pTSH); the latter
was added gradually. The reaction was carried out until the maximum ratio
of optical densities at 745 and 653 nm (absorption maxima of bacteriochlo-
rin and chlorin, respectively) was reached. It was shown that this ratio is
inconstant and reaches a maximum (8-10) between 12 h and 16 h.
The layers at the water-air interface were formed on a Langmuir
setup (NT-MDT, Russia). A solution of T3PyP in dichloromethane with C
= 0.88·10-4 М was applied to the surface of bidistilled water using a sy-
ringe with a graduation value of a 1 µL (Hamilton, Sweden) at (20±1) ºC.
The solution was taken in such a volume that the initial degree of surface
coverage, assuming perpendicularity of the planes of molecules and the
water surface (ISCD cedge, the ratio of the area occupied by the molecules
of the substance to the surface area of water) was between 25 and 51%.
Also the ISCD cface value for the parallel arrangement of the molecular
planes and the surface was calculated. Surface pressure was measured
using Wilhelmi sensor with an accuracy of 0.02 mN/m. In 15 minutes after
the application of the solution, the formed layer was compressed at a rate
of v=2.3 cm2·min-1.
Langmuir-Schefer (LS) films were prepared at (17±1) ºC. For this
purpose, the barriers compressing the layer on the water surface were
stopped at the selected pressure value, and then the layers were successive-
ly transferred onto the quartz substrate by the horizontal lift method. To
obtain optimal optical density, the number K of times the substrate was in
contact with the layer was varied from 20 to 60. The electronic absorption
spectra of LS films and solutions were recorded on a Shimadzu-Uv-1800
spectrophotometer (wavelength resolution ±1 nm), fluorescence spectra
were measured with the Solar spectrophotometer.
The structure of the layers at waterair interface was analyzed by
using quantitative analysis of compression isotherms of a nanostructured
M-monolayer, based on the concept of a layer as a real two-dimensional
gas with structural elements representing two-dimensional nanoaggregates
520 nm in diameter (M-nanoaggregates).[342] The method allows determi-
nation of the critical characteristics of floating layers: the area per mole-
cule in a nanoaggregate, the number of molecules in nanoaggregates, etc.
The stable state of the layer is described by equation π(A–Amol ) = kT/n (1)
where π is the surface pressure, A is the area per molecule in the layer,
Amol is the area per molecule in the aggregate, n is the number of mole-
cules in the aggregate, k is the Boltzmann constant and T is the absolute
temperature. When plotted in πA–π axes, a compression isotherm of the
floating layer has both linear (corresponding to the single-phase states of
the layer) and nonlinear regions. The Amol and n are the main characteris-
tics of a floating monolayer; these values may be determined by approxi-
mating the πA–π plot using a linear function (the least squares method was
used and the πA error did not exceed 3%).
Geometric parameters of the molecule, determined using the Hyper-
chemistry software, are as follows: а = 1.5 nm, b = 1.4 nm, с = 0.7 nm; the
projection areas Aproj(face) = 1.55 nm2, Aproj(edge) = 0.7 nm2; the areas
of circumscribed rectangles Arec(face) = 2.2 nm2 and Arec(edge) = 1.2 nm2.
The areas per molecule in the most densely packed monolayer are
Apack(face) = 2 nm2 and Apack(edge) =1 nm2.
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 243
Although the Whitlock procedure is now the main
method for obtaining chlorins from porphyrins, its course
has not been systematically studied. In turn, understanding
the principles of this interaction would be useful for identi-
fying reaction conditions in which the target product is ei-
ther T3PyCh or T3PyBCh. To this end, we quantitatively
monitored the diimide reduction of T3PyP in the course of
the Whitlock reaction by measuring the optical density of
the reaction mass at 653 and 745 nm after adding each por-
tion of the reducing agent (pTSH).
The reaction can be divided into two steps: the reduc-
tion of porphyrin to chlorin and the formation of bacterio-
chlorin. In the first stage, chlorin is predominantly formed,
which is accompanied by an increase in absorbance at 653 nm
(Figure 20a). In the second stage, chlorin is converted to
bacteriochlorin; absorbance decreases at 653 nm and in-
creases at 745 nm (Figure 20b).
The concentration of chlorin in the mixture initially
increases, reaches a maximum after 7.5 h, and then decreas-
es. On the other hand, the concentration of bacteriochlorin
increases insignificantly in the beginning, but starts to in-
crease sharply after 6 hours. Therefore, if the target product
of the reaction is chlorin, the reaction should be stopped at
the moment when bacteriochlorin absorption starts to grow
rapidly. Maximum conversion to bacteriochlorin is expected
when the maximum ratio of optical densities at 745 nm and
653 nm is reached.
Taking this data into account, we individually isolated
the reduced products. The hydrogenated products,
T3PyCh and T3PyBCh, were isolated by column chroma-
tography.[356] Chromatographic separation was performed
on aluminum oxide, and for the first time on silica gel using
chloroform or dichloromethane and their mixtures (up to
5%) with methanol. In general, chromatography on both
sorbents leads to almost identical results, however, separa-
tion on silica gel is more efficient in terms of product yield.
Figure 21 shows the absorption and fluorescence spec-
tra of the initial porphyrin T3PyP and its reduced forms,
T3PyCh and T3PyBCh.
As can be seen, the transition from porphyrin
T3PyP to chlorin T3PyCh results in significant changes in
the electronic spectra of their solutions. In particular, the
Soret band decreases significantly and broadens with the
formation of the "shoulder", with a simultaneous increase in
the intensity and a slight bathochromic shift of the last band
in the Q-region. Changes in the spectrum of bacteriochlorin
T3PyBCh solution are even more noticeable: intensive
absorption bands appear in the short-wave region at 355 nm
and 378 nm and in the long-wave region at 745 nm.
а
b
Figure 20. Electronic absorption spectra (DCM) of the reaction mixture during the first 6 h (а) and the last 6 h (b) of the reaction.
a
b
300 400 500 600 700 800
0
100000
200000
300000
400000
500000
450 500 550 600 650 700 750 800
0
20000
40000
60000
80000
100000
120000
140000
, нм
, нм
T3PyP
T3PyCh
T3PyBCh
Figure 21. UV-Vis absorbance spectra (DCM) (a) and emission spectra (DCM) (b) at 520 nm of T3PyP, T3PyCh and T3PyBCh.
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
244 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
a b
Figure 22. (a) The π-А isotherm of tetra(pyridine-3-yl)porphine. Point A shows the conditions of layer transfer onto the solid support. (b)
UV-Vis absorption spectra of solution in dichloromethane (C=1·10-6 М, dotted line) and of the LS film formed from nanostructured layers
(solid line).
As part of the search for a solution to the problem of
formation of nanoarchitectures of bioactive porphyrin-type
compounds, studies have begun on the possibility of form-
ing nanoparticles of these compounds. The compression
isotherm for the layer formed at cedge=51% is presented in
Figure 22a. The structure of the layers was analyzed within
the framework of the author's model of a nanostructured M-
monolayer using the quantitative method of isotherm analy-
sis [342]. It was found that T3PyP forms nanostructured lay-
ers at the water-air interface. The conditions for the for-
mation and the main characteristics of monomolecular lay-
ers obtained at small values of the ISCD are determined. In
particular, at low surface pressures (less than 1 mN/m) the
number of molecules (n) of T3PyP in the M-nanoaggregate
is 270-330; aggregate diameter (Daggr) is 2-28 nm; the area
per molecule in the nanoaggreagate mol) is 1.1 nm2; the
pressure range of existence of a stable monolayer is 0.1-
0.5 mN/m; water content in the nanoaggregate (when mole-
cules are arranged vertically in stacks) 25%; the degree of
surface coverage by aggregates at the beginning of the re-
gion of stable monolayer is 83 %.
Three-dimensional tetralayer nanoaggregates formed
on the water surface were transferred to a quartz plate by
the Langmuir-Schaefer method at the surface pressure of
50 mN/m (point A in Figure 22a). The positions of the
main bands in the absorption spectrum of T3PyP film
(Figure 22b, solid line) are shifted batochromically com-
pared to the spectrum of the monomer solution (dash-and-
dot line in Figure 22b): the Soret band by 16 nm and Q
bands by 8-12 nm. The half-width of the Soret band in the
film spectrum is twice as large as in the monomer solution.
The ratio of intensities of Q bands has also changed. These
results indicate intermolecular interactions stronger than the
van der Waals ones in the nanostructures formed within the
films of tetra(pyridine-3-yl)porphine.
Thus, 5,10,15,20-tetra(pyridine-3-yl)chlorin (T3PyCh)
and 5,10,15,20-tetra(pyridine-3-yl) bacteriochlorin
(T3PyBCh), the products of 5,10,15,20-tetra(pyridine-3-
yl)porphine reduction, were synthesized and fully charac-
terized, and their photophysical properties were studied.
The first stable T3PyP nanostructures, whose properties
differ from properties of original substance were obtained
by its controlled self-organization within layers at the air-
water interface and films on solid supports.
8. Derivatives of meso-Formylporphyrins:
Synthesis and Application as Components
of Optical Sensors and Photosensitizers
The production of porphyrin materials with target spe-
cific structures is based on various methodologies, differing
in that natural or synthetic porphyrins are used. Synthetic
porphyrins such as
-octaethylporphyrin (OEP) and meso-
tetraphenylporphyrin (TPP) are considered as a good alter-
native to the natural porphyrins due to their availability by
simple synthesis. The porphyrin core, which is easily ob-
tained by tetrapyrrole condensation,[357] then needs to be
functionalized to impart the required properties to the por-
phyrin molecule.[358-360] The free meso-positions of
-
octaethylporphyrin and meso-arylporphyrins can be func-
tionalized by a variety of methods, among which the
formylation is especially prolific.[361] The advantage of this
functionalization methodology is based on that the aldehyde
group is rich in the possibilities of further transformations
leading to the addition of functional fragments. There are
reported examples of formylporphyrins utilization in a wide
variety of reactions including, but not limited to the Wit-
tig,[362-364] Grignard,[364-366] McMurry,[367] cycloaddition,[368]
Knoevenagel[369] reactions and Schiff bases preparation.[370]
The products of the formylporphyrins transformations are
potential optical sensors[371] and photosensitizers.[46, 372,373]
meso-Functionalization of porphyrins by electrophilic
formylation of tetrapyrrole macrocycle
Although a formyl group can be inserted into a
tetrapyrrole macrocycle by various ways, the “old-
fashioned’’ classical aromatic electrophilic substitution
reaction, namely the Vilsmeier-Haack formylation reaction
stands out, as it allows the formyl group to be inserted into
the porphyrin core with simple procedure and affordable
reagents (DMF/POCl3 (Scheme 44).[361,374] The Vilsmeier
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 245
reagent is an electrophilic agent, the iminium cation,
formed by the interaction of POCl3 with DMF. To promote
the reaction of an electrophilic Vilsmeier reagent with a
nucleophilic porphyrin substrate, the tetrapyrrole aromatic
system is activated by the formation of metal complexes in
which the porphyrin core has a formal charge of -2, and
thus it is enriched with electrons. Porphyrin complexes with
Ni(II), Cu(II), Pd(II), Pt(II) resistant to HCl released during
the reaction, are usually formylated.[361]
meso-Formylporphyrins formed during the formyla-
tion of
-substituted porphyrins differ significantly in phys-
ical and chemical properties from β-formylporphyrins -
products of the formylation of β-unsubstituted porphy-
rins.[374] This is explained by the fact that the aldehyde
group in β-formyl derivatives is conjugated with the
tetrapyrrole macrocycle and exhibits all the properties of
arylaldehydes, while the aldehyde group in meso-
formylporphyrin is largely out of conjugation with the mac-
rocycle due to steric hindrances caused by neighboring pyr-
role substituents.[361] There is an additional wide long-
wavelength band in the region of 650-670 nm in the UV-
Vis spectra of meso-formylporphyrins.
In the process of formylation with a Vilsmeier reagent,
prepared from 3-(dimethylamino)acrolein instead of DMF,
the corresponding acrolein derivatives of porphyrins were
formed. These derivatives were easily cyclized to form an-
nelated six-membered cycles, when heated in an acidic
medium. Smith et al. obtained Ni(II) meso-acroleinyl-β-
octaethylporphyrin 1, which was cyclized by the action of
sulfuric acid at room temperature for two hours into benzo-
chlorin 2 (Scheme 45).[375]
The absorption spectrum of such annulled porphyrins
is significantly bathochromically shifted, which makes
them promising photosensitizers for photodynamic therapy
(PDT).[46,372,373,376] Thus, the product of the double cycliza-
tion of meso-bis-acroleinyl-β-octaethylporphyrin 3 diben-
zobacteriochlorin 5 [375] possesses a strong absorption band
in the region of 752 nm, which fully corresponds to the
tissue transparency window (Scheme 46).[373,378] The conju-
gates of the similar benzochlorin with carbohydrates were
screened using the galectin-binding-ability assay and exhib-
ited an enhancement of about 300-400-fold compared to
lactose. All conjugates were also shown to possess good
photosensitizing efficacy with fibrosarcoma tumor cells.[379]
The McMurry reaction
The McMurry reaction, first described in 1974, is a re-
action of an organometallic derivative of aldehydes and
ketones to form alkenes, which is based on the use of low-
valent titanium. Peripherally carbonyl-substituted metal
porphyrins were subjected to this reaction with the for-
mation of the variety of bis-porphyrins and their homo- and
hetero-bimetallic complexes.[367] Cu(II) meso-formyl-
-
octaethylporphyrin (CuOEP-CHO) was dimerized under the
action of TiCl3 and Zn/Cu to form of the copper complex of
bisporphyrin 6 bound by the ethene bridge with a 64%yield in
the form of a mixture of cis and trans isomers (Scheme 47).[375]
A similar reductive dimerization reaction was carried
out with acrolein derivatives, resulting in the formation of
the dimer with a hexatriene bridge. The dimerization of the
benzochlorin acrolein derivative 4 led to the dimer 7, for
which a significant bathochromic shift of the Q band up to
706 nm was observed.
Scheme 44. The Vilsmeier-Haack formylation reaction.
Scheme 45. Transformation of Ni(II) β-octaethylporphyrin (NiOEP)
into Ni(II) benzochlorin 2.
Scheme 46. Preparation of dibenzobacteriochlorin.
Scheme 47. McMurry reaction of Ni(II) and Cu(II) meso-
formylporphyrins.[375]
Scheme 48. McMurry reaction of Ni(II) meso-acroleinylbenzochlorin.
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
246 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
Grignard and Wittig reactions
Johnson and Arnold[366] performed the Grignard reac-
tion between Ni(II) meso-formyl-
-octaethylporphyrin
(NiOEP-CHO) and MeMgI, obtaining, as expected, Ni(II)
5-(1-hydroxyethyl)-
-octaethylporphyrin 8, by eliminating
water from which Ni(II) 5-vinyl-
-octaethylporphyrin 9
was obtained (Scheme 49). Smith et al. carried out a similar
reaction with a free base and a zinc complex of meso-
formyl-
-octaethylporphyrin, which resulted in 15-
alkylated products (10), and the formyl group remained
intact (Scheme 50). A regioselective attack of the C-
nucleophile to the 15-carbon of the macrocycle of Zn(II) 5-
formyl-
-octaethylporphyrin was also observed in the case
of the methyllithium reaction, while with the free base
-
octaethylporphyrin, a product of the attack to the carbonyl
group 5-(l-hydroxyethyl)-
-octaethylporphyrin was ob-
tained. The Wittig reaction is more reliable in this regard,
with the help of which the meso-formyl group of porphyrins
is easily transformed into a meso-vinyl (Scheme 49), 2-
(ethoxycarbonyl)ethenyl, 2-cyanoethenyl, and others.[361,364]
The meso-vinyl group can further be functionalized
with the conjugation extension. The direct C-H borylation
of the meso-vinyl group in 9 was performed with Cu(II)
complex as a catalyst, yielding the meso-(2-
pinacolboronylethenyl)porphyrin 11, which was shown to
act as nucleophilic partner in the Suzuki cross-coupling
leading to porphyrin derivatives with an extended π-
conjugation through the carbon-carbon double bond.[380]
The oxidative homocoupling of the borylporphyrin 11 pro-
duced the dimer 13 (Scheme 51).[381] Thus, this strategy of
the meso-formyl group transformations allows to attach
various aromatic chromophores through the ethene and bu-
tadiene bridges. The products of couplings possess batho-
chromic shift of absorption bands.
Scheme 49. The Grignard and Wittig reaction of Ni(II) meso-formyl-
-octaethylporphyrin.
Scheme 50. The Grignard reaction of Zn(II) meso-formyl-
-
octaethylporphyrin.
Scheme 51. The borylation of the vinyl-porphyrin with the subsequent
coupling reactions.
Scheme 52. Preparation of an exocyclic derivative of the dimethyl
mesoporphyrin IX.
Lugtenburg et al. obtained a 2-cyanoethenyl derivative
12 from the dimethyl meso-formyl-mesoporphyrin IX (11),
which was then cyclized when heated in an acidic medium
to a quinoline-annelated product 13 (Scheme 52).[382] The
product 13 strongly absorbed light at 685 nm and it was
tested as a photosensitizer in the PDT of the tumor cells of
the ovary of the Chinese hamster (Cricetulus griseus),
providing a good antitumor activity.
Scheme 53. Preparation of copropurpurin I from coproporphyrin I.
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 247
The formyl group in NiOEP-CHO and Ni(II) tetrame-
thyl ester of meso-formyl-coproporphyrin I (14) was trans-
formed by Wittig reaction with ethoxycarbonylmethylenetri-
phenylphosphoran into 2-(ethoxycarbonyl)ethenyl group.[383]
After demetallation in sulfuric acid, the obtained products
were heated for 24 hours in glacial acetic acid in an argon
atmosphere, causing the cyclization with the formation
of etiopurpurin I and copropurpurin I (16), respectively
(Scheme 53).
The authors of the work conducted a study on rats
with the bladder cancer induced. The obtained etiopurpurin
I and tetramethyl copropurpurin I photosensitizers were
emulsified and injected intravenously to the rats. The PDT
of the cancer cells proceeded efficiently with both com-
pounds. However, it was noted that the copropurpurin I has
a lower cytotoxic effect, and selective accumulation of eti-
opurpurin I in tumor cells was also observed.
The Knoevenagel reactions
The Knoevenagel reaction, like the Wittig reaction, is
used to transform formylporphyrins into the corresponding
derivatives of acrylic acid. Meso-formyl-triarylporphyrin
derivatives were usually synthesized under standard condi-
tions of basic catalysis. The meso-cyanoacrylate derivative
of Zn(II) meso-formyl-tri(p-tolyl)porphyrin was obtained
by boiling in piperidine with methanol for 16 hrs.[384] meso-
Formyl-diarylporphyrin reacted with nitromethane, dime-
thylmalonate and malononitrile in a mixture of piperidine,
acetic acid and toluene.[385] The product of the reaction of
meso-formyldiarylporphyrin with malononitrile containing
meso-dicyanovinyl group was shown to act as a fluores-
cence ‘‘turn-on’’ cyanide probe.[386] meso-Nitroethylene
derivative was utilized as fluorescence turn-on probes for
biothiols as it exhibited fast fluorescence enhancement and
high selectivity towards thiols based on the Michael addi-
tion mechanism.[385] It was also successfully applied to fluo-
rescent cell imaging in the NIR wavelength range.
meso-Formyl-
-octaethylporphyrin (NiOEP-CHO) is
less reactive and also easily degraded under basic condi-
tions, therefore it requires a special approach. Activation of
the carbonyl group with TiCl4 in pyridine promoted the
reaction.[369] Using the Knoevenagel reaction with NiOEP-
CHO, heterocyclic derivatives of porphyrins were ob-
tained.[387] First, upon condensation with malonic ester, the
corresponding malonate derivative (17) was obtained,
which reacted with hydrazine in the presence of sodium
ethoxide, resulting in formation of pyrazolidine-3,5-dione
linked to the Ni(II)
-octaethylporphyrin with ethane bridge
(18) (Scheme 54).
Direct synthesis of the porphyrin conjugates with thi-
ohydantoin, and thiobarbituric acid (Scheme 55) was also
performed via the Knoevenagel reaction.
The UV-Vis spectra of the heterocyclic conjugates
contain new bands arose from the interaction of the conju-
gated chromophores as well as bathochromically shifted
original absorption bands. Particularly dramatic changes
were observed in the UV-Vis spectrum of the thiobarbituric
acid conjugate, which exhibited substantial increase of ab-
sorption in green and red spectral region. Such combina-
tions of porphyrin dyes with such heterocyclic chromo-
phores and sensing fragments can be of interest as potential
promising photosensitizers and sensor dyes.
Synthesis and properties of Schiff bases derivatives
of meso-formylporphyrins
Synthesis of Schiff bases from formylporphyrins is a
promising direction of functionalization, since a huge num-
ber of various tetrapyrrole compounds with desired proper-
ties can be obtained from azomethine derivatives, including
photosensitizers and sensor dyes with the required photo-
physical characteristics.[388] There are two main ap-
proaches to the synthesis of Schiff bases: 1) interaction
of formylporphyrins with amines; 2) interaction of amines
with the so called "phosphorus complex" formed during the
Vilsmeier-Haack reaction.[370] The classical reaction of the
preparation of Schiff bases by the reaction of amines with
the aldehyde group of porphyrin was first investigated in
the synthesis of oximes, which were formed within a few
hours from the meso-formylporphyrin with hydroxylamine
hydrochloride in boiling pyridine.[366,389] Oximes are of in-
terest as precursors for the production of cyanoporphy-
rins,[390] which can be consequently transformed to carboxyl
derivatives. Intramolecular cyclizations of meso-substituted
porphyrins often leads to porphyrinoids with fused exocy-
cles like purpurins and benzochlorins. Treatment of nickel
complex of meso-oxime 21 with lead tetraacetate (1.2 eq.)
in methylene chloride in the presence of an excess of tri-
ethylamine at room temperature for 5 min gives the product
22 with fused 1,2-oxazin ring with an overall yield of
77%.[391] When a solution of Zn(II) complex of meso-oxime
23 was stirred in methylene chloride with a small amount of
water, after a few hours the starting complex was trans-
formed to hydroxy-1,2-oxazinochlorin 24 (Scheme 56).[392]
Oxidative hydroxylation of the intermediate 1,2-
oxazinochlorin possibly occurred via a peroxide mechanism
leading to the stable hydroxychlorin. Ni(II) complex of the
oxime vinylog meso-(trans-formylvinyl)-
-octaethylporphyrin
25 underwent similar intramolecular cyclization producing
a condensed eight-membered exocycle 26 (Scheme 57).[393]
Such annulated products are of interest as photosensitizers
due to their strong absorption in red region. The electronic
absorption spectrum of 1,2-oxazocine fused porphyrinoids
26 shows a significant bathochromic shift by 50 nm of all
absorption bands compared to the spectra of the corre-
sponding chlorins.
Scheme 54. Synthesis of the conjugate of Ni(II)
-octaethylporphyrin with pyrazolidine-3,5-dione.
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
248 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
Scheme 55. Synthesis of the conjugate of Ni(II)
-octaethylporphyrin with thiohydantoin and thiobarbituric acid.
Scheme 56. Synthesis of the porphyrinoids with fused 1,2-oxazin ring via oxime cyclization.
Scheme 57. Synthesis of the porphyrinoid with fused 1,2-oxazocine ring 26 via vinylogous oxime cyclization.
Scheme 58. Preparation of azomethine derivatives of octaethylporphyrin from the phosphorus complex.
Despite the large number of examples of the utiliza-
tion of the interaction of formylporphyrins with an amino
group for preparation of Schiff bases, the reaction of the
"phosphorus complex" with amines has become more wide-
spread. Ponomarev et al. was the first to isolate phosphorus
complexes of various porphyrins.[370] Azomethine deriva-
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 249
Scheme 59. Preparation of cyclopentane-annelated derivatives.
Scheme 60. Treatment of the N-methylimine derivative of the Ni(II) -octaethylporphyrin (29) with a strong base.
tives of nickel and palladium complexes of various porphy-
rinoids including
-octaethylporphyrin, tetraalkyl esters of
coproporphyrins I and II, mesoporphyrin IX and mesochlo-
rin e6 were obtained by direct interaction of "phosphorus
complexes" with amines (Scheme 58).[388, 389,395]
The metal complexes of azomethine derivatives, ob-
tained by this way, were investigated as potential photosen-
sitizers for photodynamic therapy (PDT).[388] A number of
Schiff bases, in particular, Pt(II) and Pd(II) complexes of
-
octaethylporphyrin and tetramethyl coproporphyrin I, were
investigated as sensor dyes for determination of oxygen
concentration and acidity.[396,397] An optochemical probe for
cellular diagnostics using phosphorescent determination of
oxygen and pH in living cells was developed based on the
Pt(II) complex of meso-(N-methylimino)-
-octaethylporp-
hyrin.[398]
Upon studying the thermolysis of azomethine deriva-
tives of porphyrins, it was found that the cyclization occurs
with the formation of cyclopentane-fused derivatives
(Scheme 59).[399-401] During thermolysis of metal complexes
of meso-imines of tetraalkyl ester of coproporphyrin I the
formation of both cyclopentane and cyclopentane-lactam
bicycle was observed.[395]
Treatment the Ni(II) complexes of the azomethine deriva-
tives of
-octaethylporphyrin with t-BuOK led to the for
mation of the corresponding meso-nitrile (30), meso-amide
(31) and meso-hydroxy (32) derivatives (Scheme 60). The
latter was demetalated with sulfuric acid (Scheme 61), re-
sulting in the phlorin 33 with strong light absorption in the
region of 700 nm which could be useful for the photosensi-
tizing applications.[395]
meso-Hydrazone derivatives were obtained by reac-
tion of meso-formyl derivatives of
-octaethylporphyrin
and coproporphyrin I with hydrazine and N-substituted hy-
drazines catalyzed by trifluoroacetic acid (Scheme 62).[402]
Hydrazones 34, 35 were formed as a mixture of E- and Z-
isomers.
Scheme 61. Demetalation of the Ni complex of meso-hydroxy-
-
octaethylporphyrin (32).
Scheme 62. Synthesis of hydrazones from formyl porphyrins.
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
250 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
Scheme 63. Preparation of cyclopentane fused porphyrins by thermolysis of meso-tosylhydrazones.
Scheme 64. Synthesis of the porphyrin dyads linked with the imino group.
Thermolysis of N-tosylhydrazones in the presence of a
base produced meso-carbenes that were cyclized leading to
the corresponding cyclopentane fused porphyrinoids via
intramolecular carbene CH insertion.[4 03] The cyclopentane
fused products 27, 37 obtained were the same as in the
thermolysis of azomethines, however, the second product in
the N-tosylhydrazone of the coproporphyrin I thermolysis
was 6-member ring fused 38 instead of bicyclic lactam
28,[395] and yields of the products in the carbene based reac-
tion were higher (Scheme 63).
By the interaction of metal complexes of meso-
formyltriarylporphyrin 39 and meso-aminotriarylporphyrin
40, dyads 41, linked via an imino group, were obtained
(Scheme 64).[404] However, due to the low activity of the
meso-amino group and its degradation under sufficiently
harsh reaction conditions, yields were low. A satisfactory
yield of 57% was achieved for the zinc complex by cataly-
sis with a tenfold excess of ZnBr2. Such dyads can be used
in the two-photon excited PDT, which provides a highly
targeted treatment.[405]
Unsubstituted meso-hydrazones of
-octaethylporphyrin
and
-octaethylchlorin were used in the preparation of az-
ines 42 by reaction with various aldehydes (Scheme 65).[406]
Scheme 65. Preparation of azines from the Ni(II) complex of the me-
so-hydrazone of
-octaethylporphyrin.
In using chlorins containing an aldehyde group (me-
thyl pyropheophorbide-a (PPPa) and methyl pyropheophor-
bide-d (PPPd)) as aldehyde components in the reaction with
Ni(II) meso-hydrazone of
-octaethylporphyrin 34a, por-
phyrinoid dyads 43, 44 were obtained (Scheme 66).[406] In
the excited state of the dyads the energy was efficiently
transferred from the OEP moiety to the pyropheophorbide
chromophore.
One-pot formylation, hydrazone and azine formation
was realized for the meso-amino substituted octaethylpor-
phyrin.[407] Amino-group was preliminarily protected with
trifluoroacyl group. It was found that under the condi-
tions of formylation of meso-(trifluoroacetamido)-β-
octaethylporphyrin (45), the amide fragment was oxidized
to form hydroxamic acid 49 (Scheme 67). Combination of
trifluoroacetamide and arylazine groups in the products 47-
49 led to the strongly increased absorption near 500 nm and
considerably red-shifted Q-bands up to 650 nm.
Further expansion of the -electron system of the az-
ine functionalized disubstituted
-octaethylporphyrin was
achieved through the conjugation with another tetrapyrrole.
Interaction of the hydrazone 46a with methyl pyrophe-
ophorbide-d (PPPd), containing formyl group at
-position
of the tetrapyrrole ring resulted in formation of the azine
bridged porphyrin-chlorin conjugate 50 (Scheme 68). The
resulting compound features the substantial growth of the
Q-band intensity as well as red-shifting Soret band. The
azine bridged dyad could be of interest as potential photo-
sensitizers, sensor dyes and biologically active compounds.
Porphyrins with meso-fused heterocycles
Interaction of the aldehyde group with 1,2-dithiol is
often used to protect it by the formation of the 1,3-
dithiolane. A family of pushpull quinoidal porphyrin was
obtained from a meso-formyl porphyrin through the at-
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 251
tachment of 1,3-dithiolane (benzo-1,3-dithiolane) and
malononitrile fragments at the opposite meso-positions of
the 5,15-diarylporphyrin (Scheme 69).[408]
Porphyrins with meso-fused 2-imidazolyl heterocycles
53 were obtained from the meso-formylporphyrins as car-
bonyl components in the heterocyclic condensation. 5-
formyl-10,20-diarylporphyrins as well as their Cu(II) com-
plexes reacted with phenanthrene- or phenanthroline-5,6-
dione and ammonium acetate leading to the correspond-
ing imidazolyl-porphyrin conjugates with high yields
(Scheme 70).[409] Ruthenium complex of the free base por-
phyrin-imidazo[4,5f]phenanthroline conjugate 56 showed
good binding ability to DNA, which facilitated DNA pho-
tocleavage.[410] Thus, it could be a potential photosensitizer
for PDT.
Scheme 66. Synthesis of porphyrin-chlorin dyads linked by the azine bridge.
Scheme 67. Synthesis of trifluoroacetamide and arylazine oppositely substituted β-octaethylporphyrin.
Scheme 68. Synthesis of the azine bridged dyad 50.
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
252 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
Scheme 69. Synthesis of the porphyrins with meso-fused 2-imidazolyl heterocycles.
Scheme 70. Synthesis of the porphyrins with meso-fused 2-imidazolyl heterocycles.
Scheme 71. Synthesis of the meso-linked porphyrin fullerene conjugate 58.
Scheme 72. Synthesis of the porphyrin-corrole dyads 60.
meso-Formylporphyrins can form azomethine ylide af-
ter interaction with N-methylglycine. 1,3-dipolar cycloaddi-
tion of the intermediate azomethine ylide to the double
bond leads to the porphyrins with meso-fused heterocycles.
The porphyrin fullerene conjugate 58 was obtained by
this way from meso-formyltriarylporphyrin 57, N-
methylglycine and C60 fullerene (Scheme 71).[368] The ex-
citation of the dyad led to the extremely fast formation of
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 253
the exciplex due to the strong interaction between the por-
phyrin and C60 moieties placed at close proximity.
Directly linked porphyrin-corrole dyads 60 were ob-
tained by the condensation of the meso-formyltriarylporphyrin
59 and dipyrromethane (Scheme 72).[411] The strong exciton
coupling between porphyrin and corrole and reversible en-
ergy transfer were shown to exist in the dyads. When 5,15-
bisformylporphyrin was used in the condensation, the cor-
roleporphyrincorrole triad was formed.[412] Similar direct-
ly meso-meso linked porphyrin dimers and oligomers were
obtained using condensation of meso-formylated porphyrins
with pyrrole.[413] Such porphyrin dimers and oligomers
were shown to act as prospective photosensitizers.[4 14]
To sum up, meso-functionalization of porphyrins with
formyl group provides powerful tool for development of the
diverse porphyrin derivatives possessing valuable proper-
ties. In particular, promising photosensitizers with strong
red-shifted absorption bands, including NIR bands, were
obtained from the meso-formyl porphyrins via formation of
the annulated cycles such as benzochlorins and dibenzobac-
teriochlorins. meso-Imino derivatives were applied as sen-
sor dyes in the multi-modal, multi-analyte optochemical
sensing platform for cell diagnostics. Easily formed with
the help of the formyl group porphyrin conjugates with het-
erocycles can be used as biologically active compounds and
in sensing applications. Imino- and azino- bridges represent
two alternatives for bonding porphyrins into dyads, utiliz-
ing various pathways for energy transfer between the chro-
mophores. Currently, the post-derivatization of the meso-
formylporphyrins is under intense development.
9. Interaction of Multiporphyrin Complexes and
Nanoassemblies with Molecular Oxygen in
Solutions: Mechanisms and Some Specific Effects
Together with the formation of supramolecular com-
plexes of tetrapyrrole photosensitizers, which have already
found application in biomedicine,[72-76] another approach to
the formation of possible platforms of tetrapyrrole photo-
sensitizers of the third generation can be implemented by
combining tetrapyrroles of various structures with function-
al nanocarriers of organic or inorganic nature. Both ap-
proaches are promising for increasing the stability of PSs
and their targeted delivery to the tumor.
From the photophysical background it should be noted
that in both approaches, the first necessary step is connected
with the quantitative experimental analysis of the influence
of molecular oxygen O2 on deactivation of excited singlet
and triplet states of the given PS which is included in mul-
timolecular complex or attached to nanocarrier. Keeping in
mind this idea, within German-Belarus scientific collabora-
tion such comparative study was carried out for various
multicomponent nanoassemblies containing tetrapyrrolic
macrocycles.[74,75,79,80,88] Herein, we aim to review main
results of this study and provide a comprehensive under-
standing of O2 interaction with multicomponent species in
liquid solutions at ambient temperature. At the first step,
excited S1- and T1-states quenching by molecular oxygen
was studied for a set of Zn-porphyrin and Zn-chlorin chem-
ical dimers in which -conjugated macrocycles are cova-
lently linked via spacers of various nature and flexibility
(-CH2-CH2- bond or phenyl ring in mesoposition, cyclodi-
mers) in non-polar toluene with and without pyridine ad-
mixture. At the second step, self-organized multiporphyrin
triads and pentads were formed via non-covalent binding
interactions of Zn-porphyrin or Zn-chlorin chemical dimers
with di- and tetrapyridyl containing porphyrin or chlorin
extra-ligands, and the main peculiarities of the interaction
of O2with the triads and pentads of various and controlled
geometry were evaluated. Then, we consider perspectives
of semiconductor quantum dots (QD) as well as their nano-
assemblies with porphyrin molecules as possible PDT pho-
tosensitizers, and provide quantitative comparative results
on efficiencies of the singlet oxygen generation by QD-
porphyrin nanoassemblies. Finally, as far as in many exper-
iments the intensity of the singlet oxygen (1О2 or 1Δg) emis-
sion at max = 1.27 is measured in various solvents, some
specific reasons (described in literature) are discussed
which may change the rate constant of the radiative transi-
tion 1g 3Σ-g in 1О2 molecule.
All objects under study (including monomeric porphy-
rins, chemical dimers, triads, pentads as well as QD-
porphyrin nanoassemblies) were synthesized, prepared and
characterized according to methods comprehensively de-
scribed in our earlier publications,[74,75,79,80,88,416 and references
herein] the details of experimental setup, steady-state and
time-resolved spectral measurements, direct detection of
intensity and kinetics for singlet oxygen IR-emission as
well as theoretical calculations one may find in these publi-
cations also. Below some necessary information in this re-
spect as well as the corresponding structures will be pre-
sented directly in appropriate places of the text.
Interaction of Zn-porphyrin and Zn-chlorin chemical
dimers with molecular oxygen
Typically, in degassed liquid solutions for monomeric
Zn-porphyrins and Zn-chlorins as well as for chemical di-
mers on their basis the deactivation of excited S1-states
takes place within S0=1.2-1.6 ns,[416,417] and, thus these ex-
cited states are hardly influenced by the presence of oxy-
gen at normal atmospheric pressure at 293 K (see data in
Table 3). It is not the case with long-lived excited T1-states,
and correspondingly, Table 3 collects experimental T val-
ues and bimolecular rate constants kT showing the specifici-
ty of T1-states quenching by molecular oxygen for the stud-
ied dimers and their complexes with pyridine.
For all dimers collected in Table 3, it was shown[416,417]
that at 77-293 K fluorescence and intersystem crossing
quantum yields as well as the rate constants of the internal
conversion S1S0 are hardly dependent on the interactions
between coupled macrocycles. In addition, it follows from
Table 3 that coordination of pyridine to the central Zn ion
in the dimers moiety has not a substantial effect on fluores-
cence quantum yields and lifetimes. It means that additional
non-radiative deactivation processes of S1-states are not
enhanced in pyridinated dimers. In contrast, there are some
noticeable features for triplet states for pyridinated dimers:
(i) phosphorescence lifetimes and quantum yields exhibit a
decrease by 1.5 - 2.5 times compared to uncomplexed di-
mers;[416,417] (ii) an additional T1-state quenching is found
for pyridinated complexes which diminishes upon the tri-
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
254 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
plet level lowering for a set of the investigated dimers,
(ZnOEP)2Ph Zn-31,31-cyclodimer (ZnOEP)2
(ZnHTPP)2 (ZnOEChl)2. In the result, the basic con-
clusion is as follows. The dimers (ZnOEP)2Ph, Zn-31,31-
cyclodimer and (ZnOEP)2 with high-energy triplet states
and relatively long phosphorescence decays Ph are sensi-
tive to the coordination with pyridine leading to enhancing
non-radiative deactivation of T1-states. This quenching is
attributed to several reasons: accepting role of the extra-
ligand vibrations, the enhanced spin-orbit coupling due to
the singlet ,* states energy lowering and the out-of-plane
distortion of porphyrin macrocycle. However, for the di-
mers (ZnHTPP)2 and (ZnOEChl)2 with low-energy triplet
states and relatively short lifetimes Ph the rate constants of
intrinsic non-radiative deactivation processes T1S0 (due
to Frank-Condon factor presumably) are so large that addi-
tional channels of the non-radiative deactivation of T1-
states caused by the extra-ligation seem in comparison not
very effective.
According to[417,418] the process of T1-states quenching
by O2 is realized via non-radiative transitions between elec-
tronic states in an intermediate short-living weak collision
complex formed as a result of a diffusional contact of a
given excited organic molecule with an oxygen molecule.
From this point of view, the quenching of T1-state of the
organic molecule (3M1) may take place via different ways
depending on the spin state of the collision complex:
Δ
dis
kg
1
0
1
dis
k
g
1
2
1
0
1
1
2
3
1
3
1
diff
k
1
g
2
3
1
3kΔMΔO...M~~~O...MOM
, (I)
TS
dis
k2
3
0
1
dis
k
2
3
0
1
3
2
3
1
3
3
diff
k
3
g
2
3
1
3kOMO...M~~~O...MOM
. (II)
Table 3. Lifetimes of S1- and T1-states and bimolecular rate constants for quenching of excited T1-states by molecular oxygen O2 found
for monomeric precursors and chemical dimers in toluene (TOL) and toluene+pyridine (TOL+PYR) at 293 К. For clarity, the structures of
the compounds under study are presented also, central Zn ions are not shown.
Compound
Structure
Solvent
S, ns
T,
ns
kT, a)
109 M-1s-1
kT(lig)/ kT b)
ZnOEP-CH3
TOL
TOL+PYR
1.6
1.45
330
285
1.65
1.95
1.18
(ZnOEP)2
TOL
TOL+PYR
1.3
1.2
440
205
1.2
1.9
1.6
ZnOEP-Ph
TOL
TOL+PYR
1.5
1.4
335
370
1.6
1.5
0.94
(ZnOEP)2Ph
TOL
TOL+PYR
1.2
1.1
495
490
1.1
1.1
1.0
ZnHTPP
TOL
TOL+PYR
1.6
1.5
570
530
1.0
1.05
1.05
(ZnHTPP)2
TOL
TOL+PYR
1.5
1.4
590
500
0.95
1.05
1.10
ZnOEChl-CH3
TOL
TOL+PYR
1.3
1.25
290
265
1.9
2.1
1.10
(ZnOEChl)2
TOL
TOL+PYR
1.2
1.2
350
285
1.6
2.0
1.25
ZnOEP-
cycle=CH2
TOL
TOL+PYR
2.6
2.5
310
275
1.9
2.0
1.05
Zn-31,51-cyclo-
dimer
TOL
TOL+PYR
2.5
2.5
355
295
1.6
1.9
1.2
a) The corresponding values of bimolecular rate constants of the excited S1-and T1-states quenching by molecular oxygen were calculated
using oxygen solubility in methylcohexane and toluene (TOL) at 293 K and the following expression kTS = [(T,S)-1 - [(0T,S)-1]/[O2], where
T and S are the corresponding decays of T1- and S1-states of the given compound in the presence of molecular oxygen, 0T and 0S are
decays in degassed solutions, [O2] is the concentration of dissolved molecular oxygen at 293 K.
b) kT(lig) is the rate constant of triplet state quenching for extra-ligated compound in toluene+pyridine (TOL+PYR) solution.
.
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 255
Here, kdiff is a diffusion rate constant, kST is a non-
radiative intersystem crossing rate constant, and k is a the
singlet oxygen generation rate constant. g1=1/9 and g3=1/3
are spin-statistical factors reflecting the rate constants of
the formation of the complex states 1[3M1...3O2] and
3[3M1...3O2], correspondingly; kdis is the rate constant of the
complex dissociation. In the case of porphyrins and chlorins
kdiskS where kS is the bimolecular rate constant of S1-state
quenching by O2.[418] From the considerations presented
in[417,418] it follows that at the same quenching mechanism
(exchange and donor-acceptor interactions) the quenching
scheme may differ. Scheme (I) with generation of singlet
oxygen (1O2 or 1g) corresponds to the case when the ratio
= kT/kS = 1/9, while scheme (II) without 1O2 generation is
operative at = kT/kS = 1/3.
It follows from the data collected in Table 3 that in
toluene at 293 K, all non-pyridinated dimers exhibit a no-
ticeable decrease (by 15-30%) of bimolecular rate constants
kT compared to with those known for the corresponding
precursor monomers with the same side substituents. The
reason of that is connected with the fact[74,415] that in chemi-
cal dimers containing Zn-porphyrin subunits coupled by the
phenyl spacer, the non-radiative exchange triplet-triplet
energy transfer realizes at distances RDA=12.6-17.0 Å with
rate constants of kET5106 s-1, while rate constants of T1-
state quenching by dissolved O2 for porphyrin monomers
are of kq= kT[O2] 3106 s-1 in these conditions. Corre-
spondingly, the T-T energy transfer between porphyrin
halves in the dimers competes with quenching of T1-excited
macrocycle by molecular oxygen leading to the relative
decrease of the experimental bimolecular rate constants kT
for the dimers with respect to those found for monomeric
precursors at the same conditions.
It is seen also from Table 3 that kT values for the di-
mers increase sequentially in the following set: (ZnOEP)2Ph,
(ZnHTPP)2 (ZnOEP)2 Zn-31,31-cyclodimer, (ZnOEChl)2.
