ISSN 0006-2979, Biochemistry (Moscow), 2023, Vol. 88, No. 11, pp. 1732-1738 © Pleiades Publishing, Ltd., 2023.
Published in Russian in Biokhimiya, 2023, Vol. 88, No. 11, pp. 2101-2108.
1732
REVIEW
Potential Geroprotectors – From Bench to Clinic
A l e x e y A . M o s k a l e v
Institute of Biogerontology, Lobachevsky University, 603950 Nizhny Novgorod, Russia
e-mail: amoskalev@list.ru
Received July 20, 2023
Revised September 20, 2023
Accepted September 20, 2023
AbstractGeroprotectors are substances that slow down aging process and can be used for prevention of age-related diseas-
es. Geroprotectors can improve functioning of various organ systems and enhance their homeostatic capabilities. We have
developed a system of criteria for geroprotectors and proposed their classification based on the mechanisms of their action
on the aging processes. Geroprotectors are required to reduce mortality, improve human aging biomarkers, have minimal
side effects, and enhance quality of life. Additionally, there are approaches based on combining geroprotectors targeted
to different targets and mechanisms of aging to achieve maximum effectiveness. Currently, numerous preclinical studies
are being conducted to identify new molecular targets and develop new approaches to extend healthy aging, although the
number of clinical trials is limited. Geroprotectors have the potential to become a new class of preventive medicines as they
prevent onset of certain diseases or slow down their progression.
DOI: 10.1134/S0006297923110056
Keywords: geroprotector, aging, biomarker, biological age, criteria
INTRODUCTION
Geroprotectors are compounds affecting the mech-
anisms of aging and slowing it down. They could be used
for prophylactics and delay of onset of age-related dis-
eases and extension of active period of life. Geroprotec-
tors could become and important part of modern pro-
phylactic and personalized medicine [1], and, first of all,
of the emerging medicine of healthy longevity [2].
Aging is the main cause of numerous age-related
diseases such as cardiovascular diseases, cancer, dia betes,
and neurodegenerative diseases. Studying of geroprotec-
tors could shed light on the main biological processes
underlying aging. This could help us to understand what
molecular and cellular changes occur in an organism
with aging, and what mechanisms could be modulated
to achieve geroprotection. Investigation of geroprotec-
tors could help us to develop strategies for preventing
or slowing down onset of these diseases. This is of tre-
mendous importance for improving quality of life of
individuals and to reduce economic burden on health-
care systems. However, it must be mentioned that inves-
tigation of geroprotectors is a complicated task. There
are multiple factors that could affect the results of such
studies, such as differences in experimental conditions,
genotypes and species of model organisms, problems
with short-lived controls, and irreproducibility of data.
This means that further studies and developments are
required to establish efficiency and safety of potential
geroprotectors for humans.
POTENTIAL GEROPROTECTORS
We have developed a set of criteria for geroprotec-
tors with the goal to describe to the fullest the desired
features and properties of a geroprotector and facilitate
its introduction into medical practice for prophylactics
of aging. We identified main criteria (ensuring effec-
tiveness and safety) and auxiliary criteria (accelerating
translation into practice) [3].
Main criteria for geroprotector. Geroprotector must
increase lifespan. Whether the substance is geroprotector
or not is judged by its ability to increase lifespan of the
model organism under the used experimental conditions
or decrease general human mortality. Our open on-line
database (geroprotectors.org) compiled based on 2408
literature sources includes 259 compounds that increase
lifespan at least at one concentration in one of 13 model
organisms from yeasts to humans [4]. Recently a data-
base within the framework of the DrugAge project was
created with our participation that includes more than
POTENTIAL GEROPROTECTORS 1733
BIOCHEMISTRY (Moscow) Vol. 88 No. 11 2023
1000 compounds effects of which on lifespan were in-
vestigated [5].
