Anticancer Activity of Postbiotic Mediators
Derived from Lactobacillus Rhamnosus GG and
Lactobacillus Reuteri on Acute Lymphoblastic
Leukemia Cells
Morteza Banakar1,2, Shahroo Etemad-Moghadam1, Roza Haghgoo2, Majid Mehran2, Mohammad Hossein Yazdi3,
Hadiseh Mohamadpour1, Mahdiyar Iravani Saadi4, Mojgan Alaeddini1
1 Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran
2 Department of Pediatric Dentistry, Faculty of Dentistry, Shahed University, Tehran, Iran
3 Biotechnology Research Center, Tehran University of Medical Sciences, Tehran, Iran
4 Hematology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
GMJ.2023;12:e3096
www.gmj.ir
Correspondence to:
Mojgan Alaeddini, Dental Research Center, Dentistry
Research Institute, Tehran University of Medical Sci-
ences, Tehran 14176-14411, Iran.
Telephone Number: +989122879667
Email Address: malaeddini@sina.tums.ac.ir
Received 2023-05-05
Revised 2023-06-14
Accepted 2023-06-30
Abstract
Background: Leukemia remains a global health challenge, requiring the exploration of al-
ternative therapies with reduced side eects. Probiotics, particularly Lactobacillus species,
have gained attention because of their potential anticancer properties. This study investigat-
ed the anticancer and cytotoxic eects of postbiotic mediators (PMs) derived from Lactoba-
cillus rhamnosus GG (LGG) and Lactobacillus reuteri (LR) on acute lymphoblastic leuke-
mia (ALL) cells and peripheral blood mononuclear cells (PBMCs). Materials and Methods:
The PMs were prepared by culturing LGG and LR strains and isolating the supernatant. The
MTT assay assessed cell viability on ALL Jurkat cells and PBMCs, and apoptosis analysis was
conducted using ow cytometry. Quantitative real-time PCR was also performed to analyze
BAX, BCL-2, BCLX, FAS, and p27 gene expression levels. Results: The results showed that
PMs derived from LGG and LR signicantly reduced cell viability in Jurkat cells (P<0.05)
but not PBMCs (P>0.05). Apoptosis analysis revealed an increase in apoptotic cells after PMs
treatment. Nevertheless, gene expression analysis revealed no statistically signicant dier-
ence between the treated and untreated groups in BAX, BCL-2, BCLX, FAS, and p27 gene ex-
pression levels (P>0.05). Conclusion: Findings suggest that specic PMs derived from LGG
and LR possess anticancer properties against ALL cells. This research highlighted the prom-
ise of PMs as a cutting-edge and less toxic adjuvant therapeutic strategy in cancer treatment.
[GMJ.2023;12:e3096] DOI:10.31661/gmj.v12i0.3096
Keywords: Cancer; Acute Lymphoblastic Leukemia; Probiotic; Postbiotic; Anticancer
Introduction
Cancer continues to pose a substantial
health burden worldwide, as evidenced
by the approximately 19.3 million new cas-
es and 10 million fatalities in 2020 [1]. Acute
lymphoblastic leukemia (ALL) is the predom-
inant form of childhood cancer, constituting
approximately 25% of all malignancies diag-
nosed in the pediatric population [2]. Despite
advances in understanding cancer biology and
developing novel therapeutic strategies, the
eectiveness of cancer treatment is still hin-
dered by the toxicity and enduring adverse
GMJ
Copyright© 2021, Galen Medical Journal.
This is an open-access article distributed
under the terms of the Creative Commons
Attribution 4.0 International License
(http://creativecommons.org/licenses/by/4.0/)
Email:info@gmj.ir
Banakar M, et al. Anticancer Eects of Postbiotic Mediators from Lactobacillus Species on Acute Lymphoblastic Leukemia
2GMJ.2023;12:e3096
www.gmj.ir
eects of traditional chemotherapy drugs, as
well as the development of drug resistance
[3]. As the prognosis of cancer has increased,
the focus of treatment has shifted from rescu-
ing patients at all costs to saving patients at
the lowest cost [4].
Consequently, there is a growing interest in
exploring alternative and complementary
therapies that possess anticancer properties
with reduced adverse eects on peripheral
blood mononuclear cells (PBMCs).
Probiotics, living microorganisms provid-
ing health advantages to the host when given
appropriately, have been extensively studied
for their potential therapeutic applications in
various diseases, including cancer [5]. Probi-
otics, particularly lactic acid bacteria (LAB)
including Lactobacillus species, given their
well-documented safety prole and benecial
eects on the human gut microbiome, have
attracted considerable attention due to their
benecial eects on host health, including a
potential role in preventing cancer and adju-
vant cancer therapy [6, 7]. Among these, Lac-
tobacillus rhamnosus GG (LGG) and Lacto-
bacillus reuteri (LR) have shown promise in
reducing inammation, modulating the im-
mune system, and the growth of pathogenic
bacterial [8, 9].