It is known [415,416,418] that T1-state quenching by O2 is an
electron donor-acceptor by nature [M+…O2] and thus cor-
relates with one electron oxidation potentials (Eox1/2) of por-
phyrins and chlorins. According to[419] Eox1/2 values (meas-
ured vs SCE) for Zn-meso-phenylporphyrins are higher
than Eox1/2 =0.63 V for ZnOEP, and values Eox1/2 =0.20 V for
ZnOEChl and Zn-cyclopentaneporphyrins are lower than
that for ZnOEP. Therefore, the observed sequential increase
of kT values in the presented set of the dimers reflects quali-
tatively the corresponding Eox1/2 values lowering. Finally,
the most pronounced oxygen-induced T1-state quenching
(by 60%, Table 3) is found for pyridinated dimer (ZnOEP)2
with a flexible -CH2-CH2- bridge. There are two factors that
come into play here. Firstly, electron-donating ethane
bridge in the dimer may strengthen the pyridine action in
oxygen-induced T1-state quenching (effect of Eox1/2 de-
crease). Secondly, it is known[415,416] that pyridine-
coordinated dimer (ZnOEP)2 is in the opened fully stag-
gered conformation. It means that the cross-section of the
dimer-oxygen interaction becomes larger thus leading to the
increase of the experimentally detected quenching.
Finally, based on results obtained in[415,416] one may
conclude that quantum efficiencies of singlet oxygen 1Δg
generation for both monomeric precursors and chemical
dimers of Zn porphyrins coincide practically (within exper-
imental errors) with the corresponding values of quantum
efficiencies γТ of the non-radiative intersystem crossing
S1>T1. It means that for Zn-porphyrin dimers, T1-states
quenching by oxygen O2 is realized according to scheme (I)
followed by the formation of singlet oxygen 1Δg molecule.
Interaction of porphyrin triads and pentad with molecular
oxygen
The formation of self-assembling porphyrin triads and
pentads (based on a “key-hole” principle) was realized via
two-fold extra-ligation of both central Zn ions of Zn-
porphyrin chemical dimers with two nitrogens of meso-
pyridyl rings in the corresponding di- and tetra-pyridyl-
containing porphyrin free bases.[74,420-422] The structures of
monomeric and dimeric precursors as well as optimized
geometries of multiporphyrin arrays under study are pre-
sented in Figure 23. For these multiporphyrin arrays with
known morphology, the main pathways and mechanisms of
the primary photoinduced relaxation processes were evalu-
ated using steady-state and time-resolved lumines-
cence/absorption measurements and theoretical analysis,
and are properly described in our earlier publications.[420-422]
Based on this results, photophysical events leading to the
formation of the final excited states of the triad or pentad
subunits are the following. The fast non-radiative relaxation
of the dimer S1-state (within 1.6 ps) is caused by two com-
peting processes: (i) the resonance energy S-S transfer di-
merextra-ligand, and (ii) the photoinduced electron trans-
fer from the dimer to the extra-ligand leading to the singlet
radical ion pair state formation. It means that the popula-
tion of the dimer T1-state via intersystem crossing
S1(1dimer*)T1(3dimer*) with a rate constant of kISC(5-
7)107s-1 is low probable. In its turn, because of charge
transfer events, the extra-ligand T1-state in triads and pen-
tads cannot be populated via simple S1T1 process in
ligand subunit. The population of the locally excited T1-
state of porphyrin extra-ligands in triads and pentads
(3H2P*) takes place from the upper-lying triplet radical ion
pair state 3(dimer+...H2P-) or directly from the singlet radical
ion pair state 1(dimer+...H2P-). Concluding, after any excita-
tion of the triad or pentad the final lowest locally excited
state is the extra-ligand T1-state. Namely this state should
be taken into account upon examination of the interaction
of triads and pentad with O2 in liquid solutions.
The obtained experimental data are collected in Table 4.
It is seen that at 293 K in degassed solutions, fluorescence
quantum yields F0 and lifetimes S0 do practically not differ
for all pyridyl substituted porphyrin free base derivatives
(exta-ligands). In these conditions T1-state decays are prac-
tically the same (T0= 1.2-1.4 ms) for individual extra-
ligands, triads and pentad. It was shown also[423] that in tri-
ads and pentads containing dimers (ZnHTPP)2 and
(ZnTPP)2, the locally excited T1-state (ET=1.44 eV) of ex-
tra-ligands lies essentially lower than the CT state. There-
fore, in these multiporphyrin arrays the upper lying CT-
state does not influence the deactivation of the locally ex-
cited T1-state of the extra-ligand.
In the presence of oxygen, both S1- and T1-states
quenching is observed for individual pyridyl substituted
porphyrin free bases and those including in triads and pen-
tad. Notably, Table 4 shows that for all extra-ligands and
multiporphyrin arrays under consideration the ratio
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
256 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
= kT/kS is within 0.11-0.15. It corresponds to the spin-
statistical factor of g=1/9 reflecting the situation[417,418]
where triplet states of the compounds are quenched by tri-
plet molecular oxygen, 3O2 (see Scheme 73).
The role of structural organisation of triads and pen-
tads is most readily understood upon their interaction with
molecular oxygen in liquid solutions at 293 K. The com-
parative analysis of data summarised in Table 4 shows that
both kS and kT values for extra-ligands in the triads and
pentad are smaller than those obtained for the correspond-
ing individual pyridyl substituted porphyrins. Therefore, it
is reasonable to connect the relative decrease of kS and kT
values upon transition from monomeric extra-ligands to
those including in the triads and pentad with screening
effects depending on the mutual arrangement of subunits
in multiporphyrin complexes. In fact, the Zn-porphyrin
dimer in the triad (without any populated excited states)
plays a screening role limiting the access of oxygen mole-
cule to the excited extra-ligand. For the “opened” triad
(ZnHTPP)2H2P-(p^Pyr)2 (see Figure 23) this screening
effect is minimal, and the measured decrease of kT value
for this triads is minimal with respect to that obtained for
individual H2P-(p^Pyr)2. In contrast, for “closed” triads,
(ZnHTPP)2H2P-(m^Pyr)2 and (ZnHTPP)2H2P-(m-Pyr)2,
the access of oxygen molecule to the excited extra-ligand
becomes more limited thus resulting in more pronounced
decrease of the observed kS and kT values in comparison
with those found for the corresponding monomers. Con-
sistent with this interpretation is the fact that for pentad
2(ZnOEP)2PhH2P-(m-Pyr)4 where screening effects due
two dimers (ZnOEP)2 are higher with respect to those in
triads, the relative decrease of kT values is maximal in com-
parison with the triads at the same experimental conditions
(see Table 4).
Finally, quantum yields of singlet oxygen generation
by triad (= 0.80) and pentad (=0.70) are high in toluene
and comparable with those known for monomeric free ba-
ses (= 0.68 - 0.73[418]). In fact, for triads or pentads it
means that the quenching of extra-ligands excited states by
O2 takes place with the same efficiency practically like for
monomers, but with smaller bimolecular constants because
of screening effects. In fact, the observation of singlet oxy-
gen generation by triads and pentads in liquid solutions at
room temperature serves the independent direct proof of the
existence of locally excited triplet states in these arrays.
Figure 23. A: Structures of chemical dimers with phenyl spacer, (ZnOEP)2Ph, (ZnHTPP)2, and pyridyl-containing monomeric porphy-
rinsH2P(m^Pyr)2-(iso-PrPh)2, H2P(p^Pyr)2(Ph)2 and H2P(m-Pyr)4 used as extra-ligands upon formation of triads and pentad. The corres-
ponding extra-ligands were used: i) porphyrin, H2P containing two meso-phenyls and two meso-pyridyl rings with different positions of
both pyridyl rings (adjacent) and pyridyl nitrogens N (meta and para): i) adjacent meta-pyridyl rings - H2P(m^Pyr)2-(iso-PrPh)2 and adja-
cent para-pyridyl rings - H2P(p^Pyr)2(Ph)2; ii) porphyrin, H2P containing four meso-pyridyl rings with meta-nitrogens. B: Mutual ar-
rangement of the dimers, (ZnHTPP)2, (ZnOEP)2Ph and various extra-ligands in triads and pentad (optimized geometry, HyperChem soft-
ware package, release 4, semiempirical methods AM1 and PM3). For simplicity, mesophenyl rings in (ZnHTPP)2 are omitted. Mark
denotes what porphyrin subunits are included in self-assembled triads and pentad.
H2P-(m^Pyr)2: R1,R2= m-Pyr, R3,R4=iso-PrPh H2P-(p^Pyr)2 : R1,R2= p-Pyr, R3,R4=Ph H2P-(m-Pyr)4 : R1,R2,R3,R4= m-Pyr
A
R
R
R
R
R
R
R
R
R
R
R
R
R
R
R
R
Zn Zn
N
N
N
N
N
N
N
N
Zn Zn
N
N
N
N
N
N
N
N
R'
R'
R' R'
R'
R'
H2P-(m^Pyr)2
H2P-(p^Pyr)2
H2P-(m-Pyr)4
(ZnOEP)2Ph R=C2H5
(ZnHTPP)2 R´ = HexPh:
B
(ZnHTPP)2 Н2Р(m^Pyr)2 (ZnHTPP)2 Н2Р(p^Pyr)2 2(ZnOEP)2PhН2Р(m-Pyr)4
(ZnOEP)2Ph Н2Р(m^Pyr)2
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 257
Table 4. Photophysical parameters and bimolecular rate constants of excited S1- and T1-states quenching by molecular oxygen for
pyridyl-substituted porphyrin free bases, triads and pentad in methylcyclohexane (MCH) and toluene (TOL) at 293K
Compound
Solvent
S, ns
S0, ns
kS, 10-9
M-1s-1
F
F0
T, ns
kT,10-9
M-1s-1
=kT/kS
T (77K),
ms
H2P-(m^Pyr)2
MCH
8.9
11.4
9.3
0.070
0.104
275
1.40
0.14
4.2
H2P-(p^Pyr)2
MCH
9.1
12.8
12.0
0.064
0.098
280
1.37
0.11
4.2
H2P-(m-Pyr)4
MCH
8.4
10.5
9.3
0.071
0.098
260
1.45
0.15
4.1
(ZnHTPP)2 H2P-(p^Pyr)2
MCH
8.0
10.4
11.3
-
-
340
1.15
0.10
4.1
(ZnHTPP)2 H2P-(m^Pyr)2
MCH
8.2
10.3
9.6
-
-
365
1.05
0.12
3.1
H2P-(m^Pyr)2
TOL
9.5
-
-
0.075
-
380
1.45
-
-
(ZnOEP)2Ph H2P-(m^Pyr)2
TOL
7.7
-
-
0.05
-
550
1.0
-
-
2(ZnOEP)2Ph H2P-(m-
Pyr)4
TOL
-
-
-
0.04
-
680
0.80
-
-
S, F and S0, F0 are fluorescence decays and quantum yields in the presence and absence of O2 in solution, correspondingly; kS and kT
are bimolecular rate constants of the excited S1-and T1-states quenching by molecular oxygen (for definition see Table 3); T (77K) is T1-
states decay in rigid solutions at 77 K. Mark denotes which porphyrin subunits are included in self-assembled triads and pentad.
Figure 24. Structural properties of nanoassemblies based on semiconductor CdSe/ZnS QDs capped with n-trioctylphosphine oxide
(TOPO)and H2P(m-Pyr)4 molecules. A: Schematic presentation of QD-porphyrin nanoassemblies. B: An optimized geometry for Cd33Se33
+ H2P(m^Pyr)2 (B, optimization by HyperChem 7.0; simulations by ab initio density functional theory, DFT, with the VASP code[424]). C:
the scales of CdSe core, ZnS shell, porphyrin and TOPO molecules corresponding to relative sizes of the main components of the arrays.
Interaction of “Quantum Dot – Porphyrin” nanoassemblies
with molecular oxygen
Like for multiporphyrin complexes described in the
previous section, a controllable formation of nanoassem-
blies based on semiconductor quantum dots (QD) and por-
phyrins was realized using also a “key-hole” principle upon
quantitative titration experiments in toluene at ambient
conditions.[74,75,79,80] In this case, “QD-Porphyrin” nanoas-
semblies with various porphyrin/QD ratio were successfully
prepared upon two-point coordination interactions of two
nitrogens of meso-pyridyl rings in the tetra-pyridyl-
containing H2P(m-Pyr)4 or H2P(p-Pyr)4 porphyrin free ba-
ses, and surface Zn ions of colloidal core/shell CdSe/ZnS
QDs passivated by tri-n-octyl phosphine oxide (TOPO)
(Figure 24).
From the photochemical point of view, the generation
of singlet oxygen by “QD-porphyrin” nanoassemblies
should involve two steps. First, after excitation of QD the
energy transfer QDporphyrin should be organized result-
ing in S1-state formation followed by the intersystem cross-
ing into porphyrin T1-state via the intersystem crossing
S1T1 pathway. Second, for porphyrin acting as PS,
the diffusional controlled triplet-triplet energy transfer
PS(T1)oxygen(3O2) should take place resulting in 1O2
generation in the surrounding media. This section provides
a description of the obtained basic results and a discussion
of all necessary steps what should be realized upon quanti-
tative analysis of QD PL quenching processes in “QD-
porphyrin” nanoassemblies and efficiencies of the singlet
oxygen generation.
Typically, the formation and “QD-porphyrin” nanoas-
semblies manifests itself in a pronounced quenching of QD
photoluminescence (PL).[74,79,80,424] Correspondingly, for
nanoassemblies consisting of TOPO-capped CdSe/ZnS
QDs (dCdSe = 3.0 nm, 2 ZnS monolayers, CQD = 410−7 M)
and H2P(m-Pyr)4 molecules in toluene, the direct quantita-
tive comparison of QD PL quenching results and sensitiza-
tion data for porphyrin fluorescence was carried out using a
complete set of titration points. Our results showed[79,80,424]
that in “QD-porphyrin” nanoassemblies under consideration,
Foerster resonance energy transfer (FRET) QDporphyrin
B
A
C
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
258 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
occurs with low quantum efficiency of FRET =0.10-0.15,
while the main part of the observed QD PL quenching is
due to non-FRET nature (non-FRET =0.85-0.90). We quanti-
tatively argued for the first time that a mechanism clearly
distinct from the photoinduced charge transfer and FRET
may be responsible for the QD PL quenching in QD-
porphyrin nanocomposites upon the coordinative attach-
ment of the extra-ligand on QD surface.[74,425] This process
is correlated with the extension of the exciton wave func-
tion beyond the interface between the QD and the attached
porphyrin molecule. Based on comparison of experimental
data and quantum mechanical calculations it follows that
the specificity of the exciton non-radiative decay in QD-
porphyrin nanoassemblies (that is QD PL non-FRET
quenching) is connected with the charge tunneling through
ZnS barrier in the conditions of quantum confinement.
Keeping in mind these results, a comparative study-
ing the quantum yields of the singlet oxygen generation
by alone QDs and “CdSe/ZnS QD-porphyrin” nanoassem-
blies was performed using the direct spectral-kinetic meas-
urements of 1O2 near-IR emission.[79,80,424] It follows from
these experiments (Figure 25B) that at ambient temperature
most of alone QDs are hardly perspective to the direct
generation of singlet oxygen (the quantum efficiency
γΔ =1.5%), and without attached organic PSs cannot be
used in clinical practice.
Figure 25 evidently shows that upon laser excitation at
the same experimental conditions and concentrations in air-
saturated toluene, “QD-porphyrin” nanoassemblies do in-
deed produce singlet oxygen with essentially higher effi-
ciency in comparison with alone QDs or even with alone
H2P(m-Pyr)4 molecules. The radiative relaxation of 1О2 de-
tected at reg=1270 nm is characterized by the same decay
value of (1g) = 30-31 s that is typical for 1О2 emission in
non-polar liquid solutions.[415,416] It seems to be reasonable
to connect this findings with FRET QDporphyrin in
nanoassemblies. Thus, for clear understanding the role of
FRET namely in the increase of the quantum yields of 1O2
generation by nanoassemblies one should take into account the
role of the excitation conditions and then carry out the quanti
tative analysis of the whole experimental data.
Typically, photoluminescence measurements are car-
ried out at lower excitation energies with respect to experi-
ments with essentially higher laser pulse energies being
used for 1О2 generation, and this thing was taken into ac-
count. From Fig. 25C it is evidently seen that the PL inten-
sity for QDs in “QD-porphyrin” nanoassemblies (curve 1,
em=558 nm) and for alone CdSe/ZnS QDs is not a linear
function of the laser pulse energy. In the case of alone por-
phyrin solutions, the above dependence is linear practically
(not shown), while in contrast, this dependence for porphy-
rin counterpart in the QD-porphyrin” nanoassembly is
non-linear (curve 2, em=720 nm). The linear dependence
for alone porphyrin solutions in the whole range of nano-
second laser energies is explained by the minor role of the
singlet-singlet and triplet-triplet annihilation processes at
low solute concentrations[426] (C=10-5-10-6 M in the given
case). Non-linear dependence obtained for alone QDs and
QDs in “QD-porphyrin” nanoassemblies reflects the well-
documented pump-intensity dependence of the amplitudes
of the single-exciton caused by non-radiative intraband Au-
ger processes mediated by Coulomb electron-electron inter-
actions in the conditions of the spatial confinement.[427] In
this respect, the non-linear dependence of fluorescence meas-
ured for porphyrin being attached to QD surface (Figure 25C,
curve 2, em=720 nm) presents itself a direct proof of the
realization of namely FRET process QDporphyrin com-
peting with both radiative electron-hole recombination and
non-radiative Auger process in the conditions of powerful
excitation.
The above results evidently show that the relative in-
crease of the efficiency of 1О2 generation by “QD-
porphyrin” nanoassemblies with respect to that for alone
QDs coincides with the observed sensitized increase of the
fluorescence intensity for the attached porphyrins caused by
FRET. Therefore, understanding and ultimately control-
ling the energy of laser excitation we have carried out
the quantitative estimation of FRET efficiency in the
nanoassemblies based on singlet oxygen generation ex-
periments presumably.[79]
Figure 25. Singlet oxygen detection upon laser excitation of ZnS/CdSe QDs and “QD-porphyrin” (1:4) nanoassemblies in air-saturated
toluene. A: 3D presentation of spectrum of singlet oxygen 1О2 emission. B: Comparative presentation of singlet oxygen 1О2 luminescence
decay sensitized by individual CdSe/ZnS QDs (1) with respect to that measured for nanoassemblies (2) at the same molar concentrations
of QDs and laser excitation conditions (exc=532 nm, pulse duration t1/2= 0.7 ns, reg=1270 nm). C: Dependencies of the normalized
emission intensities on the energy of laser excitation for CdSe/ZnS QDs (1, em=558 nm) and H2P(m-Pyr)4 molecules (2, em=720 nm)
being coupled in nanoassemblies.
050000 100000 150000
1000
2000
3000
4000
5000
1O2 Emission Intensity, rel. units
2
Time, ns
1
B
0 5 10 15 20
0,0
0,2
0,4
0,6
0,8
1,0 em = 720 nm
C em = 558 nm
Energy/pulse, J
Normal counts
1
0 5 10 15 20
0,0
0,2
0,4
0,6
0,8
1,0
2
A
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 259
Scheme 73.
Our findings have shown that within the experimental
accuracy the for these nanoassemblies, the values of FRET
efficiencies obtained via the direct 1О2 emission measure-
ments at low laser excitation (FRET = 0.12 0.03), are in
a good agreement with FRET efficiencies found from the
direct sensitization data for porphyrin fluorescence (FRET =
= 0.14 0.02, discussed above). Such quantitative analysis
was done for the first time and shows that namely FRET
process QDporphyrin is a reason of singlet oxygen gen-
eration in the given nanoassemblies according to the fol-
lowing Scheme 73.
To our knowledge, such an approach is a first example
showing quantitatively the role of FRET effects in the sin-
glet oxygen generation by QD PS nanoassemblies. The
obtained results may be considered as a direct proof of the
realization of namely FRET QDporphyrin process fol-
lowed by the singlet oxygen generation via porphyrin triplet
states. This conclusion has been proven also for the same
nanoassemblies using two-photon excitation.[428] All these
facts raise the conclusion that “CdSe/ZnS QD-porphyrin”
nanoassemblies may be considered as PSs of a novel type
for PDT. However, nanoassemblies based on TOPO capped
CdSe/ZnS QDs and porphyrins are not soluble in water and
contain toxic elements, what limits their biological applica-
tions. Recently, we examined water-soluble glutathione
capped AgInS/ZnS QDs with electrostatically attached Zn-
porphyrins and showed that upon UV excitation, these
nanoassemblies are capable to generate 1g (via FRET
QDporphyrin in ps time scale).[88] These preliminary re-
sults together with a specific dependence of spectral-kinetic
parameters of AgInS /ZnS QDs on pH and local polarity,
studied by us also,[429] make these nanoassemblies more
perspective in various biomedical applications (drug delivery
carriers, the distant testing the local pH, PDT treatment, etc.).
Finally, taking into account that in a lot of investiga-
tions the direct experimental measurements of the singlet
oxygen (1О2 or 1Δg) emission at max = 1.27 are realized in
various solvents, one should pay attention to some specific
effects which may change the rate constant of the radiative
transition 1g 3Σ-g in 1О2 molecule. Luminescence of sin-
glet oxygen 1Δg in solvents is difficult to observe[430,431]
because of fast quenching of 1О2 molecules by solvent vi-
brations, and the rate constant of the radiative transition
1g 3Σ-g in 1О2 molecule is strongly dependent on the
solvent properties (e.g. refractive index and polarizabil-
ity)[432-434] including interface boundaries[435] and biological
dielectrical nanoobjects.[436] Correspondingly, it is im-
portant to know all physico-chemical factors determining
the singlet oxygen 1Δg lifetime (or the rate constant of the
radiative transition 1g 3Σ-g in 1О2 molecule) in various
environments including systems in vivo. In[437] radiative rate
constants kr of the radiative transition 1g 3Σ-g have been
determined for the photoluminescence of singlet oxygen
1O2 in solutions with regularly changed refractive indexes
using laser kinetic spectroscopy. The observed changes in
the radiative rate constants kr are shown to be caused both
by inherent properties of the emitting 1O2 molecule and by
characteristics of an external environment, which defines
the local field factor and the density of photon states of the
field. In addition, the dielectric dipole moment of the transi-
tion 1g 3Σ-g increases significantly as a result of the
contact of 1O2 molecule with solvent molecules.
Nevertheless, in spite of numerous experimental stud-
ies in this direction during last decade, some peculiarities of
electronic mechanisms of 1О2 luminescence decay remain
still under the question.[438] Recently,[439] the molecular
modeling of the influence of 10 solvents on the radiative
rate constant kr was carried out. The data obtained indicate
that the correlation of kr values with molecular polarizabil-
ity that occurs in a number of cases is associated, on the one
hand, with its effect on the strength of dispersion interac-
tions in the complex, and, on the other hand, with the fact
that, to certain extent, it reflects the position of the upper
occupied orbitals of the solvent molecule. Both factors af-
fect the degree of mixing of -orbitals of 1O2 molecule,
which promotes the activation of the radiative transition
1g 3Σ-g . Only taking into account all the above factors
one may give a consistent explanation what happens with
the intensity of 1O2 in various solvents.
Conclusions
The interaction of molecular oxygen O2 with various
multicomponent complexes and nanoassemblies containing
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
260 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
tetrapyrrolic compounds was quantitatively analyzed using
set of steady-state and time-resolved experimental data ob-
tained in liquid solutions at ambient temperature. It was
found that for Zn-porphyrin and Zn-chlorin chemical di-
mers, the extra-ligation of central Zn ions of the porphyrin
macrocycles by pyridine leads to the enhancement of the
non-radiative T1S0 intersystem crossing rate constant
(the most pronounced for the dimers with higher lying T1-
states). This effect is attributed to several reasons: accepting
role of the extra-ligand vibrations, the enhanced spin-orbit
coupling due to the singlet ,* states energy lowering and
the out-of-plane distortion of porphyrin macrocycle. For
pyridinated dimers, their T1-states quenching by molecular
oxygen depends on the spacer flexibility and donor-
acceptor interactions with pyridine.
In the case of self-assembled triads and pentads con-
taining Zn-porphyrin dimers and pyridyl substituted por-
phyrin free bases (extra-ligand), the cooperative existence
of energy and electron transfer processes results in the pop-
ulation of the final locally excited S1- and T1-states of por-
phyrin extra-ligands. The quenching of these states in triads
and pentads by O2 is noticeably weakened with respect to
that for individual pyridyl substituted porphyrins, and is
caused by the screening action of a strongly quenched Zn-
porphyrin dimer subunit in triads and pentads limiting the
access of oxygen molecule to the excited extra-ligand.
These facts have to be taken into account upon the analysis
of natural dark and photoinduced reactions of porphyrin
supramolecular systems with molecular oxygen (oxygena-
tion and deoxygenation in haemoglobin, singlet oxygen
generation in tissues and PDT of cancer).
For inorganic-organic nanoassemblies based on
TOPO-capped semiconductor quantum dots CdSe/ZnS and
tetra-pyridyl porphyrins and formed via a two-fold coordi-
nation Zn….N-pyr “key-hole” principle in toluene at ambi-
ent conditions, the photoluminescence quenching for QD
with superficially attached porphyrin molecules is caused
by two main competitive reasons: the electron tunneling in
the conditions of quantum confinement (the efficiency of
0.85-0.90) as well FRET QDporphyrin (the efficiency of
0.10-0.15). On the basis of near-IR photoluminescence de-
tection of singlet oxygen 1O2 emission, it was proven that
the experimental efficiencies of 1O2 generation by “QD-
porphyrin nanoassemblies are essentially higher than
those obtained for alone QDs. It was found that in “QD-
porphyrin” nanoassemblies (1:4), FRET efficiencies
FRET = 0.12 0.03 obtained via the direct 1О2 emission
measurements at low laser excitation, are in a good coinci-
dence with FRET efficiencies FRET = 0.14 0.02 evaluat-
ed from the direct sensitization data for porphyrin fluores-
cence in nanoassemblies. It means that namely FRET pro-
cess QDporphyrin is a reason of singlet oxygen genera-
tion by QD-porphyrin nanoassemblies. Taking into account
experimental data and structure of excitonic states for QDs,
it was concluded that at ambient temperature most of alone
QDs are hardly perspective to the direct generation of sin-
glet oxygen. On the other hand, “QD-porphyrin” or “QD-
chlorin” nanoassemblies may be considered as good candi-
dates for ‘‘see and treat’’ PDT or for oxygen sensing over
physiological oxygen ranges.
Concluding, dealing with the direct quantitative meas-
urements of the singlet oxygen 1Δg emission (max = 1.27 )
in various solvents, one should take into account changes in
the rate constants kr of the radiative transition 1g 3-g in
1Δg molecule which may be caused as discussed in[417-419]
by inherent properties of the emitting singlet oxygen mole-
cule and by characteristics of an external environment (na-
ture of solvent).
10. Hydrophilic-Lipophilic Balance
and Interaction of Chlorin Type Photosensitizers
with a Transport Proteins of Blood
The trend towards the search of new effective photo-
sensitizers, best meeting the requirements applied to drugs
is of current interest for decades and up to the present
time.[6,10,440448] The ability to generate ROS is one of the
most important characteristics of potential PS designed for
PDT. However, the singlet oxygen as a highly reactive oxi-
dative form has a very short lifetime (≤ 40 ns) limiting its abil-
ity to diffuse inside biological tissues (20~40 nm).[6,12,448] Ac-
cordingly, pharmacodynamics of PS and, namely, mecha-
nisms of its biodistribution in the human body including
intracellular localization, play a crucial part in of PDT effi-
cacy increase.[6,448-451] These biological features are fairly
determined by the structure and hydrophilic-lipophilic bal-
ance (HLB) of the PS molecule, or, in fact, by the pigment
affinity to a cell membrane and ability to penetrate it.[444, 452-
454] In this regard, design of a new antitumor drug for PDT
should involve an estimation of the affinity and accumula-
tion selectivity of PS in malignized or pathogenic cells, as
well as the control of specificity of passive transport of
the drug in vivo and its localization on the target cell
sites.[84,455,456]
Our scientific group is involved in extensive and con-
tinuing series of multidisciplinary studies on the devel-
opment and investigation of new and already known po-
tential sensitizers for antitumor and antimicrobial
PDT[6,9,11,13,75,444,451,453,457-463] based on the porphyrin, chlorin
and some other macroheterocyclic platforms. Our experi-
mental efforts in this field are aimed at finding the ways to
improve efficacy of PDT in terms of pharmacodynamics
and selective biodistribution of potential PSs. Current work
is a brief of literature and own recently obtained results,
related to the influence of the structural features and am-
phiphilicity of the chlorin photosensitizers on the mecha-
nisms of these drugs distribution and transport by proteins
of human blood.
The main emphasis on PSs of chlorophyll-like nature
is made here due to several factors. Naturally derived chlo-
rins have an intense absorption (lgε = 4.5-4.6) in a long-
wavelength part of visible spectrum (~665 nm), a sufficient
quantum yield of singlet oxygen,[6,10-13,373,448] a good am-
phiphilicity[453,454] due to asymmetric location of polar and
non-polar substituents in the molecule and a susceptibility
of macrocycles to chemical modification.[6,373,464,465] Some
chlorin-based drugs like Foscan, Talaporfin, Radachlorin,
Photoditazine or Photoran e6 are currently used in clinical
oncology as antitumor PSs.[6,9,10]
Hydrophilic-lipophilic balance of naturally derived chlorin
photosensitizers
The balance of hydrophilic and lipophilic properties of
PS plays an important part both at the stage of drug admin-
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 261
istration in its dosage form when the covalent or non-
covalent binding of PS to a different type of nanocarriers is
often used[84,451,455-460] and during its delivery to the surface
of the target cell.[6,448,465] The interaction of photosensitizer
with a cellular constituents starts from the main primary
target membrane of the atypical cell.[466] The generation
of ROS by irradiated cell-localized PSs results in oxidation
of membrane lipids and proteins. The important chemical
targets for photoactivated PS are double bonds of mem-
brane unsaturated phospholipids.[10,467] The accumulation of
lipid hydroperoxides and oxidased forms of cholesterol in
membrane is a trigger for the lateral phase separation of
lipids, perturbation of their functions and, as a result, a de-
crease of membrane permeability and cell death.[468-470] Alt-
hough the mechanism of oxidative transformation of lipid
membranes during PDT remains still poorly established[466]
the most of authors are inclined to state a key role of 1O2,
formed at the photochemical reaction of PS with molecular
oxygen, in the oxidation of unsaturated membrane phos-
pholipids.[467] However, regardless of the type of photooxi-
dation mechanism,[10,75,373,447] the efficacy of a sensitizer in
the deactivation of a malignant or pathogenic cells depends
on the proximity of molecular contact between the PS
and the membrane lipid.[469] Therefore optimized PS mol-
ecule have to possess a certain HLB, to be well soluble in
water or aqueous solutions of carriers,[84,455-457] but not to be
too hydrophilic and not weaken its affinity and transfer
through lipid membranes of cells.[84,444,454]
The human body can be represented as a set of lipid-
like membrane barriers divided by water-like environ-
ment.[26,28,30] The P coefficient[3,452-454,469-475] reflecting parti-
tion of the substance between two immiscible phases is the
main parameter for the comparative quantitative estimation
of the hydrophilic-lipophilic balance (HLB) of any potential
drug.[146,452,471,472] P value of the later give an important in-
formation about its ability to interact and penetrate cell
membranes and, furthermore, about drug bioavailability and
biodistribution pathways. The results of experimental de-
termination of P values for tetrapyrrole photosensitizers and
their theoretical evaluation obtained by various computa-
tional methods are widely presented in the literature.[3,473,474]
Usually the hydrophobic core of lipid membrane is
modeled by 1-octanol (OctOH) and phosphate buffered
saline (PBS, pH 7.4) is used to mimic water-like media for
simulating transfer through the surface of lipid membranes
and experimental evaluation of the hydrophilic-lipophilic
balance of the drug.[452,454,461,462] To calculate partition coef-
ficients (P) the isothermal saturation method is normally
applied. This quantity is determined from the ratio of the
equilibrium concentrations of PS in the lipid-like phase and
in the PBS according to equation (1):
, (1)
where Cm OctOH and Cm aq are equilibrium solute molal con-
centrations in 1-octanol and aqueous phase, respectively.
Our experiments to determine P values were carried
out in thermostatically controlled 50 ml glass cells filled
with a PS solution with the initial concentration within 20 -
90 µmol/kg in a phosphate saline buffer or OctOH. Then
the second component of the immiscible solvent system
was added in a volume ratio of OctOH and PBS equal
40:60. After 36 hours of magnetic stirring at 298 K the so-
lution was kept until complete phase separation before the
probe was taken from the lipid-like layer. The equilibrium
concentration of PS was analyzed by the spectrophotomet-
ric method using previously obtained calibration graphs.
The solute concentration in the aqueous phase was estimat-
ed as the difference between the initial and lipid-like phase
equilibrium concentration of the pigment. Values of the
partition coefficients calculated according to the eq. (1) are
presented in the Table 5.
All the chlorin PSs investigated (compounds 2-9) were
synthesized by chemical modification of methylpheophor-
bide a (1).[451,454,476-478] Methylpheophorbide a and clinically
used photosensitizing agents as Fotoditazin” (8) and
Fotoran e6” were purchased from companies “CHLORIN”,
“VETA-GRANT LLC” and “RANFARMA”, respectively.
Trisodium salt of chlorin e6 (9) was obtained by precipita-
tion of “Fotoran” from aqueous solution of HCl Н ~ 6)
followed by solid powder dissolution in diluted NaOH solu-
tion (рН ~ 8) and further complete water evaporation.[451]
All the compounds were identified using 1H NMR spectros-
copy and mass spectrometry technics.
The analysis of the previously measured at 298 K and
newly obtained partition coefficients of chlorin PSs at the
interface of OctOH and PBS clearly demonstrate these
quantities differentiate strongly depending on the substitu-
tion pattern of the macroheterocyclic molecule. Thus, a
metal-free chlorophyll a appeared to be the most hydropho-
bic natural chlorin studied (P = 326.0[453]) with a highly
pronounced affinity to the lipid surrounding. Transesterifi-
cation of this PS with the replacement of a hydrophobic
phytyl tail with a methyl group (methylpheophorbide a, 1)
leads to a rather significant decrease in the P value down to
210.1.[444,454,476] An additional structural factor contributing
to the chlorin hydrophilic properties is the opening of the
cyclopentanone ring accompanied by the transformation of
phorbine (1) to chlorin (2-9) molecular structure.
Table 5. Partition coefficients (P, 298K) for compounds 19 in the 1-octanol/phosphate saline buffer biphasic system and PSs distribution
between transport proteins of human blood (%).
Photosensitizers
1
2
3
4
5
6
7
8
9
Partition in OctOH PBS system
P
210.1±
6.0 [444]
29.3 ± 0.9
[454]
20.1 ± 0.9
[451]
8.7 ± 0.2
[476]
1.04 ± 0.2
[476]
0.97 ± 0.3
[476]
19.0 [56]
2.03 ±
0.21 [9]
1.88 ±
0.09 [9]
Proteins
Serum proteins binding, %
LDL
-
52 [479]
45
53
NB
NB
38 [479]
2
1
HDL
-
31 [479]
53
40
NB
NB
44 [479]
4
5
Albumin
-
17 [479]
2
7
NB
NB
18 [479]
94
94
NB no binding
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
262 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
Figure 26. Molecular structures of PSs studied: 1 Pheophor-
bide a 17(3)-methyl ester, 2 Chlorin e6 13(1)-N-methylamide-
17(3)-(1’-(2’-hydrohymethyl-3’,8’-dioxy-quinoxalinyl)methyl
ester)-15(2)-methyl ester, 3 Chlorin e6 13(1)-N-methylamide-
15(2)-methyl ester-17(3)-O-6'-galactopyranosyl ester, 4 Chlo-
rin e6 13(1)-N-(2-N’,N’,N’- trimethylammonioethyl iodide) amide-
15(2),17(3)-dimethyl ester, 5 Chlorin e6 3(1),3(2)-bis(N,N,N-
trimethylaminomethyl iodide)-13(1)-N’-methylamide-15(2),17(3)-
dimethyl ester, 6 Chlorin e6 3(1),3(2)-bis(N,N,N-trimethylamino-
methyl iodide)-13(1)-N′-(2-N″,N″,N″-trimethylammonioethyl io-
dide) amide-15(2),17(3)-dimethyl ester, 7 Chlorin e6
13(1),15(2)-dimethyl ester, 8 Chlorin e6 dimeglumine salt, 9
Chlorin e6 trisodium salt.
For instance, at the disclosure of phorbine exo-cycle in
the molecule of macrocyclic PS conjugate with diethylene
glycol an intense decrease of the partition coefficient from
77.7 to 32.6 is observed at 298K.[444]
Any conjugation of one or higher number of hydro-
philic groups to a chlorin macroheterocycle decreases P
parameter (Table 5). At the same time, the incorparation of
a single non-ionic or zwitterionic substituent, such as rese-
dues of glycol[444] or carbohydrate,[451] N-oxide[454] or amino
acid[451] into the PS structure promotes an amphiphilicity of
the molecule without essential increase in water solubility.
Usually, the naturally derived chlorins carrying one of the
substituents listed above (Table 5, compounds 2, 3),
demonstrate partition coefficients ranged from 15 to 30.
Meantime, chlorin molecules containing one or more ionic
groups (4-6, 8, 9) and characterized by P values less than
10 acquire solubility in water to achieve therapeutic con-
centrations of PS.[6,7,9,10] So, the solubility of dicationic
chlorin (5) reaches to 0.04 mol/kg at 298K. [477] Comparison
of partition coefficients of cationic (5, 6) and anionic (8, 9)
type PSs with two and three charged groups in the OctOH
PBS system in both cases demonstrates the proximity of the
P values within pairs of PSs with a positively and negative-
ly charged groups (Table 5). At the same time, these quanti-
ties are slightly lower for cationic PS 5 and 6 revealing
more hydrophilic character of trimethylammonia groups
(P = 0.97-1.04)[476] as compared to carboxylic ones (P = 1.9-
2.0).[9] The HLB of the monocationic chlorin (4, P = 8.6)
differs significantly from di- and three charged deriva-
tives.[457] Meantime, having moderate amphiphilicity, comp.
4 retains a sufficiently high solubility in water. The mono-
carbaxylate derivative of chlorin e6 (7) is significantly less
hydrophilic (P = 19.0[478]), localized mainly in a lipid-like
phase and almost insoluble in water. An increase in the
number of charged groups in the PS molecule provides the
pronounced enhancement of macrocycle affinity towards a
water-like pool. Nevertheless, even tricationic chlorin e6 (6)
is still equally distributed between lipid-like octanol and
water-like PBS phase[476] due to the bulky non-polar macro-
cyclic part of the molecule.
Hydrophilic chlorin PSs[476] in contrast to more hydro-
phobic ones[444,451,454] demonstrate only modest Arrhenius
dependence already at P < 30. So, for such a compounds
partition coefficients measured at standard temperatures can
be attributed to physiological conditions.[476] It should also
be taken into account the P parameter of chlorin PSs with
an acidic groups depends on pH significantly.[475] In par-
ticular, it is found, that an affinity to a water-like compart-
ment in the case of anionic carboxylic derivatives of chlorin
e6 decreases with pH.[475] This information could be im-
portant for tumor-localized anionic PSs.
Thus, the P value is a reliable quantitative characteris-
tics of the hydrophilic-lipophilic balance of porphyrin and
chlorin-type macroheterocyclic PSs. Knowledge about the
HLB of photosensitizing agents is critically important for
understanding mechanisms of their delivery and pharmaco-
dynamics.[95,449,450,4 9,32] Partition coefficients provide not
only the information about PS affinity towards a water-like
or lipid-like surrounding but also an ability to bind to a dif-
ferent kind of blood proteins determining the mechanisms
of drug biodistribution in organism and, finally, the efficacy
of PDT. That is why in the next section of the paper the
effect of the structure and HLB of chlorin PS on the mech-
anisms of their transport by blood proteins, as well as on
their affinity to the membranes of atypical cells will be ana-
lyzed.
Equilibrium distribution of chlorin PSs between transport
proteins of blood
Extensive material devoted to the study of the interac-
tion of PSs of various structure with blood proteins using a
wide range of spectroscopic, chromatographic and quantum
chemical methods [17,58-63] are accumulated in the literature.
Entering biological tissues the drug participates in the pro-
cess of equilibrium redistribution on the molecules of bio-
environment, primarily on the blood transport proteins
“light” albumins (M = 66.5 kDa) or “heavy” lipoproteins of
low and high densities (LDL and HDL, M = >230 kDa and
~180, respectively).[478] Redistribution occurs regardless of
whether the PS is taken as a pure compound or as molecular
complex of the drug with any non-covalently bound deliv-
ery vehicle.[17,31-33,64]
It is known that the selectivity of PS for tumor cells is
several times higher compared to healthy mammalian
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 263
ones.[6,448] The most of photosensitizers are mainly hydro-
phobic molecules, therefore, in human circulatory system
they bind predominantly to the lipophilic protein fraction
like low-density lipoproteins (LDL).[6,296,483,486,487] Lipopro-
teins play an important role in supplying cells with choles-
terol, which is used to form membranes during cell divi-
sion. Since tumor cells have an abnormal growth rate, they
require a large excess of cholesterol and the number of LDL
receptors in them per surface unit increases significantly
compared to healthy cells.[488] Thus, the binding of PS to
LDL in the bloodstream is one of the main reasons of the
pigments tumor selectivity due to their targeted delivery.[486]
High-density lipoproteins (HDL) and albumins are al-
so used as a transport proteins of human blood.[449] HDL are
able to deliver the drug to the tumor tissue, but, compared
to LDL, they are accumulated in tumor macrophages, which
can negatively influence the efficacy of the same PS.[487] In
turn, albumins have a less pronounced affinity for malig-
nant cells, so PS associated with this type of transport pro-
teins will be accumulated both in target tumor and healthy
mammalian cells, providing less selectivity.
PS molecules of various structures have different
pharmacodynamics and bind to transport proteins in indi-
vidual ways. It can be expected that essentially hydrophilic
PS bind mainly to albumins, the amphiphilic ones interact
with both HDL and albumins and hydrophobic molecules
make a complex selectively with LDL.[449,451,478,489] This
hypothesis is in a good agreement both with a previously
published results and our data (Table 5).[451,479] Thus, three
types of transport proteins of the circulatory system com-
pete for binding with PS molecule, therefore, and the effi-
cacy of the antitumor agent mainly depends on its hydro-
philic-lipophilic balance (Table 5).
There are a number of ways to determine the nature of
PS binding to various types of proteins, including transport
ones. One of the most common is the titration of the PS
solution with a specific protein using spectral methods to
obtain both equilibrium constants and thermodynamic pa-
rameters of the process, as well as structural characteristics
such as binding sites and geometrical features of the com-
plex.[75,490,491] However, in relation to the study of PS inter-
action with transport proteins of blood the spectral approach
has some disadvantages. Albumins are commercially avail-
able and chemically stable proteins, therefore titration using
spectral measurements is the most preferable in this
case.[296,480,481] At the same time, low- and high-density
blood lipoproteins are hardly separable from each other and
because of the low stability require immediate use after the
sample preparation. In addition, the titration using spectro-
scopic methods in the case of LDL and HDL is also of little
use because it requires separate experiments for different
types of proteins and does not allow to work with a mixture
to assess the competitive distribution of PS between
transport proteins. At the same time, information about the
competitive interaction of potential PS with proteins is im-
portant for understanding the real picture of their redistribu-
tion and transport in vivo.
To solve the above problems, the gel filtration chro-
matography (GFC) capable to separate complexes of
transport human serum proteins with PS divided into frac-
tions according to the size of macromolecules is used.