Geroprotector must reverse processes associated with
age-related diseases. Clinical trials or use of geroprotector
in modern medical practice is impossible without some
symptoms associated with the diseases developed in an
individual. Hence, geroprotector should provide certain
help with the symptoms of age-related diseases in order
to have chance to be accepted in medical practice. Nev-
ertheless, geroprotectors could be potentially assigned
to a new class of medicinal drugs, prophylactic agents,
because they not only treat the symptoms of certain dis-
eases, but also create background for resistance to the de-
velopment of certain diseases or slowing down their onset
and progression. Vladimir Dilman, expert in gerontol-
ogy, named aging a universal disease[6]. According to
Mikhail Blagosklonnyi, age-related diseases (type 2 dia-
betes, Alzheimer’s disease, cardiovascular diseases) are
symptoms of aging, same as smoke is a sign of fire[7].
Hence, effectiveness of a geroprotector should be as-
sessed on its ability to delay onset of several age-related
diseases in humans (prevent multimorbidity) and extent
healthy lifespan (age before the first chronic disease
develops).
Geroprotector should provide positive effect on bio-
markers of human aging. Biomarkers of aging are molec-
ular, cellular, physiological, and psychological parame-
ters of an organism, which are known to change reliably,
qualitatively, and quantitatively in aging [8]. The gero-
protector candidate should change these parameters to
the state typical for a younger age. The criteria of gero-
protectors associated with aging biomarkers are of spe-
cial importance for translation of the results into medi-
cal practice. Investigation of lifespan of a human treated
with geroprotector candidate are very lengthy and cost-
ly. Hence, analysis of lifespan could be performed with
animals, and effects on the human aging could be elu-
cidated based on the wide spectrum of changes of bio-
markers resulting from the geroprotective therapy. Cur-
rently there have been many attempts to use estimates
of biological age as a complex biomarker of human ag-
ing in the randomized placebo-controlled clinical trials.
Inparticular, change in the thickness of the intima-me-
dia complex of carotid artery and rigidity of the arterial
wall after treatment with the complex terpene prepara-
tion was demonstrated [9].
Geroprotector should not exhibit any pronounced
side effects and should improve quality of life. Consider-
ing that some geroprotectors could exhibit prophylactic
effect only at a relatively high concentration, toxic and
most effective (from the point of view of slowing down
aging) concentrations should differ as much as possible.
Some compounds that extent lifespan of animals at cer-
tain doses, exhibit side effects in humans. Achieving a
geroprotective effect implies long-term (many years) ad-
ministration of the preparation. Use of such preparation
would require to compromise between the expected and
side effects. Hence, it is preferably that the number and
manifestation of side effects for the geroprotector candi-
date would be minimal. Efficiency of slowing down ag-
ing under the influence of geroprotectors, mostly likely,
would be noticeable only after many years of taking the
preparation on the regular basis. Hence, it is important
that they would be capable of improving quality of life
form the very beginning of their administration– facil-
itate elimination of digestive problems, affect favorably
sleeping patterns, prevent development of depression or
memory loss.
We applied the criteria for geroprotectors to a wide
range of potential geroprotectors. The criteria are met
for some natural terpenoids [10] and compounds exhib-
iting protective effect on the genome [11].
Professor V. N. Anisimov classified the life-extend-
ing compounds into three groups [12]. The first group in-
cludes geroprotectors that are equally useful for all mem-
bers of population, because they could delay the onset
of aging. The second group includes geroprotectors that
reduce mortality of individuals at advanced age. Theyal-
low to increase maximum life expectancy by slowing
down the process of population aging. The third group
are geroprotectors that increase the chances of survival of
humans at middle age without increasing maximum life
expectancy in the population. This means that they speed
up the process of aging, but at the same time increase the
probability for the individual to live to old age.
Aging could be defined as gradual reduction of the
capability of the organism’s systems to maintain stability
of inner milieu, which leads to onset of age-related dis-
eases and death. That is why the main strategy in fight-
ing aging and preventing wearing out of the body could
be maintenance of homeostatic capabilities at all levels
of organization of a live system.