The mechanisms of probiotic activity against
cancer are multifactorial, involving immuno-
modulation, suppression of tumor cell prolif-
eration, and the production of bioactive me-
tabolites [10].
Recently, research has shifted focus from the
use of live probiotic bacteria to the applica-
tion of their metabolic byproducts, called
postbiotics or postbiotic mediators (PMs).
PMs are bioactive molecules including a het-
erogeneous group of compounds, consisting
of proteins, peptides, cell wall components,
and other metabolites, mediating the bene-
cial eects of probiotics on host physiology
[11, 12].
PMs oer several advantages over live pro-
biotics, including enhanced stability, reduced
risk of bacterial translocation, absence of an-
tibiotic resistance concerns, reduced immune
response, and lower risk of infection, especial-
ly in immunocompromised such as patients
with ALL [12, 13]. Recent evidence suggests
that certain PMs derived from Lactobacillus
species may possess anticancer properties
[14]. For example, LGG-derived proteins and
exopolysaccharides have antigenotoxic and
cytotoxic potential to inhibit the prolifera-
tion and inltration of colon cancer cells [15].
Similarly, LR-produced reuterin suppresses
proliferation and induces cell death in various
human cancerous cells [16, 17]. However, the
potential anticancer activity of PMs derived
from LGG and LR against ALL remained
largely unexplored. This study aimed to ex-
amine the anticancer and cytotoxic properties
of PMs derived from LGG and LR on ALL
cancer cells and PBMCs. We hypothesize
that specic PMs may selectively target ALL
cells without causing signicant cytotoxicity
to normal cells, thus representing a promising
therapeutic approach for ALL treatment. By
elucidating these PMs’ underlying molecular
mechanisms of action, we hope to contribute
to the increasing body of evidence supporting
the potential application of PMs as eective,
novel, and less toxic adjuvant therapeutic
strategies in cancer therapy.
Materials and Methods
Ethics Approval and Consent to Participate
The present study was carried out under the
authorization and oversight of the National
Institute for Medical Research Ethics Com-
mittee (IR.NIMAD.REC.1400.148) and the
Ethics Committee of Tehran University of
Medical Sciences (IR.TUMS.DENTISTRY.
REC.1400.191). The procedures were con-
ducted following the appropriate guidelines
and regulations.
Preparation of Postbiotics
LGG (ATCC 53103) and LR (ATCC 23272)
were purchased from the Pasteur Institute of
Iran (Tehran, Iran) and maintained on MRS
agar (Merck, Darmstadt, Germany). The Lac-
tobacillus strains were grown in MRS broth
(Merck, Darmstadt, Germany) for 48 hours at
37±1 °C in a CO2 incubator before each ex-
periment. After incubation, the supernatant
and cell pellet from Lactobacillus cultures
were separated by centrifugation at 10,000×g
(8,000 rpm) for 10 minutes at 4 °C. The su-
pernatant was ltered through a 0.22 µm poly-
ethersulfone membrane syringe lter (Milli-
Anticancer Eects of Postbiotic Mediators from Lactobacillus Species on Acute Lymphoblastic Leukemia Banakar M, et al.
GMJ.2023;12:e3096
www.gmj.ir
3
pore, Burlington, USA) and neutralized with
5 M sodium hydroxide to achieve a physio-
logical pH of 7.2-7.4 [18, 19].
Cell Culture and Maintenance
Immortalized ALL (ATCC TIB-152) cell line
was prepared from the Pasteur Institute, Teh-
ran. The cell line was maintained in a culture
medium consisting of RPMI, L-glutamine, 10
mM HEPES, 23.8 mM sodium carbonate, and
10% fetal bovine serum (FBS). Incubation
took place at 37°C, with an environment of
95% humidity and 5% CO2. The culture con-
ditions were monitored and adjusted daily if
necessary.
PBMCs were isolated from 5 mL of peripheral
blood samples obtained from healthy donors.
Written and informed consent was obtained
from the volunteer. Buy coats were diluted
with phosphate-buered saline (PBS) in the
rate of 1:1, and 3ml of colEX (DNA biotech,
Tehran, Iran) was added to the buy coat for
centrifuging at 400× g for 20 min. This stage
consisted of four layers. The isolation of PB-
MCs involved subjecting the second layer to a
triple wash using PBS at 100×g for a duration
of 10 minutes at ambient temperature. The
puried PBMCs were then cultured at 37°C
with 5% CO2, utilizing RPMI-1640 medium
(Sigma, St Louis, USA) enriched with 10%
(v/v) heat-inactivated FBS and 100 U/ml pen-
icillin-streptomycin.
MTT Assay for Cytotoxicity Evaluation
The Jurkat cell line was plated in 96-well mi-
croplates at a density of 25,000 cells/mL and
incubated at 37°C with a 5% CO2 atmosphere
for cytotoxicity assessment. Following 24
hours of incubation, a serial dilution of the
percentage of a wide range from 100% (V/V)
to 25% (V/V) post-biotic mediators (PM) pro-
duced by LGG and LR strains was added to
the complete growth medium. Cells not treat-
ed with PM served as controls.