[478,479,482,483,485,489,492] (Figure 27) Spectrophotometric or
fluorescence analysis of fractions gives the gel-filtration
chromatogram or separation curve which is the dependence
of the relative optical density (A/Amax) measured on the
absorption maxima of tryptophan (280 nm, available in all
types of transport proteins), and naturally derived chlorin
PS, absorbing at ~665 nm, from the volume of the solution
(V, mL) eluting from the chromatographic column. Figure 28
demonstrates an example of gel chromatographic separation
on the column (1.670 cm) filled with polyacrylamide gel
Acrilex P-200. Three pronounced peaks of blood proteins
are observed usually on the gel-chromatogram (curves 1,
Figure 28), where the first one corresponds to LDL (with an
admixture of HDL), while the second and third represent
HDL and albumins, respectively. This finding is con-
firmed by the biochemical analysis of the obtained frac-
tions (Figure 27).[478,489] The area under the PS curve on the
gel chromatogram, measured for individual peaks corre-
sponding to specific types of blood proteins, allows to cal-
culate the percentage of pigment in each fraction (Table 5).
25 30 35 40 45 50 55 60 65 70 75 80 85
0.0
0.2
0.4
0.6
0.8
1.0
1.2
1.4
A/Amax
V, ml
1
2
3
Figure 27. Detection of protein components of human blood by biochemical analyzer “COBAS” (left, 1 blood protein sample, 2 low-
density lipoproteins, 3- albumins) and the principle of gel-filtration chromatographic separation of proteins depending on the size of the
molecule (right).
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
264 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
25 30 35 40 45 50 55 60 65 70 75
0.0
0.2
0.4
0.6
0.8
1.0
2
a
A/Amax
V, ml
1
30 35 40 45 50 55 60 65 70 75
0.0
0.2
0.4
0.6
0.8
1.0
A/Amax
V, ml
b
2
1
Figure 28. Gel filtration chromatograms of 70% blood serum (1 mL) with addition of 9∙10-5 mol/kg of PSs 4 (a) or 8 (b): 1 main protein
(absorption at 280 nm), 2 - analyzed PS (665 nm). Column elution rate is 0.2 mL/min, the volume of each collected fraction is 2.5 mL.
As mentioned above, the degree of amphiphilicity af-
fects significantly the biodistribution of drugs, including
PS, using blood transport system.[478,489] As can be seen
from the literature and our data[9,475,478,479] (Table 5), hydro-
philic chlorin molecules 8 and 9 containing two and three
anionic groups, respectively, are accumulated to a greater
extent in the albumin structure (94%). An increase in the
partition coefficients P and affinity of PS to a lipid-like
compartments, on the contrary, leads to preferential binding
of drug molecule to a less polar lipoprotein fractions. So,
more hydrophobic compound 7 (P = 19.0 56), containing the
only carboxyl group is about equally distributed between
both LDL and HDL (Table 5) with a total binding to lipo-
proteins of 82%. Amphiphilic conjugates of chlorin e6 with
an antimicrobial drug “Dioxidine” (2) and D-galactose (3)
behave in approximately the same way due to a relatively
similar HLB (P = 20 30). Binding of the compound 3 to
lipoproteins is overwhelming and reaches 98% (Table 5).
Based on close P values and HLB of di- and tri-
anionic (8, 9) and di- and tricationic PSs (5, 6) an identical
picture of blood proteins binding could be expected for
them. However, it was found that molecules 5 and 6 carry-
ing two and three cationic trialkylammonia groups, respec-
tively, do not demonstrate interaction to any types of
transport proteins during GFC. The reason of that is the
most important characteristic affecting the equilibrium dis-
tribution of PS across transport proteins is not only the
number, but also the sign of the charged substituents pre-
sented in the molecule. As known from the literature[478] the
at the pigment interaction with albumin, the nonpolar mac-
rocyclic core is embedded into the hydrophobic pocket of
the transport protein surrounded by positively charged
groups.[478] Therefore, anionic PSs strongly bind to albu-
mins, whereas cationic pigments such as compounds 4, 5 or
6, are repulsed electrostatically from these transport mole-
cules. At the same time, the moderate hydrophobicity of
monocationic chlorin 4 (P = 8.3) allows it to form stable
complex with blood lipoproteins (in total 93%, see Table 5).
Mainly hydrophilic (P ~ 1) PSs 5 and 6 are not able to be
transported by lipid transport proteins due to its high affini-
ty for the water-like phase.
Thus, based on the fact that LDL is the most preferred
PS transport agent, it can be assumed that cationic pigments
with an appropriate HLB estimated by partition coefficient
of about 10, such as the compound 4, should demonstrate a
good efficacy as photosensitizing agents in antitumor PDT.
In addition, due to the amphiphilic structure and the pres-
ence of a cationic group in the molecule PS 4 is more versa-
tile compared to neutral or anionic chlorins, since it can be
used both in antitumor and antimicrobial PDT. [3,5,6,75,447,476]
Amphiphilic PSs with non-ionic or zwitterionic func-
tional groups (2, 3) interact effectively with LDL and HDL
(Table 5), however, provide negligible solubility in water
and pronounced aggregation in aqueous solutions. The later
factor affects negatively their ability to generate ROS and
reduces PS bioavailability.[10-13,373,447] The application of
micellar, polymer and other types of nanocarriers for PS
binding and solubilization is an effective way to achieve its
therapeutic concentrations in aqueous solutions.[84,471]
The results presented above allow us to draw the fol-
lowing important conclusions:
1. The structural features of photosensitizing mole-
cules designed for PDT determine their polarity, am-
phiphilic properties and significantly affect the biodistribu-
tion of pigments, mechanisms of their transport in the sys-
tem bloodstream and the selectivity of accumulation in the
membranes of malignazed or pathogenic cells.
2. PS distribution coefficients reflecting the state of
its hydrophilic-lipophilic balance are correlated with the
type of their distribution between blood transport proteins.
Thus, highly hydrophilic anionic-type chlorins give a stable
complex with blood albumins, whereas a decrease in the
affinity of PS to the aqueous phase leads to the predominant
binding to lipoprotein fractions of the blood. Cationic PSs
carrying two or more charged groups do not interact with
any type of transport proteins.
3. Gel filtration chromatography is a reliable method
of primary evaluation of the mechanism of PS biodistribu-
tion between proteins of the circulatory system.
Chlorin-type monocationic pigments with moderate
HLB (P ~10) are the most promising photosensitizing
agents, which, on the one hand, maintain the solubility in
water, and, on the other hand, are transported mainly by
lipoproteins in the system bloodstream of human. Thus,
due to the number and sign of the charges PS 4 demon-
strates higher versatility compared to neutral or anionic
chlorins and can be applied both in antitumor and antimi-
crobial PDT.
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 265
11. Sulfur-Containing Derivatives
of Natural Bacteriochlorophyll a as Promising
Photosensitizers for PDT of Cancer
The development of antitumor drugs is one of the
most important areas in the modern medicinal chemistry.[1]
The World Health Organization (WHO) defines cancer as a
broad group of pathological processes that can begin in
almost any organ or tissue and can be accompanied by un-
controlled division of abnormal cells. Despite the variety of
existing therapeutic agents, all of them have a number of
drawbacks, including non-selectivity of action, high toxici-
ty, and resistance of tumors to these agents. Since oncologi-
cal diseases continue to threaten the people’s health and
quality of life around the world, there is a need for innova-
tive targeted therapeutics aiming at specific targets in tumor
cells in order to implement a personalized approach in on-
cology.[2]
The development of sulfur-containing pharmaceutical
substances plays an important role in the development of
medicinal chemistry. There are more than 300 sulfur-
containing drugs approved by the US Food and Drug Ad-
ministration (FDA), the most popular of which include sul-
fonamides, thioethers, sulfones, penicillin, etc.[493] Photo-
sensitizers that acquire new useful properties upon incorpo-
ration of sulfur-containing groups or individual sulfur at-
oms into their structure also attract attention.
One of the most desirable groups of PS includes natu-
ral bacteriochlorins with absorption maxima in the near-
infrared spectral range, which expands the capabilities of
treating deep-seated tumors.[494] Moreover, they have a rela-
tively high yield of singlet oxygen and low toxicity along
with increased tropism to tumor tissues.[106,495]
The glutathione system is one of the key protective
cell systems affecting the efficiency of its photoinduced
death.[496] This system is vitally important for the existence
of mammalian cells, including tumor ones. It is known that
the level of enzymes in this system is elevated in various
types of malignant neoplasms.[497] The thiol-containing
tripeptide glutathione (γ-glutamylcysteinylglycine, GSH)
plays an important role in the detoxification of xenobiotics,
free radical control, maintenance of the cell redox balance,
formation of multiple drug resistance (MDR), etc.[498] This
makes it promising to aim pharmacological compounds at
the glutathione system as a biological target in order to
damage the enzymes of this system, which would favor the
vulnerability of tumor cells to oxidative stress.[499]
Two main enzymes are responsible for the biosynthe-
sis of glutathione in a cell: first, γ-glutamylcysteine ligase
(GCL, EC 6.3.2.2), which catalyzes the reaction between
glutamate and cysteine, followed by the addition of a gly-
cine residue to the resulting dipeptide with involvement of
glutathione synthetase (GSS, EC 6.3.2.3).[500,501] Cellular
glutathione participates in binding to various xenobiotics
with involvement of enzymes of the glutathione S-
transferase family, being an important regulator of intracel-
lular metabolism.[502] Glutathione binding to reactive oxy-
gen species (ROS), which is necessary for the protection
against oxidative stress and regulation of the cell redox bal-
ance, is catalyzed by GST or by specific enzymes of the
glutathione peroxidase family (GPx, EC 1.11.1.9).[503] Upon
interaction with ROS, glutathione is converted to an oxi-
dized dimeric form (GSSG), which, under the action of
glutathione reductase (GR, EC 1.8.1.7), is reduced into the
GSH monomer, thus closing the redox cycle.[504,505]
An important metabolic pathway that uses glutathione
for detoxification of metabolites is the glyoxalase system
consisting of glyoxalase I (GloI, EC 4.4.1.5) and glyoxalase
II enzymes (GloII, EC 3.1.2.6).[506-508] This system catalyzes
the conversion of methylglyoxal, a highly toxic glycolysis
product, to D-lactate via the S-D-lactoylglutathione inter-
mediate. The energy metabolism of the majority of tumor
cells is based on the Warburg effect, i.e., aerobic glycolysis
where glucose is converted to lactate in the presence of
oxygen. This process in tumor cells is characterized by
much higher activity (up to 200-fold) in comparison with
normal cells.[509,510] Such a rate of glycolytic transfor-
mations results in a higher formation rate of glycolysis side
products, including methylglyoxal (MG), a cytotoxic me-
tabolite. Therefore, tumor cells are more sensitive to MG
compared to healthy cells, and the glyoxalase system is a
promising target for anticancer agents.
A number of inhibitors of glutathione system enzymes
are currently known that make it possible to control its
function at different stages of the metabolic pathway. GCL
is the most promising target for the disruption of glutathi-
one biosynthesis in the course of anti-tumor therapy by hin-
dering and disruption of oncogenesis.[500] The best known
inhibitors of γ-glutamylcysteine ligase include methionine
and buthionine sulfoximines (MSO and BSO, respectively).
In vitro and in vivo studies have shown the prospects of
using MSO and BSO in combination with the known
chemotherapeutic agents.[511] Moreover, clinical studies
showed the safety of BSO administered intravenously.[512]
The inhibition of the glyoxalase system mainly in-
volves the inhibition of the rate-limiting enzyme GloI.
The best known GloI inhibitors include S-conjugated
glutathione derivatives such as S-alkylglutathiones, S-p-
bromobenzylglutathione, etc.[513] It was shown in in vitro
studies that S-p-bromobenzylglutathione made it possible to
overcome multiple drug resistance (MDR) and recover the
sensitivity of tumor cells to therapy.[514] Subsequent studies
have shown that tumor cell lines with high GloI expression
are resistant to current clinical antitumor drugs but sensitive
to GloI inhibitors.[515,516]
One of the promising ways for increasing the PDT ef-
ficiency is to create combined-action drugs comprising a
photosensitizer and a chemotherapeutic agent.[92,517] Exam-
ples are known where the chemotherapeutic agent in a
combined drug manifests pro-oxidant action that includes
the inhibition of the tumor cell defense system against oxi-
dative stress.[518,519]
The purpose of this work was to synthesize and study
the photoinduced cytotoxicity of photosensitizers based on
natural bacteriochlorins with sulfur-containing molecules
capable of affecting the glutathione and glyoxalase systems
for photodynamic efficiency improvement. All experi-
mental details are presented in Supporting Information.
The sulfur-containing proteinogenic amino acids cys-
teine and cystine are among the key compounds involved in
the redox processes in malignant cells. They undergo re-
versible enzymatic reactions with glutathione, thus weaken-
ing the antioxidant defense of tumor cells. The use of sul-
fur-containing amino acids in the development of antitumor
drugs may shift the redox equilibrium in a tumor cell to-
ward the generation of oxidative stress, which increases the
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
266 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
PDT efficiency.[524,525] Unlike cysteine and cystine, methio-
nine has a methylated thiol group that cannot directly inter-
act with the molecules of the cell’s glutathione antioxidant
system. However, some tumor cell lines are characterized
by enhanced consumption of this amino acid, a phenome-
non called “methionine dependence”.[626] Therefore, creat-
ing a conjugate with this amino acid would increase the
tropism of the entire molecule to tumor tissues. Moreover,
methionine derivatives, such as L-methionine- and L-
butyonine-sulfoximine, are selective inhibitors of the gluta-
thione antioxidant system, including γ-glutamylcysteine
synthase. Based on all of the above, it is obvious that creat-
ing bacteriochlorin conjugates with sulfur-containing amino
acids (cysteine, cystine, methionine and its derivatives) may
enhance the photoinduced effect of antitumor therapy due
to the action on the redox system of malignancies and en-
hanced tropism to the latter.
Previously we reported a synthesis of derivatives of
O-propyloxime-N-propoxybacteriopurinimide with cysteine
(2a), cystine (2b), methionine (3), methioninesulfoximine
(4), and buthioninesulfoximine (5),[520,521] that involved
addition of the above aminoacids and their derivatives to
the carboxyl group of the propionic residue at macrocycle
position 17 (Scheme 74).
At the first stage of this work, we optimized the condi-
tions for the preparation of DPBP conjugates with esters of
sulfur-containing amino acids presented in Scheme 74. The
reaction yield was taken as the optimization criterion. To
increase it, various agents activating the carboxyl group
were studied (Table 6).
Table 6. Reaction conditions in the synthesis of DPBP conjugates
with sulfur-containing derivatives (all in inert argon atmosphere
during 15 min).
Compound
Reaction conditions, i
Yield, %
DPBP-Cys (2a)
EEDQ, CH2Cl2, 25oC
27
NHS, CH2Cl2, 0oC
19
DCC, CH2Cl2, 0oC
17
EDC, CH2Cl2, 0oC
25
DCC, NHS, CH2Cl2, 0oC
21
EDC, NHS, CH2Cl2, 0oC
41
DPBP-Cys-Cys-
DPBP (2b)
EEDQ, CH2Cl2, 25oC
33
EDC, CH2Cl2, 0oC
28
EDC, NHS, CH2Cl2, 0oC
56
DPBP-Met (3)
EEDQ, CH2Cl2, 25oC
29
EDC, CH2Cl2, 0oC
28
EDC, NHS, CH2Cl2, 0oC
46
DPBP-MSO (4)
EEDQ, CH2Cl2, 25oC
31
EDC, CH2Cl2, 0oC
28
EDC, NHS, CH2Cl2, 0oC
48
DPBP-BSO (5)
EEDQ, CH2Cl2, 25oC
24
EDC, CH2Cl2, 0oC
27
EDC, NHS, CH2Cl2, 0oC
50
Scheme 74. Reagents and conditions: i: provided in the Table 6; ii: NH2-R, CH2Cl2, inert argon atmosphere, 25oC, 48 h.
Figure 29. A study photoinduced activity of compounds 1, 2a,b, 3 in vitro on HeLa tumor cell line. All compounds were used as Kolliphor ELP
4% micellar solutions: A. Exposure in the presence of PS in the incubation medium. The time of incubation with photosensitizers before exposure
to light was 4 h. The power density was 21.0 ± 1.0 mW/cm2 and the calculated light dose was 10 J/cm2; B. Exposure without PS in the incubation
medium. The time of incubation with photosensitizers before exposure to light was 4 h. The power density was 21.0 ± 1.0 mW/cm2 and the calcu-
lated light dose was 10 J/cm2; C. Dark cytotoxicity.
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 267
The combination of 1-ethyl-3-(3-dimethylaminop-
ropyl)carbodiimide (EDC) and N-hydroxysuccinimide (NHS)
as the activating agents was found to be the best. They were
considerably superior to N-еthoxycarbonyl-2-ethoxy-1,2-
dihydroquinoline (EEDQ) and 1,3-dicyclohexylcarbodiimide
(DCC) and allowed the reaction yield to be increased by a
factor of 1.5-2. Due to the high solubility of EDC in water,
multiple extractions can be avoided, while addition of NHS
produces a stable activated derivative of O-propyloxime-N-
propoxybacteriopurpurinimide with N-succinimide.
In the previous model experiments, we observed a re-
versible transition of the free thiol group to the oxidized
disulfide form.[520] Therefore, in this work we were the first
to estimate the in vitro antitumor photodynamic efficacy of
DPBP-Cys (2a), DPBP-Cys-Cys-DPBP (2b), and DPBP-
Met (3) derivatives that can affect the glutathione system of
tumor cells.
Study of photo-induced antitumor activity of DPBP-Cys (2a),
DPBP-Cys-Cys-DPBP (2b), and DPBP-Met (3) in vitro
The HeLa cell line with elevated levels of intracellular
glutathione was selected for in vitro biological tests.[497] The
time of incubation with compounds before light exposure
was 4 h, while the incubation time of cells after irradiation
was 24 h. The cells were irradiated with a standard light
dose of 10 J/cm2 with an exposure time of 13 min. The
starting compound, O-propyloxime-N-propoxybacteriopur-
purinimide (1), was chosen as the reference compound. The
survival of tumor cells was assessed both visually using an
inverted microscope and by the MTT test. The results of the
latter are shown in Figure 29.
The half-maximum inhibition concentration (IC50) of
compounds (2a) and (3) after 4 hours of cell incubation was
2-3 times smaller than that of the reference compound O-
propyloxime-N-propoxybacteriopurinimide (1), both under
exposure to light in the presence of PS in the incubation
medium (Figure 29A) and under exposure to light after PS
removal from the incubation medium (Figure 29B). The
IC50 of the dimeric compound (2b) was slightly larger and
comparable to that of the original compound (1). The dark
cytotoxicity of compounds (2a) and (2b) was slightly high-
er than that of derivative (3) (Figure 29C).
Since the photoinduced IC50 of DPBP-Cys-Cys-DPBP
(2b) was higher than that of cysteine (2a) and methionine
(3), only DPBP-Cys (2a) and DPBP-Met (3) were selected
for in vivo biological studies.
Study of the photoinduced antitumor activity of DPBP-Cys
(2a), DPBP-Met (3) in vivo
The accumulation kinetics of DPBP-Cys (2a) and
DPBP-Met (3) in a tumor compared to healthy tissues was
studied on a tumor model of mouse sarcoma S37 using lo-
cal electron spectroscopy. After intravenous injection, both
drugs showed accumulation of the compounds of interest
in the tumor with the maximum accumulation at 180 and
270 min, respectively (Figure 30). The fluorescence con-
trast in the accumulation maxima was 3.09 and 5.86 for
DPBP-Cys (2a) and DPBP-Met (3), respectively.
The data obtained indicate that accumulation of
DPBP-Cys (2a) and DPBP-Met (3) in the tissues of malig-
nant neoplasms occurs, which, in combination with the re-
stricted area of light exposure, provides an efficient tool for
tumor targeting. Moreover, the maximum accumulation
times of the compounds lie in the range convenient for the
photodynamic therapy procedure.
At the next step, the photoinduced antitumor activity
of DPBP-Cys (2a) and DPBP-Met (3) was studied in vivo
using various irradiation modes. The dose of both photo-
sensitizers was 5 mg/kg; DPBP (1) whose accumulation
kinetics was studied previously was taken as the reference
drug.[527] To select the optimal light dose, two irradiation
modes with light doses of 150 J/cm2 and 300 J/cm2 at a ra-
diation power density of 0.2 W/cm2 were used. The PDT
procedure was performed at the time points when the max-
imum accumulation of drugs took place (15 min, 180 min,
and 270 min after intravenous injection of DPBP (1),
DPBP-Cys (2a), and DPBP-Met (3), respectively).
Analysis of the growth dynamics of the primary tumor
node showed that the highest antitumor effect was shown
by the DPBP-Met compound 3 (Figure 31). The optimal
PDT conditions for this compound are: a photosensitizer
dose of 5 mg/kg, an interval of 270 min between its injec-
tion and the beginning of irradiation, and a light dose of
300 J/cm2.
Figure 30. Accumulation kinetics of compounds 2a (A) and 3 (B). The content of compounds in the samples was determined in vivo by
local electron spectroscopy on sarcoma S37 tumor-bearing mice. The ratio of areas under the curves of fluorescence and laser reflection
peaks was estimated.
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
268 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
Figure 31. The growth dynamics of the primary tumor node in irradiation modes of 300 J/cm2 (A) and 150 J/cm2 (B) and at a photosensi-
tizer dose of 5 mg/kg. The animals were monitored for 15 days after the PDT procedure. The anti-tumor effect was estimated from the
dynamics of primary tumor growth in the experimental and control groups.
Figure 32. Tumor growth inhibition in experimental and comparison groups in irradiation modes of 300 J/cm2 (A) and 150 J/cm2 (B) and
at a photosensitizer dose of 5 mg/kg. The animals were monitored for 15 days after the PDT procedure.
Using this PDT mode with DPBP-Met (3), complete
resorption of the tumor node was achieved on day 4 after
the treatment. The figure shows the tumor growth inhibi-
tion (TGI) data in two irradiation modes, 150 J/cm2 (A)
and 300 J/cm2 (B) (Figure 32).
Thus, the in vivo studies on the biological properties
of DPBP-Cys (2a) and DPBP-Met (3) indicated that the
methionine methyl ester derivative (DPBP-Met (3)) has a
two times higher selectivity of accumulation in the tumor
tissue than the cysteine derivative 2a. Moreover, the pho-
toinduced antitumor activity of the DPBP-Met derivative
(3) at a light dose of 300 J/cm2 was higher than that of
DPBP (1) and DPBP-Cys (2a).
Synthesis of DPBP conjugates with sulfur-containing inhib-
itors of enzymes of the glutathione and glyoxalase systems
Inhibitors of γ-glutamylcysteine synthase and glyoxa-
lase I were used as sulfur-containing molecules capable of
affecting the glutathione and glyoxalase systems and pos-
sessing antitumor activity. Inhibition of these enzymes
should increase the efficiency of photoinduced antitumor
therapy and reduce dark toxicity through the intracellular
targeted impact on tumor cells.
Methionine and buthionine sulfoximines were chosen
as potent γ-glutamylcysteine synthase inhibitors. Their con-
jugation with DPBP (1) was performed as described in our
previously studies and resulted in conjugates DPBP-MSO
(4) and DPBP-BSO (5).
S-Hexylglutathione dimethyl ester (8) was chosen as the
glyoxalase I inhibitor. The esters of this class of compounds
are known to have greater antitumor activity than the ana-
logues with free carboxyl groups.[528] Initial attempts to
obtain S-hexylglutathione dimethyl ether (8) by a conven-
tional technique in acid medium failed to give the expected
result, which seems to be due to the formation of a side
product of S-hexylglutathione cyclization. Therefore, dime-
thyl ether (8) was obtained using diazomethane (Scheme 75).
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 269
Scheme 75. Synthesis of S-hexylglutathione dimethyl ether (8). Reagents and conditions: i: 1-iodohexane, 1M aqueous NaOH, EtOH, inert argon
atmosphere, 25°C; ii: CH2N2, Et2O.
Scheme 76. Reagents and conditions: i: compound 8, EDC, NHS,
CH2Cl2, inert argon atmosphere, 25oC.
At the next step, S-hexylglutathione dimethyl ether
was added to the free carboxyl group of O-propyloxime-N-
propoxybacteriopurinimide (1) using the optimized reaction
conditions shown in Table 6 (Scheme 76).
The spectral characteristics of the pigment thus ob-
tained did not differ from those of the original DPBP (1).
The absorption maximum in the long-wave spectrum region
for all the sulfur-containing conjugates (2-5, 9) was 800 nm.
Study of the photoinduced antitumor activity of DPBP-GS-
Hex, DPBP-MSO and DPBP-BSO in vitro
At the final stage of this work, the photoinduced cyto-
toxicity of all the resulting conjugates of O-propyloxime-N-
propoxybacteriopurinimide with sulfur-containing amino
acids (2-5) and with S-hexylglutathione dimethyl ether (9)
was studied in vitro on the Hela tumor cell line. The time of
incubation with compounds before light exposure was 4 h,
while the incubation time after irradiation was 24 h. Cells
were irradiated with a standard light dose of 10 J/cm2, the
exposure time was 13 min. The starting compound, O-
propyloxime-N-propoxybacteriopurpurinimide (1), was
chosen for comparison. Tumor cell survival was estimated
both visually using an inverted microscope and by the MTT
test whose results are presented in Table 7.
The highest photoinduced cytotoxicity in vitro was
observed for compounds DPBP-MSO (4), DPBP-BSO (5),
and DPBP-GS-Hex (9) than have 5-6 times lower IC50 than
O-propyloxime-N-propoxybacteriopurinimide (1) and 2-
3 times lower IC50 than the conjugates with cysteine (2a)
and methionine (3), both upon exposure to light in the pres-
ence of PS in the incubation medium and upon exposure to
light followed by removal of PS from the incubation medi-
um. The lowest cytotoxicity without light exposure (dark
cytotoxicity) was shown by DPBP-GS-Hex (9).
In conclusion, DPBP derivatives with sulfur-
containing amino acids were obtained previously in our
works.[520,521] In this work, we have optimized the reac-
tion conditions for the amidation of O-propyloxime-N-
propoxybacteriopurinimide with sulfur-containing amino
acids and modified glutathione, which allowed us to increase
the yield of the target derivatives by a factor of 1.5-2.
Table 7. Photoinduced cytotoxicity in vitro of sulfur-containing compounds on HeLa tumor cells.
Compound
Exposure in the presence of PS
in the incubation medium
Exposure with subsequent
removing PS from in the incu-
bation medium
Dark cytotoxicity
IC50 value, nM
DPBP (1)
111 ± 8
129 ± 24
2141 ± 763
DPBP-Cys (2a)
50 ± 2
62 ± 6
1260 ± 72
DPBP-Cys-Cys-DPBP (2b)
104 ± 10
117 ± 8
1355 ± 136
DPBP-Met (3)
44 ± 0.4
46 ± 2
785 ± 63
DPBP-MSO (4)[]
16 ± 1
20 ± 5
232±27
DPBP-BSO (5)[]
20 ± 0.7
21 ± 2
485±63
DPBP-GS-Hex (9)
24 ± 4
27 ± 3
2042±981
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
270 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
The in vivo biological studies of solubilized DPBP
conjugates with methyl esters of cysteine 2a and methio-
nine 3 on tumor-bearing mice with subcutaneously inocu-
lated sarcoma S37 were performed for the first time. Methi-
onine derivative 3 was found to have a 1.9-fold higher se-
lectivity of accumulation in tumor tissue than cysteine de-
rivative 2a. Moreover, the photoinduced antitumor activity
of compound 3 under the conditions used (5 mg/kg, light
dose 300 J/cm2) was found to be higher than that of the refer-
ence compound DPBP 1 and cysteine-based pigment 2a.
A new derivative 9 of natural bacteriochlorophyll a
with an inhibitor of the glutathione-dependent enzyme gly-
oxalase I, was obtained.
All the sulfur-containing bacteriochlorin conjugates
obtained were studied in vitro on Hela tumor cells, and their
photoinduced cytotoxicity was shown to increase in com-
parison with the parent compound 1. This pattern shows a
beneficial role of sulfur-containing compounds in the mani-
festation of photodynamic activity and suggests that the
development of such PS for antitumor PDT is a promising
approach.
12. Antitumour Efficacy of Photodynamic Therapy
of Experimental Laboratory Animal Tumours
with a New Photosensitizer of Chlorin Series
Most photosensitizers used for photodynamic therapy
in clinical practice are derivatives of chlorin e6. To date,
research aimed at increasing the anti-tumour efficiency of
PS continues. It is known that only high-purity chlorin e6
has the most pronounced photodynamic properties, and the
presence of related chlorine and the other ingredients in the
dosage form significantly impairs the properties of the pho-
tosensitizer. Concomitant chlorines have a much lower
quantum output of singlet oxygen and less accumulation in
tumour tissues. This requires the application of a higher
light dose and leads to blurring of the clear boundary during
the PDT session and, as a result, significant damage to
healthy tissues adjacent to the tumour node.[529] The pres-
ence of polyvinylpyrrolidone in the composition of the me-
dicinal form of the photosensitizer leads to increased vis-
cosity of the injectable solutions and makes it impossible to
apply a rapid and convenient method of jet administration,
replacing it with only drip intravenous administration. Thus
the presence of impurities reduces the effectiveness of chlo-
rine PSs. As a result, the improvement of PS is now on
course to achieve the highest possible purity of chlorin e6 in
a sustainable form.
The new photosensitizer Heliochlorin, developed by
Professor G.V. Ponomarev and co-authors,[70] is available in
a lyophilically dried form, including trismeglumin salt of
chlorin e6 and meglumin as a cryostabilizer. Heliochlorin is
a PS based on highly purified chlorin e6 from methyl pheo-
pharbide a, with a high core content of 93-98%.
The study of the antitumor efficacy of PDT with the
Heliochlorin on the models of murine Erlich carcinoma and
rat sarcoma M-1 showed that treatment at the optimal time
after intravenous administration of PS with certain parame-
ters of laser exposure allowed to achieve complete regres-
sion of tumour nodes and no recurrence within 90 days after
therapy in 100% of animals.
The animal experiments were conducted in a strict ac-
cordance with the Guidelines for the Care and Use of Labora-
tory Animals of the National Medical Research Radiologi-
cal Centre of the Ministry of Health of the Russian Federa-
tion and in accordance with the rules and requirements of
the European Convention ETS/STE No. 123 and interna-
tional standard GLP (OECD Guide 1:1998).
Table 8. Experiment Design.
Tumour
Test system
Group
PS dose, mg/kg
Mode of
administration
Laser action parameters
E, J/cm2
Ps, W/cm2
Ehrlich’s
carcinoma
mice
1 group-PDT
2.5
intravenously
150
0.48
1 control
-
-
-
-
2 group-PDT
5.0
intravenously
150
0.48
2 control
-
-
-
-
Sarcoma M-1
rats
3 group-PDT
2.5
intravenously
150
0.48
3 control
-
-
-
-
4 group-PDT
5.0
intravenously
150
0.48
4 control
-
-
-
-
This study was performed using sarcoma M-1 bearing
outbred female rats (totally 32 animals) at three months of
age weighting on average 180-200 g and 48 female outbred
mice two months old, 19-20 g weight with implanted Erlich
carcinoma. The animals were housed in T-3, T-4 cages un-
der the natural light conditions with the forced ventilation
of 16 times/h, at a room temperature and relative humidity
of 5070%. The animals had free access to water and PK-
120-1 feed for rodents (Laboratorsnab Ltd., Moscow, Rus-
sia). The tumour strains was obtained from the tumor bank
of the N.N. Blokhin National Medical Research Center of
Oncology of the Ministry of Health of Russian Federation.
Sarcoma M-1 was implanted subcutaneously as a 1.0 mm3
piece of donor tumour into the outer side of the left thigh.
The experiment was started 8-9 days after transplantation,
when the largest diameter of the tumour nodes reached 0.8-
1.0 cm. Erlich’s ascitic carcinoma liquid with 2.5 × 106 tumour
cells from the donor mice was injected subcutaneously into
the area of the lateral surface of the left thigh of mice. The
experiment was started 4 days after transplantation, when
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 271
the largest diameter of the tumour nodes reached 0.4-0.6 cm.
The experimental and control groups containing eight rats
and twelve mice each. As the series with the different
treatment regimens were not performed simultaneously,
each series had its own control. As a control, we studied
tumour-bearing animals without any exposure. The session
of photodynamic therapy was performed, as described pre-
viously.[530] The animals of the experimental groups re-
ceived the PS in doses of 2.5 and 5.0 mg/kg into the caudal
vein. The tumours were exposed to the laser irradiation
using an Atkus-2 semiconductor device (Atkus, Russia;
λ = 662 nm, diameter of the light spot = 1 cm for mice and
1.5 cm for rats). The following doses of laser irradiation
were tested: E = 150 J/cm2; Ps = 0.48 W/cm2 (Table 8). In
our preliminary studies the optimal time of beginning of
laser action after administration of the drug (drug-light in-
terval) was determined: 45-60 min after intravenous admin-
istration of PS to mice and 60-75 min to rats.[531] The results
were analyzed in the seventh, fourteenth, twenty-first and
ninetieth days and compiled in Table 9. The antitumor effi-
cacy of the PDT was assessed according to the criteria de-
scribed earlier.[532,533]
After the PDT, there was a pronounced swelling of
soft tissue in the irradiation zone, which is a natural reac-
tion to photodynamic tumor damage with any known PS.
The swelling was accompanied by hyperemia of the radia-
tion zone and surrounding soft tissue. Tissue alteration was
observed and, depending on the size of the surface to be
irradiated, a thick scab of dark color was formed for 2-4 days
after treatment.
Up to 21 days after photodynamic exposure (with a PS
dose of 5.0 mg/kg and laser exposure parameters:
E=150 J/cm2; Ps=0.48 W/cm2) an inhibition effect was ob-
served on murine Erlich carcinoma in all experimental ani-
mals, and remained up to 3 months in 100% of animals.
When the PS dose was reduced to 2.5 mg/kg at the same
light dose, the complete tumour regression was observed to
21 days. Later on, some animals relapsed into tumour for
90 days after therapy, however, the percentage of cured
animals was 66.7% (Table 9) and 33.3% of mice with re-
lapse tumors increased their life expectancy by 45% com-
pared to control (Table 10). All the control groups men-
tioned above demonstrate continuous tumor growth (see
Table 9).
After PDT of rat sarcoma M-1 with the following pa-
rameters: the PS doses 2.5 and 5.0 mg/kg and dose of light:
E=150 J/cm2; Ps=0.48 W/cm2 up to 90 days after therapy total
regression of tumor in 100% of animals was observed (Table. 9).
Table 9. Anti-tumour efficacy of murine Erlich carcinoma and rat sarcoma M-1 PDT with intravenous administration of Heliochlorin
(laser action parameters E = 150 J/cm2; Ps = 0.48 W/cm2).
No. gr.
PS dose,
mg/kg
Observation period, days of
7
14
21
90
Erlich’s carcinoma of mice
1
2.5
(2 and 3) 100%
(4) 67%
2
5.0
(2 and 3) 100%
(4) 100%
Control 1+2
(1) 0.09±0,02
(1) 0.94±0,16
(1) 4.05±2.43
-
Sarcoma M-1 rats
3
2.5
(2 and 3) 100%
(4) 100%
4
5.0
(2 and 3) 100%
(4) 100%
Control 3+4
(1) 2.7±0.5
(1) 10.2±2.1
(1) 17.1±4.2
-
Note: (1) Tumor volume (V, cm3); (2) Tumor growth inhibition (TGI, %); (3) Percentage of animals with full tumor regression (R); (4)
Percentage of completely cured animals.
Table 10. The life expectancy of animals (days) and an increase in
the mean life span of animals (%) after PDT compared to control.
PS dose, mg/kg
Life expectancy
Increase in the mean life
span of animals, %
Mice
2.5
86.0±4.0
45
5.0
>90
>100*
control
59.3±3.0
-
Rats
2.5
>90
>100*
5.0
>90
>100*
control
37.3±2.2
-
*Significant increase in the mean life span of animals compared to
control;
>90 days and > 100% of animals with full tumour regression, no
recurrence until 90 days after PDT, are removed from the experi-
ment with the help of essential anesthesia.
Thus, the new chlorin e6 derivative drug - Heliochlorin
is an effective photosensitizer. Photodynamic therapy of
experimental malignant tumours: murine Erlich carcinoma
and rat sarcoma M-1 showed high photodynamic activity of
the drug. Treatment involving intravenous administration of
Heliochlorin at doses of 5.0 mg/kg (murine Erlich carcino-
ma) and 2.5 mg/kg (rat sarcoma M-1) at optimal time be-
tween administration of the drug and laser treatment with
energy density 150 J/cm2 and power density 0.48 W/cm2
allowed to achieve the maximum therapeutic effect - com-
plete tumor regression in 100% of animals and no recur-
rence of neoplasms up to 90 days after treatment. It has thus
been shown that Heliochlorin has a pronounced antitumor
activity for photodynamic therapy when administered at
lower doses and at lower laser effects than other known
chlorin drugs, clinical.
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
272 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
13. Clinical Application of Photodynamic Therapy
To date, in clinical practice, new methods of treatment
based on the photochemistry achievements, photobiology
and quantum physics are increasingly spreading. Photody-
namic therapy (PDT) is a completely new technology used
in the treatment of various diseases.[534-536]
Photodynamic therapy is highly effective and side ef-
fects and complications are minimal at the same time. This
method is based on the selective accumulation of a photo-
sensitizing substance in the tissue of a malignant tumor,
followed by the chemical reaction development that causes
the destruction of sensitized substances under the influence
of a laser beam of a certain wavelength and power. PDT has
no negative effect on nearby tissues and organs. Healing
occurs according to the type of natural reparative processes;
therefore, the method is organ-preserving, well-tolerated
and permissible with repeated use.[537,538]
In the last decade, an increasing number of specialists
have turned to studying the possibilities of this method in
various fields of medicine.
Photodynamic therapy is widely and successfully used
in purulent surgery; there is a pronounced antibacterial and
anti-inflammatory effect. Resistance to PDT in the treat-
ment of chronic infectious processes in pathogenic micro-
organisms does not develop. Recently, PDT of purulent
wounds has been carried out using photosensitizers of the
chlorin series and has a positive effect on the course of the
wound process which is manifested by a pronounced anti-
bacterial effect accelerating the cleansing of wounds from
purulent-necrotic detritus and shortening the healing time of
wound defects.[539]
A.V. Heinz and co. A clinical study was conducted on
the treatment of 120 patients with purulent and long-term
non-healing soft tissue wounds of various etiologies and
localization using the photoditazin photosensitizer (VETA-
GRAND LLC, Russia, registration certificate No. LS
001246 of 18.05.2012) in the form of a gel. According to
the results, it was concluded that this method of treatment
contributes to reducing the time of wound cleansing from
purulent-necrotic detritus, the appearance of granulations
and reducing the timing of the epithelialization onset by
1.52 times. It also reduces the time of complete healing of
purulent wounds by 5-7 days compared with traditional
treatment.
It is important to note that the developed PDT method
of purulent wounds using photosensitizers of the chlorin
series provides a reduction in the time of granulation and
complete healing of purulent wounds.
The effectiveness of PDT does not depend on the
spectrum of sensitivity of pathogenic microorganisms to
antibiotics and has a detrimental effect on antibiotic-
resistant strains of Staphylococcus aureus, Pseudomonas
aeruginosa and other bacteria. Photodynamic damage is
caused by the cytotoxic effect of singlet oxygen and free
radicals, therefore, the development of resistance of micro-
organisms to PDT is minimal. It should be noted that with
prolonged treatment of local infectious processes, the anti-
microbial effect of PDT does not weaken, and the bacteri-
cidal effect is local in nature and does not have a systemic
detrimental effect on the saprophytic flora of the body.
In the field of endocrinology, the results of treatment
of 72 patients with trophic ulcers who underwent PDT with
photoditazine in the form of a gel, both in an independent
version and in combination with NO-plasma therapy were
analyzed. It was concluded that PDT accelerates the cleans-
ing of wounds from devitalized tissues, normalizes micro-
circulatory disorders and stimulates fibroblast proliferation
and granulation tissue maturation. It also enhances phago-
cytosis promotes the epithelization of ulcerative defect on
the 12th-15th day.[540]
Of no small importance is the use of PDT in arthros-
copy for the arthrosis treatment and other inflammatory
processes in large human joints. The results of treatment of
11 patients with deforming arthrosis, synovitis and bursitis
who underwent PDT with the drug photoditazine have been
published. The drug was administered intravenously at the
rate of 0.05 mg/kg of the patient's body weight. After PDT,
the swelling in the projection of the lesion decreased; the
intensity of the severity of the pain syndrome and the local
temperature reaction of the skin was normalized.[543] Treat-
ment results analysis of 86 patients aged 4 to 17 years with
a clinical diagnosis of rheumatoid arthritis, who underwent
PDT with the drug photoditazine in the form of a gel,
showed that this method eliminates the main clinical symp-
toms and is highly effective in the treatment of inflammato-
ry diseases of the joints.
Clinical trials have been successfully launched in
many countries of the world to study the effectiveness of
PDT in the treatment of ophthalmic diseases, in particular,
such as neovascularization.
The results of PDT treatment effectiveness study in
Russia with photosens (FSUE "SSC "NIOPIC", Russia,
registration certificate RN000199/02 dated 04.03.2010) as
monotherapy and combination of PDT with anti-VEGF in
38 patients with age-related macular degeneration have
been published. According to the angiographic picture of
the fundus and the average thickness of the retina in the
foveolus, patients have increased visual acuity, the activity
of the subretinal neovascular membrane has decreased after
treatment.[541]
These guides indicate the effectiveness of the PDT
method with the drug photosens in the treatment of patients
with subretinal neovascular membrane against the back-
ground of age-related macular degeneration.
PDT is used in otorhinolaryngology to improve the
treatment effectiveness of patients with ear throat and nose
purulent-inflammatory diseases, in conditions of reduced
immunity and increased resistance to antibiotics. The re-
sults of PDT in the treatment of purulent sinusitis in 41 pa-
tients with the photosensitizer radachlorin (LLC "RADA-
PHARMA", Russia, registration certificate No.LS-001868
dated 12/16/2011) have been published. Treatment results
analysis showed high efficiency of PDT. 38 patients had a
complete cure; in 3 patients, a significant decrease in the
titers of the detected pathogen was noted in the absence of
clinical signs of the disease.[542]
Data from a number of studies confirm that the use of
PDT in the toxic-allergic form allows to achieve stable re-
mission and in simple forms of chronic tonsillitis to com-
plete cure of the disease.
For many years, specialists have been investigating
the problem of respiratory papillomatosis. Laryngeal papil-
loma is a benign tumor but is prone to rapid growth and
frequent recurrence. Etiotropic therapy of this disease does
not exist to this day. Photodynamic therapy is actively used
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 273
as an alternative method of treating this disease. Russian
authors report on the results of treatment of 19 patients di-
agnosed with respiratory papillomatosis by PDT with rad-
achlorin. After PDT, therapeutic effect was registered in all
patients; complete eradication of PVI was 77%. The ap-
pearance of scars in the larynx and trachea or the aggra-
vation of the existing scarring process has not been rec-
orded.[543]
Currently, the main role in the pathogenesis of the
precancerous and tumor pathology of the cervix develop-
ment belongs to HPV of high oncogenic risk, namely 16,
18, 31, 33, 45 and 52 types. Russian authors report on the
treatment results of 104 patients diagnosed with dysplasia
of the II-III stage and cancer in situ of the cervix associated
with highly oncogenic HPV genotypes with the drug pho-
togem. After PDT, complete eradication of HPV DNA was
achieved in 94% of cases and partial eradication of HPV
was diagnosed in 6% of cases. This is the evidence of a
pronounced antiviral effect of treatment.[544]
The incidence of epithelial malignant neoplasms of the
skin is increasing annually. Basal cell skin cancer (BCC)
accounts for a large proportion of them. One of the most
effective, minimally invasive and organ-preserving methods
of skin epithelial malignant neoplasms treatment is PDT.