Classification of geroprotectors according to their
ability to maintain homeostasis of an organism [13].
Correction of the consequences of homeostasis disruption.
Disruption of homeostasis with age is manifested by de-
viation from the healthy level of such vital parameters as
blood acid-base balance, arterial blood pressure, level of
glucose and cholesterol in blood. Hence, preparations
preventing development of such age-related states could
be considered as geroprotectors. Geroprotectors that
correct consequences of homeostasis disruption could
include antidiabetic, antiarrhythmic, hypolipidemic, vas-
cular, and antihypertension preparations. Indeed, such
preparations as antidiabetic biguanides (metformin) and
sodium glucose cotransporter 2 inhibitors (SGLT2l) are
capable of decreasing cardiovascular mortality, beyond
the limits of secondary prevention[14, 15]. Bisphospho-
nates (inhibitors of bone resorption) also decrease overall
mortality of the patients [16]. Stable reduction of mortal-
ity was demonstrated for the 65 years old and older pa-
tients taking statins [17].
MOSKALEV173 4
BIOCHEMISTRY (Moscow) Vol. 88 No. 11 2023
Table 1. Mechanisms of action of potential geroprotectors [23]
Mechanism Group of geroprotectors Examples
Prevention
of free radical oxidation
free radical scavengers or antioxidants;
transition metal chelators;
fatty acids resistant to lipid peroxidation
vitamins C, A, E,
deuterated polyunsaturated
fatty acids
Maintenance
of mitochondrial functions
mitohormetins and inhibitors
of mitochondrial electron transport chain;
inducers of mitophagy;
activators of PPARy/PGC-1α and mitoproliferation;
NAD
+
precursors
urolithin A,
NMN, NR, NA
Maintenance of proteostasis agent preventing glycation, and inhibitors
of crosslinking of advanced glycation end
products (AGEs);
antagonists of AGEs receptors, RAGE;
anti-amyloid compounds;
stimulators of extracellular matrix metabolism;
inducers of autophagy;
proteasome activators;
transient translation inhibitors
spermidine
Senolytics/senostatics compounds facilitating selective elimination
of senescent cells or slowing down their emergence
quercetin + dasatinib
Hormetins compounds inducing moderate damage
and stimulating activation of defence mechanisms
of stress resistance
sulforaphane, curcumin
Suppressors
of genome instability
antimutagen compounds;
telomere stabilizers;
inhibitors of retrotransposition
inhibitors of reverse
transcriptase
Epigenetic regulators inhibitors of HDACs, HATs;
activators of SIRTs
butyrate,
beta-hydroxybutyrate,
resveratrol
Inhibitors
of ageing-associated pathways
inhibitors of mTORC1; inhibitors of PI3K;
inhibitors of Ras; inhibitors of Myc;
inhibitors of AT1; activators of AMPK;
Klotho activators
rapamycin, torin2,
wortmannin, LY294002
Anti-inflammatory preparations
inhibitors of NF-κB; activators of NRF2
ibuprofen, aspirin
Gut microbiota optimizers prebiotics, metabiotics, and enterosorbents dietary fibers, lignans
Anti-fibrotic agents inhibitors of fibrosis-associated signaling pathways inhibitors
of TGF-βALK5
p-Smad2,3 pathway
Neurotrophic factors plat role in development and maintenance
of structures in central and peripheral nervous systems
BDNF mimetics
Factors preventing disruption
of barrier function
inhibitors of matric metalloproteinases,
activators of synthesis of tight junction proteins
MMP9 inhibitors
Immunomodulators factors facilitating thymus regeneration
and preservation of the pool of naïve T-cells
regulators
of JAKSTAT pathway
POTENTIAL GEROPROTECTORS 1735
BIOCHEMISTRY (Moscow) Vol. 88 No. 11 2023
Table 2. Clinical trials of potential geroprotectors
Group Indication References
Inhibitor of mTOR,
RTB101
immunity to viral infection in elderly [32]
AMPK modulator,
metformin
cardiovascular diseases [33]
Activator of Nrf2-pathway,
sulforaphane