Photodynamic damage effectively destroys the tumor and
pathological tissues preserving the surrounding healthy
ones as much as possible.[545]
The positive effect in the treatment of skin cancer by
PDT varies between 70-100% and depends on the localiza-
tion of the tumor process and the stage, the dose of the pho-
tosensitizer and the chemical structure as well as the pa-
rameters of laser irradiation. Complete regression of tumors
is registered in 75-80% of patients, and the duration of the
relapse-free period ranges from 2 months to 5 years.[546,547]
The PDT method is successfully used for inconvenient
localization of tumors with the absence of cosmetic defects
in the form of gross deformation. PDT is indicated for tu-
mors resistant to previously conducted methods of tradi-
tional therapy.
Along with primary and recurrent malignant tumors, a
special place is occupied by metastatic lesions of the skin.
According to the literature, the frequency of metastases of
cancer of internal organs to the skin ranges from 0.29 to
3.3%.[548] Of the metastatic malignant skin tumors, the larg-
est group is breast cancer.
A group of authors performed PDT with domestic
photosensitizers in 36 patients with intradermal metastases.
When using prolonged PDT in patients with intradermal
metastases of breast cancer and melanoma, complete re-
gression of tumors was obtained in 39.3% and 38%, respec-
tively, partial in 46% and 52.4%. Against the background
of previously conducted traditional therapy, tumor re-
sistance to traditional treatment was noted in all pa-
tients.[549]
Kaposi's sarcoma is a rare angioproliferative disease
that is associated with human type 8 herpesvirus. PDT is
the most promising method of its treatment. According to
the results of treatment of 15 patients with intra-tumor ad-
ministration of domestic photosensitizers using PDT, a pos-
itive clinical effect was noted by improving the general
condition of patients and reducing the area of tumor lesion,
which in turn leads to an improvement in the patients life
quality.
Lung cancer occupies the first place in the structure of
morbidity and mortality from malignant neoplasms. The
surgical method of treatment is used for early central lung
cancer. At the same time, about 20-50% of patients are in-
operable. Photodynamic therapy can be used in combina-
tion with classical methods; therefore, it is more promis-
ing.[550] Some authors argue that the effectiveness of PDT in
early central lung cancer (CRL) depends on the form of
growth and size of primary bronchial cancer. Thus, with a
superficial type of tumor (up to 5 mm), complete regression
with PDT is achieved in 91% of cases, from 5 to 10 mm
in 89.4%. In nodular and polypoid types of tumors with a
diameter of up to 5 mm, complete regression was noted in
93.7% of cases, and in tumors from 5 to 10 mm in
62.0%.[551] A number of other authors after PDT in 99 pa-
tients with early CRL in art. IA with tumors up to 1 cm in
diameter, complete regression was observed in 95% of pa-
tients, with tumors of 2 cm or more in 46%. An excel-
lent PDT result was indicated when the tumor size was up
to 1 cm when the distal border of the tumor was clearly
visible during bronchoscopy. In this case, full regression
was achieved in 98% of the observations. Tumor recurrence
was diagnosed in 13% of patients.[552] Experts from the UK
noted that 517 patients had a 5-year survival rate of 70%
with complete regression of the tumor. In the USA, in one
of the PDT clinics, complete tumor regression was achieved
in 69.5% of cases. A new focus of metachronous lung can-
cer was detected in 24% during further follow-up.[553]
A number of Russian researchers report the results of
PDT of early CRL in 37 patients with domestic photosensi-
tizers (photogem, radachlorin, photosens). In 87% of pa-
tients, complete regression of the tumor was noted.
According to the data of many authors, PDT CRL is a
highly effective method and has no equal analogues, espe-
cially in patients with primary multiple bronchial lesions
as well as with a high risk of complications of surgical
treatment.
PDT is an alternative method for the stenosing malig-
nant neoplasms treatment of the respiratory tract, such as
lung atelectasis and pneumonia. There is information about
PDT with domestic photosensitizers in 55 patients with
stenosing malignant lung tumors of central localization.
This group of patients had a positive clinical effect after
treatment.
Recently, intrapleural prolonged PDT with the drug
photosens has been used to treat tumor pleurisy in primary
and metastatic lesions of the pleura. According to some
authors, conducting such therapy in patients with mesothe-
lioma and metastatic pleural lesion makes it possible to
achieve a stable cessation of intrapleural exudation in 92%
of patients with a follow-up period of up to 3.5 years.
To date, serious experience has been accumulated in
the use of PDT in the early cancer treatment of the esopha-
gus and stomach. This method allows to be applied repeat-
edly in the form of multi-course treatment with an interval
of several months. Conducting a multi-course PDT is pos-
sible for many years.
In 116 patients (121 tumor foci), photodynamic thera-
py was performed for initial cancer of the esophagus and
stomach with intravenous administration of domestic pho-
tosensitizers. Complete regression was achieved in 90
(74.4%) foci and partial regression was achieved in 31
(25.6%) tumor foci.[554]
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
274 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
Based on the above, it can be concluded that with the
use of PDT, high oncological results can be achieved in the
treatment of initial forms of esophageal and stomach cancer.
Tumor lesion of the digestive tract aggravates the
condition of patients and reduces the quality of life. PDT is
an alternative method of palliative care in inoperable pa-
tients with stenosing cancer of the upper digestive tract. The
only method of eliminating tumor stricture is photodynamic
therapy for recurrent dysphagia after stenting due to tumor
germination through the walls of the prosthesis or tumor
growth above or below the stent. The recanalization effect
lasts on average 3 months. Repeated PDT also has a benefi-
cial effect on the recurrence of dysphagia. To achieve the
highest quality of life in patients of this group, a multi-
course PDT is recommended.[555,556]
Stomach cancer ranks second in the structure of can-
cer mortality. At the time of diagnosis, in 70% of patients,
the tumor process is locally widespread and is the cause of
high mortality in the first year after diagnosis. A search is
underway for new methods of specialized therapeutic ef-
fects on the area of the surgical field and peritoneum. In-
traoperative PDT is one of such methods. The researchers
note that the use of intraoperative PDT with domestic pho-
tosensitizers in patients with peritoneal dissemination in
disseminated gastric cancer in combination with palliative
surgical treatment to increase the duration of the relapse-
free period and the overall survival of patients in this group
is effective and safe.
Among all malignant neoplasms of the genitourinary
system, bladder cancer accounts for 70%. Surgical treat-
ment in the volume of transurethral resection within healthy
tissues to the muscle layer is the main method of treatment.
But at the same time, the percentage of relapses does not
decrease and is 40-90% in the first year of follow-up. Cur-
rently, PDT is one of the most promising areas of relapse
prevention.
A number of experts indicate that when using PDT
with 5-aminolevulinic acid as adjuvant therapy, the re-
lapse-free survival was more than 2.3 years in every sec-
ond patient.[557]
Some authors report surgical treatment (transurethral
resection of the bladder) followed by adjuvant PDT with
domestic photosensitizers and intravesical chemotherapy in
35 patients with non-muscularly invasive bladder cancer.
During the observation period, no signs of progression or
recurrence of the tumor process were found.[558]
Therefore, as an adjuvant treatment, PDT in combina-
tion with chemotherapy or bladder TUR is a promising
method that allows achieving high oncological results in the
treatment of initial forms of bladder cancer.
The high frequency of precancerous diseases and early
cervical cancer in the structure of gynecological pathology
in young women significantly disrupts reproductive func-
tion and is an urgent problem. The human papilloma-
virus (HPV) plays a major role in the carcinogenesis of
the cervix.
One of the newest approaches to the treatment of cer-
vical pathology, combining optimal therapeutic effect and
the absence of undesirable complications, is PDT. This
method has both antitumor and antiviral effects aimed at
both the lesion site and the source of permanent HPV infec-
tion of the epithelial layers.
In the study of B. Monk, PDT of dysplastic changes in
the cervical epithelium and preinvasive cancer was per-
formed with local application of photophrine with an expo-
sure time of 24 h and an energy density of 100-140 J/cm2.
Out the 11 patients (73%), 8 showed complete regression of
pathological changes.
Russian researchers conducted a comparative analysis
of PDT results in 195 women with dysplastic changes and
early cervical cancer with intravenous administration of
domestic photosensitizers. All patients were diagnosed with
HPV types 16/18. Antitumor and antiviral effects were
evaluated after PDT. A complete regression of the tumor
was diagnosed in 178 people, partial in 6, stabilization -
in 11 people. Complete eradication of the virus was diag-
nosed in 182 people, partial in 8 people and no effect
in 5 patients.
It is important to note that PDT for precancerous and
early cervical cancer is an effective organ-sparing treatment
method that allows the patient to be cured, as well as con-
tributes to her medical and social rehabilitation.
In addition, PDT can be considered as a method of
secondary prevention of cervical cancer in viruspositive
women and used as an independent method of treatment in
this contingent of patients. PDT is an alternative method of
treatment of precancerous and initial tumor pathology of the
cervix with preservation of anatomical and functional integ-
rity of the organ, which is important for the realization of
reproductive function in women.
An urgent problem of modern clinical oncogynecolo-
gy is the treatment of dystrophic diseases, intraepithelial
neoplasia and initial vulvar cancer. Vulvar cancer among
malignant tumors of the female genital organs is 5-8%.
Vulvar cancer is most often preceded by a variety of back-
ground and precancerous diseases. Surgical intervention is
complicated by the fact that the tumor is often localized in
close proximity to important anatomical structures (urethra,
vagina, rectum) or passes to them. The use of photodynam-
ic therapy has expanded the possibilities of therapy for can-
cer and precancerous vulva.
In one of the works of Russian authors, the results of
patients treatment with dystrophic diseases of the vulva,
who underwent PDT using domestic photosensitizers, were
analyzed. Treatment was carried out in 30 patients with
vulvar lesions. By the nature of the revealed pathology of
the vulva, 20 patients (66.7%) had verified sclerotic lichen
of the vulva, 8 (26.6%) had squamous cell hyperplasia of
the vulva, 2 patients (6.7%) had mixed dystrophy. The age
of the patients ranged from 33 to 80 years, the average age
was 56.5 years. Complete clinical remission in the vulvar
pathology group was noted in 27 (90%) of 30 patients. In 3
(10%) patients with vulvar lichen sclerosis, a second PDT
session was required, after which a clinical cure was diag-
nosed. In all cases, a good cosmetic effect was recorded which
is especially important for women of reproductive age.[559]
According to another clinical study, the results of PDT
in 67 patients were analyzed. In 36 patients (53.7%), vulvar
lichen sclerosis was verified, in 16 (23.9%) squamous cell
hyperplasia of the vulva and in 15 patients (22.4%) in-
taepithelial neoplasia of the vulva I-III art. Complete clini-
cal remission was noted in 59 (88.1%) of 67 patients. 8
(11.9%) patients had partial remission, which required a
repeat PDT session. After that clinical cure was diagnosed.
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 275
PDT of the affected vulva area attacked by the dys-
trophic process prevented the progression of the disease and
achieved clinical recovery in 92.5% of patients.[560]
The analysis of the obtained PDT results using domes-
tic photosensitizers in the treatment of dystrophic diseases
and intraepithelial neoplasia of the vulva showed high ther-
apeutic efficacy with minimal side effects and no complica-
tions after the treatment. The PDT method allows perform-
ing organ-preserving treatment in patients with vulvar dis-
eases without deterioration of their quality of life. All this
indicates the high efficiency of this method in the treatment
of patients with vulvar diseases and the prospects for fur-
ther development of this direction in oncogynecology.
14. PDT for Activation
of Antitumor Immune Response
Cancer is one of the leading causes of death world-
wide, being the cause of death for nearly 10 million people
in 2020, or nearly one in six deaths.[561] The global cancer
burden is expected to be 28.4 million cases in 2040, a 47%
increase from 2020.[562] Despite significant scientific and
financial efforts to improve five-year survival rates, statis-
tics for many types of cancer are improving only slightly.
This includes the bronchial, lung, esophageal, stomach,
pancreas, liver, bile ducts, and brain cancers.[563] Other
types of cancer have better survival parameters, which ex-
perts explain either by a good response to chemotherapy or
by late activation of metastasis. This makes it possible to
identify the tumor with instrumental methods before the
metastatic stage and surgically remove it. According to the
American authors, the minimum tumor size diagnosed with
nuclear medicine techniques is 15 mm on average in U.S.
clinics.[564] Impossibility of early detection is a major reason
for treatment failures of tumors that begin to metastasize at
smaller sizes.
In light of this, an important scientific challenge is
finding sensitive methods and tools to diagnose minimally
sized tumors. It is important for early-stage tumors diagno-
sis. Another major scientific problem is the development of
minimally invasive method for evaluating the direction of
the tumor process evolution and the possibility of changing
this direction towards to the destruction of the tumor and
metastases. This is especially important for actively metas-
tasizing tumors, which include most of the types of tumors
with a low five-year survival.
Increasingly popular optical diagnostic techniques us-
ing absorptive and fluorescent dyes have significantly ad-
vanced 5-year survival rates for several types of can-
cer.[565,566] Tissue spectral and video fluorescence analysis
devices, with fluorescence excitation in the red and near-
infrared ranges of the spectrum, have significantly ad-
vanced the diagnostic capabilities of optical methods.[567,568]
For example, in neurosurgical intraoperative navigation
during tumor removal, small bleeding completely blocks
the possibility of fluorescence registration with excitation in
the blue range, whereas with red range radiation, fluores-
cence registration is possible even under a layer of blood up
to 1 mm.[569] The most commonly used near-infrared fluor-
ophores in clinical practice today are protoporphyrin IX
induced by 5-aminolevulinic acid,[570] methylene
blue,[571,572] fluorescein[573,574] and indocyanine green.[575]
One of the main directions of PDT development is the crea-
tion of photosensitizers that allow using infrared laser radia-
tion for excitation, which has a greater depth of penetration
into biological tissues. Currently promising infrared photo-
sensitizers are products of bacteriochlorin series, the ab-
sorption maximums of which lie in the range of 760
820 nm,[57 657 8] products based on nanostructured forms of
phthalocyanine derivatives.[579,580] Indocyanine green with
an absorption maximum around 780 nm, widely used for
fluorescent blood flow studies and identification of diseased
lymph nodes,[581] also has photodynamic effects, as demon-
strated in experimental models of skin cancer[582,583] and
infectious pneumonia.[584]
Tumor stroma
It is known that any primary tumor consists not only
of neoplastic cells, but also of a supporting stroma, the
main elements of which are cells of the hematopoietic sys-
tem (in particular neutrophils and macrophages).[585] A
change in the phenotype of cells in the tumor microenvi-
ronment gives the tumor the opportunity to grow and in-
vade. That is why the study of the transformed cells interac-
tions with the stroma seems relevant and clinically im-
portant. Despite the emergence of various targeted drugs on
the market, which have made a big breakthrough in the
treatment of certain types of oncology, but such diseases as
cancer of the stomach, liver, bile ducts, brain and spinal
cord are still in the shadow of scientific progress and can
not be treated with most drugs. The same applies to the skin
melanoma. To date, particularly relevant is the search for
new approaches to therapy, namely, new specific methods
of influencing tumor stromal cells.[586]
Over the past decade, it has been shown that the stro-
ma is an active and decisive component of tumor growth
and its progression. Stromal cells, such as immune, mesen-
chymal and endothelial cells, can account for more than
half of the cellular contents of the tumor. Their activation
and function are the determining factor for regression or,
conversely, growth of tumors. Further research should pro-
vide a better understanding of the interaction between tu-
mors and the immune system, and this knowledge should
be used to optimize cancer treatments.[587,588]
By secreting immunosuppressive cytokines or other
pro-tumor molecules, tumor cells can create a microenvi-
ronment that inhibits antitumor immunity and promotes
malignancy. Among the cells recruited by the tumor, mac-
rophages are particularly numerous and are present on all
stages of tumor progression. In the primary tumor, macro-
phages (so-called tumor-associated macrophages or TAM)
can stimulate angiogenesis and enhance the invasion of
tumor cells, their mobility and the ability to penetrate into
the vessels.[589] During metastasis, metastasis-associated
macrophages (MAM) contribute to extravasation of tumor
cells, their survival and sustained growth.[590] Tumor-
associated macrophages also suppress immunity by pre-
venting the attack by natural killers and T-cells on tumor
cells during tumor progression and after recovery from
chemo- or immunotherapy. Thus, macrophages are attrac-
tive targets for cancer treatment.
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
276 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
Selective accumulation of PS by macrophages
According to literature, the amount of tumor-
associated macrophages in the tumor microenvironment is
20 to 80% of the total number of cells,[591] while the accu-
mulation of a photosensitizer in them can be up to 9 times
higher (for porphyrin series) than in tumor cells.[592] This is
due to the fact that during intravenous administration the
photosensitizers bind to low-density lipoproteins, on the
capture of which macrophages are oriented.
The scientific community already have developments
on the selective accumulation of photosensitizers in tumor-
associated macrophages. A group led Professor Hamblin
conducted studies of targeted photodynamic therapy aimed
at tumor-associated macrophages. A photosensitizer was
developed based on chlorin e6 bound to albumin, which
selectively accumulated in TAM with intracutaneous ad-
ministration and provided high PDT efficiency.[593,594] An-
other group of Japanese researchers suggested using chlorin
conjugated with mannose for selective accumulation in
TAM (M-chlorin), selectivity of the accumulation was
demonstrated on cells in vitro.[595] Particularly successful in
terms of accumulation in macrophage cells are nanoparti-
cles. As such, it was shown that the use of photosensitizers
in the nanoform allows selective accumulation in tumor-
associated macrophages and tumor cells and the effective-
ness of photodynamic therapy in melanoma B16F10 was
demonstrated.[596]
In recent years, the possibility of using photosensitiz-
ers in the form of molecular nanocrystals for fluorescent
diagnosis and photodynamic therapy has been investigat-
ed.[597] When such molecular aluminum phthalocyanine
nanocrystals came in contact with the surrounding biologi-
cal cells, the effect of fluorescence "kindling" and singlet
oxygen generation was recorded.[598] he manifestation of the
photoactive properties of mTHPC nanoparticle colloid, as
well as the ability to change cellular functions, has been
demonstrated in interaction with polarized macrophages.[599]
PDT-induced immune response
Although the researchers initially focused their efforts
on increasing the direct cytotoxicity for tumor cells subject-
ed to photodynamic effects (including increasing the gener-
ation efficiency of singlet oxygen by photosensitizers),
there has recently been growing interest in developing
methods and approaches for further studying and enhancing
the immunostimulatory properties of PDT. During PDT,
apoptosis and / or necrosis of tumor cells, destruction of the
tumor-associated vasculature, as well as inflammation that
can induce an immune response in the patient, which subse-
quently also leads to the death of tumor cells due to their de-
struction by their own immune cells, arises directly.[600,601]
PDT induced immune response not only increases the
effectiveness of the therapy performed by systemically
eliminating residual tumor lesions and thus controlling the
primary growth of the tumor, but also has a certain potential
to fight cancer cells at the metastatic stage and relapse of
the disease.[602] Phototoxic damage to tumor cells and ves-
sels induces the release of mediators provoking a local in-
flammatory reaction, resulting in the activation of innate
immunity and the recruitment of phagocytes (macrophages,
neutrophils, dendritic and mast cells), and natural killer
(NK) that capture damaged and / or dying tumor
cells.[603,604] Mediators of inflammation are perceived by
immune cells as danger signals and can be recognized and
neutralized by phagocytes, which leads to induced cytotoxic
activity of immune cells against tumor cells.[605] In the
event that a disrupting tumor cell is captured by an antigen -
presenting cell (for example, dendritic cell), an effective
antitumor immune response is triggered by activating a
specific T-cell response: converting naïve T-cells into cyto-
toxic tumor-specific T-lymphocytes.[606]
From the point of view of fundamental medicine,
when approaching anti-tumor therapy as the management of
the immune response, many questions arise: how the PDT
affects the polarization of macrophages, which populations
of macrophages die first during PDT and whether the sur-
viving populations of cells change their functions, whether
anti-tumor immunity is acquired as a result of PDT, is it
possible to evaluate non-invasively the quantitative ratio of
macrophages of different polarizations before and after the
PDT is performed directly in the tumor in vivo, how to neu-
tralize the microglia of the brain activated as a result of
PDT into the proinflammatory phenotype after tumor re-
sorption, etc.
Vascular effect of PDT, oxygenation, effect of hypoxia on
immune antitumor response
As a result of the vascular mechanism of PDT and the
destruction of the tumor-associated vascular network, the
nutrition of the tumor ceases, which contributes to its re-
gression.[607] However, since in practice, during the treat-
ment, an evaluation of the state of the vessels is not carried
out, their destruction is often incomplete. An important tool
for evaluating the vascular effect of PDT should be the
measurement of tissue oxygenation during and after thera-
py, with the possibility of evaluating deep-seated tumor
layers.[608] At the moment, separate studies are under way to
monitor the oxygenation of tissues during PDT. The studies
on experimental animals demonstrated the possibility of
monitoring changes in the vascular bed and saturation with
oxygen using photoacoustic microscopy during PDT with
Verteporfin at a resolution of 60 μm.[609] Similar studies are
conducted using optical coherent angiography.[610] In the
past few years, there have been reports of laser speckle vis-
ualization in real time, a method developed for non-
invasive monitoring of in vivo blood flow dynamics and
vascular structure with high spatial and temporal resolu-
tion.[611] The possibility of conducting classical PDT in
combination with the use of specific PET radiopharmaceu-
ticals to evaluate the effect on cells (apoptosis, necrosis,
proliferation, metabolism) or vascular damage was demon-
strated.[612] However, to be widespread clinical practice, the
method of monitoring blood flow parameters should be
simple in management, inexpensive and allow assessing not
only the dynamics of blood vessels of the tumor, but also
their oxygenation which is relevant for PDT with non-
vascular drugs.
Premise of PDT activation of immunity in brain tumors
Fluorescent diagnosis of brain tumors received wide
development in the clinical practice of the Russian Federa-
tion, especially to conduct intraoperative navigation.[613,614]
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 277
Towards PDT methods Russian and Western doctors exhib-
it a more cautious attitude. This is due to the fact that mi-
croglia cells, acting as phagocytic macrophages in the acti-
vated state, can stay in this state for a long time and phago-
cytize healthy cells.[615] Microglia cells are activated by
mechanical, radiation, or photodynamic effects on brain
tissue.[616] One of the first photosensitizers, methylene blue,
found neuroprotective effects on the central nervous system
in traumatic brain injury by increasing autophagy, reducing
brain edema and inhibiting microglia activation.[617,618] It
should be noted that the photodynamic properties of meth-
ylene blue (and it has a rather significant photodynamic
effect[619]) have not been used or discussed. Another point
of caution in the use of PDT for brain tumors is that, in ac-
cordance with the effect mechanism, the irradiation zone
will be destroyed and this will facilitate the migration of
non-killed cancer cells to other areas for continued growth.
In fact, PDT appears to be an assistant to tumor-associated
microglial cells that express matrix metalloproteinases
(MMPs) to destroy the matrix in the microenvironment of
tumor cells, facilitating their migration and, consequently,
metastasis. To mitigate these effects, it is necessary to eval-
uate the accumulation of a photosensitizer in microglia cells
of different polarization and to select the light doses neces-
sary for their deactivation. When the blood-brain barrier is
compromised, which often occurs with malignant growth,
monocytes enter the tumor stroma, which may polarize into
pro-tumor or pro-inflammatory phenotypes upon extravasa-
tion, which also requires a separate study of their interac-
tion with photosensitizers.[620]
Thus, PDT, depending on the time after the introduc-
tion of the photosensitizer and its redistribution between the
cells of the stroma and the tumor, as well as the irradiation
mode, stimulates a cascade of response reactions, the
strength of which can already be controlled at the irradia-
tion stage. For brain tumors, especially for stage 3,4 glio-
mas, precise and long-term monitoring of the tumor bed
condition is necessary not only during surgery, but also
during remission. The most promising for this purpose are
fiber-optic implants, installed for a long time in the projec-
tion of the tumor bed.[621]
15. Multifunctional Agents Based on Tetrapyrroles
for Magnetic Resonance Imaging Guided
Photodynamic Therapy
Despite significant progress in the study of the cancer
biochemical mechanisms and the development of antitumor
drugs, cancer remains one of the leading causes of morbidi-
ty and mortality worldwide.[622] Traditional methods sur-
gery, chemotherapy and radiotherapy are widely used to
treat oncological diseases, but these methods don’t always
allow the tumors to be completely removed and have seri-
ous side effects. There is an extensive search for safe and
cost-effective approaches to improve the effectiveness of
traditional cancer therapies and to find new alternative ap-
proaches to its treatment.[623-625] Early diagnosis and detec-
tion of the pathological mass are the key to successful can-
cer therapy. Diagnostic imaging implies selective detection
of pathological processes at the cellular and molecular lev-
els, as well as their real-time monitoring with high sensi-
tivity.[627] Over the years, cancer imaging techniques have
progressed significantly from traditional methods providing
structural information to more advanced and relatively new
functional and molecular imaging technologies. In this re-
gard, the concept of theranostics assuming application of
the advantages of molecular imaging agents and tumor
therapy with a single drug is a promising strategy.[628, 629]
Integration of optical imaging and phototherapy meth-
ods have recently initiated phototheranostics.[629-631] Thus,
the combination of optical fluorescence imaging (FLI) in
the near infrared (NIR) (650-1700 nm) window of "optical
tissue permeability", and PS action could be a promising
approach in clinical practice. The combination of PDT and
the magnetic resonance imaging procedure (MRI), which is
widely used in clinical diagnostics due to its high three-
dimensional spatial resolution and ability to penetrate into
deep tissues, is of particular interest.[632,633] The relevance of
MRI is due to its non-invasiveness, non-ionizing, and non-
radiating modality, its ability to provide detailed infor-
mation on tissue anatomy, function, and metabolism in vivo
with excellent anatomical resolution.[634] Non-specific con-
trast agents based on paramagnetic metals (Gd(III), Mn(II),
Fe(III)) are effective for improving sensitivity in clinical
diagnostics, they account for approximately 25-30% of all
MRI scans.[635-637] However, the application of such drugs is
associated with the danger of increased nephrotoxicity due
to the release of metal ion from the chelating complex,
therefore, one of the most important tasks of the creation of
contrast agents is to achieve stable metal chelation. In con-
nection with the above, the design of multifunctional diag-
nostic systems for the combined diagnosis and therapy of
cancer using PDT and MRI methods is a promising ap-
proach in clinical practice.
Figure 33. The chemical structure of theranostic agents for MRI
guided PDT.
The introduction of contrast agent and PS as individu-
al objects leads to limitations in their pharmacodynamics
and pharmacokinetics, and, as a consequence, to differences
in imaging based on their biodistribution. The inclusion of
MRI contrast agent and PS in one theranostic agent has
been shown to overcome this limitation, when the contrast
agent and PS have the same biodistribution patterns, thus
PDT treatment is further optimized.[638] Furthermore, this
strategy of bimodal agents combining within a single mo-
lecular object provides the additional benefits of enhanced
PS hydrophilicity in biological fluids and improved proton
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
278 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
relaxation for MRI efficiency due to the increased molecu-
lar weight.[639] In addition, the growing interest in the de-
velopment of effective PS-based contrast agents is related
to the fact that no commercially available contrast agent is
effective enough for accurate detection of malignant neo-
plastic tissue.
In the past few years, agents for phototheranostics
based on tetrapyrrole structures that could combine MRI
diagnosis and PDT have been actively developed.[631,640-643]
The limited cavity size of tetrapyrrole macrocycles cannot
ensure stable binding to the large radius metal ions used in
MRI, it was proposed to introduce an external chelating
fragment into the PS molecule to create the drug-
theranostics (Figure 33).[644-646]
This review is devoted to recent advances in the appli-
cation of tetrapyrrole compounds for MRI guided PDT. It
emphasizes that the introduction of an external chelating
fragment into the structure of tetrapyrroles leads to genera-
tion of promising agents for phototheranostics. Data on the
available contrast agents as well as PS based on
tetrapyrroles containing Gd(III) for MR diagnostics are
presented, methods of inclusion of metal atoms into such
systems are analyzed.
Types of contrast probes used in MRI diagnosis
The principle of MRI is based on the phenomenon of
nuclear magnetic resonance and proton spin relaxation in
the magnetic field divided into 2 types: 1) longitudinal re-
laxation called T1 relaxation or spin-lattice relaxation aris-
ing from the dissipation of absorbed radiofrequency pulse
energy in the surrounding tissue; 2) transverse relaxation,
called T2 relaxation or spin-spin relaxation arising from the
loss of phase coherence during spin precession. Longitudi-
nal and transverse relaxations are two synchronous magnet-
ic processes, each of which provides its own type of con-
trast: T1 recovery and T2 attenuation have brightening
(positive contrast) and darkening (negative contrast) effects,
respectively.[639,647]
The amount of intrinsic contrast between tissues pro-
duced on an MR image depends on the differences in T1
and/or T2 relaxation times of the observed tissues. In some
cases, the difference in signals between diseased and
healthy tissue is difficult to detect on the MR image. Patho-
logical tissues may have no significant difference in T1 or
T2 signals from the surrounding normal tissues. This signal
distinction can be increased by introduction of an exoge-
nous contrast agent (CA contrast agent).[632] The exogenous
contrast agents improve MR images by locally decreasing
T1 and T2 proton relaxation times.
Two main types of magnetic resonance probes are
paramagnetic and superparamagnetic agents with unpaired
electrons, and the corresponding magnetic moments engage
in dipolar and scalar interactions with nearby water pro-
tons.[639] Then the local water protons relax and quickly
exchange with other unrelaxed water protons reducing the
average relaxation time. The amplification of the signal
contrast can be achieved in two ways: either by higher tis-
sue vascularization, which, for example, is typical for tu-
mors, or by increased affinity of CA to the target area.
The contrast agent (CA) should have high relaxation
ability, stability, specific biodistribution, rapid clearance,
low osmolality and viscosity, as well as low toxicity. All
these contrast agents have their advantages and draw-
backs.[636] The typical CA is a chelated metal ion with a
constant magnetic moment.[632,636, 639,643] The acute and
chronic toxic side effects caused by the metal ion as well as
the chelating agent are significantly reduced due to com-
plexation.
T1 contrast agents are called positive CAs, and they
are mostly compounds with paramagnetic metals. The in-
vestigated tissues absorbing such contrast agents become
bright on T1-weighted images and can be distinguished
from other pathological and normal physiological states,
T1-weighted images are usually more preferable to T2 con-
trast agents. T1-type agents contain paramagnetic ions
(Gd3+ and Mn2+), which have high electron spin and con-
stant magnetic moment due to seven and five unpaired elec-
trons, respectively. T2 contrast agents are classified as neg-
ative CAs, they are derived from iron(III) compounds in
most cases. The negative contrast agents reduce T2 signals
by shortening the T2 relaxation time, which leads to a
strong decrease in the MR signal and thus to the negative
contrast in vivo.[29,632,636,639,647]
Gadolinium-based contrast agents
Gadolinium-based CAs are the basis of clinically used
MRI contrast agents due to its properties: the high magnetic
moment and the long electron spin relaxation time.[648-649]
The contrast agents containing gadolinium reduce the T1
and T2 relaxation times of neighboring water protons,
which increases the signal intensity of T1-weighted images
and decreases the signal intensity of T2-weighted images.
The contrast agents containing gadolinium reduce the T1
and T2 relaxation times of adjoining water protons, that
increases the signal intensity of T1-weighted images and
decreases the signal intensity of T2-weighted images. How-
ever, there are some physical restrictions, for example, the
short half-life complicates the differentiation of benign and
malignant tumors. In addition, gadolinium binds weakly to
serum proteins, its salts are usually hydrolyzed and con-
verted to hydroxides, which are absorbed by the reticuloen-
dothelial system (RES) and accumulate in the body, espe-
cially in the liver, spleen and bones, thereby imparting po-
tential toxicity to the drug. To partially overcome these
problems, Gd is introduced in the chelated forms. Currently
nine Gd contrast compounds with multidentate chelating
ligands have been approved for the clinical practice. Gener-
ally, the cyclic ligand is more stable than the linear ligand.
The ionic compounds are slightly more stable than non-
ionic compounds, but they have a higher osmolality at the
same time. The ionic and hydrophilic complexes comprise
gadolinium (III) diethylentriamine pentaacetate (Gd-
DTPA, referred to as gadopentetate dimeglumine),
Gd(III) 1,4,7,10-tetrazacyclododecane-1,4,7,10-tetraacetate
(Gd-DOTA, gadoterate) and polyaspartate Gd(III). The
nonionic hydrophilic gadolinium (III) chelates are Gd(III)-
diethylene-triamine pentaacetate-bis(methylamide) (Gd-
DTPA-BMA, gadodiamide) and macrocyclic chelate Gd-
DOTA, in which the acetic acid functional group is substi-
tuted with 2-propanol radical (Gd-HP-DO3A, gadoteridol).
The ionic and lipophilic gadolinium complexes comprise
Gd-benzyl-oxymethyl derivative of dimethylglucamine salt
of diethyltriaminepentaacetate (Gd-BOPTA, gadobenate
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 279
dimeglumine) and Gd-ethoxybenzyl diethylentriaminepent-
aacetate (Gd-EOB-DTPA, gadoxetate) (Figure 34).[648]
Although clinically used Gd chelates are relatively
safe and generally tolerated by patients well, there have
been some recent concerns about nephrogenic systemic
fibrosis and nephrogenic fibrosing dermopathy in some
patients, as well as retention of small quantities of Gd in
bone and brain tissue has been found. These side effects
have been identified in patients with impaired renal func-
tion and patients, who have been exposed to MRI scans
with contrast repeatedly. However, there is no evidence that
the application of approved gadolinium-based contrast
agents can lead to pathological changes in the body.
Manganese-based contrast agents
Great attention has been recently paid to the develop-
ment of the contrast agents based on Mn, which is an en-
dogenous metal and has less toxicity. Mn(II) forms a
Mn(II) high-spin d5-cation (S = 5/2) and Mn(III) d4-cation
(S = 2). Mn(II) can act as a strong paramagnetic ion gener-
ating T1 signal amplification in vivo.[650,651] Manganese
dipyridoxyldiphosphate (Mn-DPDP, Teslascan) and man-
ganese(II) chloride tetrahydrate (LumenHance) are the only
clinically approved Mn-based contrast agents.[652] Manga-
nese in ionic form and manganese chelates have a short
half-life. Manganese compounds are excreted primarily
through the gastrointestinal tract and the biliary tract unlike
gadolinium contrast agents. However, high concentrations
of manganese have neurotoxic effects similar to Parkinson's
disease. Mn-based contrast agent, manganese-N-picolyl-
N,N',N'-trans-1,2-cyclohexenediamin triacetate (Mn-Py-
C3A), has been recently proposed, and it provides high spin
relaxation and resistance to Mn dissociation simultaneous-
ly. The application of Mn-Py-C3A exhibited the tumor con-
trast enhancement comparable to gadoterate meglumine in
an animal model of breast cancer, and its partial hepatobili-
ary elimination allowed visualization of liver tumors in a
model of the metastasis (Figure 35).[653] However, despite
the rather high efficiency of the proposed Mn(II)-based
MRI contrast agents, the problems of thermodynamic sta-
bility and kinetic inertness of these complexes remain unre-
solved, that calls into question their clinical application.
Iron-based contrast agents
The iron-based preparations were proposed as a poten-
tial alternative to gadolinium contrast compounds more
than thirty years ago. It has led to extensive preclinical and
clinical studies, but today the only commercially available
contrast agent for MRI is Combidex® (iron oxide nanopar-
ticles), approved exclusively in the Netherlands. Magnetic
nanoparticles of iron (III) oxide are T2 contrast agent, and
also Fe3+ ion has a significant T1 contrast enhancement ef-
fect.[639] The magnetic properties of Fe3+ are due to external
unpaired electrons unlike the Gd3+ ion, so they depend
strongly on the coordination method of the metal with the
ligand. Iron is a widespread and endogenous element in the
human body, so high-spin Fe(III) complexes are very prom-
ising as contrast agents for MRI, they have the best bio-
compatibility and safety profiles as compared to other metal
complexes.[654]
Figure 34. Gadolinium-based contrast agents approved for clinical application.[648]
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
280 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
Figure 35. Liver imaging of an orthotopic mouse model of colorectal liver metastasis with complex Mn-Py-C3A at 4.7 T. (A and B) T1-
weighted axial images of the liver at the level of the tumor (arrow) prior to and during the hepatocellular phase after injection of Mn-Py-
C3A, respectively. (C) Liver parenchyma vs. tumor CNR prior to and following treatment with Mn-Py-C3A. Reprinted with permission
from [653]. Copyright 2018 American Chemical Society.[653]
Figure 36. Potential MRI contrast agents based on Fe(III) complexes.[657]
There are two types of iron oxide-based contrast
agents: superparamagnetic iron oxide (SPIO) and ultra-
small superparamagnetic iron oxide (USPIO), which are
colloidal suspensions of iron oxide nanoparticles. SPIO and
USPIO have been successfully used to diagnose liver tu-
mors in some cases. The superparamagnetic iron oxide par-
ticles or their ultra-small forms are removed from the blood
by the reticuloendothelial system of the liver, spleen and
lymph nodes after intravenous injection. Due to this specif-
ic biodistribution, these preparations can be used for the
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 281
organ imaging and the evaluation of metastasis spread, the
macrophage labeling in the vulnerable atherosclerotic
plaques, and the detection of inflammatory lesions after
transplantations. This explains the recent increased interest
in the application of iron-based preparations for MRI.[653] In
terms of the development of new contrast agents, Schellen-
berger et al. have shown that low molecular weight Fe(III)
complexes such as Fe-CDTA (Magnevist®) provide signif-
icant contrast imaging in vivo and exhibit enhancement
kinetics similar to the data for the clinically used agent Gd-
DTPA (Figure 36).[656,657]
The main problem of iron nanoparticles application in
medical practice is related to the fact that nanoparticles tend
to accumulate in vessels, causing their embolization and
blocking the blood flow [654, 657].
Design of tetrapyrroles conjugates with Gd(III)
for phototheranostics
Tetrapyrrole compounds can become a versatile plat-
form for creating such agents due to their structure, func-
tionalization ability, photophysical properties, and affinity
for the tumor. The conjugation of tetrapyrrole PS with
MRI-contrasts seems to be a particularly promising ap-
proach for the theranostics purposes.[658] Since unsubstitut-
ed tetrapyrroles are poorly soluble in aqueous media, the
presence of hydrophilic complexes of Gd(III) and other
paramagnetic metals makes PS soluble under physiological
conditions and improves tissue distribution and excretion in
vivo. In this regard, the molecular design of the therapeutic
molecule, in particular, the selection of the optimal number
and type of paramagnetic metal complexes within the con-
jugate to fulfill all the requirements for a drug and diagnos-
tic drug is a critical point. PS for PDT, in turn, should have
an amphiphilic structure, be non-toxic and have a strong
absorption in the window of biological transparency to
achieve the required depth of the tissue penetration and
accumulation in the tumor.[659] In addition, the growing
interest in the development of effective PS-based contrast
agents is related to the fact that, to date, no commercially
available contrast agent is sufficiently effective for the ac-
curate detection of malignant neoplastic tissues.
Despite the well-known coordination abilities of
tetrapyrroles, there are limitations in obtaining stable com-
plexes with large radius paramagnetic metals, such as
Gd(III) ions. The limited cavity size of the porphyrin mac-
rocycle makes it very difficult to obtain stable compounds
with metals used in MRI. Therefore, the introduction of an
external chelating fragment into the structure of the mole-
cule became necessary to realize the porphyrin-based
theranostics concept, and most of the publications involve
the conjugation of the tetrapyrrole cycle with an external
chelating agent.
Previously, bifunctional agent 1 (Gd2(DTPA)4TPP)
based on meso-5,10,15,20-tetrakis(4-aminophenyl)porphyrin
bound to 4 DTPA complexes, two of which were metallated
with Gd(III) ions, was first proposed in 1993.[660] It was
found that the relaxation value for the obtained conjugate
with Gd(III) was twice as high as for GdDTPA at 20 MHz,
and a significant enhancement of the contrast image of the
tumor compared to the adjacent normal tissues in mice was
indicative of the affinity of the compound to the tumor tis-
sue (Figure 37). Photoinduced toxicity studies performed
Figure 37. Structure of a new bimodal agent.[660]
Figure 38. Chemical structure of the leader theranostic
compound 2.[661]
on two cell lines (HT29 and L1210) under multi-
wavelength laser irradiation with λ=488 nm and 514 nm
showed phototoxicity of the Gd2(DTPA)4TPP conjugate
comparable to values for the commercially available PS
hematoporphyrin. This Gd2(DTPA)4TPP conjugate was the
first bimodal agent prototype developed for the MRI-
controlled PDT.
More than ten years later Pandey et al. developed and
investigated several theranostic agents based on the natural
porphyrin pyropheoforbide with substituents of lipophilic
or hydrophilic nature.[661] These porphyrins were conjugat-
ed to one, two, three, or six GdDTPA complexes. The con-
jugate design included the creation of a chelating agent
bond by an additional branched linker away from the mac-
rocycle, and the stability of the GdDTPA core was main-
tained by chelation with five anionic carboxyl groups.
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
282 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
Figure 39. Magnetic resonance (MR) images of rat (Fischer) with Ward's colon tumors before (left) and 24 h after (right) injection of
HPPH-3-GdDTPA conjugate (dose: 10 μmol/kg). Reprinted with permission from [661]. Copyright 2018, American Chemical Society.[661]
Theranostics containing one or two Gd complexes required
the use of a liposomal composition to impart water solubili-
ty. With three (compound 2) and six GdDTPA fragments in
the conjugate molecule, water solubility was significantly
improved. Compound 2 (HPPH-3GdDTPA) with three che-
lating fragments (Figure 38), was found to be the best can-
didate with respect to imaging and treatment results. This
conjugate showed a marked enhancement of MR contrast in
tumors in mice 24 h after injection at a dose 10 times lower
than the clinical dose used with Magnevist® (Figure 39).
Fluorescence imaging obtained from the emission of
light by the HPPH derivative also showed maximum inten-
sity 24 h after injection. Also, this compound showed an
effective PDT effect after a single irradiation at 665 nm
(70 J/cm2) 24 h after injection.
In the following work, conjugates of tetra- and mono-
substituted meso-arylporphyrins 3 with GdDTPA residues
were obtained (Figure 40).[662] Measurement of the longitu-
dinal relaxation capacity of the products showed 154% and
251% enhancement over the commonly used Gd-DTPA for
the mono- and tetrasubstituted derivatives, respectively, and
their relaxation times were r1 7.82 mM-1s-1 and 14.8 mM-1s-1.
Research [663] describes the production of potential
theranostic drugs based on zinc phthalocyanine complexes
conjugated with one molecule of the GdDO3A complex
(Figure 41). To reduce the degree of aggregation in aqueous
media and increase the solubility, the PS macrocycles were
modified with peripheral hydrophilic substituents. The
ZnPht-1Gd conjugate 4 showed a lower proton relaxation
value (1.43 mM-1s-1 at 128 MHz) compared to the commer-
cial preparation Omniscan™ (3.23 mM-1s-1 at 128 MHz).
This phenomenon may be due to steric hindrances that
make it difficult for water molecules to access the metal
center. At the same time, the ZnPht-1Gd compound pro-
duced cytotoxic singlet oxygen quite efficiently in DMSO
medium when irradiated with a quantum yield of 0.67.[663]
Figure 40. Structure of the new monosubstituted contrast agent 3.[662]
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 283
Figure 41. Chemical structure of 4 (ZnPht-1Gd) [663] and 5
(ZnPz-1Gd).[664]
Figure 42. T1 weighted MR image of WI-38 VA13 cells
incubated with 0.50, and 100 μM of Zn-Pz-1Gd(III) for 24 h at 4.7
T. Copyright © 2010, American Chemical Society.[663]
The zinc-based porphyrazine complex PS was bound
to one, four, and eight GdDO3A complexes to form bifunc-
tional conjugates (ZnPz-Gd) (Figure 41). These compounds
showed a strong increase in relaxation (up to 12.8 mM-1s-1
for ZnPz-8Gd at 60 MHz and 37 °C) with an increase in the
number of Gd-complexes attached to the PS (Figure 42).