alleviation of symptoms of mild and moderated depression in the patients
with cardiological surgeries in anamnesis
[34]
Reduction of NAD
+
supplementing with NAD
+
precursors (nicotinamide, nicotinamide riboside,
nicotinamide mononucleotide) positively affect biomarkers of lipid profile
in the patients with cardiovascular diseases and dyslipidemia
[35]
Senolytics
quercetin + dasatinib decrease the level of inflammation in adipose tissues
and improve systemic metabolic function in elderly
[36]
Enhancement of capabilities of homeostatic systems
of an organism. At the cellular level the key homeostatic
role is played by the stress response proteins. Nutrient
deficiencies, hypoxia, DNA damage, disruptions of ho-
meostasis are perceived by the cell as stress. Activation
of the stress-resistance systems could not only alleviate
damages, but also could transform the system to the
higher level of defence against the new spontaneous er-
rors and damages [18]. Induction of the stress-resistance
mechanisms could be realized by the factors causing
moderate stress that do not induce damage, but capa-
ble of activating defence. This phenomenon was termed
“hormesis” [19], and compounds causing it were named
hormetins [20].
Neutralization of the agents causing disruptions in
homeostasis in external and internal environment. This
is a wide group of compounds including, for example,
chelators of transition metals Cu and Fe participating in
the Fenton, Haber–Weiss, and Maillard reactions; scav-
engers of reactive oxygen species; compounds cleaving
crosslinks; preparations preventing protein aggregation
(anti-amyloid compounds).
Suppression of excessive homeostatic reaction causing
further loss of homeostasis. Hyperactivation of some ho-
meostatic reaction in response to stress could cause even
more severe damage than the initial damage. Accord-
ing to the hypothesis of geroconversion suggested by
M. B. Blagosklonny, after the cell cycle arrest the aging
cells continue their growth. Geroconversion results in
hypersecretion, hypertrophy, and proinflammatory cell
phenotype, which depends on activity of the mTOR ki-
nase [21]. Inflammaging postulated by Franceschi [22]
could be considered as such excessive homeostatic reac-
tion. Preparations targeting specific targets hyperactivat-
ed in aging (mTORC1, NF-κB, PARP1, iNOX, COX2,
p38, S6K, TGF-β, AT1, IGFR, HIF-1) could also be
also considered as inhibitors of hyperactivation.
At present the number of investigated targets of po-
tential geroprotectors is very limited, and their efficien-
cy in prolonging life leaves much to be desired. One of
the potential approaches to expand the list of effective
targets is to include in consideration a wider spectrum
of molecular, cellular, and systemic processes associated
with age-related diseases (Table 1). However, it must be
mentioned that each individual compound could affect
several pathways, and each geroprotector could belong
to several subclasses.
To increase the number of potential effective targets
of geroprotectors it has been suggested to use comput-
erized search algorithm based on comparison of tran-
scriptomic signature of old and young cells [24]. Real-
ization of such approach allowed establishing potential
geroprotective properties in some FDA approved drugs
such as pioglitazone (PPARγ agonist)[25]. New targets
for potential drugs with anti-aging properties and as-
sociated with treatment of 14 age-related diseases were
identified with the help of the specialized computer pro-
gram, PandaOmics, that uses a set of artificial intelli-
gence algorithms. In particular, several gene targets were
found that play an important role in the processes of in-
flammation and in rigidity of extracellular matrix in the
organism tissues. These genes include CASP3, VEGFA,
and MMP9[26].
Another way to increase efficiency of geroprotection
is combination of several targets associated with aging in
one intervention. Combining rapamycin with wortman-
nin increased drosophila lifespan by 23.4% [27]. Prepa-
rations affecting TGF-β and IGF-1 pathways synergis-
tically increased the lifespan of Caenorhabditis elegans
up to 2-fold [28]. Simultaneous inhibition of TGF-β
and treatment with oxytocin enhance neurogenesis,
decrease neuroinflammation, improve cognitive func-
tions, rejuvenate liver and muscles, and decrease the
number of aging cells expressing p16 in old mice [29].
MOSKALEV1736
BIOCHEMISTRY (Moscow) Vol. 88 No. 11 2023
Year-long treatment of the patients with recombinant
human growth hormone together with dehydroepi-
androsterone(DHEA) and metformin changes epigen-
etic age by approximately 1.5 years [30].
Problems associated with application of geropro-
tectors should be also considered [31]. Majority of gero-
protectors investigated in the model organisms increase
lifespan only marginally or only in the animals of one sex.
Aging is not recognized as a disease or pathological state,
which explains low number of clinical trials of pharma-
ceuticals, practically limiting them to investigation of
natural compounds in composition of biologically ac-
tive supplements. Absence of the generally accepted set
of biomarkers of human aging also complicates clinical
studies of potential geroprotectors.
The available clinical data are quite limited and
provide information only on usefulness of potential
geroprotectors with regards to surrogate health markers
and mainly for the patients with age-related diseases,
but not for the healthy individuals (Table2).
Considering that the main goal of application of
geroprotectors is extension of human healthspan, pro-
spectively treatment should start before the development
of the first chronic age-related disease. At present we
can only talk about gerosuppressors, because they help
to prevent or to slow down some manifestations of ag-
ing; so far there is no clinically proven strategies to re-
verse aging.
CONCLUSIONS
Despite the existence of numerous known mecha-
nisms for prophylactics of aging, many geroprotectors
have been poorly investigated even at the preclinical
stage. It is difficult to draw any far-reaching conclusions,
because there are no sufficient data to examine if poten-
tial geroprotectors meet the required criteria. Side effects
could be more significant than the potential benefits in
the long term. Some potential geroprotectors showed
very promising results in the studies with animal mod-
els, but their efficiency must be confirmed in the pla-
cebo-controlled, blind, multi-center clinical trials using
biomarkers of human aging and mortality data.
The goal of using geroprotector is extension of
healthy life, and treatment should start before mani-
festations of chronic diseases in order to delay onset of
the first are-related chronic disease. However, it is very
difficult to obtain approval for such clinical trial with-
out declaration of any symptoms/indications, especially
due to the serious side effects of some preparations with
pre-clinically proven geroprotective activity, such as
EDTA and rapamycin. It seems more promising, in our
opinion, to examine biologically active supplements in
clinical setting using biomarkers of aging as check points
as well as methods for evaluation of biological age.
At present we can talk not about geroprotectors,
but about gerosuppressors, because we can slow down
but not prevent development of some manifestations of
aging. However, new methods fighting aging could ap-
pear in future, and we hope for development of the new
branch of medicine, medicine of healthy longevity, in
our country, similar to the recently created clinical cen-
ters in Singapore, Hong Kong, and USA.
Acknowledgments. The author is grateful to his col-
leagues and collaborators participating in our studies of
geroprotectors cited in this review.
Funding. This work was financially supported by
the Program “Prioritet 2030” of the Nizny Novgorod
State University “Healthy Generation”.
Ethics declarations. The author declares no conflict
of interests in financial or any other sphere. This article
does not contain any studies with human participants or
animals performed by the author.
REFERENCES
1. Fomenko, A. M., Proshkina, E. N., Fedintsev, A. Yu.,
Tsvetkov, V. O., Shaposhnikiv, M. V., and Moskalev,
A. A. (2016) Potential Geroprotectors, Evropeisky Dom,
St.-Petersburg.
2. Bischof, E., Scheibye-Knudsen, M., Siow, R., and Moska-
lev, A. (2021) Longevity medicine: upskilling the physi-
cians of tomorrow, Lancet Healthy Longev., 2, e187-e188,
doi:10.1016/S2666-7568(21)00024-6.