Cellular uptake was observed only for compounds contain-
ing a single Gd complex (ZnPz-1Gd). A noticeable photo-
toxic effect (with 50% cell destruction) was observed after
10-min white light irradiation.[664]
Porphyrin derivatives incorporating the Gd(III) ion in-
to their cavity are an attractive agents due to their simpli-
fied structure and lower synthetic costs. However, numer-
ous studies in vivo demonstrate problems associated with
the reduced stability of these compounds caused by the dis-
sociation of gadolinium ions from the porphyrin cavity [665].
Nevertheless, two compounds based on substituted porphy-
razines 6-7 (GdPz1 and GdPz2), differing in the nature of
the peripheral groups and containing a Gd(III) ion in the
macrocycle cavity were obtained (Figure 43)[665]. Significant
accumulation of the agent in the tumor in vivo was con-
firmed by fluorescence and MRI methods for both com-
pounds. A good relaxation value under very strong magnet-
ic field conditions (4.67 mM-1 s-1 at 9.4 Tesla) was obtained
for compound GdPz1, and cellular uptake, photodynamic
activity and fluorescence in vivo were also studied. PDT
activity was evaluated on CT26 cell line when irradiated
with 615-635 nm light (10-20 J/cm2, incubation concentra-
tion from 10-7 to 10-4 M). Moderate tumor death was ob-
served, so further optimization of various parameters such
as drug dose and light application was necessary.
The use of the multifunctionalized PS for the treat-
ment of tumors with PDT and chemotherapy, as well as for
MRI imaging, has also been reported.[49] Functionalized
meso-arylporphyrin including gadolinium ion in the macro-
cycle cavity was covalently bound to chemotoxic plati-
num(II) complexes. The Gd/Pt-P1 compound 8 was ob-
tained from 5,10,15,20-tetra(4-pyridyl)porphyrin (P1) coor-
dinated with four Pt(II) complexes (Pt-P1) and one Gd(III)
ion (Figure 44). The new polyfunctional agent showed al-
most doubled relaxation capacity at 3 Tesla compared to
commercial GdDTPA. The phototoxic effect was studied
on the C6 cell line after a 10-minute irradiation at 630 nm.
A synergistic chemophotodynamic antitumor effect was
observed in cells from C6 tumor-bearing mice.
Figure 43. Molecular structure of GdPz1 and GdPz2.[665]
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
284 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
Figure 44. Molecular structure of 8.[666]
Figure 45. Chemical structure of the theranostic compound 9
developed by Chen et al.[667]
Increasing the number of coordinated water molecules
near Gd(III) contrast agents is particularly attractive strate-
gy to receive agents with high relaxation in a strong mag-
netic field. In this case, careful design of the structure of
hepta- or hexadentate ligands is necessary in order to obtain
Gd(III) complexes with good thermodynamic stability and
kinetic inertness and to avoid the formation of ternary com-
plexes with endogenous molecules. Chen et al. proposed a
new structure of potential theranostics 9 (Figure 45) con-
sisting of a central core-tetraphenylporphyrin bound to four
GdDTTA complexes.[667]
This compound showed a high relaxation value
(14.1 mM-1s-1 at 0.55 Tesla), which doubled in the presence
of human serum albumin, indicating a high affinity of the
obtained conjugate for this blood transport protein (Figure 46).
Uptake of H1299 cell line was confirmed by confocal mi-
croscopy. Effective formation of singlet oxygen was ob-
served under irradiation at 650 nm. The results demonstrat-
ed high potential of this compound as a contrast agent for
multimodal (MR and luminescence) imaging and as a PS
for PDT.[667]
In the following work, a number of theranostics based
on zinc porphyrin complexes with an extended π-system
with the external chelate ligand GdDOTA 10-12 were ob-
tained, and the properties of the synthesized single- and
two-photon activated PS as imaging probes were studied
(Figure 47).[668,669] The attachment of GdDOTA to PS was
realized using the commercial ligand DOTAGA (1,4,7,10-
tetraazacyclodododecane-1-glutar-4,7,10-triacetic acid),
which provided local flexibility while maintaining the sta-
bility of the GdDOTA fragment.
High relaxation r1 values were obtained for 10-12,
which were 19.32, 19.94, and 14.33 mM-1-s-1 at 20 MHz,
respectively, which is 4-5 times higher for known contrast
agents. A 20% increase in relaxation was also observed in
the presence of BSA. Compound 10 had a strong single-
photon absorption capacity (εmax = 41000 M-1 cm-1 at
667 nm in water).
Figure 46. (A) T1-weighted phantom MR images for complex 9 with a concentration of 0−50.0 μM. (B) T1-weighted phantom MR imag-
es for complex 9. Copyright © 2014, American Chemical Society.[667]
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 285
Figure 47. Chemical structure of theranostics 10-12 developed by Heitz et al.[668,669]
Figure 48. Structure of complexes with Gd(III), Fe(III), Mn(III).
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
286 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
The high PDT effect evaluated in HeLa cells was ob-
served with single-photon excitation at 660 nm (1 h, 1 μM
incubation), and a moderate two-photon PDT effect was
achieved at 930 nm (300 scans, 1 μM incubation). High
PDT effect was observed in HeLa cells after single-photon
irradiation at 740 nm (30 min, incubation concentration
of 1 μM). The effect of two-photon PDT was observed at
910 nm (300 scans, incubation concentration 2 μM) as a
function of light power, and 100% cell death was observed
at an average power of 108 mW.
In our work, terpyridine derivatives were used as an
external chelating agent to obtain conjugates of meso-
arylporphyrins with Gd(III), Fe(III), and Mn(III).[670] Ter-
pyridine belongs to the type of linear pincer (claw-shaped)
ligands and forms coordination chelates of the tridentate
type with various metal cations in an almost flat geometry
due to the presence of three conjugated nitrogen atoms in
the molecule.[671] We have developed synthetic approaches
for the preparation of conjugates based on porphyrins of
A2B2 and A3B type with an active functional groups (NH2,
COOH, OH, Py) (Figure 48).
The highest yields were achieved when the amide
bond between the porphyrin and terpyridine fragments was
created. Thus, the yields of conjugates 15-17 were 90-95%.
The conditions for obtaining conjugates 13-17 with para-
magnetic ions Gd(III), Fe(III), Mn(III) for the subsequent
creation of MRI-agents were established. Thus, it appeared
that conjugate complexes with Fe(III) and Mn(III) salts
were formed under mild conditions in an inert atmosphere
at room temperature with the addition of sodium acetate as
a catalyst, while the introduction of gadolinium under mild
conditions was not effective enough and required harsher
reaction conditions. To obtain Gd(III) complexes, the reac-
tion was carried out in an inert box in an autoclave when
heated well above the boiling point of the solvents (80°C).
To assess the safety of the obtained conjugates, it was
important to evaluate the effect of the newly synthesized
compounds on the ability to disrupt the integrity of intercel-
lular contacts (test for inhibition of metabolic cooperation
or for promoter activity). The study demonstrated that com-
pounds 13-17 do not exhibit promoter activity in the given
concentration ranges, i.e., they do not disrupt intercellular
contacts. Thus, when exposed to compound 14 at concen-
trations of 4.5 μM; 2.25 μM; 1.125 μM and 0.56 μM, the
degree of cell cooperation was 109%, 105%, 101%, 111%
relative to untreated cells (Figure 49). At the same time, the
degree of cell cooperation in the samples treated with TPA
at a final concentration of 5 μg/ml was 15% to 20%, which
is statistically significantly different from the control
(p<0.01). Figure 49 shows microphotographs of the cell
monolayer in the scratch region from the control and exper-
imental samples.
The dark and light-induced toxicity of the obtained
conjugates was evaluated in vitro on the Hep-2 cell line
with and without irradiation for 1.5 and 24 h. The absence
of dark and light-induced toxicity of the obtained com-
pounds after 1.5 h was shown. However, an increase in
dark-induced toxicity was observed with prolonged incuba-
tion. Moreover, for the conjugates linked by an amide bond,
the toxicity was lower. Thus, for conjugate 13 after 24 h of
incubation the IC50 was 2.47±0.233 μM (irradiation dose
8.073 J/cm2) and 2.55±0.28 μM (no irradiation). For conju-
gate 15, the IC50 was 1.55±0.15 μM (irradiation dose 8.073
J/cm2) and 4.17±0.251 μM (no irradiation) (Figure 50).
Such an effect may be related to the better biocompatibility
(lower toxicity) of the amide bond for cells.
Figure 49. Micrographs of cell monolayer in the scratch area of control and experimental samples. A. Negative control (dH2O). B. Posi-
tive control (TPA, 5 μg/ml). Compound 12: 4.5 μM (C); 2.25 μM (D); 1.125 μM (E) and 0.56 μM (F). LV propagation length in 4 minutes.
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 287
Figure 50. Effect of compounds on the viability of Hep2 cell line. Cells were irradiated for 90 min using the Medical Therapy Philips TL
20W/52 lamp (irradiation dose of 8.073 J/cm2). Incubation of cells with the compound without irradiation in the dark for 90 min. * statisti-
cally significant differences in cell survival relative to values with zero concentration of the compound (p<0.01) were noted. A) conjugate
13; B) conjugate 15.
In conclusion, increasing interest in the development
of effective contrast agents for MRI diagnostics based on
tetrapyrrole PS is related to the fact that, to date, no com-
mercially available contrast agent is effective enough for
accurate detection of malignant neoplastic tissues. Analysis
of the literature has shown that the development of multi-
functional theranostic systems for combined diagnosis and
therapy of cancer using PDT and MRI methods is a promis-
ing approach in clinical practice. Tetrapyrrole PSs are a
suitable chemical platform for the design of multimodal
agents for PDT. The strategy of combining bimodal agents
in the structure of a single molecular object provides addi-
tional advantages of increased hydrophilicity of the PS in
biological fluids and improved proton relaxation for MRI
efficiency due to the increased molecular weight.
16. Effect of Amphiphilic Polymers
and Polysaccharides on the Photosensitizing
Activity of Porphyrin and Non-Porphyrin Dyes
One of the most important priorities of the Strategy for
Scientific and Technological Development of the Russian
Federation is the "Transition to personalized medicine,
high-tech healthcare and health-saving technologies, includ-
ing through the rational use of drugs (primarily antibacterial
drugs)". Antibacterial photodynamic therapy (aPDT) can be
considered not only in connection with the problem of the
growth of resistance of pathogenic microorganisms to anti-
microbial drugs, but also as a certain niche in antibiotic
therapy, the relevance of which is associated with the loss
of antibiotic effectiveness, as a result of which “common
infections and minor injuries that have been curable for
decades can now kill again” (opinion of WHO Assistant
Director-General for Health Security Dr. Keiji Fukuda). It
is precisely because of this circumstance that researchers
from scientific and medical centers in Japan, the USA,
South Korea, France, Australia, etc., begin to develop ideas
about the use of aPDT instead of antibiotic therapy in the
treatment of dental and some other types of chronic in-
flammatory processes. Approaches to the use of aPDT in
the fight against wound infections, trophic ulcers and bed-
sores are also being developed. This is evidenced by the
growing number of publications and relevant international
symposiums. There is also a growing awareness that photo-
dynamic therapy (PDT), including aPDT, should be under-
stood not only as photoinduced effects on pathological tis-
sues with necrosis or apoptosis of affected cells, but also as
a targeted initiation of the effective dynamics of subsequent
regenerative processes of tissue granulation and epitheliali-
sation.[448,673] Such processes are initiated, among other
things, by the body's immune system in the tissues adjacent
to the pathologically developing tissues of the affected ob-
jects. And the task of PDT should be to choose the optimal
regimes of photodynamic effects on pathological tissues so
that the regenerative systems of the body can show their
activity to the full extent. It is for this reason that the level
of energy effects during PDT of affected objects should be
minimized to a certain level in order to initiate, but not sup-
press, the possibilities of the body's regenerative systems.
First of all, a controlled decrease in the level of light expo-
sure during aPDT can be associated with a certain decrease
in the concentration of photosensitizers (PS) introduced into
the corresponding drugs, which is extremely important for
reducing the phototoxicity of such dosage forms. As it was
shown earlier,[674] the solution of such problems can be
achieved by using PS with amphiphilic polymers (AP), with
fragments of which PS molecules can form conformational
complexes. In this case, the specific activity of the PS in the
composition of such complexes with AP can exceed the
specific activity of the initial PS molecules by an order of
magnitude or more (“polymer effect”). Since photoinduced
effects on pathological tissues during PDT are associated
with photosensitized (with the participation of PS mole-
cules, usually porphyrins, their chlorine derivatives, phthal-
ocyanines) generation of singlet oxygen and its other reac-
tive forms (ROS), conclusions about the "polymer effect"
were made preliminary on the basis of the values of effec-
tive rate constant keff of photooxidation of a model sub-
strate, tryptophan, and confirmed by aPDT of infected
wounds in laboratory animals.[674,675] The idea of using PS
together with polymeric components in the preparation of
medicinal drugs for aPDT lies in the development of an
integrated approach to the creation of effective photosensi-
tizing systems. This is, first of all, the search for new com-
ponents for the developed photosensitizing compositions
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
288 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
PS-AP, where PS is a photosensitizer; AP is an amphiphilic
polymer. At the same time, it is possible to use not only
traditional PS, such as phthalocyanine, porphyrin and chlo-
rine derivatives, but also non-porphyrin compounds - dyes
that not only show a high activity in the photogeneration of
reactive oxygen species (ROS), primarily singlet oxygen
1О2 with a quantum yield ФΔ =0.75-0.76, but also exhibit-
ing their own bactericidal activity (which is traditionally
used in medical practice).[320,676,677] This, in particular, ap-
plies to methylene blue (MB) and rose bengal (RB).[678680]
These PS are currently used in aPDT of bacterial, viral and
fungal infections, and their cost is significantly less than the
cost of porphyrin compounds. It was shown in [673] that MB
and RB dyes, as well as porphyrin compounds, can become
the basis for the search for effective polymer-containing PS
systems for aPDT. Methylene blue (Figure 51,a) is an or-
ganic dye of the thiazine dyes group, which has long been
known as an effective photogenerator of singlet oxygen.
Recently, it has attracted attention as a promising photosen-
sitizer for PDT of cancer and infectious diseases.[681,682]
Methylene blue is used as a PS for the destruction of My-
cobacterium tuberculosis by аPDT.[683] However, unlike
porphyrin derivatives, which require a long incubation peri-
od for photosensitization, phenothiazine derivatives, such as
MB, do not require such a long pretreatment. Rose bengal
(Figure 51,b) is a dianionic xanthene dye, also used in pho-
todynamic therapy of certain types of cancer and fungal
diseases.[684,685] The structure of MB and RB is represented
by polycyclic aromatic compounds, which tend to form
stable aggregates with low PS activity in aqueous solutions.
In addition, due to its anionic nature, RB hardly penetrates
through the cell walls of bacteria.
Studies have shown that ternary block copolymers of
oxyethylene and oxypropylene, pluronics F127 and F108,
polyvinylpyrrolidone, polyethylene glycol (PEG), and pol-
yvinyl alcohol, may be of interest as AP for PS systems for
aPDT. Naturally, the necessary (preliminary) condition for
the use of such AP in PS systems is their promoting role in
the model process of tryptophan photooxidation, which
occurs during the photosensitized energy transfer of triplet
excited PS molecules to molecular oxygen with the for-
mation of singlet oxygen in aqueous media. The kinetic
regularities of tryptophan photooxidation catalyzed by
methylene blue and rose bengal in the presence (and ab-
sence) of the indicated AP, as well as an assessment of the
effect of AP on the absorption and fluorescence spectra of
the dyes, are presented below.
As shown earlier, amphiphilic polymers can initiate
the deaggregation of porphyrins or dyes in an aqueous me-
dium, thereby increasing the proportion of PS molecules
that are effectively involved in the process of photosensitiz-
ing oxidation of substrate molecules. It is known that the
aggregation of porphyrins in aqueous media, which reduces
their photosensitizing activity due to the internal dissipation
of photoexcitation energy, is due to hydrophobic interac-
tions of their macrocycles, while the aggregation of these
dyes is associated with their substituted anthracene struc-
ture. The magnitude of the effect of AP on the state of PS
associates in all cases was analyzed by the value of the ef-
fective rate constant keff of the rate of tryptophan photooxi-
dation in the presence of these PS and AP. At the same
time, the measure of the ability of amphiphilic polymers to
deaggregate the associates of these dyes when interacting
with MB and RB, a kind of activity series” of AP in the
process of deaggregation, does not coincide with the similar
series of AP activity during the destruction of aggregates of
water-soluble porphyrin PS.[686 -688]
Thus, according to the ability to cause deaggregation
of porphyrin water-soluble PS, in particular, photoditazine
and dimegin, that is, to increase the value of keff, am-
phiphilic polymers are arranged as follows: PVP > F127 >
> F108 > PEG > PVA. A similar pattern for MB and RB
looks somewhat different: PVA > F108 > F127 > PEG >
> PVP. These differences in the nature of the interaction of
amphiphilic polymers with porphyrin PS and dyes are ap-
parently related to the difference in the structures of
macrocyclic PS and anthracene derivatives, which have
small sizes.
The main interactions that determine the supramolecu-
lar structural organization of macrocyclic porphyrin com-
pounds (including photoditazine and dimegine) in aqueous
solutions are associated with hydrophobic, hydrogen, and
donor-acceptor bonds. At the same time, the nature of in-
termolecular interactions of anthracene derivatives should
be greatly influenced by the presence of positively charged
(MB) or electronegative (RB) groups. Indeed, anionic RB,
depending on the pH of the medium, can exist in cationic,
neutral (lactone), and anionic forms (Figure 52). In a neu-
tral photoinactive form, rose bengal exists in a slightly acid-
ic environment (pH < 5) (Figure 52,II). In a neutral and
weakly alkaline medium (pH 7.27.6), RB has a character-
istic absorption spectrum with two bands (550 and 515 nm),
corresponding to the dianionic form of RB (Figure 52,III).
Thus, RB works as a PS only in a neutral and weakly alka-
line medium (рН 7.2-7.6).[689,690] Another issue that is usu-
ally discussed in connection with the development of effec-
tive PS systems for aPDT is associated with the simultane-
ous use of biologically active molecules - proteins, proteo-
lytic enzymes, polysaccharides with their own bactericidal
activity during aPDT procedures. However, such biopoly-
mers can reduce and even suppress the activity of dyes in
1О2 photogeneration. It is shown below that one of the pos-
sible ways to solve these problems, specifically, when using
natural polysaccharides with wound healing ability - chi-
tosan (CHT) and sodium alginate (SA) in аPDT procedures
- is the formation of conjugates of PS (in this case, MB and
RB) with the indicated polysaccharides. Such conjugates
provide better penetration of PS through bacterial cell
walls, the structure of which includes polysaccharides and
their complexes with proteins,[691] directly into the cell.
a
b
Figure 51. Structural formula of methylene blue (a) and rose
bengal (b).
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 289
Cation
Lactone
Dianion
I
II
III
Figure 52. Tautomeric forms of rose Bengal.
Figure 53. Schematic representation of the formation of the RB conjugate - a fragment of CHT.
Among microbial pathogens, gram-negative bacteria
have the highest antibiotic resistance, and therefore the dis-
eases caused by such microflora are extremely difficult to
treat with antimicrobial therapy. They are also considered
to be sufficiently resistant to photodynamic effects,[692]
which is associated with the low permeability of their outer
membrane for PS. It is known that cationic PS can most
effectively interact with gram-negative bacteria.[210] Cur-
rently, PS covalently bound to polymeric polycations (natu-
ral and synthetic) are also used in aPDT. Thus, it was
shown in [693] that the treatment of deuteroporphyrin with
the polycationic peptide polymyxin B promotes the penetra-
tion of the formed complex through the membrane of gram-
negative bacteria. The use of conjugates of anionic dyes
(phthalocyanines) with cationic polylysine leads to an in-
crease in their photobactericidal activity.[694] It was also
shown that the binding of photosensitizers used in medical
practice, in particular, photohem and photoditazine, with
chitosan gel reduced the toxicity of the photosensitizer and
significantly reduced hemorrhagic phenomena in the
wound.[695-697] In this case, the interaction of polycationic
chitosan and porphyrin PS significantly reduces their activi-
ty in the processes of generation of singlet oxygen.[698] It
was shown that such a decrease is due to the aggregation of
porphyrin molecules near the protonated amino groups of
chitosan.[699,700] Moreover, as we demonstrated earlier, the
activity of PS-chitosan systems in 1О2 photogeneration in
the aqueous phase can be increased by adding AP, specifi-
cally, polyvinylpyrrolidone or Pluronic F127.[701,702] It was
shown by the proton NMR method that in the presence of
the indicated AP, the photosensitizer is coordinated with the
molecules of the amphiphilic polymer (formation due to
hydrophobic and hydrogen bonds), which promotes PS dis-
aggregation and, obviously, prevents its interaction with
chitosan molecules.[686]
At the same time, the anionic dye RB interacts with
protonated amino groups of chitosan macromolecules to
form ionically bound conjugates (Figure 53), which leads to
a decrease in the value of the effective photooxidation rate
constant keff by 2.53 times.
In addition, it was shown that RB molecules that do
not interact with chitosan macromolecules retain their pho-
tocatalytic activity. It should be emphasized that this equi-
librium can exist only in a buffer solution, where the pH for
the RB-CHT system cannot be lower than 4.54.0. When
this reaction is carried out in an aqueous solution, the intro-
duction of chitosan almost completely inhibits the process
of photocatalytic oxidation of tryptophan. Since chitosan
becomes soluble only in a slightly acidic medium (0.2
0.5% acetic acid, pH 4.5), it is also necessary to establish
the role of acetic acid in the observed effect of chitosan on
the PS activity of porphyrin and nonporphyrin photosensi-
tizers, as well as the effect of acetic acid on the structure of
the amphiphilic polymer, which determines the activity of
PS-AP systems under such conditions. It turned out that in
the case of rose bengal in an acetic acid medium, a slight
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
290 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
decrease in the value of keff (by approximately 1.3 times) is
observed, i.e. the main role in reducing the photocatalytic
activity of RB belongs to chitosan. At the same time, in the
presence of PVP, the photocatalytic activity of RB is practi-
cally restored to its original value. A similar effect on the
value of keff in the presence of chitosan is observed when
MB is used as a PS. In this case, the drop in keff is 25-35%.
A different picture is observed for PS of porphyrin and
chlorine nature. It is shown that in a solution of 0.2% acetic
acid (pH~4) both the photocatalytic activity of PPS and
their complexes with AP and the spectral properties of
dimegin and chlorin e6 change. Under these conditions, PS
is protonated to form a monoprotonated form. The photo-
catalytic activity of the monoprotonated form of DMG in-
creases by ~1.4 times, while the photoactivity of protonated
Се6 decreases by ~1.2 times.[701] Due to the decrease of the
keff value of tryptophan photooxidation catalyzed by MB
and RB in the presence of chitosan, experiments on the use
of sodium alginate as a polysaccharide (a biopolymer with
wound healing properties) were carried out; Pluronic F127,
PVA, CHT and PVP were used as amphiphilic polymers.
The obtained results are shown in Table 11.
Table 11. Values of keff for MB, RB systems in the presence of
polymers. The concentrations of polymers indicated in parentheses
are given in mass fractions. [PS] = 5·10-6 M, [Trp] = 1·10-4 M,
Trp-tryptophan.
As follows from the data in Table 11, sodium alginate,
as well as CHT, can significantly reduce the photocatalytic
activity of MB, while the addition of amphiphilic polymers
to the CHT-PS mixture can almost completely neutralize
the effect of polysaccharides. The most active in these sys-
tems are PVP, F127, and PVA. It is interesting to note that
sodium alginate in the case of RB practically does not re-
duce the value of keff. Possibly, this is due to the absence
of interaction between the SA polyanion and the anionic
RB. It should also be noted that in PS systems based on
porphyrin photosensitizers, polyvinyl alcohol is completely
inactive. Thus, in order to obtain effective PS polymer
compositions containing polysaccharides and amphiphilic
polymers, it is necessary, starting from the choice of PS, to
carry out a targeted selection of AP and polysaccharide
components.
Acknowledgements. The review was financially supported
by a number of foundations, programs and grants: Section 1
(Koifman O.I. et al.) by Ministry of Science and Higher
Education of the Russian Federation (grant agreement No
FZZW-2020-0008); Section 2 (Fedorov A.Yu. et al.) by
the Russian Science Foundation under Grant No. 21-73-
10230 (https://rscf.ru/en/project/21-73-10230/) and by State
assignment via the Research scientific laboratory of
“Chemistry of natural products and their synthetic ana-
logues” of Scientific Educational Centre “Technoplatform
2035” (FSWR-2021-014); Section 3 (Belykh D.V.) The
work was carried out within the framework of the state
tasks of the Institute of Chemistry of the Komi Research
Center of the Ural Branch of the Russian Academy of Sci-
ences (Syktyvkar) No. 122040600073-3; Section 4 (Leb-
edeva N.Sh. et al.) by the Russian Science Foundation
(grant no. 21-73-20140); Section 5 (Gorbunova Yu.G. et
al.) by the Russian Science Foundation (project no. 19-
13-00410-P); Section 6 (Dudkin S.V.) by the Ministry of
Science and Higher Education of the Russian Federation
(Contract/agreement No. 075-00697-22-00); Section 7
(Lyubimtsev A.V., Maiorova L.A. et al.) by the grant of
the Russian Science Foundation (20-12-00175), and Minis-
try of Science and Higher Education of the Russian Federa-
tion (FZZW-2020-0008, synthesis of the compounds); Sec-
tion 8 (Tyurin V.S. et al.) by the Russian Science Founda-
tion, grant 22-23-00903; Section 9 (Zenkevich E.I.) by
the BSPSR program “Photonics and Electronics for Innova-
tions (2021-2025, Belarus)”, Volkswagen Foundation (Pro-
ject “New Functionalities of Semiconductor Nanocrystals
by Controllable Coupling to Molecules”), and Visiting
Scholar Program of TU Chemnitz, Germany (E.Z., 2020-
2021); Section 10 (Berezin D.B. et al.) by Russian Foun-
dation for Basic Research, project number 20-03-00153.
Authors are grateful to the Centers for Shared Use of Scien-
tific Equipment of ISUСT (support of the Ministry of Sci-
ence and Higher Education of Russia, project number 075-
15-2021-671) and ISC RAS. The authors are grateful to Dr.
I.I. Hludeev and Dr. V.P. Zorin (Belorussian State University)
for their assistance in realization of gel-chromatographic ex-
periments. Section 11 (Grin M.A. et al.) Chemical syn-
thesis and article preparation was performed as a part of the
project “Radiopharmaceuticals” during realization of the
RTU MIREA university development program (Federal
academic leadership program Priority 2030). Biological
studies were supported by the Ministry of Science and
Higher Education of the Russian Federation (project
0706-2020-0019). NMR spectra were performed using the
equipment of the Shared Science and Training Center for
Collective Use RTU MIREA and supported by the Ministry
of Science and Higher Education of the Russian Federation.
Section 14 (Loschenov V. B. et al.) by the Ministry of
Education and Science of the Russian Federation, grant for
the creation and development of world-class research cen-
ters No. 075-15-2022- 315 “Photonics” and by the Russian
Foundation for Basic Research, grant 20-02-00928. Section
15 (Zhdanova K.A. et al.) by the Russian Science Founda-
tion (project no. 22-73-10176); Section 16 (Solovieva A.N. et
al.) by state order No. 122040400099-5 of the N.N. Se-
menov Federal Research Center for Chemical Physics RAS.
The composition of the system in the
order of mixing
keff, L/(mol·s)
MB
1500
MB, SA (0.001)
400
MB, SA (0.0001)
900
MB, PVA (0.04), SA (0.001)
800
MB, PVP (0.04), SA (0.001)
500
MB, F127 (1.30) SA (0.001)
800
MB, F127 (1.30) SA (0.0001)
1450
MB, CHT (0.01)
200
MB, F127 (1.30) CHT (0.01)
700
RB
1600
RB, SA (0.01)
1550
RB, SA, PVA
1800
RB, CHT (0.01)
50
RB, PVP (1.30), CHT (0.01)
1400
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 291
References
1. Sausville E.A. Chapter 1: Anticancer Drug Development: An
Introduction. In: Cancer Pharmacology: An Illustrated
Manual of Anticancer Drugs, Springer Publishing Company,
2019, 1-10, DOI: 10.1891/9780826162045.0001.
2. Laxmikeshav K., Kumari P., Shankaraiah N. Medicinal
Research Reviews, 2022, 42(1), 513-575, DOI:
10.1002/med.21852.
3. Liu Y., Qin R., Zaat S.A.J., Breukink E., Heger M. J. Clin.
Transl. Res. 2015, 1, 140-167.
4. Algorri J.F., Ochoa M., Roldán-Varona P., Rodríguez-Cobo
L., López-Higuera J.M. Cancers (Basel). 2021, 13.
5. Hamblin M. Curr. Opin. Microbiol. 2016, 33, 67-73.
6. Kustov A.V., Berezin D.B., Strel'nikov A.I., Lapochkina N.P.
Antitumor and antimicrobial photodynamic therapy:
mechanisms, targets, clinical and laboratory studies. A guide.
/ Ed. by A.K. Gagua. Moscow: Largo, 2020, 108 p. (in
Russian).
7. Van Straten D., Mashayekhi V., De Bruijn H.S., Oliveira S.,
Robinson D.J. Cancers 2017, 9, 1-54.
8. Caterino M., D'Aria F., Kustov A.V., Belykh D.V.,
Khudyaeva I.S., Startseva O.M., Berezin D.B., Pylina Ya.I.,
Usacheva T.R., Amato J., et al. J. Biol. Macromolecules
2020, 145, 244-251.
9. Kustov A.V., Privalov O.A., Strelnikov A.I., Koifman O.I.,
Lubimtsev A.V., Morshnev Ph.K., Moryganova T.M.,
Kustova T.V., Berezin D.B. J. Clin. Med 2022, 11, 233.
10. Bonnett R. Chem. Soc. Rev. 1995, 24, 19-33.
11. Venediktov E.A., Tulikova E.Yu., Rozhkova E.P., Belykh
D.V., Khudyaeva I.S., Berezin D.B. Macroheterocycles 2017,
10, 295-300.
12. Zenkevich E., Sagun E., Knyukshto V., Shulga A., Mironov
A., Efremova O., Bonnett R., Songca S.P., Kassem M.J.
Photochem. Photobiol. B 1996, 33, 171-180.
13. Kustov A.V., Morshnev Ph.K., Kukushkina N.V.,
Krestyaninov M.A., Smirnova N.L., Berezin D.B., Kokurina
G.N., Belykh D.V. Comptes Rendus Chimie 2022, 25, 97-102.
14. Bonnett R. Chemical Aspects of Photodynamic Therapy.
Amsterdam: Science Publishers, 2000.
15. Allison R.R., Moghissi K. Clin. Endosc. 2013, 46, 2429.
16. Gogoi A., Kao F.-J., Liu Y.-L., Zhuo G.-Y. Front. Phys.
2022. doi: 10.3389/fphy.2022.977683.
17. Kou J., Dou D. and Yang L. Oncotarget, 2017, 8, 81591-81603.
www.impactjournals.com/oncotarget/.
18. Ochsner M. J. Photochem. Photobiol. B. 1997; 39:118.
19. Zenkevich E., Sagun E., Knyukshto V., Shulga A., Mironov
A., Efremova O., Bonnett R., Pinda Songca S., Kassem M. J.
Phochem. Photobiol. B: Biol. 1996, 33, 171-180.
20. Isakau H.A., Parkhats M.V., Knyukshto V.N., Dzhagarov
B.M., Petrov E.P., Petrov P.T. J. of Photochem. Photobiol. B:
Biology. 2008, 92, 165174.
21. Parkhats M.V., Galievskiy V.A., Stasheuski A.S., Truka-
cheva T.V., Dzhagarov B.M. Optics and Spectroscopy. 2009,
107, 10261032.
22. Dadeko A., Murav’eva T.D, Starodubtsev A.M., Gorelov S.I.,
Dobrun M.V., Kris’ko T K., Bagrov I.V., Belousova I.,
Ponomarev G.V. Optics and Spectroscopy. 2015, 119, 633-637.
23. Zhang J., Jiang C., Longo J.P.F., Azevedo R.B., Zhang H.,
L.A. Muehlmann. Acta Pharmaceutica Sinica B. 2018, 8,
137146.
24. Amos-Tautua B.M., Pinda Songca S., Oluwafemi O.S. Mole-
cules. 2019, 24, 2456. doi:10.3390/molecules24132456.
25. Krasnovsky Jr. А.А., Kozlov A.S., Benditkis A.S. Macrohet-
erocycles. 2019,12, 171-180.
26. Soy R.C., Babu B., Oluwole D.O., Nwaji N., Oyim J., Amu-
haya E., Prinsloo E., Mack J., Nyokong T. J. Porphyrins
Phthalocyanines. 2019, 23, 3445.
27. Szurko A., Rams-Baron M., Montforts F.P., Bauer D., Kozub
P., Gubernator J., Altmann S., Stanek A., Sieron A., Ratuszna
A. Photodiagnosis and Photodynamic Therapy. 2020, 30,
101799. doi:https://doi.org/10.1016/j.pdpdt.2020.
28. Zhang Q., He J., Yu W., Li Y., Liu Z., Zhou B., Liu Y. RSC
Med. Chem., 2020, 11, 427437. DOI: 10.1039/c9md00558g.
29. Xiao-An Z., Lovejoy K. S., Alan J. Lippard S. J., Proc. Natl.
Acad. Sci. U. S. A., 2007, 104, 1078010785.
https://doi.org/10.1073/pnas.070239310.
30. Xiaolong L., Xiaoda L., Lijia J., Xiuli Y., Zhifei D. Bio-
materials, 2014, 35, 63796388. DOI:
10.1016/j.biomaterials.2014.04.094.
31. Lovell J. F., Pui-Chi L., Theranostics, 2012, 2, 815816. doi:
10.7150/thno.5128.
32. Huang H., Song W., Rieffel J., Lovell J. F., Front. Phys.,
2015, 3, 2329. https://doi.org/10.3389/fphy.2015.00023.
33. Bhupathiraju N. V. S. D. K., Rizvi W., Batteas J. D., Drain C.
M. Org. Biomol. Chem., 2016, 14, 389-408.
doi:10.1039/c5ob01839k.
34. Mamardashvili G.M., Mamardashvili N.Zh., Koifman O.I.
Russ. Chem. Rev. 2008, 77, 5975.
35. Koifman O.I., Mamardashvili N.Zh. Nanotechnologies in
Russia 2009, 4, 253261.
36. Beletskaya I., Tyurin V.S., Tsivadze A.Y., Guilard R., Stern
C. Chem. Rev. 2009, 109, 16591713.
37. Handbook of Porphyrin Science, ed. K. M. Kadish, K. M.
Smith and R. Guilard, World Scientific Publishing Co., Sin-
gapore, 2010, vol. 4.
38. Handbook of Porphyrin Science, ed. K. M. Kadish, K. M.
Smith and R. Guilard, World Scientific Publishing Co., Sin-
gapore, 2014, vol. 27.
39. P. M. R. Pereira, J. P. C. Tomé and R. Fernandes, Molecular
Targeted Photodynamic Therapy for Cancer, in.
40. Handbook of Porphyrin Science, ed. K. M. Kadish, K. M.
Smith and R. Guilard, World Scientific Publishing Co., Sin-
gapore, 2016, vol. 39, pp. 127169.
41. M. Lupu, C. D. Thomas, F. Poyer, J. Mispelter, V. Rosilio
and P. Maillard, Photobiology and Photochemistry Handin-
Hand in Targeted Antitumoral Therapies, in Handbook of
Porphyrin Science, ed. K. M. Kadish, K. M. Smith and R.
Guilard, World Scientific Publishing Co., Singapore, 2016,
vol. 39, pp. 171356.
42. Mfouo-Tynga I.S., Dias L.D., Inada N.M., Kurachi, C. Pho-
todiagnosis and Photodynamic Therapy, 2021, 34, 102091,
DOI: 10.1016/j.pdpdt.2020.102091.
43. Patent RU 2 128 993 C1, 1999.
44. Lovell J. F., Liu T. W. B., Chen J., Zheng G. Chem. Rev.,
2010, 110, 28392857. doi:10.1021/cr900236h.
45. Celli J. P., Spring B. Q., Rizvi I., Evans C. L., Samkoe K. S.,
Verma S., Pogu B.W., Hasan T. Chem. Rev., 2010. 110,
27952838. doi:10.1021/cr900300p.
46. Ethirajan M., Chen Y., Joshi P., Pandey R.K. Chem. Soc.
Rev., 2011, 40, 340362.
47. Ormond A.B., Freeman H.S. Materials 2013, 6, 817-840.
doi:10.3390/ma6030817.
48. Habermeyer B., Guilard R. Photochem. Photobiol. Sci., 2018,
17, 6751690. DOI: 10.1039/c8pp00222c.
49. Gomes A.T.P.C., Neves M.G.P.M.S., Cavaleiro J.A.S. An
Acad Bras Cienc 2018. 90 (1 Suppl. 2): 993-1026.
http://dx.doi.org/10.1590/0001-3765201820170811.
50. Li X., Lee S., Yoon J. Chem. Soc. Rev., 2018, 47, 11741188.
DOI: 10.1039/c7cs00594f.
51. Zhao X., Liu J., Fan J., Chao H., Peng X. Chem. Soc. Rev.,
2021, 50, 41854219. DOI: 10.1039/d0cs00173b.
52. Tian J., Huang B., Nawaz M.H., Zhang W. Coord. Chem.
Rev., 2020, 420, 213410.
53. Wang B., Zu G., Sun Y., Zhang Y., Wang X., Han J., Zhang
C., J. Liaon. Shihua Univ. 2020, 40 2938. DOI:
10.3969/j.issn.1672-6952.2020.06.007.
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
292 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
54. Park W., Cho S., Han J., Shin H., Na K., Lee B., Kim D.H.
Biomater. Sci., 2018, 6, 79-90. DOI: 10.1039/c7bm00872d.
55. Rajora M.A., Louac J.W.H., Zheng G. Chem. Soc. Rev.,
2017, 46, 64336469. DOI: 10.1039/c7cs00525c.
56. Fernandes S.R.G., Fernandes R., Sarmento B., Pereira P. M.
R., Tomé J.P.C. Org. Biomol. Chem., 2019, 17, 25792593.
DOI: 10.1039/c8ob02902d.
57. Zhou J., Rao L., Yu G., Cook T.R., Chen X., Huang F. Chem.
Soc. Rev., 2021, 50, 2839-2891. DOI: 10.1039/d0cs00011f.
58. H. Zhang, J. Han Org. Biomol. Chem., 2020, 18, 48944905.
DOI: 10.1039/d0ob00763c.
59. Hao M., Chen B., Zhao X., Zhao N., Xu F.-J. Mater. Chem.
Front., 2020, 4, 25712609 DOI: 10.1039/d0qm00323a.
60. Escudero, A., Carrillo-Carrión, C., Castillejos, M. C., Romero-
Ben, E., Rosales-Barrios, C., & Khiar, N. A. Mater. Chem.
Front., 2021, 5, 37883812. DOI: 10.1039/d0qm00922a.
61. Zheng, Q., Liu, X., Zheng, Y., Yeung, K. W. K., Cui, Z.,
Liang, Y., Li Z., Zhu S., Wang X., Wu, S. Chem. Soc. Rev.,
2021, 50, 50865125. DOI: 10.1039/d1cs00056j.
62. Policard A., Compt. Rend. Soc. Biol., 1924, 91, 14231424.
63. Auler H., Banzer G., Z. Krebsforsch., 1942, 53, 6568.
64. Lipson R.L., Pratt J.H., Baldes E.J., Dockerty M.B., Obstet.
Cynecol., 1964, 24, 7884.
65. Dougherty T.J., Kaufman J.E., Goldfarb A., Weishaupt K.R.,
Boyle D., Mittleman A., Cancer. Res., 1978, 38, 26282635.
66. Filonenko E. V. Russ. J. Gen. Chem., 2015, 85, 211216.
DOI: 10.1134/S1070363215010399.
67. Patent RU 2183635 C2, 1999.
68. Patent RU 2183956, 2002.
69. Patent RU 2276976, 2004.
70. Patent RU 2523380, 2014.
71. Morozova N.B., Pankratov A.A., Plotnikova E.A., Vorontso-
va M.S., Makarova E., Lukyanets E.A., Kaprin. A. Res.&
Prac. Medi. J. 2019, 6 DOI:10.17709/2409-2231-2019-6-4-7.
72. Fukuzumi S., Lee Y.-M., Nam W. ChemPhotoChem 2018, 2,
121 135.
73. Lee H., Hong K.I., Jang W.D. Coordination Chemistry Re-
views. 2018. 354, 46-73.
74. Zenkevich E.I., von Borczyskowski C. Photoinduced relaxa-
tion processes in self-assembled nanostructures: multiporphy-
rin complexes and composites “CdSе/ZnS quantum dot-
porphyrin”. In: “Multiporphyrin Arrays: Fundamentals and
Applications” (D. Kim, Ed.) Singapore: Pan Stanford Pub-
lishing Pte. Ltd., 2012, Chapter 5, 217-28.
75. Koifman O.I., Ageeva T.A., Beletskaya I.P., Averin A.D.,
Yakushev A.A., Tomilova L.G., Dubinina T.V., Tsivadze
A.Yu., Gorbunova Yu.G., Martynov A.G., Konarev D.V.,
Khasanov S.S., Lyubovskaya R.N., Lomova T.N., Korolev
V.V., Zenkevich E.I., Blaudeck T. , Ch. von Borczyskowski,
Zahn D.R.T., Mironov A.F., Bragina N.A., Ezhov A.V.,
Zhdanova K.A., Stuzhin P.A., Pakhomov G.L., Rusakova
N.V., Semenishyn N.N., Smola S.S., Parfenyuk V.I., Vashu-
rin A.S., Makarov S.V., Dereven’kov I.A., Mamardashvili
N.Zh., Kurtikyan T.S., Martirosyan G.G., Burmistrov V.А.,
Aleksandriiskii V.V., Novikov I.V., Pritmov D.A., Grin
M.A., Suvorov N.V., Tsigankov A.A., Fedorov A.Yu.,
Kuzmina N.S., Nyuchev A.V., Otvagin V.F., Kustov A.V.,
Belykh D.V., Berezin D.B., Solovieva A.B., Timashev P.S.,
Milaeva E.R., Gracheva Yu.A., Dodokhova M.A., Safronen-
ko A.V., Shpakovsky D.B., Syrbu S.A., Gubarev Yu.A.,
Kiselev A.N., Koifman M.O., Lebedeva N.Sh., Yurina E.S.
Macroheterocyclic Compounds a Key Building Block in
New Functional Materials and Molecular Devic-
es Macroheterocycles 2020, 13, 311-467.
76. Jing H., Rong J., Taniguchi M., Liey J.S. Coord. Chem. Rev.
2022, 456, 214278. https://doi.org/10.1016/j.ccr.2021.214278.
77. Huang L., Asghar S., Zhu T., Ye P., Hu Z., Chen Z., Xiao
Y. Expert Opinion on Drug Delivery. 2021, 18, 1473-1500.
78. Chen J., Fan T., Xie Z., Zeng Q., Xue P., Zheng T., Chen Y.,
Luo X., Zhang H. Biomaterials. 2020, 237, 119827. doi:
10.1016/j.biomaterials.2020.119827.
79. Zenkevich E.I., Sagun E.I., Knyukshto V.N., Stasheuski A.S.,
Galievsky V.A., Stupak A.P., Blaudeck T., von Borczyskow-
ski C. J. Phys. Chem. C. 2011, 115, 21535-21545.