3. Moskalev, A., Chernyagina, E., Tsvetkov, V., Fedint-
sev,A., Shaposhnikov, M., Krut’ko, V., Zhavoronkov,A.,
and Kennedy, B.K. (2016) Developing criteria for eval-
uation of geroprotectors as a key stage toward transla-
tion to the clinic, Aging Cell, 15, 407-415, doi: 10.1111 /
acel.12463.
4. Moskalev, A., Chernyagina, E., de Magalhaes, J. P., Ba-
rardo, D., Thoppil, H., Shaposhnikov, M., Budovsky,A.,
Fraifeld, V. E., Garazha, A., Tsvetkov, V., Bronovitsky,E.,
Bogomolov, V., Scerbacov, A., Kuryan, O., Gurinovich,R.,
Jellen, L. C., Kennedy, B., Mamoshina,P., Dobrovolska-
ya, E., Aliper, A., etal. (2015) Geroprotectors.org: a new,
structured and curated database of current therapeutic in-
terventions in aging and age-related disease, Aging (Albany
NY), 7, 616-628, doi:10.18632/aging.100799.
5. Barardo, D., Thornton, D., Thoppil, H., Walsh, M.,
Sharifi, S., Ferreira, S., Anzic, A., Fernandes, M., Mon-
teiro, P., Grum, T., Cordeiro, R., De-Souza, E. A.,
Budovsky, A., Araujo, N., Gruber, J., Petrascheck, M.,
Fraifeld, V. E., Zhavoronkov, A., Moskalev, A., and
deMagalhaes, J. P. (2017) TheDrugAge database of ag-
ing-related drugs, Aging Cell, 16, 594-597, doi: 10.1111/
acel.12585.
6. Dilman, V. M. (1986) The Grand Biological Clock. Introduc-
tion to Integral Medicine [in Russian], Znaniye, Moscow.
POTENTIAL GEROPROTECTORS 1737
BIOCHEMISTRY (Moscow) Vol. 88 No. 11 2023
7. Blagosklonny, M. V. (2009) TOR-driven aging: speeding
car without brakes, Cell Cycle, 8, 4055-4059, doi:10.4161/
cc.8.24.10310.
8. Moskalev, A. (2020) The challenges of estimating biologi-
cal age, Elife, 9, e54969, doi:10.7554/eLife.54969.
9. Maganova, F., Voevoda, M., Popov, V., and Moska-
lev, A. (2023) A prospective randomized comparative
placebo-controlled double-blind study in two groups
to assess the effect of the use of biologically active addi-
tives with Siberian fir terpenes for the biological age of a
person, Front. Pharmacol., 14, 1150504, doi: 10.3389/
fphar.2023.1150504.
10. Proshkina, E., Plyusnin, S., Babak, T., Lashmano-
va,E., Maganova, F., Koval, L., Platonova, E., Shaposh-
nikov,M., and Moskalev, A. (2020) Terpenoids as potential
geroprotectors, Antioxidants (Basel), 9, 529, doi:10.3390/
antiox9060529.
11. Proshkina, E., Shaposhnikov, M., and Moskalev, A.
(2020) Genome-protecting compounds as potential
geroprotectors, Int. J. Mol. Sci., 21, 4484, doi:10.3390/
ijms21124484.
12. Anisimov, V. N. (2001) Life span extension and can-
cer risk: myths and reality, Exp. Gerontol., 36, 1101-1136,
doi:10.1016/s0531-5565(01)00114-0.
13. Moskalev, A., Chernyagina, E., Kudryavtseva, A., and
Shaposhnikov, M. (2017) Geroprotectors: aunified con-
cept and screening approaches, Aging Dis., 8, 354-363,
doi:10.14336/AD.2016.1022.