80. Zenkevich E., von Borczyskowski C. (Eds.), Self-Assembled
Organic-Inorganic Nanostructures: Optics and Dynamics.
Singapore: Pan Stanford, 2016.
81. Martynenko, I.V., Orlova, A.O., Maslov V.G., Baranov A.V.,
Fedorov A.V., Artemyev M. Beilstein J. Nanotechnol. 2013,
4, 895902.
82. Rakovich Yu.P. “Organic-Inorganic Hybrid Nanosystems for
Photodynamic Therapy”. The 2nd International Symposium
on Physics, Engineering and Technologies for Biomedicine,
KnE Energy & Physics, 2018, 416419. doi:
10.18502/ken.v3i2.1845.
83. Moghassemi S., Dadashzadeh A., Azevedo R.B., Feron O.,
Amorim C.A. J. Control Release. 2021, 339, 75-90.
84. Yakavets I., Millard M., Zorin V., Lassalle H.P., Bezdetnaya
L. J. Control Release. 2019, 304, 268-287.
85. Sun C.Y, Cao Z., Zhang X.J., Sun R., Yu C.S., Yang X.
Theranostics. 2018, 18, 2939-295.
86. Sewid F.A., Annas K.I., Dubavik A., Veniaminov A.V.,
Maslov V.G., Orlova A.O. RSC Advances. 2022, 12, 899-906.
87. Rybkin A.Y., Belik A.Y., Goryachev N.S., Mikhaylov P.A.,
Kraevaya O.A., Filatova N.V., Parkhomenko I.I., Peregudov
A.S., Terent'ev A.A., Larkina E.A., Mironov A F., Troshin
P.A., Kotelnikov A.I. Dyes and Pigments, 2020, 180,
108411. doi.org/10.1016/j.dyepig.2020.108411.
88. Zenkevich E., Blaudeck T., Sheinin V., Kulikova O., Sely-
shchev O., Dzhagan V., Koifman O., von Borczyskowski C.,
Zahn D.R.T. J. of Molec. Structure. 2021, 1244, 131239.
doi.org/10.1016/ j.molstruc. 2021. 131239.
89. Yu X.-T., Sui S.-Y., He Y.X., Yu C.-H., Peng Q. Biomater
Adv. 2022, 135, 212725. doi: 10.1016/j.bioadv.2022.212725.
90. Mokhtari R. B., Homayouni T. S., Baluch N., Morgatskaya E.,
Kumar S., Das B., Yeger H. Oncotarget. 2017, 8, 3802238043.
https://doi.org/10.18632/oncotarget.16723.
91. Khdair A., Chen D., Patil Y., Ma L., Dou Q.P., Shekhar
M.P., Panyam J. J. Control. Release. 2010, 141, 137144.
https://doi.org/10.1016/j.jconrel.2009.09.004.
92. Otvagin V.F., Kuzmina N.S., Kudriashova E.S., Nyuchev
A.V., Gavryushin A.E., Fedorov A.Yu. J. Med. Chem. 2022,
65 16951734. https://doi.org/10.1021/acs.jmedchem.1c01953.
93. Hu X., Ogawa K., Li S., Kiwada T., Odani A. Chem. Lett.
2017, 46, 764766. https://doi.org/10.1246/cl.170095.
94. Thapa P., Li M., Bio M., Rajaputra P., Nkepang G., Sun Y.,
Woo S., You Y. J. Med. Chem. 2016, 59 32043214.
https://doi.org/10.1021/acs.jmedchem.5b01971.
95. Kwiatkowski S., Knap B., Przystupski D., Saczko J.,
Kędzierska E., Knap-Czop K., Kotlińska J., Michel O., Ko-
towski K., Kulbacka Ju. Biomed. Pharmacother. 2018, 106,
10981107. https://doi.org/10.1016/j.biopha.2018.07.049.
96. Hiyama K., Matsui H., Tamura M., Shimokawa O., Hiyama M.,
Kaneko T., Nagano Y., Hyodo I., Tanaka J., Miwa Y., Ogawa
T., Nakanishi T., Tamai I. J. Porphyrins Phthalocyanines. 2013,
17 3643. https://doi.org/10.1142/S1088424612501192.
97. Chen Y., Zheng X., Dobhal M.P., Gryshuk A., Morgan J.,
Dougherty T.J., Oseroff A., Pandey R.K., Methyl Pyrophe-
ophorbide-a Analogues: Potential Fluorescent Probes for the
Peripheral-Type Benzodiazepine Receptor. Effect of Central
Metal in Photosensitizing Efficacy, J. Med. Chem. 2005, 48,
36923695. https://doi.org/10.1021/jm050039k.
98. Dąbrowski J.M., Pucelik B., Regiel-Futyra A., Brindell
M., Mazuryk O., Kyzioł A., Stochel G., Macyk W., Ar-
naut L.G. Coord. Chem. Rev. 2016, 325, 67101.
https://doi.org/10.1016/j.ccr.2016.06.007.
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 293
99. Ye Y., Wang L.-X., Zhang D.-P., Yan Y.-J., Chen Z.-L. J.
Innov. Opt. Health Sci. 2015, 8, 1540001.
https://doi.org/10.1142/S1793545815400015.
100. Dąbrowski J.M., Arnaut L.G. Photochem. Photobiol. Sci.
2015, 14, 17651780. https://doi.org/10.1039/C5PP00132C.
101. Pinto da Silva L., Magalhães C.M., Núñez-Montenegro A.,
Ferreira P.J.O., Duarte D., Rodríguez-Borges J.E., Vale N.,
Esteves da Silva J.C.G. Biomolecules, 2019, 9, 384.
https://doi.org/10.3390/biom9080384.
102. Li Q., Li W., Di H., Luo L., Zhu C., Yang J., Yin X., Yin H.,
Gao J., Du Y., You J. J. Control Release. 2018, 277, 114125.
https://doi.org/10.1016/j.jconrel.2018.02.001.
103. Ulfo L., Costantini P.E., Di Giosia M., Danielli A., Cal-
varesi M., Pharmaceutics. 2022, 14, 241.
https://doi.org/10.3390/pharmaceutics14020241.
104. Nyuchev A.V., Otvagin V.F., Gavryushin A.E., Roma-
nenko Y.I., Koifman O.I., Belykh D.V., Schmalz H.-G.,
Fedorov A.Yu. Synthesis. 2015, 47, 37173726.
https://doi.org/10.1055/s-0034-1378876.
105. Hu X., Ogawa K., Li S., Kiwada T., Odani A. Bull. Chem. Soc.
Jpn. 2019, 92 790796. https://doi.org/10.1246/bcsj.20180382.
106. Cheruku R.R., Cacaccio J., Durrani F.A., Tabaczynski
W.A., Watson R., Siters KMissert., J.R., Tracy E.C., Dukh
M., Guru K., Koya R.C., Kalinski P., Baumann H., Pan-
dey R.K. J. Med. Chem. 2021, 64, 741−767.
https://doi.org/10.1021/acs.jmedchem.0c01735.
107. Huang L., Wei G., Sun X., Jiang Y., Huang Z., Huang Y.,
Shen Y., Xu X., Liao Y., Zhao C. Eur. J. Med. Chem. 2018,
151, 294303. https://doi.org/10.1016/j.ejmech.2018.03.077.
108. Simões J.C.S., Sarpaki S., Papadimitroulas P., Therrien B.,
Loudos G. J. Med. Chem. 2020, 63, 1411914150.
https://doi.org/10.1021/acs.jmedchem.0c00047.
109. Zhao X., Ma H., Chen J., Zhang F., Jia X., Xue J. Eur. J. Med.
Chem. 2019,182, 111625. https://doi.org/10.1016/j.ejmech.
2019.111625.
110. Otvagin V.F., Nyuchev A.V., Kuzmina N.S., Grishin I.D.,
Gavryushin A.E., Romanenko Y.V., Koifman O.I., Belykh
D.V., Peskova N.N., Shilyagina N.Y., Balalaeva I.V., Fedo-
rov A.Yu. Eur. J. Med. Chem. 2018, 144, 740750.
https://doi.org/10.1016/j.ejmech.2017.12.062.
111. Otvagin V.F., Kuzmina N.S., Krylova L.V., Volovetsky
A.B., Nyuchev A.V., Gavryushin A.E., Meshkov I.N., Gor-
bunova Y.G., Romanenko Y.V., Koifman O.I., Balalaeva
I.V., Fedorov A.Yu., J. Med. Chem. 2019, 62, 1118211193.
https://doi.org/10.1021/acs.jmedchem.9b01294.
112. Huang K., Niu Y., Yuan G., Yan M., Xue J., Chen J. Sens.
Actuators B Chem. 2022, 355, 131275.
https://doi.org/10.1016/j.snb.2021.131275.
113. Musallam L., Ethier C., Haddad P.S., Bilodeau M. EGF me-
diates protection against Fas-induced apoptosis by depleting
and oxidizing intracellular GSH stocks, J. Cell. Physiol.
2004, 198, 6272. https://doi.org/10.1002/jcp.10389.
114. Zhang W., Lu J., Gao X.N., Li P., Zhang W., Ma Y., Wang
H., Tang B. Angew. Chem. Int. Ed. 2018, 57, 4891-4896.
https://doi.org/10.1002/anie.201710800.
115. Brave S.R., Odedra R., James N.H., Smith N.R., Marshall G.B.,
Acheson K.L., Baker D., Howard Z., Jackson L., Ratcliffe K.,
Wainwright A., Lovick S.C., Hickinson D.M., Wilkinson R.W.,
Barry S.T., Speake G., Ryan A.J. Int. J. Oncol. 2011, 39,
271278. https://doi.org/10.3892/ijo.2011.1022.
116. Leriche G., Chisholm L., Wagner A. Bioorganic Med. Chem.
2012, 20, 571582. https://doi.org/10.1016/j.bmc.2011.07.048.
117. Krylova L.V., Peskova N.N., Otvagin V.F., Kuzmina N.S.,
Nyuchev A.V., Fedorov A.Yu., Balalaeva I.V., Opera Med.
Physiol. 2022, 9, 514. https://doi.org/10.24412/2500-2295-
2022-3-5-14.
118. Kuzmina N.S., Otvagin V.F., Krylova L.V., Nyuchev A.V.,
Romanenko Yu.V., Koifman O.I., Balalaeva I.V., Fedorov
A.Yu. Mendeleev Commun. 2020, 30, 159161.
https://doi.org/10.1016/j.mencom.2020.03.009.
119. Singh S., Aggarwal A., Bhupathiraju N.V.S.D.K., Arianna
G., Tiwari K., Drain C.M., Glycosylated Porphyrins, Phthal-
ocyanines, and Other Porphyrinoids for Diagnostics and
Therapeutics, Chem. Rev. 2015 ,115, 1026110306.
https://doi.org/10.1021/acs.chemrev.5b00244.
120. Zaki M., Arjmand F., Tabassum S. Inorganica Chim. Acta.
2016, 444, 1-22122. https://doi.org/10.1016/j.ica.2016.01.006.
121. Muhammad N., Guo Z. Curr Opin Chem Biol. 2014, 144153.
https://doi.org/10.1016/j.cbpa.2014.02.003.
122. Hadi A.G., Jawad K., Ahmed D.S., Yousif E. Syst. Rev. Pharm.
2019, 10, 2631. https://doi.org/10.5530/srp.2019.1.5.
123. Khan A., Parveen S., Khalid A., Shafi S. Inorg. Chim. Acta.
2020, 505, 119464. https://doi.org/10.1016/j.ica.2020.119464.
124. Shaheen F., Sirajuddin M., Ali S., Rehman Z.-U., Dyson P.J.,
Shah N.A., Tahir M.N. J. Organomet. Chem. 2018, 856, 1322.
https://doi.org/10.1016/j.jorganchem.2017.12.010.
125. Tikhonov S., Ostroverkhov P., Suvorov N., Mironov A.,
Efimova Yu., Plutinskaya A., Pankratov A., Ignatova A.,
Feofanov A., Diachkova E., Vasil’ev Yu., Grin M. Int. J. Mol.
Sci. 2021, 22, 13563. https:// doi.org/10.3390/ijms222413563.
126. Witt O., Deubzer H. E., Milde T., Oehme I. Cancer Lett. 2009,
277, 821. https://doi.org/10.1016/j.canlet.2008.08.016.
127. Eckschlager T., Plch J., Stiborova M., Hrabeta J. Int. J. Mol.
Sci. 2017, 18, 1414. https://doi.org/10.3390/ijms18071414.
128. Halsall J.A., Turner B.M., BioEssays. 2016, 38, 11021110.
https://doi.org/10.1002/bies.201600070.
129. Banik D., Noonepalle S., Hadley M., Palmer E., Gracia-
Hernandez M., Zevallos-Delgado C., Manhas N., Simonyan
H., Young C.N., Popratiloff A., Chiappinelli K.B., Fernandes
R., Sotomayor E.M., Villagra A. Cancer Res. 2020, 80,
36493662. https://doi.org/10.1158/0008-5472.CAN-19-
3738.
130. Aru B., Günay A., Şenkuytu E., Yanıkkaya Demirel G., Gü-
rek A.G., Atilla D. ACS Omega. 2020, 5, 25854−25867.
https://doi.org/10.1021/acsomega.0c03180.
131. Rakha E.A., El-Sayed M.E., Green A.R., Lee A.H.S., Robert-
son J.F., Ellis I.O., Cancer. 2007, 109, 2532.
https://doi.org/10.1002/cncr.22381.
132. Aru B., Günay A., Yanıkkaya Demirel G.Y., Gürek A.G.,
Atilla D. RSC Adv. 2021, 11, 3496334978.
https://doi.org/10.1039/D1RA05404J.
133. Thomas C.J., Rahier N.J., Hecht S.M. Bioorg. Med. Chem. 2004,
12, 15851604. https://doi.org/10.1016/j.bmc.2003.11.036.
134. Zhang H.-X., Lin H.-H., Su D., Yang D.-C., Liu J.-Y., Mol.
Pharmaceutics. 2022, 19, 630641. https://doi.org/10.1021/
acs.molpharmaceut.1c00761.
135. Pettit G.R., Singh S.B., Hamel E., Lin C.M., Alberts D.S.,
Garcia-Kendal D. Experientia. 1989, 45, 209211.
https://doi.org/10.1007/BF01954881.
136. Nam N.H., Combretastatin A-4 analogues as antimitotic anti-
tumor agents, Curr. Med. Chem. 2003, 10, 16971722.
https://doi.org/10.2174/0929867033457151.
137. Bio M., Rajaputra P., Nkepang G., Awuah S.G., Hossion
A.M.L., You Y. J. Med. Chem. 2013, 56, 39363942.
https://doi.org/10.1021/jm400139w.
138. Ha S.Y.Y., Zhou Y., Fong W.P., Ng D.K.P. J. Med. Chem. 2020,
63, 85128523. https://doi.org/10.1021/acs.jmedchem.0c00893.
139. Chou T.C. Pharmacol. Rev. 2006, 58, 621681.
https://doi.org/10.1124/pr.58.3.10.
140. Gaukroger K., Hadfield J., Lawrence N.J., Nolan S.,
McGown A.T. Org. Biomol. Chem. 2003, 1, 30333037.
https://doi.org/10.1039/B306878A.
141. Hadfield J.A., McGown A.T., Mayalarp S.P., Land E.J.,
Hamblett I., Gaukroger K., Lawrence N.J., Hepworth L.A.,
Butler J., Substituted Stilbenes and Their Reactions,
US7220784B2, 2007.
142. Klán P., Šolomek T., Bochet C.G., Blanc A., Givens R., Ru-
bina M., Popik V., Kostikov A., Wirz J., Chem. Rev. 2012,
113, 119−191. https://doi.org/10.1021/cr300177k.
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
294 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
143. Kuzmina N.S., Otvagin V.F., Maleev A.A., Urazaeva M.A.,
Nyuchev A.V., Ignatov S.K., Gavryushin A.E., Fedorov
A.Yu. J. Photochem. Photobiol. A. 2022, 433, 114138.
https://doi.org/10.1016/j.jphotochem.2022.114138.
144. Lin W., Peng D., Wang B., Long L., Guo C., Yuan J. Eur. J.
Org. Chem. 2008, 2008, 793796. https://doi.org/10.1002
/ejoc.200700972.
145. Sheldon J.E., Dcona M.M., Lyons C.E., Hackett J.C., Hart-
man M.C.T. Org. Biomol. Chem. 2016, 14, 4049.
https://doi.org/10.1039/c5ob02005k.
146. Malatesti N., Munitic I., Jurak I. Biophys Rev. 2017, 9, 149168.
DOI: 10.1007/s12551-017-0257-7.
147. Feng Y., Tonon C.C., Ashraf Sh., Hasan T. Adv. Drug Delivery
Rev. 2021, 177, 113941. DOI: 10.1016/j.addr.2021.113941.
148. Pucelik B., Sułek A., Dabrowski J. M. Coord. Chem. Rev.
2020, 416, 213340 DOI: 10.1016/j.ccr.2020.213340.
149. Sharma S. K., Dai T., Kharkwal G.B., Huang Y.-Y., Huang
L., Bil De Arce V. J., Tegos G. P., Hamblin M. R. Curr.
Pharm. Des. 2011, 17(13), 13031319. DOI:
10.2174/138161211795703735.
150. Sobotta L., Skupin-Mrugalska P., Piskorz J., Mielcarek J.
Eur. J. Med. Chem. 2019, 175, 72-106. DOI:
10.1016/j.ejmech.2019.04.057.
151. Dharmaratne P., Sapugahawatte D.N., Wang B., Chan C.L., Lau
K.-M., Lau C.B.S., Fung K.P., KP D., IP M. Eur. J. Med. Chem.
2020, 200, 112341. DOI: 10.1016/j.ejmech.2020.112341.
152. Takahashi T., Ogasawara Sh., Shinozaki Y., Tamiaki H.
Eur. J. Org. Chem. 2019, 37, 6333-6340. DOI:
10.1002/ejoc.201901172.
153. Suvorov N., Pogorilyy V., Diachkova E., Vasil’ev Y.,
Mironov A., Grin M. Int. J. Mol. Sci. 2021, 22, 6392. DOI:
10.3390/ijms22126392.
154. Kustov A.V., Morshnev P.K., Kukushkina N.V., Smirnova
N.L., Berezin D.B., Karimov D.R., Shukhto O.V., Kustova
T.V., Belykh D.V., Mal’shakova M.V., Zorin V. P., Zorina T.
E. Int. J. Mol. Sci. 2022, 23, 5294. DOI:
10.3390/ijms23105294.
155. Cai J.-Q., Liu X.-M., Gao Z.-J., Li L.-L., Wang H. Materials
Today D 2021, 45, 77-92. DOI: 10.1016/j.mattod.2020.11.001.
156. Awad M., Thomas N., Barnes T.J., Prestidge C.A. J. Con-
troll. Release 2022, 346, 300316. DOI:
10.1016/j.jconrel.2022.04.035.
157. Anas A., Sobhanan J., Sulfiya K.M., Jasmin C., Sreelakshmi
P.K., Biju V. Journal of Photochemistry and Photobiology,
C: Photochemistry Reviews 2021, 49, 100452. DOI:
10.1016/j.jphotochemrev.2021.100452.
158. Aroso R. T., Piccirillo G., Arnaut Z. A., Gonzalez A.C.S., Ro-
drigues F.M.S., Pereira M. M. Journal of Photochemistry
and Photobiology 2021, 7, 100043 DOI:
10.1016/j.jpap.2021.100043.
159. Luciano M., Brückner C. Molecules 2017, 22, 980. DOI:
10.3390/molecules22060980.
160. Wiehe A., O’Brien J. M., Senge M. O. Photochem. Photobi-
ol. Sci. 2019, 18, 2565-2612 DOI: 10.1039/c9pp00211a.
161. Huang L., Wang M., Huang Y.-Y., El-Hussein A., Wolf L.,
Chiang L. Y., Hamblin M. R. Photochem Photobiol Sci.
2018, 17, 638651. DOI: 10.1039/c7pp00389g.
162. Tegos G. P., Anbe M., Yang Ch., Demidova T. N., Satti M.,
Mroz P., Janjua S., Gad F., Hamblin M. R. Antimicrobial
Agents and Chemotherapy 2006, 50, 14021410. DOI:
10.1128/AAC.50.4.14021410.2006.
163. Le Guern F., Ouk T.-S., Grenier K., Joly N., Lequartb V., Sol
V. J. Mater. Chem. B 2017, 5, 69536962 DOI:
10.1039/c7tb01274h.
164. Yin R., Dai T., Avci P., Jorge A. E. S., CMA de Melo W.,
Vecchio D., Huang Y.-Y., Gupta A., Hamblin M. R. Curr.
Opin. Pharmacol. 2013, 13, 132. DOI:
10.1016/j.coph.2013.08.009.
165. Huang L., Huang Y.-Y., Mroz P., Tegos G. P., Zhiyentayev
T., Sharma S. K., Lu Z., Balasubramanian T., Krayer M.,
Ruzie C., Yang E., Kee H. L., Kirmaier C., Diers J. R., Boci-
an D. F., Holten D., Lindsey J. S., Hamblin M. R. Antimicro-
bial Agents and Chemotherapy 2010, 38343841 DOI:
10.1128/AAC.00125-10.
166. Taima H., Okubo A., Yoshioka N., Inoue H. Tetrahedron
Lett. 2005, 46, 41614164 DOI: 10.1016/j.tetlet.2005.04.069
167. Ryazanova O., Voloshin I., Dubey I., Dubey L., Zozulya V.
Ann. N.Y. Acad. Sci. 2008, 1130, 293299. DOI:
10.1196/annals.1430.033.
168. Miyatake T., Hasunuma Y., Mukai Y., Oki H., Watanabe M.,
Yamazaki S. Bioorg. Med. Chem. 2016, 24, 11551161.
DOI: 10.1016/j.bmc.2016.01.054.
169. Miyatake T., Tamiaki H., Shinoda H., Fujiwara M., Matsu-
shita T. Tetrahedron 2002, 58, 998910000. DOI:
10.1016/S0040-4020(02)01328-5.
170. Saenz C., Ethirajan M., Tracy E. C., Bowman M.-J., Cacac-
cio J., Ohulchanskyy T., Baumann H., Pandey R. K. J. Pho-
tochem.& Photobiol., B: Biology 2022, 227, 112375 DOI:
10.1016/j.jphotobiol.2021.112375.
171. Mironov A.F., Nechaev A.V. Russ. J. Bioorg. Chem. 2003,
29, 9698. DOI: 10.1023/A:1022299007111.
172. Mironov A.F., Nechaev A.V. Russ. J. Bioorg. Chem. 2001,
27, 120123. DOI: 10.1023/A:1011337304402.
173. Nechaev A.V., Mironov A.F. Russ. J. Bioorg. Chem. 2008,
34, 245251. DOI: 10.1134/S1068162008020167.
174. Gushchina O.I., Gramma V.A., Larkina E.A., Mironov
A.F. Mendeleev Commun. 2017, 27, 5052. DOI:
10.1016/j.mencom.2017.01.015.
175. Mal’shakova M.V., Frolova L.L., Alekseev I.N., Kutchin A.
V., Patov S. A., Belykh D. V. Russ. Chem. Bull. 2018, 67,
14671475. DOI: 10.1007/s11172-018-2241-1.
176. Khudyaeva I.S., Shevchenko O.G., Belykh, D.V. Russ.
Chem. Bull. 2020, 69, 742750. DOI: 10.1007/s11172-020-
2827-2.
177. Gradova M. A., Movchan T. G., Khudyaeva I. S.,
Chernyad’ev A. Yu., Plotnikova E. V., Lobanov A. V., Be-
lykh D. V. Macroheterocycles 2020, 13, 23-32 DOI:
10.6060/mhc191072g.
178. Pylina Y. I., Khudyaeva I. S., Startseva O. M., Shadrin D.
M., Shevchenko O. G., Velegzhaninov I. O., Kukushkina N.
V., Berezin D. B., Belykh D. V. Macroheterocycles 2021, 14,
317-322. DOI: 10.6060/mhc210944b.
179. Gushchina O. I., Larkina E. A., Mironov A. F. Macrohetero-
cycles 2014, 7, 414-416. DOI: 10.6060/mhc140931g.
180. Li J., Zhang X., Liu Y., Yoon I., Kim D.-K., Yin J.-G., Wang
J.-J., Shim Y. K. Bioorg. Med. Chem. 2015, 23, 16841690.
DOI: 10.1016/j.bmc.2015.03.021.
181. Gushchina O.I. «Sintez i svoystva amidnykh proizvodnykh
khlorinovogo ryada», diss. kand. khim. nauk, 02.00.10
Bioorganicheskaya khimiya. Moskva, 2019, 130 s (in Russ).
182. Mironov A.F., Ruziev R.D., Lebedeva V.S. Russ. J. Bioorg.
Chem. 2004, 30, 466476. DOI: 10.1023/B:RUBI.000004
3791.62162.42.
183. Pantiushenko I.V., Rudakovskaya P.G., Starovoytova A.V. et
al. Biochemistry Moscow 2015, 80, 752762. DOI:
10.1134/S0006297915060103.
184. Mironov A. F., Grin M. A., Tsiprovskii A. G., Titeev R. A.,
Nizhnik E. A., Lonin I. S. Mendeleev Commun. 2004, 14,
204-207. DOI: 10.1070/MC2004v014n05ABEH001941.
185. Grin M.A., Mironov A.F. Russ. Chem. Bull. 2016, 65, 333349.
DOI: 10.1007/s11172-016-1307-1.
186. Mal’shakova M. V., Belykh D. V. J. Porphyrins Phthalocya-
nines 2022, 26, 403406. DOI: 10.1142/S108842462250033X.
187. Takahashi T., Ogasawara Sh., Shinozaki Y., Tamiaki H.
Bull. Chem. Soc. Jpn. 2020, 93, 467476. DOI:
10.1246/bcsj.20190367.
188. Ogasawara S., Tamiaki H. Bull. Chem. Soc. Jpn. 2018, 91,
1724-1730. DOI: 10.1246/bcsj.20180243.
189. Pandey R. K., Smith N. W., Shiau F.-Y., Dougherty T. J., Smith
K. M. J. Chem. Soc., Chem. Commun. 1991, 1637-1638. DOI:
10.1039/C39910001637.
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 295
190. Pandey R.K., Shiau F.U., Smith N.N., Dougherty D.J., Smith
K.M. Tetrahedron 1992, 48, 7591-7600. DOI:
10.1016/S0040-4020(01)90371-0.
191. Pavlov V.Y., Ponomarev G.V. Chem. Heterocycl. Comp. 2004,
40, 393-425. DOI: 10.1023/B:COHC.0000033531.25694.2e.
192. Ponomarev G.V. Chem. Heterocycl. Compounds 1997, 33,
11271166. DOI: 10.1007/BF02290864.
193. Belykh D.V. Russ. J. Gen. Chem. 2019, 89, 26042649. DOI:
10.1134/S1070363219120430.
194. Belykh D.V., Tarabukina I.S., Gruzdev I.V., Kodess M.I.,
Kutchin A.V. J. Porphyrins Phthalocyanines 2009, 13, 949956.
DOI: 10.1142/S1088424609001133.
195. Belykh D.V., Tarabukina I.S., Gruzdev I.V., Kuchin A.V.
Macroheterocycles 2010, 3, 145-149. DOI: 10.6060/mhc2010.2-
3.145.
196. Belykh D. V., Khudyaeva I. S., Startceva O. M., Gruzdev I.
V., Romanenko Y. V. Macroheterocycles 2016, 9, 366-372.
DOI: 10.6060/mhc160540b.
197. Tarabukina I.S., Startseva O.M., Patov S.A., Belykh D.V.
Macroheterocycles 2015, 8, 168-176. DOI:
10.6060/mhc150456b.
198. Kustov A.V., Kustova T.V., Belykh D.V., Khudyaeva I.S.,
Berezin D.B. Dyes and Pigments 2020, 173, 107948. DOI:
10.1016/j.dyepig.2019.107948.
199. Stamati I., Kuimova M. K., Lion M., Yahioglu G., Phillipsb
D., Deonarain M. P. Photochem. Photobiol. Sci. 2010, 9,
1033-1041. DOI: 10.1039/C0PP00038H.
200. Phillips D. Pure Appl. Chem. 2011, 83, 733748. DOI:
doi:10.1351/PAC-CON-11-01-05.
201. Stamati I. Targeted Photodynamic Therapy of cancer using
photoimmunoconjugates based on pyropheophorbide a deriv-
atives. A thesis submitted in fulfilment of the requirements
for the degree of Doctor of Philosophy and the Diploma of
Imperial College London. Division of Cell and Molecular
Biology Faculty of Natural Sciences Imperial College, Lon-
don September 2010.
202. Li J.Z., Wang J.J., Yoon I., Cui B.C., Shim Y.K. Bioorg.
Med. Chem. Lett. 2012, 22, 18461849. DOI:
10.1016/j.bmcl.2012.01.088.
203. Casteel M. J., Jayaraj K., Gold A., Ball L. M., Sobsey M. D.
Photochem. Photobiol. 2004, 80, 294.
204. Costa L., Alves E., Carvalho C., Tomé J. P., Faustino M. A.,
Neves M. G., Tome A. C., Cavaleiro J. A., Cunha Â., Al-
meida A. Photochem. Photobiol. Sci. 2008, 7, 415.
205. Martins D., Mesquita M. Q., Neves M. G., Faustino M. A.,
Reis L., Figueira E., Almeida A. Planta 2018, 248, 409.
206. Lebedeva N. S., Koifman O. Russ. J. Bioorg. Chem. 2022,
48, 1.
207. Lebedeva N., Gubarev Y., Koifman M., Koifman O. Mole-
cules 2020, 25, 4368.
208. Ergaieg K., Seux R. Desalination 2009, 246, 353.
209. Merchat M., Bertolini G., Giacomini P., Villaneuva A., Jori
G. J. Photochem. Photobiol. B: Biol. 1996, 32, 153
210. Hamblin M. R., Hasan T. Photochem. Photobiol. Sci. 2004,
3, 436.
211. Costerton J., Ingram J., Cheng K. Bacteriol. Rev. 1974, 38,
87.
212. Nikaido H. Rev. Infect. Dis. 1988, S279.
213. Hamblin M. R., O’Donnell D. A., Murthy N., Rajagopalan
K., Michaud N., Sherwood M. E., Hasan T. J. Antimicrob.
Chemother. 2002, 49, 941.
214. Lebedeva N. S., Yurina E. S., Lubimtsev A. V., Gubarev Y.
A., Syrbu S. A. Chem. Phys. Lett. 2022, 140090.
215. Lebedeva N. S., Yurina E. S., Gubarev Y. A., Syrbu S. A.
Spectrochim. Acta, Part A 2018, 199, 235.
216. Rapacka-Zdończyk A., Woźniak A., Michalska K., Pierański
M., Ogonowska P., Grinholc M., Nakonieczna J. Front. Med.
2021, 8, 642609.
217. Vaara M. Microbiol. Rev. 1992, 56, 395.
218. Carvalho C. M., Gomes A. T., Fernandes S. C., Prata A. C.,
Almeida M. A., Cunha M. A., Tomé J. P., Faustino M. A.,
Neves M. G., Tomé A. C. J. Photochem. Photobiol. B: Biol.
2007, 88, 112.
219. Alves E., Costa L., Carvalho C., Tomé J. P., Faustino M. A.,
Neves M. G., Tomé A. C., Cavaleiro J. A., Cunha Â., Al-
meida A. BMC Microbiol. 2009, 9, 1.
220. Spesia M. B., Lazzeri D., Pascual L., Rovera M., Durantini E.
N. Fems Immunol. Med. Mic. 2005, 44, 289.
221. Gyulkhandanyan A. G., Paronyan M. H., Gyulkhandanyan A.
G., Ghazaryan K. R., Parkhats M. V., Dzhagarov B. M.,
Korchenova M. V., Lazareva E. N., Tuchina E. S.,
Gyulkhandanyan G. V. J. Innov. Opt. Health Sci. 2022, 15,
2142007.
222. Reddi E., Ceccon M., Valduga G., Jori G., Bommer J. C.,
Elisei F., Latterini L., Mazzucato U. Photochem. Photobiol.
2002, 75, 462.
223. Boyle R. W., Dolphin D. Photochem. Photobiol. 1996, 64,
469.
224. Lazzeri D., Rovera M., Pascual L., Durantini E. N. Photo-
chem. Photobiol. 2004, 80, 286.
225. Marciel L., Mesquita M. Q., Ferreira R., Moreira B., PMS
Neves M. G., F Faustino M. A., Almeida A. Future Med.
Chem 2018, 10, 1821.
226. Goncalves P. J., Bezzerra F. C., Teles A. V., Menezes L. B.,
Alves K. M., Alonso L., Alonso A., Andrade M. A., Boris-
sevitch I. E., Souza G. R. J. Photochem. Photobiol. A: Chem.
2020, 391, 112375.
227. Ribeiro C. P., Gamelas S. R., Faustino M. A., Gomes A. T.,
Tome J. P., Almeida A., Lourenco L. M. Photodiagn.
Photodyn. Ther. 2020, 31, 101788.
228. Ribeiro C. P., Faustino M. A., Almeida A., Lourenço L. M.
Microorganisms 2022, 10, 718.
229. Maisch T., Bosl C., Szeimies R.-M., Lehn N., Abels C. Anti-
microb. Agents Chemother. 2005, 49, 1542.
230. Guterres K. B., Rossi G. G., Moreira K. S., Burgo T. A. L.,
Iglesias B. A. BioMetals 2020, 33, 269.
231. Guterres K. B., Rossi G. G., de Campos M. M. A., Moreira
K. S., Burgo T. A. L., Iglesias B. A. Photodiagn. Photodyn.
Ther. 2022, 38, 102770.
232. Rossi G. G., Guterres K. B., Moreira K. S., Burgo T. A. L.,
de Campos M. M. A., Iglesias B. A. Photodiagn. Photodyn.
Ther. 2021, 36, 102514.
233. Nitzan Y., Ashkenazi H. Curr. Microbiol. 2001, 42, 408.
234. Oriel S., Nitzan Y. Photochem. Photobiol. 2012, 88, 604.
235. Gubarev Y. A., Lebedeva N. S., Yurina E. S., Syrbu S. S.,
Kiselev A. N., Lebedev M. A. JPA 2021.
236. Koifman O. I., Lebedeva N. S., Gubarev Y. A., Koifman M.
O. Chem. Heterocycl. Compd. 2021, 57, 423.
237. Lebedeva N. S., Gubarev Y. A., Mamardashvili G. M.,
Zaitceva S. V., Zdanovich S. A., Malyasova A. S., Romanen-
ko J. V., Koifman M. O., Koifman O. I. Scientific reports
2021, 11, 1.
238. Koifman M., Malyasova A., Romanenko Y. V., Yurina E.,
Lebedeva N. S., Gubarev Y. A., Koifman O. Spectrochim.
Acta, Part A 2022, 279, 121403.
239. Gubarev Y. A., Lebedeva N. S., Yurina E. S., Mamardashvili
G. M., Zaitceva S. V., Zdanovich S. A., Koifman O. I. J. Bi-
omol. Struct. Dyn. 2022, 1.
240. Kiselev A. N., Syrbu S. A., Lebedeva N. S., Gubarev Y. A.
Inorganics 2022, 10, 63.
241. Kiselev A., Lebedev M., Syrbu S., Yurina E., Gubarev Y.,
Lebedeva N., Beljanina N., Shirokova I., Kovalishena O.,
Koifman O. Russ. Chem. Bull. 2022, 71 (in press).
242. Lebedeva N. S., Gubarev Y. A., Lyubimtsev A. V., Yurina E.
S., Koifman O. I. Macroheterocycles 2017, 10, 37.
243. Syrbu S.A., Kiselev A., Lebedev M.A., Gubarev Y.A., Yurina
E.S., Lebedeva N.S. Russ. J. Gen. Chem. 2022, 92, 10051010.
https://doi.org/10.1134/S1070363222060123
244. Shang J., Ye G., Shi K., Wan Y., Luo C., Aihara H., Geng
Q., Auerbach A., Li F. Nature 2020, 581, 221.
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
296 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
245. DeRosa M. C., Crutchley R. J. Coord. Chem. Rev. 2002,
233234, 351371.
246. Nyokong T., Ahsen V., Photosensitizers in Medicine, Envi-
ronment and Security, Springer, 2012.
247. Dąbrowski J. M., Pucelik B., Regiel-Futyra A., Brindell M.,
Mazuryk O., Kyzioł A., Stochel G. G., Macyk W., Arnaut L.
G., DeRosa M. C., Crutchley R. J., Gouterman R., Sekkat N.,
Van Den Bergh H., Nyokong T., Lange N., Molecules, 2012,
17, 98144.
248. Nyokong T., Pure Appl. Chem., 2011, 83, 17631779.
249. Gouterman R., J. Mol. Spectrosc., 1968, 27, 225235.
250. Martynov A. G., Mack J., May A. K., Nyokong T., Gorbuno-
va Y. G., Tsivadze A. Y., ACS Omega, 2019, 4, 72657284.
251. Pereira G. F. M., Tasso T. T., Inorganica Chim. Acta, 2021,
519, 120271.
252. Ogunsipe A., Maree D., Nyokong T., J. Mol. Struct., 2003,
650, 131140.
253. Juríček M., Kouwer P. H. J., Rehák J., Sly J., Rowan A. E., J.
Org. Chem., 2009, 74, 2125.
254. Dilber G., Durmuş M., Kantekin H., Dye. Pigment., 2019,
160, 267284.
255. Kollar J., Machacek M., Halaskova M., Lenco J., Kucera R.,
Demuth J., Rohlickova M., Hasonova K., Miletin M., Nova-
kova V., Zimcik P., J. Med. Chem., 2020, 63, 76167632.
256. Bunin D. A., Martynov A. G., Safonova E. A., Tsivadze A.
Y., Gorbunova Y. G., Dye. Pigment., 2022, 207, 110768.
257. Halaskova M., Rahali A., Almeida-Marrero V., Machacek
M., Kucera R., Jamoussi B., Torres T., Novakova V., De La
Escosura A., Zimcik P., ACS Med. Chem. Lett., 2021, 12,
502507.
258. Lioret V., Saou S., Berrou A., Lernerman L., Arnould C.,
Decréau R. A., Photochem. Photobiol. Sci., 2022, 06.
259. Schneider L., Larocca M., Wu W., Babu V., Padrutt R.,
Slyshkina E., König C., Ferrari S., Spingler B., Photochem.
Photobiol. Sci., 2019, 18, 27922803.
260. Kobayashi N., Furuyama T., Satoh K., J. Am. Chem. Soc.,
2011, 133, 1964219645.
261. Furuyama T., Satoh K., Kushiya T., Kobayashi N., J. Am.
Chem. Soc., 2014, 136, 765776.
262. Yoshida T., Furuyama T., Kobayashi N., Tetrahedron Lett.,
2015, 56, 16711674.
263. Furuyama T., Yoshida T., Hashizume D., Kobayashi N.,
Chem. Sci., 2014, 5, 24662474.
264. Kolomeychuk F. M., Safonova E. A., Polovkova M. A.,
Sinelshchikova A. A., Martynov A. G., Shokurov A. V.,
Kirakosyan G. A., Efimov N. N., Tsivadze A. Y., Gorbunova
Y. G., J. Am. Chem. Soc., 2021, 143, 1405314058.
265. Safonova E. A., Martynov A. G., Nefedov S. E., Kirakosyan
G. A., Gorbunova Y. G., Tsivadze A. Y., Inorg. Chem., 2016,
55, 24502459.
266. Dumoulin F., Durmuş M., Ahsen V., Nyokong T., Coord.
Chem. Rev., 2010, 254, 27922847.
267. Ribeiro C. P. S., Lourenço L. M. O., J. Photochem. Photobi-
ol. C Photochem. Rev., 2021, 48.
268. Anaya-Plaza E., Aljarilla A., Beaune G., Nonappa, Timonen
J. V. I., de la Escosura A., Torres T., Kostiainen M. A., Adv.
Mater., 2019, 31.
269. Nene L. C., Magadla A., Nyokong T., J. Photochem. Photo-
biol. B Biol., 2022, 235, 112553.
270. Scalise I., Durantini E. N., Bioorganic Med. Chem., 2005, 13,
30373045.
271. Sindelo A., Kobayashi N., Kimura M., Nyokong T., J. Pho-
tochem. Photobiol. A Chem., 2019, 374, 5867.
272. Meshkov I. N., Bulach V. V., Gorbunova Y. G., Gostev F. E.,
Nadtochenko V. A., Tsivadze A. Y., Hosseini M. W., Chem.
Commun., 2017, 53, 99189921.
273. Meshkov I. N., Bulach V., Gorbunova Y. G., Kyritsakas N.,
Grigoriev M. S., Tsivadze A. Y., Hosseini M. W., Inorg.
Chem., 2016, 55, 1077410782.
274. Stuzhin P. A., J. Porphyr. Phthalocyanines, 1999, 03, 500513.
275. Donzello M. P., Ercolani C., Gaberkorn A. A., Kudrik E. V.,
Meneghetti M., Marcolongo G., Rizzoli C., Stuzhin P. A.,
Chem. - A Eur. J., 2003, 9, 40094024.
276. Safonova E. A., Meshkov I. N., Polovkova M. A., Vo-
lostnykh M. V., Tsivadze A. Y., Gorbunova Y. G., Mendele-
ev Commun., 2018, 28, 275277.
277. Martynov A. G., Safonova E. A., Tsivadze A. Y., Gorbunova
Y. G., Coord. Chem. Rev., 2019, 387, 325347.
278. Chen Y., Li G., Pandey R.K. Curr. Org. Chem. 2004, 8,
1105-1134.
279. Huang Y.-Y., Luo D., Hamblin M.R. Curr. Org. Chem. 2015,
19, 948-957.
280. Martinez De Pinillos Bayona A., Mroz P., Thunshelle C.,
Hamblin M.R. Chem. Biol. Drug Design, 2017, 89, 192-206.
281. Aggarwal A., Samaroo D., Jovanovic I.R., Singh S., Tuz M.P.,
Mackiewicz M.R. J. Porphyrins Phthalocyanines 2019, 23.
282. Senge M.O., Brandt J.C. Photochem. Photobiol. 2011, 87,
1240-1296., 729-765.
283. Senge M.O. Photodiagn. PDT 2012, 9, 1700-179.
284. Hu Z., Rao B., Chen S., Duanmu J. BMC Cancer 2010, 10, 1-13.
285. Mazor O., Brandis A., Plaks V., Neumark E., Rosenbach-
Belkin V., Salomon Y., Scherz A. Photochem. Photobiol.
2005, 81, 342-351.
286. Scherz A., Salomon Y., Lindner U., Coleman J. Comprehen.
Ser. Photochem. Photobiol. Sci. 2016, 15, 461-480.
287. Gouterman M. In The Porphyrins. (Ed D.Dolphin). Academic
Press, New York, 1978, 3 1-165.
288. Juselius J., Sundholm D. Phys. Chem. Chem. Phys. 2000, 2,
2145-2151.
289. Otero N., Fias S., Rodenković S., Bultinck P., Grana A.M.,
Mandado M. Chem. Eur. J. 2011, 17, 3274-3286.
290. Fliegel H., Sundholm D. J. Org. Chem. 2012, 77, 3408-3414.
291. Moss G.P. Pure Appl. Chem. 1987, 59, 779-832.
292. Lindsey J.S. Chem. Rev. 2015, 115, 6534-6620.
293. Dudkin S.V, Makarova E.A., Lukyanets E.A. Russ. Chem.
Rev. 2016, 85, 700-730.
294. Taniguchi M., Lindsey J.S. Chem. Rev. 2017, 117, 344-535.
295. Whitlock Jr H.W., Hanauer R., Oester M.Y., Bower B.K. J.
Am. Chem. Soc. 1969, 91, 7485-7489.