14. Araldi, E., Jutzeler, C. R., and Ristow, M. (2023) Ef-
fects of antidiabetic drugs on mortality risks in individu-
als with type2 diabetes: Aprospective cohort study of UK
Biobank participants, medRxiv, doi: 10.1101/2023.05.19.
23290214.
15. Roussel, R., Travert, F., Pasquet, B., Wilson, P. W., Smith,
S. C., Jr., Goto, S., Ravaud, P., Marre, M., Porath, A.,
Bhatt, D. L., Steg, P. G., and Reduction of Atherothrom-
bosis for Continued Health Registry Investigators (2010)
Metformin use and mortality among patients with diabetes
and atherothrombosis, Arch. Intern. Med., 170, 1892-1899,
doi:10.1001/archinternmed.2010.409.
16. Bergman, J., Nordstrom, A., Hommel, A., Kivipelto, M.,
and Nordstrom, P. (2019) Bisphosphonates and mortality:
confounding in observational studies? Osteoporos. Int., 30,
1973-1982, doi:10.1007/s00198-019-05097-1.
17. Gitsels, L. A., Bakbergenuly, I., Steel, N., and Kulinska-
ya, E. (2021) Do statins reduce mortality in older peo-
ple? Findings from a longitudinal study using primary
care records, Fam. Med. Community Health, 9, e000780,
doi:10.1136/fmch-2020-000780.
18. Moskalev, A. A., Aliper, A. M., Smit-McBride, Z.,
Buzdin, A., and Zhavoronkov, A. (2014) Genetics and epi-
genetics of aging and longevity, Cell Cycle, 13, 1063-1077,
doi:10.4161/cc.28433.
19. Calabrese, E. J. (2013) Hormesis: toxicological foun-
dations and role in aging research, Exp. Gerontol., 48,
99-102, doi:10.1016/j.exger.2012.02.004.
20. Rattan, S. I. (2012) Rationale and methods of discovering
hormetins as drugs for healthy ageing, Expert Opin. Drug
Discov., 7, 439-448, doi:10.1517/17460441.2012.677
430.
21. Blagosklonny, M. V. (2014) Geroconversion: irreversible
step to cellular senescence, Cell Cycle, 13, 3628-3635,
doi:10.4161/15384101.2014.985507.
22. Franceschi, C. (2007) Inflammaging as a major character-
istic of old people: can it be prevented or cured? Nutr. Rev.,
65, S173-S176, doi:10.1111/j.1753-4887.2007.tb00358.x.
23. Moskalev, A., Guvatova, Z., Lopes, I. A., Beckett, C.W.,
Kennedy, B. K., De Magalhaes, J. P., and Makarov,
A.A. (2022) Targeting aging mechanisms: pharmacolog-
ical perspectives, Trends Endocrinol. Metab., 33, 266-280,
doi:10.1016/j.tem.2022.01.007.
24. Zhavoronkov, A., Buzdin, A. A., Garazha, A. V., Bor-
isov, N.M., and Moskalev, A.A. (2014) Signaling path-
way cloud regulation for in silico screening and ranking
of the potential geroprotective drugs, Front. Genet., 5, 49,
doi:10.3389/fgene.2014.00049.
25. Yang, J., Peng, S., Zhang, B., Houten, S., Schadt, E.,
Zhu, J., Suh, Y., and Tu, Z. (2020) Human geroprotec-
tor discovery by targeting the converging subnetworks of
aging and age-related diseases, Geroscience, 42, 353-372,
doi:10.1007/s11357-019-00106-x.
26. Pun, F. W., Leung, G. H. D., Leung, H. W., Liu, B.H.M.,
Long, X., Ozerov, I. V., Wang, J., Ren, F., Aliper, A.,
Izumchenko, E., Moskalev, A., de Magalhaes, J.P., and
Zhavoronkov, A. (2022) Hallmarks of aging-based du-
al-purpose disease and age-associated targets predicted
using PandaOmics AI-powered discovery engine, Aging
(Albany NY), 14, 2475-2506, doi:10.18632/aging.203960.