296. Konan Y. N., Gurny R., Alleman E. J. Photochem. Photobiol.
B. Biol. 2002, 66, 89-106.
297. Pushpan S.K., Venkatraman S., Anand V.G., Sankar J., Par-
meswaran D., Ganesan S., Chandrashekar T.K. Curr. Med.
Chem. Anti-Cancer Agents, 2002, 2, 187-207.
298. Allison R.R., Downie G.H., Cuenca R., Hu X.-H., Childs
C.J.H., Sibata C.H. Photodiagn. PDT. 2004, 1, 27-42.
299. Wainwright M. Photosensitisers in biomedicine. Wiley-
Blackwell, 2009, 269 p.
300. Patent RU 2479585 C1 (2013).
301. Patent US 6410586 B1 (2002).
302. Lyubimtsev A.V., Semeikin A.S., Syrbu S.A., Koifman O.I.
Book of abstracts of the 14th International conference «Syn-
thesis and application of porphyrins and their analogues(
ICPC-14)». Ivanovo, 2022, 47.
303. Riyad Y.M., Naumov S., Schastak S., Griebel J., Kahnt A.,
Hupl T., Neuhaus J., Abel B., Hermann R. J. Phys. Chem. B
2014, 118, 11646−11658.
304. Sharma S.K., Mroz P., Dai T., Huang Y.-Y., Denis T. G. St.,
Hamblin M.R. Isr. J. Chem. 2012, 52, 691-705.
305. Dąbrowski J. M. Adv. Inorg. Chem. 2017, 70, 343-394.
306. Oertel M. Schastak S.I., Tannapfel A., Hermann R., Sack U.,
Mössner J., Berr F. J. Photochem. Photobiol. B Biol. 2003,
71, 1-10.
307. Schastak S., Jean B., Handzel R., Kostenich G., Her-
mann R., Sack U., Orenstein A., Wang Y., Wiedemann P. J.
Photochem. Photobiol. B Biol. 2005, 78, 203-213.
308. Yakubovskaya R.I., Plotnikova E.A., Plyutinskaya A.D.,
Morozova N.B., Chissov V.I., Makarova E.A., Dudkin S.V.
Lukyanets E.A., Vorozhtsov G.N. J. Photochem. Photobiol. B
Biol. 2014, 130, 109-114.
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 297
309. Schastak S., Gitter B., Handzel R., Hermann R., Wiedemann
P. Methods Find. Exp. Clin. Pharmacol. 2008, 30, 129-133.
310. Schastak S, Ziganshyna S, Gitter B, Wiedemann P, Claudep-
ierre T. PLoS ONE 2010, 5, e11674.
311. Patent RU 2663900 C1 (2018).
312. Pankratov A.A., Andreeva T.N., Plotnikova E.A., Morozova
N.B., Vorontsova M.S., Merkulova I.B., Abramova T.V.,
Starkova N.N., Kalinichenko V.N., Yakubovskaya R.I. Russ.
J. Biotherapy, 2017, 16(s1), 60.
313. Plyutinskaya A.D., Plotnikova E.A., Stramova V.O., Yaku-
bovskaya R.I. Russ. J. Biotherapy, 2017, 16(s1), 63.
314. Maklygina Yu.S., Sharova A.S., Kundu B., Balla V.K., Stei-
ner R., Loschenov V.B. Biomedical Photonics 2016, 5, 4-12.
315. Patent RU 2537759 C1 (2013).
316. Patent RU 2535097 C1 (2014).
317. Koifman O.I., Ponomarev G.V., Sergeeva T.V., Ivanov A.V.,
Tsitrin E.B. Russ. J. Biotherapy, 2017, 16(s1), 43.
318. Sergeeva T.V., Zharov E.V., Bogatyrev O.P., Ivanov A.V.,
Zinov’ev S.V., Koifman O.I., Krasnovsky A.A. Russ. J. Bio-
therapy, 2015, 14(1), С. 130.
319. Rzhevskii D.I., Ivanov A.V., Rodionov I.V., Dyachenko I.A.,
Murashev A.N. Biomedical Photonics, 2019, 8(4s), 35-36.
320. Ghorbani J., Rahban D., Aghamiri Sh., Teymouri A., Baha-
dor A. Laser Ther. 2018, 27(4), 293302.
https://doi.org/10.5978/islsm.27_18-RA-01.
321. Collins T.L., Markus E.A., Hassett D.J., Robinson J.B. Cur-
rent microbiology 2010, 61(5), 411-416.
322. Cieplik F., Tabenski L., Buchalla W., Maisch T. Frontiers in
microbiology 2014, 5, 405.
323. Patent RU 2523380 C1, 2014.
324. Patent RU 2647588 C1, 2018.
325. Meng Sh., Xu Z., Hong G., Zhao L., Zhao Zh., Guo J., Ji H.,
Liu T. Eur. J. Med. Chem. 2015, 92, 35-48.
326. Parthiban V., Yen P. Y. M., Uruma Y., Lai P-Sh. Bull. Chem.
Soc. Jpn. 2020, 93, 978-984.
327. Khan R., Özkan M., Khaligh A., Tuncel D. Photochem. Pho-
tobiol. Sci. 2019, 18, 1147-1155.
328. Hamblin M.R., Chiang L.Y., Lakshmanan S., Huang Y-Y.,
Garcia-Diaz M., Karimi M., et al. Nanotechnology reviews
2015, 4(4), 359-72.
329. Tutt L.W., Boggess T.F. Progress in quantum electronics
1993, 17(4), 299-338.
330. Algethami N., Sadeghi H., Sangtarash S., Lambert C. J. Nano
Lett. 2018, 18, 4482−4486.
331. Bellamy-Carter A., Roche C., Anderson H. L., Saywell A.
Sci. Rep. 2021, 11, 20388.
332. .Wang D., Niu L., Qiao Z. Y., Cheng D. B., Wang J., Zhong
Y. A. CS Nano 2018, 12, 37963803.
333. Stoffelen C., Huskens J. Soft Small 2016, 12, 96−119.
334. Ariga K., Nishikawa M., Mori T., Takeya J., Shrestha L. K.,
Hill J. P. Sci. Technol. Adv. Mater. 2019, 20, 5195.
335. Webre W. A., Gobeze H. B., Shao S., Karr P. A., Ariga K.,
Hill J. P., D’Souza F. Chem Commun. 2018, 54, 13511354.
336. Oldacre A. N., Friedman A. E., Cook T. R. J. Am. Chem. Soc.
2017, 139, 1424−1427.
337. Huang Z., Qin B., Chen L., Xu J. F., Faul C. F., Zhang X.
Macromol. Rapid Commun. 2017, 38, 1700312−1700326.
338. Yang L., Tan X., Wang Z., Zhang X. Chem. Rev. 2015, 115,
7196−7239.
339. Rubia-Payá C., De Miguel G., Martín-Romero M. T., Giner-
Casares J. J., Camacho L. Adv. Colloid Interface Sci. 2015,
225, 134−145.
340. Kuzmin S.M., Chulovskaya S.A., Parfenyuk V.I. Electro-
chimica Acta 2020, 342, 136064.
341. Mon M., Bruno R., Ferrando-Soria J., Armentano D., Pardo
E. J. Mater. Chem. A 2018, 6, 49124947.
342. Maiorova L. A., Kobayashi N., Zyablov S. V., Bykov V. A.,
Nesterov S. I., Kozlov A. V., Koifman O. I. Langmuir 2018,
34(31), 9322-9329.
343. Karlyuk M.V., Krygin Y.Y., Maiorova-Valkova L.A., Ageeva
T.A., Koifman O.I. Russ. Chem. Bull. 2013, 62(2), 471-479.
344. Vu T.T., Maiorova L.A., Berezin D.B., Koifman O.I. Macro-
heterocycles 2016, 9(1), 73-79.
345. Valkova L.A., Glibin A.S., Koifman O.I. Macroheterocycles
2011, 4(3), 222-226.
346. Valkova L.A., Erokhin V.V., Glibin A.S., Koifman O.I. J.
Porphyrins Phthalocyanines .2011, 15(9-10), 1044-1051.
347. Valkova L.A., Shabyshev L.S., Feigin L.A., Akopova O.B.
Molecular crystals and liquid crystals science and technology.
Section C, Molecular materials Print 1996, 6(4), 291-298.
348. Maiorova L.A., Koifman O.I., Burmistrov V.A., Kuvshinova
S.A., Mamontov A.O. Protection of Metals and Physical
Chemistry of Surfaces 2015, 51(1), 85-92.
349. Valkova L.A, Shabyshev L.S., Feigin L.A., Akopova O.B.
Izvestiya Akademii Nauk Seriya Fizicheskaya 1997,
61(3):631-636.
350. Kharitonova N.V., Maiorova L.A., Koifman O.I. J. Porphy-
rins Phthalocyanines 2018, 22(06), 509-520.
351. Gromova O.A., Maiorova L.A., Salnikov D.S., Demidov V.I.,
Kalacheva A.G., Torshin I.Yu., Bogacheva T.E., Gromov
A.N., Limanova O.A., Grishina T.R., Jafari S.M., Koifman
O.I. BioNanoSci. 2022, 12, 7482.
352. Erokhina S., Pastorino L., Lisa D. Di, Kiiamov A.G., Tayur-
skii D.A., Iannotta S., Erokhin V., Faizullina A.R. MethodsX
2021, 8,101230.
353. Rikhtegarana S., Katouzianbc I., Jafari S.M., Kiani H, Ma-
iorova L.A., Takbirgou H. Food Structure 2021, 30, 100227.
354. Maiorova L.A., Erokhina S.I., Pisani M., Barucca G., Marcac-
cio M. Colloids Surf. B. 2019, 182C, 110366.
355. Semeikin A.S., Koifman O.I., Berezin B.D. Chem Heterocycl
Compd 1982. 18, 10461047 /https://doi.org/10.1007/
BF00503191.
356. Dudkin S., Makarova E., Slivka L., Lukyanets E. J. Porphy-
rins Phthalocyanines 2014, 18, 107-114.
357. Koifman O. I.; Ageeva T. A. Russ. J. Org. Chem. 2022, 58 (4),
443-479 DOI: 10.1134/S1070428022040017.
358. Beletskaya I. P.; Tyurin V. S.; Uglov A.; Stern C.; Guilard R. In
Handbook of Porphyrin Science; Kadish, K. M., Smith, K. M.,
Guilard, R., Ed.; World Scientific: Singapore, 2012; Chapter 108,
Vol. 23, pp. 81-279. DOI: 10.1142/9789814397605_0010.
359. Vicente M. d. G. H.; Smith K. M. Curr. Org. synt. 2014, 11
(1), 3-28 DOI: 10.2174/15701794113106660083.
360. Senge M. O. Chem. Commun. 2011, 47 (7), 1943-1960 DOI:
10.1039/C0CC03984E.
361. Ponomarev G. V. Chem. Heterocycl. Comp. 1994, 30 (11),
1444-1465 DOI: 10.1007/BF01172868.
362. Burrell A. K.; Officer D. L. Synlett 1998, 1998 (12), 1297-1307
DOI: 10.1055/s-1998-1938.
363. Ando A.; Yamazaki M.; Komura M.; Sano Y.; Hattori N.;
Omote M.; Kumadaki I. Heterocycles 1999, 50 (2), DOI:
10.3987/com-98-s(h)90.
364. Dahms K.; Senge M. O.; Bakri Bakar M. Eur. J. Org. Chem.
2007, (23), 3833-3848 DOI: 10.1002/ejoc.200700380.
365. Runge S.; Senge M. O. Tetrahedron 1999, 55 (34), 10375-10390.
DOI: 10.1016/S0040-4020(99)00579-7.
366. Arnold D.P.; Johnson A.W.; Mahendran M. J. Chem. Soc.,
Perkin Trans. 1 1978, (4), 366-370 DOI:
10.1039/P19780000366.
367. Smith K.M. In Synthesis and Modifications of Porphyrinoids;
Paolesse, R., Ed.; Springer Berlin Heidelberg: Berlin,
Heidelberg, 2014; pp 1-34. DOI: 10.1007/7081_2013_100.
368. Tkachenko N. V.; Lemmetyinen H.; Sonoda J.; Ohkubo K.;
Sato T.; Imahori H.; Fukuzumi S. J. Phys. Chem. A 2003, 107
(42), 8834-8844 DOI: 10.1021/jp035412j.
369. Fuhrhop J.-H.; Witte L.; Sheldrick W. S. Justus Liebigs
Annalen der Chemie 1976, 1976 (9), 1537-1559 DOI:
10.1002/jlac.197619760904.
370. Ponomarev G.V. Chem. Heterocycl. Comp. 1996, 32 (11),
1263-1280 DOI: 10.1007/BF01169958.
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
298 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
371. Paolesse R.; Nardis S.; Monti D.; Stefanelli M.; Di Natale C.
Chem. Rev. 2017, 117 (4), 2517-2583 DOI:
10.1021/acs.chemrev.6b00361.
372. Nyman E. S.; Hynninen, P. H. Research advances in the use
of tetrapyrrolic photosensitizers for photodynamic therapy.
Journal of Photochemistry and Photobiology B: Biology
2004, 73 (12), 1-28 DOI: 10.1016/j.jphotobiol.2003.10.002.
373. Sternberg E. D.; Dolphin D.; Brückn C. Tetrahedron 1998,
54 (17), 4151-4202 DOI: 10.1016/S0040-4020(98)00015-5.
374. Cerqueira A. F. R.; Moura N. M. M.; Serra V. V.; Faustino M. A.
F.; Tomé A. C.; Cavaleiro J. A. S.; Neves M. G. P. M. S.
Molecules 2017, 22 (8), 1269. DOI: 10.3390/molecules22081269.
375. Vicente M. G. H.; Smith K. M. J. Org. Chem. 1991, 56 (14),
4407-4418 DOI: 10.1021/jo00014a016.
376. Glowacka-Sobotta A.; Wrotynski M.; Kryjewski M.; Sobotta
L.; Mielcarek J. J. Porphyr. Phthalocyanines 2019, 23
(01n02), 1-10 DOI: 10.1142/s108842461850116x.
377. Moreira L. M.; Vieira dos Santos F.; Lyon J. P.; Maftoum-
Costa M.; Pacheco-Soares C.; Soares da Silva N. Austral. J.of
Chem. 2008, 61 (10), 741-754 DOI: 10.1071/CH08145.
378. Morgan A.; Garbo G.; Rampersaud A.; Skalkos D.; Keck R.;
Selman S. Photodynamic Action of Benzochlorins. SPIE:
1989; Vol. 1065.
379. Li G.; Pandey S. K.; Graham A.; Dobhal M. P.; Mehta R.;
Chen Y.; Gryshuk A.; Rittenhouse-Olson K.; Oseroff A.;
Pande R. K. J. Org. Chem. 2004, 69 (1), 158-172. DOI:
10.1021/jo030280b.
380. Belyaev E. S.; Kozhemyakin G. L.; Tyurin V. S.; Frolova V.
V.; Lonin I. S.; Ponomarev G. V.; Buryak A. K.; Zamilatskov
I. A. Org. & Biomol. Chem. 2022, 20 (9), 1926-1932. DOI:
10.1039/D1OB02005F.
381. Orlova E. A.; Romanenko Y. V.; Tyurin V. S.; Shkirdova A.
O.; Belyaev E. S.; Grigoriev M. S.; Koifman O. I.;
Zamilatskov I. A. Macroheterocycles 2022, 15 (3), in press.
382. van der Haas, Richard N. S.; de Jong, Robertus L. P.;
Noushazar M.; Erkelens K.; Smijs, Threes G. M.; Liu Y.;
Gast P.; Schuitmaker Hans J.; Lugtenburg J. Eur.J. Org.
Chem. 2004, 2004 (19), 4024-4038. DOI:
10.1002/ejoc.200400366.
383. Morgan A. R.; Rampersaud A.; Garbo G. M.; Keck R. W.;
Selman S. H. J.Med. Chem. 1989, 32 (4), 904-908. DOI:
10.1021/jm00124a029.
384. Muthiah C.; Taniguchi M.; Kim H.-J.; Schmidt I.; Kee H. L.;
Holten D.; Bocian D. F.; Lindsey J. S. Photochem.&
Photobiol. 2007, 83 (6), 1513-1528. DOI: 10.1111/j.1751-
1097.2007.00195.x.
385. Wang Q.; Ma F.; Tang W.; Zhao S.; Li C.; Xie Y. Dyes Pigm.
2018, 148, 437-443 DOI: 10.1016/j.dyepig.2017.09.046.
386. Chen B.; Ding Y.; Li X.; Zhu W.; Hill J. P.; Ariga K.; Xie Y.
Chem. Commun. (Cambridge, U. K.) 2013, 49 (86), 10136-10138.
DOI: 10.1039/c3cc46008h.
387. Shkirdova A. O.; Orlova E. A.; Ponomarev G. V.; Tyurin V.
S.; Zamilatskov I. A.; Buryak A. K. Macroheterocycles 2021,
14 (3), 208-212. DOI: 10.6060/mhc211046z.
388. Tyurin, V. S.; Erzina, D. R.; Zamilatskov, I. A.; Chernyadyev, A.
Y.; Ponomarev, G. V.; Yashunskiy, D. V.; Maksimova, A. V.;
Krasnovskiy, A. A.; Tsivadze, A. Y. Macroheterocycles 2015, 8
(4), 376-383. DOI: 10.6060/mhc151199z.
389. Ponomarev G. V.; Rozynov B. V. Chem. Heterocycl.Comp.
1973, 9 (9), 1065-1068 DOI: 10.1007/BF00474772.
390. Yashunsky D. V.; Morozova Y. V.; Ponomarev G. V. Chem.
Heterocycl. Comp. 2000, 36 (4), 485-486 DOI:
10.1007/BF02269554.
391. Yashunsky D. V.; Morozova Y. V.; Ponomarev G. V.
Chem.Heterocycl. Comp. 2000, 36 (4), 487-488 DOI:
10.1007/BF02269555.
392. Yashunsky D. V.; Morozova Y. V.; Ponomarev G. V. Chem.
Heterocycl. Comp. 2001, 37 (3), 380-381 DOI:
10.1023/A:1017587906810.
393. Morozova Y. V.; Yashunsky D. V.; Maksimov B. I.;
Ponomarev G. V. Chem. Heterocycl. Comp. 2003, 39 (3),
388-389 DOI: 10.1023/A:1023935415004.
394. Volov A. N.; Zamilatskov I. A.; Mikhel I. S.; Erzina D. R.;
Ponomarev G. V.; Koifman O. I.; Tsivadze A. Y.
Macroheterocycles 2014, 7 (3), 256-261 DOI:
10.6060/mhc140274z.
395. Erzina D. R.; Zamilatskov I. A.; Stanetskaya N. M.; Tyurin V.
S.; Kozhemyakin G. L.; Ponomarev G. V.; Chernyshev V. V.;
Fitch A. N. Eur. J. Org. Chem. 2019, 2019 (7), 1508-1522 DOI:
10.1002/ejoc.201801659.
396. Papkovsky D. B.; Ponomarev G. V.; Wolfbeis O. S. J.
Photochem & Photobiol A: Chemistry 1997, 104 (1), 151-158
DOI: 10.1016/S1010-6030(97)04592-9.
397. Borchert N. B.; Ponomarev G. V.; Kerry J. P.; Papkovsky
D. B. Analyt. Chem. 2011, 83 (1), 18-22 DOI:
10.1021/ac1025754.
398. Zhdanov A. V.; Li L.; Yang P.; Shkirdova A. O.; Tang S.;
Yashunsky D. V.; Ponomarev G. V.; Zamilatskov I. A.;
Papkovsky D. B. Sensor. & Actuators B: Chemical 2022,
355, 131116 DOI: 10.1016/j.snb.2021.131116.
399. Ponomarev G. V.; Shul'ga A. M. Chem. Heterocycl. Comp.
1984, 20 (4), 383-388 DOI: 10.1007/BF00513851.
400. Ponomarev G. V.; Shul'ga A. M. Chem. Heterocycl. Comp
1987, 23 (7), 757-762 DOI: 10.1007/BF00475643.
401. Ponomarev G. V.; Shul'ga A. M.; Rozynov B. V. Chem.
Heterocycl. Comp 1993, 29 (2), 155-162 DOI:
10.1007/BF00531657.
402. Shkirdova A. O.; Zamilatskov I. A.; Stanetskaya N. M.;
Tafeenko V. A.; Tyurin V. S.; Chernyshev V. V.;
Ponomarev, G. V.; Tsivadze A. Y. Macroheterocycles 2017,
10 (4), 480-486 DOI: 10.6060/mhc171148z.
403. Kozhemyakin G. L.; Tyurin V. S.; Shkirdova A. O.; Belyaev
E. S.; Kirinova E. S.; Ponomarev G. V.; Chistov A. A.;
Aralov A. V.; Tafeenko V. A.; Zamilatskov I. A. Org. &
Biomolec. Chem. 2021, 19 (42), 9199-9210 DOI:
10.1039/D1OB01626A.
404. Harper S. R.; Pfrunder M. C.; Esdaile L. J.; Jensen P.;
McMurtrie J. C.; Arnold D. P. Eur.J. Org. Chem. 2015, 2015
(13), 2807-2825 DOI: doi:10.1002/ejoc.201500183.
405. Dahlstedt E.; Collins H. A.; Balaz M.; Kuimova M. K.;
Khurana M.; Wilson B. C.; Phillips D.; Anderson H. L. Org.
& Biomolec. Chem. 2009, 7 (5), 897-904 DOI:
10.1039/b814792b.
406. Belyaev E. S.; Shkirdova A. O.; Kozhemyakin G. L.; Tyurin
V. S.; Emets V. V.; Grinberg V. A.; Cheshkov D. A.;
Ponomarev G. V.; Tafeenko V. A.; Radchenko A. S.;
Kostyukov A. A.; Egorov A. E.; Kuzmin V. A.; Zamilatskov
I. A. Dyes and Pigments 2021, 191, 109354 DOI:
10.1016/j.dyepig.2021.109354.
407. Andreeva V. D.; Ponomarev G. V.; Shkirdova A. O.; Tyurin
V. S.; Zamilatskova I. A. Macroheterocycles 2021, 14 (3),
201-207 DOI: 10.6060/mhc213990z.
408. Smith M. J.; Blake I. M.; Clegg W.; Anderson H. L. Org. &
Biomolec. Chem. 2018, 16 (19), 3648-3654 DOI:
10.1039/C8OB00491A.
409. Birin, K. P.; Gorbunova, Y. G.; Tsivadze, A. Y. RSC Adv.
2015, 5 (82), 67242-67246 DOI: 10.1039/c5ra13532j.
410. Jiang, J.; Liu, D.; Zhao, Y.; Wu, F.; Yang, K.; Wang, K.
Appl. Organomet. Chem. 2018, 32 (9), n/a DOI:
10.1002/aoc.4468.
411. Chen, C.; Zhu, Y.-Z.; Fan, Q.-J.; Song, H.-B.; Zheng, J.-Y.
Syntheses, Chem. Lett. 2013, 42 (8), 936-938 DOI:
10.1246/cl.130324.
412. 57. Murugavel M.; Reddy R. V. R.; Sankar J. RSC Advances
2014, 4 (26), 13669-13672 DOI: 10.1039/C4RA01229A.
413. Hong, S.-K.; Jeoung, E.; Lee, C.-H. J. Porphyr.
Phthalocyanines 2005, 09 (04), 285-289 DOI:
10.1142/s1088424605000368.
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 299
414. Higashino T.; Imahori H. ECS Meeting Abstracts 2021,
MA2021-01 (16), 769-769 DOI: 10.1149/ma2021-
0116769mtgabs.
415. Sagun E.I., Zenkevich E.I., Knyukshto V.N., Shulga A.M.
Optics and Spectroscopy. 2011, 110, 251-256.
416. Sagun E.I., Ganzha V.A., Dzhagarov B.M., Shulga A.M.
Khimicheskaya Physika (in Rus.). 1991, 10, 477-484.
417. Gijzeman O.L.J., Kaufman E., Porter G. J. Chem. Soc. Far-
ad. Trans. II. 1973, 69, 708-720.
418. Dzhagarov B.M., Sagun E.I., Ganzha V.A., Gurinovich G.P.
Khimicheskaya Physika (in Rus.). 1987, 6,
419. Majranowski V.G. In: Porphyrins :Spectroscopy, Photochem-
istry, Applications (Enikolopyan N.S., Ed.) Moscow: Nauka,
1987, 127-181.
420. Zenkevich E.I., von Borczyskowski C., Shulga A.M., Bachilo
S.M, Rempel U., Willert A. Chem. Phys. 2002, 275, 185-209.
421. Zenkevich E.I., von Borczyskowski C., Shulga A.M. J Por-
phyrins Phthalocyanines. 2003, 7, 731-754.
422. Zenkevich E.I. Macroheterocycles 2016, 9, 121-140.
423. Zenkevich E.I., Willert A., Bachilo S.M., Rempel U., Kilin
D.S., Shulga A.M., von Borczyskowski C. Materials Science
and Engineering C. 2001, 18, 99-111.
424. Zenkevich E.I., von Borczyskowski C. Surface Photochemis-
try of Quantum Dot-Porphyrin Nanoassemblies for Singlet
Oxygen Generation. In: “Photoinduced Processes at Surfaces
and in Nanomaterials”, ACS Symposium Series (Kilin D.,
Ed.) Washington: American Chemical Society. 2015, Vol.
1196, Chapter 12, 235272.
425. Blaudeck T., Zenkevich E.I., Cichos F., von Borczyskowski.
C. J. Phys. Chem. C. 2008, 112, 20251-20257.
426. Lakowicz J.R. Principles of Fluorescence Spectroscopy. New
York: Kluwer Academic/Plenum Publishers, 1999, 2nd edi-
tion, p. 245.
427. Klimov V. Nanocrystall Quantum Dots. Washington: CRS
Press LLC. 2010, p. 410.
428. Lemon C.M., Karnas E., Bawendi M.G., Nocera D.G. Inorg.
Chem. 2013, 52, 10394−10406.
429. Motevich I. G., Zenkevich E.I., Stroyuk O.L., Raievska O.E.,
Kulikova O.M., Sheinin V.B., Koifman О.I., Zahn D.R.T.,
Strekal N.D. J. of Applied Spectroscopy. 2021, 87, 1057-1066.
430. Salokhiddinov K., Byteva I., Dzhagarov B. Opt. Spectrosc.
1979, 47, 881-884.
431. Krasnovsky A., Jr. Photochem. Photobiol. 1979, 29, 29-34.
432. Krasnovsky A., Jr. Chem. Phys. Lett. 1981, 81, 443-445.
433. Ogilby P. R., Foote. C. S. J. Am. Chem. Soc. 1982, 104, 7,
20692070.
434. Minaev B.F., Lunell S., Kobzev G.I. J. Mol. Strucr. 1993,
284, 19.
435. Gaponenko S.V. Introduction to Nanophotonics, Cambridge:
Cambridge University Press. 2010.
436. Mogilevtsev D., Maloshtan A., Lepeshkevich S.V, Dzhaga-
rov B.M. J. Fluoresc. 2012, 22, 14151419.
437. Dzhagarov B.M., Zharnikova E.S., Stasheuski A.S., Galiev-
sky V.A., Parkhats M.V. J. Applied Spectroscopy. 2012, 79,
869-874.
438. Minaev B. Chemistry and Chemical Technology. 2016, 10,
519-530.
439. Ivashin N.V., Shchupak Е.Е. Optics and Spectroscopy. 2018,
124, 3242.
440. Pham T.C., Nguyen V.N., Choi Y., Lee S., Yoon J. Chem.
Rev. 2021, 121, 13454-13619.
441. Maisch T. J. Photochem. Photobiol. B: Biol. 2015, 150, 2-10.
442. Nguyen V.N., Yan Y., Zhao J., Yoon J. Acc. Chem. Res.
2021, 54, 207-220.
443. Moura N.M.M., Monteiro C.J.P., Tome A.C., Neves
M.G.P.M.S., Cavaleiro J.A.S. Arkivoc 2022, 2, 54-98.
444. Kustov A.V., Belykh D.V., Startseva O.M., Kruchin S.O.,
Venediktov E.A., Berezin D.B. Pharm. Anal. Acta. 2016, 7,
1000480.
445. Sobotta L., Skupin-Mrugalska P., Mielcarek J., Goslinski T.,
Balzarini J. Mini Rev. Med. Chem. 2015, 15(6), 503-521.
446. Sobotta L., Skupin-Mrugalska P., Piskorz J., Mielcarek, J.
Dyes Pigm. 2019, 163, 337-355.
447. Sobotta L., Skupin-Mrugalska P., Piskorz J., Mielcarek J.
Eur. J. Med. Chem. 2019, 175, 72-106.
448. Castano A.P., Demidova T.N., Hamblin M.R. Photodiagn.
Photodyn. Ther. 2004, 1, 279-293. https://doi.org/10.1016/
S1572-1000(05)00007-4.
449. ????????
450. Castano A.P., Demidova T.N., Hamblin M.R. Photodiagn.
Photodyn. Ther. 2005, 2, 91-106.
451. Bonneau S., Vever-Bizet C. Exp. Opin. Ther. Patents 2008,
18, 1011-1025. 26. Malatesti N., Munitic I., Jurak I.
Biophys. Rev. 2017, 9, 149-168.
452. Kustov A.V., Morshnev P.K., Kukushkina N.V., Smirnova
N.L., Berezin D.B., Karimov D.R., Shukhto O.V., Kustova
T.V., Belykh D.V., Mal’shakova M.V., Zorin V.P., Zorina
T.E. Int. J. Mol. Sci. 2022, 23, 5294.
453. Smith D.A., van de Waterbeemd, Walker D.K., Mannhold R.,
Kubinyi H., Timmerman H. / In Methods and principles in
medicinal chemistry. Mannhold, R., Kubinyi, H., Timmer-
man, H., Eds. WileyVCH Verlag: Weinheim 2001, 141 p.
454. Gerola A.P, Tsubone T.M, Santana A., de Oliveira H.P.M.,
Hioka N., Caetano W. J. Phys. Chem. B 2011, 115, 7364-7373.
455. Kustov A.V., Belykh D.V., Smirnova N.L., Khudyaeva I.S.,
Berezin D.B. J. Chem. Thermodyn. 2017, 115, 302-306.
456. Kim J., Santos O.A., Park J.H. J. Contr. Release 2014, 191,
98-104.
457. Zeinali M., Abbaspour-Ravasjani S., Ghorbani M.,
Babazadeh A., Soltanfam T., Santos A.C., Hamblin M.R.
Drug Discov. Today 2020, 25, 1416-1430.
458. Berezin D.B., Kustov A.V., Krestyaninov M.A, Batov D.V.,
Kukushkina N.V., Shukhto O.V. J. Mol. Liq. 2019, 283,
532-536.
459. Berezin D.B., Makarov V.V., Znoyko S.A., Mayzlish V.E.,
Kustov A.V. Mend. Commun. 2020, 30, 621-623.
460. Kustov A.V., Krestyaninov M.A., Kruchin S.O., Shukhto
O.V., Kustova T.V., Belykh D.V., Khudyaeva I.S., Koifman
M.O., Razgovorov P.B., Berezin D.B. Mend. Commun. 2021,
31, 65-67.
461. Shukhto O.V., Khudyaeva I.S., Belykh D.V., Berezin D.B.
ChemChemTech, 2021, 64(11), 86-96.
462. Kustov A.V., Smirnova N.L., Berezin D.B., Berezin M.B. J.
Chem. Thermodyn. 2015, 83, 104-109.
463. Kustov A.V., Smirnova N.L., Berezin D.B., Berezin M.B. J.
Chem. Thermodyn. 2015, 89, 123-126.
464. Zhidomorov N.Yu., Nazarenko O.A., Demidov V.I., Kustov
A.V., Kukushkina N.V., Koifman O.I., Gagua A.K., Tomi-
lova I.K., Berezin D.B. Biomed. Photonics 2022, 11, 23-32.
465. O’Connor A.E., Gallagher W.M., Byrne A.T. Photochem.
Photobiol. 2009, 85, 1053-1074.
466. Rezende L.G., Tasso T.T., Candido P.H.S., Baptista M.S.
Photochem. Photobiol. 2022, 98, 572-590.
467. Tsubone T.M., Martins W.K., Pavani C., Junqueira H.C., Itri
R., Baptista M.S. Sci. Reports 2017, 7, 1-19.
468. Di Mascio P., Martinez G.R., Miyamoto S., Ronsein G.E.,
Medeiros M.H.G. Chem. Rev. 2019, 119, 2043-2086.
469. Itri R., Junqueira H.C., Mertins O., Baptista M.S. Biophys.
Rev. 2014, 6, 47-61.
470. Bacellar I.O.L., Oliveira M.C., Dantas L.S., Costa E.B., Jun-
queira H.C., Martins W.K., Durantini A.M., Cosa G., Di
Mascio P., Wainwright M., Miotto R., Cordeiro R.M.,
Miyamoto S., Baptista M.S. J. Am. Chem. Soc. 2018, 140,
9606-9615.
471. Tsubone T.M., Baptista M.S., Itri R. Biophys. Chem. 2019,
254, 106263.
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
300 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
472. Berezin D.B., Solodukhin T.N., Shukhto O.V., Belykh D.V.,
Startseva O.M., Khudyaeva I.S., Kustov A.V. Russ. Chem.
Bull. 2018, 67(7), 1273-1279.
473. Belykh D.V., Startseva O.M., Patov S.A. Macroheterocycles
2014, 7(4), 401-413.
474. Kępczyński M., Pandian R.P., Smith K.M., Ehrenberg B.
Photochem. Photobiol. 2002, 76, 127-134.
475. Abraham M.H., Acree W.E. New J. Chem. 2016, 40, 9945-
9950.
476. Čunderlıková B., Gangeskar L., Moan J. J. Photochem.
Photobiol. B 1999, 53, 81-90.
477. Kustov A.V., Belykh D.V., Smirnova N.L., Venediktov E.A.,
Kudayarova T.V., Kruchin S.O., Berezin D.B. Dyes Pigm.
2018, 149, 553-559.
478. Kustov A.V., Berezin D.B., Kruchin S.O., Batov D.V. Russ.
J. Phys. Chem. 2022, 96(4), 793-799.
479. Khludeev I.I., Kozyr’ L.A., Zorina T.E., Zorin V.P. Bull.
Exp. Biol. Med. 2015, 160, 208-212.
480. Berezin D.B., Kustov A.V., Kukushkina N.V., Morshnev
Ph.K., Karimov D.R., Belykh D.V., Belykh E.S., Zorina T.E.,
Zorin V.P. Intern. Conf. Porph. Phthaloc. (ICPP-12),
Madrid, 2022, 331.
481. Samaroo D., Zahran M., Wills A.C., Guevara J., Tatonetti A.
Porph. Sci. Women 2020, 3, 266-281.
482. Ghosh S., Chakrabarty S., Bhowmik D., Kumar G.S.,
Chattopadhyay N. J. Phys. Chem. B 2015, 119, 2090-2102.
483. Hamblin M.R., Newman E.L. J. Photochem. Photobiol. B.
1994, 26, 45-56.
484. Hamblin M.R., Newman E.L. J. Photochem. Photobiol. B.
1994, 26, 147-157.
485. Polo L., Valduga G., Jori G., Reddi E. Intern. J. Biochem.
Cell Biol. 2002, 34, 10-23.
486. Reshetov V., Zorin V., Siupa A., D’Hallewin M.A.,
Guillemin F., Bezdetnaya L. Photochem. Photobiol. 2012,
88, 1256-1264.
487. Ossoli A., Wolska A., Remaley A.T., Gomaraschi M.
Biochim. Biophys. Acta (BBA) - Molec. Cell Biol. Lipids
2022, 1867, 159068.
488. Hadi T., Ramseyer C., Gautier T., Bellaye P.S., Lopez T.,
Schmitt A., Lirussi F. JCI Insight 2020, 5(24), e140280.
489. Cruz P.M.R., Mo H., McConathy W.J., Sabnis N., Lacko
A.G. Front. Pharmacol. 2013, 4, 119.
490. Jori G. In vivo transport and pharmacokinetic behavior of
tumor photosensitizers. Photosensitizing compounds: their
chemistry, biological and clinical use. Wiley, Chichester (Ci-
ba foundation symposium). 2007, 78-94.
491. Williamson M.P. Prog. Nucl. Magn. Reson. Spectrosc. 2013,
73, 1-16.
492. Barut B., Demirbaş Ü., Şenocak A., Özel A., Kantekin H.
Synth. Met. 2017, 229, 22-32.
493. Duong-Ly K.C., Gabelli S.B. Methods in enzymology.
Academic Press 2014, 541, 105-114.
494. Feng, M., Tang, B., H Liang, S., Jiang, X. Current topics in
medicinal chemistry, 2016, 16(11), 1200-1216, DOI:
10.2174/1568026615666150915111741.
495. Plekhova N., Shevchenko O., Korshunova O., Stepanyugina
A., Tananaev I., Apanasevich V. Bioengineering, 2022, 9(2),
82, DOI: 10.3390/bioengineering9020082.
496. Kessel D., Smith K.M., Pandey R.K., Shiau F.Y., Henderson
B. Photochemistry and photobiology, 1993, 58(2), 200-203,
DOI: 10.1111/j.1751-1097.1993.tb09549.x.
497. Balendiran G. K., Dabur R., Fraser D. Cell Biochemistry and
Function: Cellular biochemistry and its modulation by active
agents or disease, 2004, 22(6), 343-352, DOI: 10.1027/cbf.1149.
498. Gamcsik M.P., Kasibhatla M.S., Teeter S.D., Colvin O.M.
Biomarkers, 2012, 17(8), 671-691, DOI:
10.3109/1354750X.2012.715672.
499. Lu, S.C. Molecular aspects of medicine, 2009, 30(1-2), 42-59,
DOI: 10.1016/j.mam.2008.05.005.
500. Brozovic A., Ambriović-Ristov A., Osmak M. Critical re-
views in toxicology, 2010, 40(4), 347-359, DOI:
10.3109/10408441003601836.
501. Asantewaa G., Harris I.S. Current Opinion in Biotechnology,
2021, 68, 292-299, DOI: 10.1016/j.copbio.2021.03.001.
502. Kalinina E.V., Gavriliuk L.A. Biochemistry (Moscow), 2020,
85(8), 895-907, DOI: 10.1134/S0006297920080052.
503. Townsend D.M., Findlay V.L., Tew, K.D. Methods in enzy-
mology, 2005, 401, 287-307, DOI: 10.1016/S0076-
6879(05)01019-0.
504. Flohé L., Toppo S., Orian L. Free Radical Biology and Medi-
cine, 2022, 187, 113-122, DOI: 10.1016/j.freeradbiomed.
2022.05.003.
505. Couto N., Wood J., Barber J. Free radical biology and medicine,
2016, 95, 27-42, DOI: 10.1016/j.freeradbiomed.2016.02.028.
506. Miller, C. G., & Schmidt, E. E. (2019). British Journal of
Pharmacology, 176(4), 532-543, DOI: 10.1111/bph.14498.
507. Thornalley P.J. Biochemical Society Transactions, 2003,
31(6), 1343-1348, DOI: 10.1042/bst0311343.
508. He Y., Zhou C., Huang M., Tang C., Liu X., Yue Y., Qingchun
D., Zhebin Z., Liu, D. Biomedicine & Pharmacotherapy, 2020,
131, 110663, DOI: 10.1016/j.biopha.2020.110663.
509. Morgenstern J., Campos M., Nawroth P., Fleming T. Antiox-
idants, 2020, 9(10), 939, DOI: 10.3390/antiox9100939.
510. Liberti M.V., Locasale J.W. Trends in biochemical sciences,
2016, 41(3), 211-218, DOI: 10.1016/j.tibs.2015.12.001.
511. Yu L., Chen X., Sun X., Wang L., Chen S. Journal of Can-
cer, 2017, 8(17), 3430, DOI:10.7150/jca.21125.
512. Bailey H. H. Chemico-biological interactions, 1998, 111,
239-254, DOI:10.1016/S0009-2797(97)00164-6.
513. Bailey H.H., Mulcahy R.T., Tutsch K.D., Arzoomanian R.Z.,
Alberti D., Tombes M.B., Wilding G., Pomplun M., Spriggs
D.R. Journal of clinical oncology, 1994, 12(1), 194-205,
DOI: 10.1200/JCO.1994.12.1.194.
514. Vince R., Daluge S., Wadd W. B. Journal of medicinal chem-
istry, 1971, 14(5), 402-404, DOI: 10.1021/jm00287a006.
515. Thornalley P.J., Edwards L.G., Kang Y., Wyatt C., Davies
N., Ladan M. J., Double J. Biochemical pharmacology, 1996,
51(10), 1365-1372, DOI: 10.1016/0006-2952(96)00059-7.
516. Santarius T., Bignell G.R., Greenman C.D., Widaa S., Chen
L., Mahoney C.L., Butler A., Edkins S., Waris S., Thornalley
P.J., Futreal A.P., Stratton M.R. Genes, Chromosomes and
Cancer, 2010, 49(8), 711-725, DOI: 10.1002/gcc.20784.
517. Rabbani N., Thornalley P.J. Int. J. Molecular Sci.
2022, 23(5), 2453, DOI: 10.3390/ijms23052453.
518. Grin M., Suvorov N., Ostroverkhov P., Pogorilyy V., Kirin
N., Popov A., Sazonova A., Filonenko E. Biophysical Rev.,
2022, 1-23, DOI: 10.1007/s12551-022-00962-6.
519. Tikhonov S., Ostroverkhov P., Suvorov N., Mironov A.,
Efimova Y., Plutinskaya A., Pankratov A., Ignatova A.,
Feofanov A., Diachkova A., Vasil’ev Y., Grin M. Int.l J. Molec-
ular Sci., 2021, 22(24), 13563, DOI: 10.3390/ijms222413563.
520. Hu F., Yuan Y., Mao D., Wu W., Liu B. Biomaterials,
2017, 144, 53-59, DOI: 10.1016/j.biomaterials.2017.08.018.
521. Grin M.A., Pogorilyy V.A., Noev A.N., Tikhonov S.I., Ma-
jouga A.G., Mironova A.F. Macroheterocycles, 2018, 11(1),
89-94, DOI: 10.6060/mhc180176p.
522. Mironov A.F., Ostroverkhov P.V., Tikhonov S.I., Pogorilyy
V.A., Kirin N.S., Chudakova O.O., Tsygankov A.A., Grin M.
A. Fine Chemical Technologies, 2021, 15(6), 16-33, DOI:
10.32362/2410-6593-2020-15-6-16-33.
523. Liu L., Huang J., Li K., Hu X., Sun C. J. chromatography. B,
Analytical technologies in the biomedical and life sciences,
2011, 879, 56-60.
524. Percie du Sert N., Hurst V., Ahluwalia A., Alam S., Avey
M.T., Baker M., Würbel H. Journal of Cerebral Blood Flow
& Metabolism, 2020, 40(9), 1769-1777, DOI:
10.1113/EP088870.
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 301
525. Berk M., Copolov D., Dean O., Lu K., Jeavons S., Schapkaitz
I., Anderson-Hunt M., Judd F., Katz F., Katz P., Ording-
Jespersen S., Little J., Conus P., Cuenod M., Do Q.K., Bush
A.I. Biological psychiatry, 2008, 64(5), 361-368, DOI:
10.1016/j.biopsych.2008.03.004.
526. Yu M., Liu Y., Duan Y., Chen Y., Han J., Sun L., Yang
X. Biochemical and biophysical research communications,
2017, 484(1), 56-63, DOI: 10.1016/j.bbrc.2017.01.072.
527. Cavuoto P., Fenech M. F. Cancer treatment reviews,
2012, 38(6), 726-736, DOI: 10.1016/j.ctrv.2012.01.004.
528. Patent RU 2521327C1, 2012.
529. Levy E.J., Anderson M.E., Meister A. Proceedings of the
National Academy of Sciences, 1993, 90(19), 9171-9175,
DOI: 10.1073/pnas.90.19.9171.