27. Danilov, A., Shaposhnikov, M., Plyusnina, E., Kogan, V.,
Fedichev, P., and Moskalev, A. (2013) Selective anticancer
agents suppress aging in Drosophila, Oncotarget, 4, 1507-
1526, doi:10.18632/oncotarget.1272.
28. Admasu, T. D., Chaithanya Batchu, K., Barardo, D., Ng,
L. F., Lam, V. Y. M., Xiao, L., Cazenave- Gassiot, A.,
Wenk, M.R., Tolwinski, N.S., and Gruber, J. (2018) Drug
synergy slows aging and improves healthspan through
IGF and SREBP lipid signaling, Dev. Cell, 47, 67-79.e65,
doi:10.1016/j.devcel.2018.09.001.
29. Mehdipour, M., Etienne, J., Chen, C. C., Gathwala, R.,
Rehman, M., Kato, C., Liu, C., Liu, Y., Zuo, Y., Con-
boy, M. J., and Conboy, I. M. (2019) Rejuvenation of
brain, liver and muscle by simultaneous pharmacological
modulation of two signaling determinants, that change
in opposite directions with age, Aging (Albany NY), 11,
5628-5645, doi:10.18632/aging.102148.
30. Fahy, G. M., Brooke, R. T., Watson, J. P., Good, Z.,
Vasanawala, S. S., Maecker, H., Leipold, M.D., Lin, D.
T. S., Kobor, M.S., and Horvath, S. (2019) Reversal of
epigenetic aging and immunosenescent trends in humans,
Aging Cell, 18, e13028, doi:10.1111/acel.13028.
31. Moskalev, A. (2020) Is anti-ageing drug discovery be-
coming a reality, Expert Opin. Drug Discov., 15, 135-138,
wdoi:10.1080/17460441.2020.1702965.
MOSKALEV1738
BIOCHEMISTRY (Moscow) Vol. 88 No. 11 2023
32. Mannick, J. B., Teo, G., Bernardo, P., Quinn, D., Russell,
K., Klickstein, L., Marshall, W., and Shergill, S. (2021)
Targeting the biology of ageing with mTOR inhibitors to
improve immune function in older adults: phase 2b and
phase 3 randomised trials, Lancet Healthy Longev., 2,
e250-e262, doi:10.1016/S2666-7568(21)00062-3.
33. Dihoum, A., Rena, G., Pearson, E. R., Lang, C.C., and
Mordi, I. R. (2023) Metformin: evidence from preclin-
ical and clinical studies for potential novel applications
in cardiovascular disease, Expert Opin. Invest. Drugs, 32,
291-299, doi:10.1080/13543784.2023.2196010.
34. Ghazizadeh-Hashemi, F., Bagheri, S., Ashraf-
Ganjouei, A., Moradi, K., Shahmansouri, N., Mehr-
pooya, M., Noorbala, A.A., and Akhondzadeh, S. (2021)
Efficacy and safety of sulforaphane for treatment of mild
to moderate depression in patients with history of cardiac
interventions: Arandomized, double-blind, placebo-con-
trolled clinical trial, Psychiatry Clin. Neurosci., 75, 250-
255, doi:10.1111/pcn.13276.
35. Zhong, O., Wang, J., Tan, Y., Lei, X., and Tang, Z. (2022)
Effects of NAD
+
precursor supplementation on glucose
and lipid metabolism in humans: a meta-analysis, Nutr.
Metab. (Lond), 19, 20, doi:10.1186/s12986-022-00653-9.
36. Islam, M. T., Tuday, E., Allen, S., Kim, J., Trott, D.W.,
Holland, W.L., Donato, A.J., and Lesniewski, L.A. (2023)
Senolytic drugs, dasatinib and quercetin, attenuate adipose
tissue inflammation, and ameliorate metabolic function
in old age, Aging Cell, 22, e13767, doi:10.1111/acel.13767.