530. Belyii Yu.A., Tereshchenko A.V., Volodin P.L. et al. Refrac-
tive surgery and ophthalmology. 2006, 6, 55-57.
531. Abramova O.B., Demyashkin G.A., Drozhzhina V.V. Mole-
cules. 2022. 27, DOI 10.3390/molecules27113445.
532. Abramova O.B., Drozhzhina V.V., Kozlovtseva E.A. Macro-
heterocycles. 2021. 14, 312-316. DOI 10.6060/mhc224208a.
533. Abramova O.B., Yuzhakov V.V., Kaplan M.A. et al Bull Exp
Biol Med. 2021. 170, 479484. DOI: 10.1007/s10517-021-
05092-9.
534. Abramova O.B., Kaplan M.A., Yuzhakov V.V. et al. Bull
Exp Biol Med. 2021. 171, 468471. DOI 10.1007/s10517-
021-05252-x.
535. Sokolov V.V., Chissov V.I., Filonenko E.V. et al. Proceed-
ings of SPIE - The International Society for Optical Engi-
neering, 1995, 2325, 364366. doi: 10.1117/12.199168.
536. Sokolov V.V., Filonenko E.V., Telegina L.V. et al. Quantum
Electronics, 2002, 32, 963969. doi: 10.1070/QE2002v032n11A
BEH002329.
537. Filonenko E.V. Bio-medical Photonics, 2021, 10, 422 (in
Russian). doi: 10.24931/2413943220219-4-4-22.
538. Zharkova N.N., Kozlov D.N., Smirnov V.V., Sokolov V.V.,
Chissov V.I., Filonenko E.V., Sukhin G.M., Galpern M.G.,
and Vorozhtsov G.N. Proceedings of SPIE - Photodynamic
Therapy of Cancer II, 1995, 2325, 400-403. doi:
10.1117/12.199176.
539. Chissov, V.I., Skobelkin O.K., Mironov A.F., Smirnov V.V.,
Sokolov V.V., Sukhin G.M., Filonenko E.V., Litvin D.G.,
Stranadko, E.F., Kolobanov, A.S., Astrakhankina, T.A., No-
kel Yu., A., Zharkova N.N., Kozlov D.N. Khirurgiya, 1994,
70, рр. 3-6.
540. Tolstykh P.I., Derbenov V.A., Kuleshov I.Yu. Khirurgiya,
2010, 12, 17-22. (in Russian).
541. Tamrazova O.B., Molochkov A.V., Bagramova G.E., Pomer-
antsev O.N. Photodynamic therapy of venous stasis ulcers,
Klinicheskaya dermatologiya i venerologiya, 2013, 62-67. (in
Russian).
542. Hopley C., Salkeld G. Br. J. Ophthalmol., 2004, 88, 982-987.
543. Isaev V.M., Nasedkin A.N., Ashurov Z.M., Reshetnikov
A.V. Rossiiskaya otorinolaringologiya, 2004, 5, 76-79. (in
Russian).
544. .Nasedkin A.N., Grachev N.S., Logunova E.V. Foto-
dinamicheskaya terapiya i fotodiagnostika, 2013, 3, 59. (in
Russian).
545. Trushina O.I., Novikova E.G., Sokolov V.V., Filonenko
E.V., Chissov V.I., Vorozhtsov G.N. Photodiagnosis and
Photodynamic Therapy, 2008, 5 256-259. doi:
10.1016/j.pdpdt.2008.09.005. (in Russian).
546. Kapinus V.N., Kaplan M.A., Kudryavtseva G.T., Khnychev
S.S., Yakovlev V.V., Romanko Yu.S. Sovremennye metody
fotodinamicheskoi (flyuorestsentnoi) diagnostiki i foto-
dinamicheskoi terapii. Sbornik nauchnykh trudov. Obninsk,
2001, 36-41.
547. Romanko Yu.S., Kaplan M.A., Popuchiev V.V. Ros. zhurn.
kozh. i ven. boleznei, 2004, 6, 6-10. (in Russian).
548. Lehmann P. Br J Dermatol, 2007, 156 793-801.
549. Kaplan M.A., Kapinus V.N., Goranskaya E.V. Opukholi
zhenskoi reproduktivnoi sistemy, 2011, 4, 28-31. (in Russian).
550. Filonenko E.V., Okushko A.N., Sukhin D.G., Yanikova A.G.
Onkologiya. Zhurnal im. P.A. Gertsena, 2012, 3, 52-54. (in
Russian).
551. Simone C.B., Friedberg J.S., Glatstein E., Stevenson J.P.,
Sterman D.H., Hahn S.M., Cengel K.A. J Thorac Dis, 2012,
4. 63-75.
552. Hayata, Y, Kato, H, Konaka, C., Ono J., Takizawa N. Chest,
1982, 81, 269-277.
553. Mathur P.N., Edell E., Sutedja T., Vergnon J.M., Chest,
2003, vol. 123(1), pp. 176-180.
554. Cortese D.A., Edell E.S., Kinsey J.H. Mayo Clin Proc, 1997,
72, 595-602.
555. Filonenko E.V., Vashakhmadze L.A., Khomyakov V.M.
Sibirskii onkologicheskii zhurnal, 2012, 2, 84-89. (in Rus-
sian).
556. Sokolov V.V., Pavlov P.V., Karpova E.S., Pirogov S.S. Multi-
course photodynamic therapy through self-expanding stent for
obstructing cancer of upper gastrointestinal tract, Fotodinamich-
eskaya terapiya i fotodiagnostika, 2014, No. 1, pp. 33-34. (in
Russian).
557. Filonenko E.V. Photodynamic therapy of early cancer of
hollow organs, Fotodinamicheskaya terapiya i fotodiagnosti-
ka, 2015, No. 1, pp. 22-25. (in Russian).
558. Berger A.P., Steiner H., stenzi A., Akkad T., Bartsch G.,
Holtl L. Photodynamic therapy with intravesical instilla-
tion of 5-aminolevulinic acid for patients with recurrent
superficial bladder cancer: a single-center study, Urology,
2003, vol. 61(2), pp. 338-41.
559. Zubkov A.Yu., Nuriev I.R., Sitdykov E.N. Role of adjuvant
intravesical chemotherapy in combined organ-sparing treat-
ment of non-muscle-invasive bladder cancer, Onkourologiya,
2014, No. 2, pp. 26-28. (in Russian).
560. Monk A., Brewer C., Van Nostrand K., Bems M. Photody-
namic therapy using topically applied dihematoporphyrin
ether in the treatment of cervical intraepithelial neoplasia,
Gynecol Oncol, 1997, vol. 64(1), pp. 70-5.
561. Novikova E.G., Sokolov V.V., Sidorova I.S., Chulkova E.A.
Fluorescence diagnosis and photodynamic therapy of back-
ground and pre-cancer vulvar diseases with 20% Alasens
ointment, Ross. onkologicheskii zh., 2009, No. 2, pp. 12-19.
(in Russian).
562. Ferlay, J., Ervik, M., Lam, F., et al. Global Cancer Observa-
tory: Cancer Today (2020), (available at
http://gco.iarc.fr/today).
563. Sung, H., Ferlay, J., Siegel, R. L., et al. CA: A Cancer J. Cli-
nicians 2021, 71, 209249. doi:10.3322/caac.21660.
564. Jemal, A., Ward, E. M., Johnson, C. J., et al. JNCI: J.
National Cancer Institute 2017, 109, djx030.
doi:10.1093/jnci/djx030.
565. Erdi, Y. E. Mol Imaging Radionucl Ther. 2012, 21, 2328.
doi:10.4274/Mirt.138.
566. Stummer, W., Pichlmeier, U., Meinel, T., et al. Lancet Oncology
2006, 7, 392401. doi:10.1016/S1470-2045(06)70665-9.
567. Kausch, I., Sommerauer, M., Montorsi, F., et al. Eur Urol
2010, 57, 595606. doi:10.1016/j.eururo.2009.11.041.
568. Liu, R., Xu, Y., Xu, K., Dai, Z. Aggregate. 2021, 2, e23.
doi:10.1002/agt2.23.
569. Craig, S. E. L., Wright, J., Sloan, A. E., Brady-Kalnay, S. M.
World Neurosurg 2016, 90, 154163. doi:10.1016/j.wneu.
2016.02.060.
570. Loshchenov, M., Zelenkov, P., Potapov, A., Goryajnov, S., Bo-
rodkin, A. Photonics & Lasers in Medicine 2014, 3, 159170.
doi:10.1515/plm-2013-0017.
571. Harada, Y., Murayama, Y., Takamatsu, T., Otsuji, E.,
Tanaka, H. Int J Mol Sci 2022, 23, 6478.
doi:10.3390/ijms23126478.
572. Barnes, T. G., Hompes, R., Birks, J., et al. Surg Endosc 2018,
32, 40364043. doi:10.1007/s00464-018-6219-8.
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
302 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
573. Hillary, S. L., Guillermet, S., Brown, N. J., Balasubramanian,
S. P. Langenbecks Arch Surg 2018, 403, 111118.
doi:10.1007/s00423-017-1641-2.
574. Schebesch, K.-M., Brawanski, A., Hohenberger, C., Hohne,
J. Turk Neurosurg 2016, 26, 185194. doi:10.5137/1019-
5149.JTN.16952-16.0.
575. Sun, Z., Jing, L., Fan, Y., et al. Int Rev Neurobiol 2020, 151,
139154. doi:10.1016/bs.irn.2020.03.004.
576. Morales-Conde, S., Alarcón, I., Yang, T., et al. Surg Endosc
2020, 34, 38973907. doi:10.1007/s00464-019-07159-1
577. Martinez De Pinillos Bayona, A., Mroz, P., Thunshelle, C.,
Hamblin, M. R. Chemical Biology & Drug Design 2017, 89,
192206. doi:10.1111/cbdd.12792.
578. Mroz, P., Huang, Y., Szokalska, A., et al. FASEB j. 2010, 24,
31603170. doi:10.1096/fj.09-152587.
579. Sharma, S. K., Krayer, M., Sperandio, F. F., J. Porphyrins
Phthalocyanines 2013, 17, 7385.
doi:10.1142/S108842461250126X.
580. Meerovich, I. G., Sanarova, E. V., Oborotova, N. A. Russian
Journal of General Chemistry 2015, 85.
doi:10.1134/S1070363215010430.
581. Baldea, I., Ion, R.-M., Olteanu, D. E. Medicine and Pharma-
cy Reports 2015, 88, 175180. doi:10.15386/cjmed-419.
582. Skubleny, D., Dang, J. T., Skulsky, S., Surg Endosc 2018, 32,
26202631. doi:10.1007/s00464-018-6100-9.
583. Maruyama, T., Akutsu, Y., Suganami, A. PLoS ONE 2015,
10, e0122849. doi:10.1371/journal.pone.0122849.
584. Urbanska, K., Romanowska-Dixon, B., Matuszak, Z., Acta
Biochim Pol 2002, 49, 387391.
585. Geralde, M. C., Leite, I. S., Inada, N. M. Physiological Re-
ports 2017, 5, e13190. doi:10.14814/phy2.13190.
586. Mansouri, S., Heylmann, D., Stiewe, T., Kracht, M., Savai,
R. eLife 2022, 11, e79895. doi:10.7554/eLife.79895.
587. Engblom, C., Pfirschke, C., Pittet, M. J. Nat Rev Cancer
2016, 16, 447462. doi:10.1038/nrc.2016.54.
588. Perentes, J. Y., Duda, D. G., Jain, R. K. Disease Models &
Mechanisms 2009, 2, 107110. doi:10.1242/dmm.001842.
589. Turley, S. J., Cremasco, V., Astarita, J. L. Nat Rev Immunol
2015, 15, 669682. doi:10.1038/nri3902.
590. Noy, R., Pollard, J. W. Immunity 2014, 41, 4961.
doi:10.1016/j.immuni.2014.06.010.
591. Kitamura, T., Qian, B.-Z., Pollard, J. W. Nat Rev Immunol
2015, 15, 7386. doi:10.1038/nri3789.
592. Milas, L., Wike, J., Hunter, N., Volpe, J., Basic, I. Cancer
Research 1987, 47, 10691075.
593. Korbelik, M., Krosl, G., Olive, P. L., Chaplin, D. J. Br J
Cancer 1991, 64, 508512. doi:10.1038/bjc.1991.339.
594. Anatelli, F., Mroz, P., Liu, Q. Mol Pharm 2006, 3, 654664.
doi:10.1021/mp060024y.
595. Korbelik, M., Hamblin, M. R. Photochem. Photobiol. Sci.
2015, 14, 14031409. doi:10.1039/c4pp00451e.
596. Hayashi, N., Kataoka, H., Yano, S. Molecular Cancer Thera-
peutics 2015, 14, 452460. doi:10.1158/1535-7163.MCT-14-
0348.
597. Wen, A. M., Lee, K. L., Cao, P. Bioconjugate Chem. 2016,
27, 12271235. doi:10.1021/acs.bioconjchem.6b00075.
598. Scalfi-Happ, C., Zhu, Z., Graefe, S. Photodiagnosis and Photo-
dynamic Therapy 2018, 22, 106114. doi:10.1016/j.pdpdt.
2018.03.004.
599. Makarov, V. I., Pominova, D. V., Ryabova, A. V. Pharma-
ceutics 2022, 14, 2122. doi:10.3390/pharmaceutics14102122.
600. Zhu, Z., Scalfi-Happ, C., Ryabova, A., et al. Journal of Pho-
tochemistry and Photobiology B: Biology 2018, 185, 215222.
doi:10.1016/j.jphotobiol.2018.06.015.
601. Agostinis, P., Berg, K., Cengel, K. A., et al. CA Cancer J
Clin 2011, 61, 250281. doi:10.3322/caac.20114.
602. Castano, A. P., Demidova, T. N., Hamblin, M. R. Photodiag-
nosis Photodyn Ther 2005, 2, 91106. doi:10.1016/S1572-
1000(05)00060-8.
603. Shams, M., Owczarczak, B., Manderscheid-Kern, P.,
Bellnier, D. A., Gollnick, S. O. Cancer Immunol Immunother
2015, 64, 287297. doi:10.1007/s00262-014-1633-9.
604. Jalili, A., Makowski, M., Świtaj, T., et al. Clinical Cancer
Research 2004, 10, 44984508. doi: 10.1158/1078-
0432.CCR-04-0367.
605. Saji, H., Song, W., Furumoto, K., Kato, H., Engleman, E. G.
Clinical Cancer Research 2006, 12, 25682574. doi:
10.1158/1078-0432.CCR-05-1986.
606. Piette, J. Photochem Photobiol Sci 2015, 14, 15101517. doi:
10.1039/c4pp00465e.
607. Anzengruber, F., Avci, P., de Freitas, L. F., Hamblin, M. R.
Photochem Photobiol Sci 2015, 14, 14921509. doi:
10.1039/c4pp00455h.
608. Azzouzi, A.-R., Vincendeau, S., Barret, E., et al. The Lancet
Oncology 2017, 18, 181191. doi: 10.1016/S1470-
2045(16)30661-1.
609. Mallidi, S., Anbil, S., Lee, S., et al. J. Biomed. Opt 2014, 19,
028001. doi: 10.1117/1.JBO.19.2.028001.
610. Shao, P., Chapman, D. W., Moore, R. B., Zemp, R. J. J. Bio-
med. Opt 2015, 20, 106012. doi:
10.1117/1.JBO.20.10.106012.
611. Sirotkina, M. A., Matveev, L. A., Shirmanova, M. V. Sci Rep
2017, 7, 41506. doi:10.1038/srep41506.
612. Goldschmidt, R., Kalchenko, V., Scherz, A. in Optical Meth-
ods for Tumor Treatment and Detection: Mechanisms and
Techniques in Photodynamic Therapy XXVI, D. H. Kessel, T.
Hasan, Eds. SPIE, San Francisco, California, United States,
2017, 10047, 100470M. doi: 10.1117/12.2251971.
613. Lakouas, D. K., Huglo, D., Mordon, S., Vermandel, M. Pho-
todiagnosis and Photodynamic Therapy 2017, 18, 236243.
doi: 10.1016/j.pdpdt.2017.03.002.
614. Potapov, A. A., Goryaynov, S. A., Okhlopkov, V. A., et al.
Neurosurg Rev 2016, 39, 437447. doi: 10.1007/s10143-015-
0697-0.
615. Zelenkov, P., Shevelev, I., Potapov, A., et al. Photodiagnosis
and Photodynamic Therapy 2011, 8, 163. doi:
10.1016/j.pdpdt.2011.03.133.
616. Xu, H., Wang, Z., Li, J., et al. Neural Plasticity 2017, 2017,
111. doi: 10.1155/2017/5405104.
617. von Leden, R. E., Cooney, S. J., Ferrara, T. M., et al. Lasers
Surg Med 2013, 45, 253263. doi: 10.1002/lsm.22133.
618. Zhao, M., Liang, F., Xu, H., Yan, W., Zhang, J. Mol Med Rep
2016, 13, 1320. doi: 10.3892/mmr.2015.4551.
619. Wang, Y.-C., Cui, Y., Cui, J.-Z., et al. Molecular Medicine
Reports 2015, 12, 21492154. doi: 10.3892/mmr.2015.3607.
620. Tardivo, J. P., Del Giglio, A., de Oliveira, C. S., et al. Photo-
diagnosis and Photodynamic Therapy 2005, 2, 175191. doi:
10.1016/S1572-1000(05)00097-9.
621. Chekhonin, V. P., Baklaushev, V. P., Yusubalieva, G. M.,
Volgina, N. E., Gurina, O. I. Annals of the Russian academy
of medical sciences 2012, 67, 6678. doi:
10.15690/vramn.v67i8.352.
622. Jain, A., Betancur, M., Patel, G. D., et al. Nature Mater 2014,
13, 308316. doi: 10.1038/nmat3878.
623. Siegel R. L., Miller K.D., Fuchs H.E., Jemal A. CA Cancer J
Clin 2022, 72, 733. https://doi.org/10.3322/caac.21708.
624. Wicki A., Witzigmann D., Balasubramanian V., Huwyler J.
J. Controlled Release. 2015, 200, 138157.
http://dx.doi.org/10.1016/j.jconrel.2014.12.030
625. Liu W., Liu C., Wang H., Xu L., Zhou J., Li S., Cheng Y.,
Zhou R., Zhao L., Comput. Struct. Biotechnol. J., 2022, 20,
5150-5161, https://doi.org/10.1016/j.csbj.2022.09.017.
626. Ashrafizadeh M., Aghamiri S., Tan S. C., Zarrabi A., Sharifi
E., Rabiee N., Kadumudi F. B., Pirouz A. D., Delfi M.,
Byrappa K., Thakur V. K., Kumar K. S. S., Girish Y. R.,
Zandsalimi F., Zare E. N., Orive G., Tay F., Hushmandi K.,
Kumar A. P., Karaman C., Karimi-Maleh H., Mostafavi E.,
Makvandi P., Wang, Y. Nano Today, 2022, 45, 101532,
https://doi.org/10.1016/j.nantod.2022.101532.
O. I. Koifman et al.
Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304 303
627. Jeong Y., Hwang H.S., Na K. Biomaterials research, 2022.
26:27, 1-13. https://doi.org/10.1186/s40824-022-00277-3.
628. Aoun F., Kourie H. R., Artigas C., Roumeguère T. Future oncol.
2015, 11, 22052219. https://doi.org/10.2217/fon.15.104.
629. Gao. M., Tang B.Z. Coord. Chem. Rev. 2020, 402, 213076.
https://doi.org/10.1016/j.ccr.2019.213076.
630. Ng K. K., Zheng G. Chem. Rev., 2015, 115, 1101211042.
doi:10.1021/acs.chemrev.5b00140.
631. Yin X., Cheng Y., Feng Y., Stiles W.R., Park S. H., Kang H.,
Choi H.S. Adv. Drug Delivery Rev., 2022, 189, 114483,
https://doi.org/10.1016/j.addr.2022.114483.
632. Gao D., Guo X., Zhang X., Chen S., Wang Y., Chen T., Huang
G., Gao Y., Tian Z., Yang Z., Materials Today Bio. 2020, 5,
100035, https://doi.org/10.1016/j.mtbio.2019.100035.
633. Terreno E., Castelli D.D., Viale A., Aime S., Chem. Rev. 2010,
110 (5), 30193042, https://doi.org/ 10.1021/cr100025t.
634. Zhou Z., Bai R., Munasinghe J., Shen Z., Nie L., Chen X. ACS
Nano 2017, 11 52275232, https://doi.org/10.1021/
acsnano.7b03075.
635. Ying Liu, Weiqiang Lin, Fang Yang, Tianfeng Chen. Applied
Materials Today. 2022, 28, 101520. https://doi.org/10.1016/
j.apmt.2022.101520.
636. Abakumova T.O., Nukolova N.V., Gusev E.I., Сhekhonin
V.P. Neurosci. Behav. Physiol. 2015, 115 (1), 58-65. doi:
10.17116/jnevro2015115115865.
637. Xiao Y.-D., Paudel R., Liu J., C., Zhang Z.-S., Zhou S.-
K. Int. J. Mol. Med. 2016, 38, 1319-1326. DOI:
10.3892/ijmm.2016.2744.
638. Li J., Li X., Gong S., Zhang C., Qian C., Qiao H., Sun M.,
Nano Lett. 2020, 20 48424849, https://doi.org/10.1021/
acs.nanolett.0c00817.
639. Xie J., Jiang J., Davoodi P., Srinivasan M.-P, Wang C.-H.
Chem. Eng. Sci. 2015, 125, 32.
640. Zhang Z., Zhou F.-L., Davies G.-L., Williams G.R. View,
VIEW. 2022, 3, 20200134. https://doi.org/
10.1002/VIW.20200134.
641. He M., Chen Y., Tao C., Tian Q., An L., Lin J., Tian Q.,
Yang H., Yang S. ACS applied materials & interfaces, 2019.
11(45): pр. 41946-41956. https://doi.org/10.1021/
acsami.9b15083.
642. Brewster II J.T., G. D.Thiabaud, P. Harvey, H.Zafar, J. F. Reu-
ther, S. Dell’Acqua, R. Johnson M., Root H. D., Metola P.,
Jasanoff A., Casella L., Sessler J. L. Chem. 2020, 6, 703-724
https://doi.org/10.1016/j.chempr.2019.12.016.
643. Felder, P.S., S. Keller, and G. Gasser. Advanced Therapeu-
tics, 2020. 3(1): p. 1900139. https://doi.org/10.1002/
adtp.201900139.
644. Schmitt J., Jenni S., Sour A., Heitz V., Bolze F., Pallier A.,
Bonnet C.S., Toth E., Ventura B. Bioconjugate chemistry,
2018. 29, 3726-3738. https://doi.org/10.1021/
acs.bioconjchem.8b00634.
645. Tsolekile, N., S. Nelana, and O.S. Oluwafemi. Molecules,
2019. 24(14): p. 2669. https://doi.org/10.3390/ mole-
cules24142669.
646. Grin M.A., Brusov S.S., Shchepelina E.Yu., Ponomarev P.V.,
Khrenova M.K., Smirnov A.S., Lebedeva V.S., Mironov A.F.
Mendeleev Commun. 2017, 27, 338-340. 10.1016/
j.mencom.2017.07.005.
647. Boros E., Gale E.-M., Caravan P. Dalton Trans. 2015, 44,
4804. https://doi.org/10.1039/C4DT02958E.
648. Vlaardingerbroek M.-T., Boer J. A. Magnetic resonance im-
aging: theory and practice. Springer Science & Business Me-
dia, United States, 2013.
649. Chan K. W.-Y., Wong W.-T., Coord Chem Rev, 2007, 251
(1720), 2428-2451, https://doi.org/10.1016/
j.ccr.2007.04.018.
650. Guglielmo F. F., Mitchell D. G., Gupta S. Radiologic Clinics
of North America, 2014, 52(4), 637-656, https://doi.org/
10.1016/j.rcl.2014.02.004.
651. M. Botta, F. Carniato, D. Esteban-Gómez, C. Platas-Iglesias,
L. Tei, Future Med. Chem. 2019, 11 14611483,
https://doi.org/10.4155/fmc-2018-0608.
652. Geraldes C.F.G.C., Castro M. M. C.A., Peters J. A. Coor.
Chem. Rev. 2021, 445, 214069. https://doi.org/10.1016/
j.ccr.2021.214069.
653. Rocklage S. M., Cacheris W. P., Quay S. C., Hahn F. E.,
Raymond K. N. Inorg. Chem., 1989, 28, 477485.
https://doi.org/10.1021/ic00302a019.
654. Wang J., Wang H., Ramsay I. A., Erstad D. J., Fuchs B. C.,
Tanabe K. K., Caravan P., Gale E. M. J. Med. Chem. 2018,
61, 19, 88118824 https://doi.org/10.1021/
acs.jmedchem.8b00964.
655. Jeon M., Halbert M.V., Stephen Z.R., Zhang M., Adv. Mater.
2020, 1906539 https:// doi.org/10.1002/adma.201906539.
656. Shu G., Chen M., Song J., Xu X., Lu C., Du Y., Xu M., Zhao
Z., Zhu M., Fan K., Fan X., Fang S., Tang B., Dai Y., Du Y.,
Ji J. Bioactive Materials, 2021,6(5), 1423-1435,
https://doi.org/10.1016/j.bioactmat.2020.10.020.
657. Scharlach C., Warmuth C., Schellenberger E. Magnetic Res-
onance Imaging, 2015, 33(9), 1173-1177. https://doi.org/
10.1016/j.mri.2015.06.017.
658. Wahsner J., Gale E. M., Rodríguez-Rodríguez A., Caravan P.
Chem. Rev. 2019, 119, 2, 957-1057. https://doi.org/
10.1021/acs.chemrev.8b00363.
659. Calvete M. J.F., Pinto S. M.A., Pereira M. M., Geraldes C.
F.G.C. Coord. Chem. Rev., 2017, 333, 82-107,
https://doi.org/10.1016/j.ccr.2016.11.011.
660. Mironov A.F., Zhdanova K.A Bragina N.A. Russ Chem Rev.
2018, 87(9), 859881. https://doi.org/10.1070/RCR4811.
661. Hindré, F., et al. J. Magnetic Resonance Imaging, 1993. 3(1):
p. 59-65. https://doi.org/10.1002/jmri.1880030111.
662. Li G., Slansky A., Dobhal M. P., Goswami L. N., Graham A.,
Chen Y., Kanter P., Alberico R. A., Spernyak J., Morgan J.,
Mazurchuk R., Oseroff A., Grossman Z., Pandey R. K. Bio-
conjugate Chemistry. 2005, 16 (1), 32-42. https://doi.org/
10.1021/bc049807x.
663. Haroon-Ur-Rashid M.N., Umar K., Khan M.N., Yaseen M.,
Anjum J. Struct. Chem. 2014, 55 910915.
https://doi.org/10.1134/S0022476614050163.
664. Tekdaş A. D., Garifullin R., Şentürk B., Zorlu Y., Gundogdu
U., Atalar E., Tekinay A.B., Chernonosov A.A., Yerli Y.,
Dumoulin F., Guler M.O., Ahsen V., Gürek A.G.. Photochem
Photobiol, .2014. 90, 1376-1386. https://doi.org/
10.1111/php.12332.
665. Song Y., Zong H., Trivedi E.R., Vesper B.J., Waters E.A.,
Barrett A.G.M., Radosevich J.A., Hoffman B. M., Meade T.J.
Bioconjugate chemistry, 2010. 21, 2267-2275. https://
doi.org/10.1021/bc1002828.
666. Yuzhakova D. V., Lermontova S. A., Grigoryev I. S., Mura-
vieva M. S., Gavrina A. I., Shirmanova M. V., Balalaeva I.
V., Klapshina L. G., Zagaynova E. V. Biochimica et Biophys-
ica Acta (BBA). 2017, 1861 (12), 3120-3130,
https://doi.org/10.1016/j.bbagen.2017.09.004.
667. Wu B., Li X., Huang T., Lu S., Wan B., Liao R., Li Y.,
Baidya A., Long Q., Xu H., Biomater. Sci., 2017, 5, 1746
DOI: 10.1039/C7BM00431A.
668. Luo J., Chen L.-F., Hu P., Chen Z.-N. Inorganic Chemistry
2014, 53, 4184-4191. DOI: 10.1021/ic500238s.
669. Jenni, S., Bolze F., Bonnet. C.S, Pallier A., Sour A., Toth E.,
Ventura B., Heitz V. Inorganic Chemistry, 2020. 59(19): p.
14389-14398. https://doi.org/10.1021/acs.inorgchem.0c02189
670. Schmitt J., Jenni, S.; Sour A., Heitz V., Bolze, F., Pallier A.,
Bonnet C. S., Tóth ., Ventura B. Bioconjugate Chem. 2018,
29, 3726−3738. https://doi.org/10.1021/acs.bioconjchem.
8b00634.
671. Vyalba F. Yu., Ivantsova A.V., Zhdanova K.A., Usachev
M.N., Gradova M. A., Bragina N.A. Mendeleev Commun.
2022, 32(5), 675-677. https://doi.org/10.1016/j.mencom
.2022.09.036.
Synthesis Strategy of Tetrapyrrolic Photosensitizers for Their Practical Application in Photodynamic Therapy
304 Макрогетероциклы / Macroheterocycles 2022 15(4) 207-304
672. Mefteh W.B. Touzi H.; Bessueille F.; Chevalier Y.; Kalfat
R.; Jaffrezic-Renault N. Electroanalysis. 2015, 27(1), 84-92.
https://doi.org/10.1002/elan.201400373
673. Oyama J.; Fernandes Herculano Ramos-Milaré, Á. C.; Lopes
Lera-Nonose, D. S. S.; Nesi-Reis, V.; Galhardo Demarchi, I.;
Alessi Aristides, S. M.; Juarez Vieira Teixeira, J.; Gomes
Verzignassi Silveira, T.; Campana Lonardoni, M. V. Photo-
diagn. Photodyn. Therapy. 2020, 30, 101682.
https://doi.org/10.1016/j.pdpdt.2020.101682.
674. Solovieva, A.B, Timashev S.F., Russ. Chem. Rev. 2003, 72,
965.
675. Solovieva, A.B., Rudenko, T.G., Shekhter, A.B., Glagolev,
N.N., Spokoinyi, A.L., Fayzullin, A.L., Aksenova, N.A.,
Shpichka, A.I., Kardumyan, V.V., Timashev, P.S. J. Photo-
chem. Photobiol. B: Biology. 2020., 210. 111954. DOI:
10.1016/j.jphotobiol.2020.111954.
676. Günsel, A.; Taslimi, P.; Atmaca, G. Y.; Bilgiçli, A. T.;
Pişkin, H.; Ceylan, Y.; Erdoğmuş, A.; Yarasir, M. N.; Gülçin,
İ. Novel. J. Molec. Struct. 2021, 1237, 130402.
https://doi.org/10.1016/j.molstruc.2021.130402.
677. Lee, H.-J.; Kang, S.-M.; Jeong, S.-H.; Chung, K.-H.; Kim,
B.-I. Photodiagn. Photodyn. Therapy 2017, 20, 116119.
https://doi.org/10.1016/j.pdpdt.2017.09.003.
678. Méndez D.A.C.; Gutierrez E.; Dionísio E.J.; Oliveira, T.M.;
Buzalaf, M.A.R.; Rios D.; Machado M.A.A.M.; Cruvinel T.
Lasers Med. Sci. 2018, 33, 479487. https://doi.org/10.1007
/s10103-017-2379-3.
679. Pérez-Laguna V.; García-Luque I.; Ballesta S.; Pérez-
Artiaga L.; Lampaya-Pérez V.; Samper S.; Soria-Lozano P.;
Rezusta A.; Gilaberte Y. Photodiagn. Photodyn. Therapy
2018, 21, 211216. https://doi.org/10.1016/j.pdpdt.
2017.11.012.
680. Usacheva M.N.; Teichert M.C.; Biel M.A. J. Photochem.
Photobiol. B: Biology 2003, 71 (13), 8798.
https://doi.org/10.1016/j.jphotobiol.2003.06.002.
681. Lim E.J., Oak C.-H., Heo J., Kim Y.-H. Oncology Reports,
2013, 30, 856862. doi:10.3892/or.2013.2494.
682. Dos Santos A. F., Terra L. F., Wailemann R. A. M., Oliveira
T. C., Gomes V. de M., Mineiro M. F., Meotti F.C., Bruni-
Cardoso A., Baptista M.S., Labriola L. BMC Cancer. 2017,
17,194 doi:10.1186/s12885-017-3179-7.
683. Shih MH, Huang FC. Invest Ophthalmol Vis Sci. 2011,
52,223229.
684. Atenco-Cuautle JC, Delgado-López MG, Ramos-García R.,
Ramírez-San-Juan JC, Ramirez-Ramirez J., Spezzia-
Mazzocco T. Proc. SPIE 11070, 17th International Photody-
namic Association World Congress, 11070A1 2019 7 Au-
gust; https://doi.org/10.1117/12.2525456.
685. Houang J., Perrone G.G., Pedrinazzi C., Longo L., Mawad
D., Boughton P. C., Ruys A.J., Lauto, A. Adv. Therapeutics,
2018, 1800105. doi:10.1002/adtp.201800105.
686. Aksenova A., Oles T., Sarna T., Glagolev N. N., Chernjak
A.V., Volkov V.I., Kotova S.L., Melik-Nubarov N.S.,
Solovieva A.B. Laser Physics, 2012, 22, 16421649.
687. Koifman O.I., Ageeva T.A. Porphyrin Polymers: Synthesis,
Properties, Applications. Moscow: LENAND, 2018. 304 p..
688. Koifman O.I., Ageeva T.A. Polymer Sci. Ser. C. 2004, 46,
49-72.
689. Amat-Guerri F., López-González M. M. C., Sastre R., late R.
Martinez-Utrilla. Dyes Pigments, 1990, 13, 219232.
doi:10.1016/0143-7208(90)80021-g.
690. Martin M. M.; Lindqvist L. J. Luminescence 1975, 10, 381390.
https://doi.org/10.1016/0022-2313(75)90003-4.
691. Jiang C., Brown P. J., Ducret A., Brun Y. V. Nature. 2014,
506, 489-493.
692. Malik Z., Hanania J., Nitzan Y. J. Photochem. Photobiol. B:
Biol. 1990, 5, 281-293.
693. Nitzan Y. Photochem Photobiol. 1992, 55, 89-96.
694. Strakhovskaya M.G., Belenikina N., Nikitina V., Kovalenko
S., Kovalenko I., Averyanov A., Rubin A., Galochkina T.
Clinical Practice. 2013, 4, 25-30. 10.17816/clinpract4125-30.
695. Loke W.K., Lau S.K., Yong L.L., Khor E. Sum C.K. J. Bio-
med. Mater. Res. 2000, 53, 8.
696. Fontana C. R., dos Santos D.S., Jr., Bosco J. M., Spolidorio
D. M., Marcantonio R. A., Drug Deliv. 2008, 15, 417-422.
DOI: 10.1080/10717540802007433.
697. Solovieva A.B., Rudenko T.G., Shekhter A.B., Glagolev
N.N., Spokoinyi A.L., Fayzullin A.L., Aksenova N.A.,
Shpichka A.I., Kardumyan V.V., Timashev P.S. J. Photo-
chem. Photobiol., B: Biology. 2020. 210, 111954. DOI:
10.1016/j.jphotobiol.2020.111954.
698. Aksenova N.A., Timofeeva V.A., Rogovina S.Z., Timashev
P.S., Glagolev N.N., Solov’eva A.B. Polym. Sci. Ser. B 2010,
52, 6772 https://doi.org/10.1134/S1560090410010100.
699. Glagolev N.N., Rogovina S.Z., Solov’eva A.B., Aksenova N.A.,
Kotova S.L. Rus. J. Phys. Chem. 2006, 80. Suppl. 1., S72.
700. Glagolev N.N., Aksenova N.A., Rogovina S.Z., Solovieva
A.B. Reports of the Russian Academy of Sciences, 2007, 416,
57-59.
701. Kardumyan V. V., Aksenova N. A., Glagolev N. N., Ti-
mashev P. S., Solovieva A. B. J. Chem. Phys. 2020, 152.
194901 DOI:10.1063/5.0007362.
702. Kuryanova A.S., Aksenova N.A., Savko M.A., Glagolev
N.N., Dubovik A.S., Plashchina I.G., Timashev P.S.,
Solovieva A.B. Rus. J. Phys. Chem. 2022, 96, 747-753.
Received 24.11.2021
Accepted 07.11.2022
... Some medical issues remain topical and unresolved even in the 21st century. These are, in particular, the issues of cancer and antibiotic-resistant bacterial infections [1][2][3][4][5][6][7][8]. This fact predetermines the constant search for new effective and at the same time maximally safe ways to solve these problems [7]. ...
... This fact predetermines the constant search for new effective and at the same time maximally safe ways to solve these problems [7]. Photodynamic therapy (PDT) is one of the most promising methods aimed at combating both cancer diseases and pathogenic bacterial microflora; a method associated with the use of photosensitisers (PS) [7][8][9][10]. A photosensitizer is usually an organic dye with the ability to selectively accumulate in tumour tissues or bacterial cells. ...
Article
The article considers issues concerning the reaction of exocycle opening in phorbin molecules (on the example of methylpheophorbide a) under the O nucleophiles - water and alcohols action. According to the author, under certain conditions relatively weak nucleophiles as water and alcohols can cause the opening of the exocycle in the molecule of methylpheophorbide a with the formation of chlorine derivatives e6. The study of this reaction, in addition, reveals new synthetic methods to obtain chlorine e6 esters of different degrees of substitution, and eventually - free chlorine e6 in the form of triacids. Free chlorine is a valuable photosensitiser for photodynamic therapy (PDT), and a starting compound for the preparation of other photosensitizers. The article also discusses possible synthetic approaches to obtain free chlorine e6.
... Light absorption of biological tissues in this window is minimal, thus allowing deeper light penetration through the tissue, and, as a result, higher therapeutic effectiveness. There is an increasing interest in bacteriochlorin-based photosensitizers as fluorescent probes and medicines for diagnostics and treatment, because their NIR fluorescence and efficient singlet oxygen generation properties are advantageous both for imaging and photodynamic therapy [4][5][6][7][8][9][10]. The most important characteristics of these PSs are their rapid clearance from circulation and their minimum skin-phototoxicity induced by visible light. ...
Article
Full-text available
This research presents a novel synthetic photosensitizer for the photodynamic therapy (PDT) of malignant tumors: meso-tetra(3-pyridyl) bacteriochlorin, which absorbs at 747 nm (in the long-wavelength region of the spectrum) and is stable when stored in the dark. H2Py4BC demonstrates pronounced photoinduced activity in vitro against tumor cells of various geneses (IC50 varies from 21 to 68 nM for HEp2, EJ, S37, CT26, and LLC cultured cells) and in vivo provides pronounced antitumor efficacy in the treatment of mice bearing small or large S37, Colo26, or LLC metastatic tumors, as well as in the treatment of rats bearing RS-1 liver cholangioma. As a result, total regression of primary tumor nodules and cure of 40 to 100% of the animals was proven by the experiment criteria, MRI, and histological analysis. Meso-tetra(3-pyridyl) bacteriochlorin quickly penetrates and accumulates in the tumor tissue and internal organs of mice, and after 24 h, 80% of the dye is excreted from the skin in addition to 87–92% from the liver, kidneys, and spleen.
Article
A series of molecular brushes with a polyfluorene (PF) backbone and polymethacrylic acid side chains of varying lengths were prepared by atom transfer radical polymerization. The structure and composition of the synthesized compounds were confirmed by ¹ H NMR and IR spectroscopy. Effect of the length of the backbone on spectral and conformational parameters of the macromolecules in solutions was analyzed. The grafting density of side chains was about 90%. Spectral methods have been used to determine the dependence of side chain grafting on the luminescent properties of polymer solutions, including quantum yields. It was shown that an optimal length of polymethacrylic acid side chains provides solubility of the polymer brushes. Solutions of PF‐ graft ‐polymethacrylic acid complexes with the model substance curcumin were investigated. It was established that the molecular brushes containing curcumin form monomolecular micelles. Molecular brushes with zinc phthalocyanine, potential systems for photodynamic, and photothermal therapy, were studied.
Article
The article considers issues concerning the reaction of exocycle opening in phorbin molecules (on the example of methylpheophorbide a) under the O nucleophiles - water and alcohols action. According to the author, under certain conditions relatively weak nucleophiles as water and alcohols can cause the opening of the exocycle in the molecule of methylpheophorbide a with the formation of chlorine derivatives e6. The study of this reaction, in addition, reveals new synthetic methods to obtain chlorine e6 esters of different degrees of substitution, and eventually - free chlorine e6 in the form of triacids. Free chlorine is a valuable photosensitiser for photodynamic therapy (PDT), and a starting compound for the preparation of other photosensitizers. The article also discusses possible synthetic approaches to obtain free chlorine e6.
Article
The interaction of cobalt(II) tetrasulfophthalocyanine (CoPc) with the ORF8 accessory protein of SARS-CoV-2 was studied by spectroscopy and calorimetry. The protein was found to shift the aggregation equilibrium in cobalt tetrasulfophthalocyanine solutions towards dimerization. Most probably, the CoPc dimer binds to ORF8 on the greater β-sheet side, thus causing fluorescence quenching. The protein affinity constant to CoPc dimer is 1.5 × 105. Differential scanning calorimetry data indicate that ORF8 undergoes thermally induced denaturation in the temperature range of 38–67°C. Melting of ORF8 includes two stages, which partly overlap. The complex formation of ORF8 with CoPc leads to thermal stabilization of the protein, thus preventing the second stage of protein unfolding. Denaturation of the complex proceeds between 40 and 77°C as two temperature-separated stages. According to gel electrophoresis and immunoblotting data, visible light photoirradiation of the ORF8 complex with CoPc does not induce photooxidation of the protein. It was shown that water-soluble cobalt sulfo-substituted phthalocyanine can be considered as a potential drug inhibiting the ORF8 accessory protein.
Article
In this work, screening studies of the cytotoxic effect of chlorins with fragments of di-, tri-, and pentaethylene glycol at the macrocycle periphery in relation to HeLa, A549, and HT29 cells were performed. It is shown that, despite different hydrophobicity, all the compounds studied have a comparable photodynamic effect. The conjugate of chlorin e6 with pentaethylene glycol, which has the lowest tendency to association among the studied compounds with tropism for low density lipoproteins and the best characteristics of the formation of molecular complexes with Tween 80, has a significant difference in dark and photoinduced toxicity (ratio IC50(dark)/IC50(photo) approximately 2 orders of magnitude for all cell lines), which allows to hope for a sufficiently large "therapeutic window". A study of the interaction of this compound with HeLa cells shows that the substance penetrates the cell and, after red light irradiation induces ROS appearance inside the cell, associated, apparently, with the photogeneration of singlet oxygen. These data indicate that photoinduced toxic effects are caused by damage to intracellular structures as a result of oxidative stress. Programmed type of cell death characterized with caspase-3 induction is prevailing. So, the conjugate of chlorin e6 with pentaethylene glycol is a promising antitumor PS that can be successfully solubilized with Tween 80, which makes it suitable for further in vivo studies.
Article
Full-text available
Antimicrobial photodynamic therapy (APDT) has received a great deal of attention due to its unique ability to kill all currently known classes of microorganisms. To date, infectious diseases caused by bacteria and viruses are one of the main sources of high mortality, mass epidemics and global pandemics among humans. Every year, the emergence of three to four previously unknown species of viruses dangerous to humans is recorded, totaling more than 2/3 of all newly discovered human pathogens. The emergence of bacteria with multidrug resistance leads to the rapid obsolescence of antibiotics and the need to create new types of antibiotics. From this point of view, photodynamic inactivation of viruses and bacteria is of particular interest. This review summarizes the most relevant mechanisms of antiviral and antibacterial action of APDT, molecular targets and correlation between the structure of cationic porphyrins and their photodynamic activity.
ResearchGate has not been able to resolve any references for this publication.