The cells were incubated for 24 and 48 in-
tervals. At each instance, 20 μL of 5 mg/
mL MTT solution (3-(4,5-dimethylthi-
azol-2-yl)-2,5-diphenyl tetrazolium bromide)
(Sigma-Aldrich, Steinheim, Germany) in PBS
was added to each well. The plates were in-
cubated in the dark for a duration of 4 hours
before centrifugation at 2000×g for 5 minutes
to separate formazan crystals. Subsequently,
170 μL of growth medium was extracted from
each well, and the resulting formazan crystals
were dissolved in 100 μL of dimethyl sulfox-
ide (DMSO) (Fisher Scientic, UK) for 15–30
minutes. The absorbance of the formazan dye
was measured by a plate reader (M491-Epoch
reader, Bio Tek Instruments, Inc., Winooski,
VT, USA) at 570 nm. The experiment was
carried out in triplicate and replicated three
times.
Cell viability was calculated as a percentage
according to the following equation: [(A_
sample - A_blank) / (A_control - A_blank)] ×
100%, where A_sample represents the absor-
bance of cells treated with PMs, A_blank rep-
resents the absorbance of PMs, and A_control
represents the absorbance of untreated cells.
The half-maximal inhibitory concentration
(IC50) was determined [19].
Apoptosis Analysis by Flow Cytometry
To investigate the induction of apoptosis by
PMs derived from LGG and LR, ALL cells
were treated with PMs at a concentration de-
termined to be the IC50 value for 24 hours.
After the incubation periods, the cells were
harvested by centrifugation at 2500×g for 5
minutes and rinsed twice with cold PBS. The
cell pellets were then resuspended in 100 μL
of binding buer (10 mM HEPES, pH 7.4,
140 mM NaCl, 2.5 mM CaCl2), and stained
with 5 μL of Annexin V-FITC (uorescein
isothiocyanate) and 5 μL of propidium iodide
(PI) solution (BD Biosciences, USA) for 15
minutes at room temperature while being kept
in the dark. Each sample was supplemented
with 400 μL of binding buer, and the stained
cells were subsequently analyzed using a ow
cytometer (BD FACSCanto II, USA) within a
time frame of one hour.
A minimum of 10,000 events were recorded
for each sample, and the data were analysed
using FlowJo software (BD Biosciences,
USA). The apoptotic cells were characterized
by their positive staining for Annexin V-FITC
and negative staining for PI, indicating early
apoptosis, or by their positive staining for both
Annexin V-FITC and PI, indicating late apop-
tosis. The apoptotic cell percentage in each of
the treatment groups was compared to that of
the control group to assess the pro-apoptotic
4GMJ.2023;12:e3096
www.gmj.ir
Banakar M, et al. Anticancer Eects of Postbiotic Mediators from Lactobacillus Species on Acute Lymphoblastic Leukemia
impact of the PMs on ALL cells. The experi-
ments were conducted in triplicate and repli-
cated thrice to ensure reproducibility.
RNA Isolation, cDNA Synthesis, and Quanti-
tative Real-Time PCR Analysis
The extraction of total RNA from both treat-
ed and untreated Jurkat cells was performed
using the RNeasy Mini Kit (GeneAll Biotech-
nology Co, Korea) according to the manufac-
turers instructions.
The concentration and purity of RNA were
evaluated using a NanoDrop instrument
(M491-Epoch reader, Bio Tek Instruments,
Inc., Winooski, VT, USA). A Revert Aid First
Strand cDNA Synthesis Kit (Thermo Fish-
er Scientic, US) was utilized to synthesize
complementary DNA (cDNA) from 2μg of
total RNA following the instructions provided
by the manufacturer. The cDNA synthesis was
conducted using a 20 μL volume comprising
1 μg of total RNA, 2 μL of 10x reverse tran-
scription (RT) buer, 0.8 μL of 25x deoxyri-
bonucleotide triphosphate (dNTP) mix (100
mM), 2 μL of 10x RT random primers, 1 μL
of MultiScribe Reverse Transcriptase (50 U/
μL), 1 μL of RNase inhibitor (20 U/μL), and
nuclease-free water.
The reverse transcription reaction was con-
ducted using a thermal cycler under the speci-
ed conditions: incubation at 25°C for a dura-
tion of 10 minutes, followed by incubation at
37°C for 60 minutes, and nally, incubation at
85°C for 5 minutes. The experimental proce-
dure involved the utilization of the PowerUp
SYBR Green Master Mix (Takara, Kyoto,
Japan) Detection System and Software (Re-
al-time PCR Roche Light Sycler 96) to per-
form quantitative reverse transcription poly-
merase chain reaction (qRT-PCR) analysis.
The reaction mixture for qRT-PCR consisted
of 20 μL in total.
This included 10 μL of PowerUp SYBR Green
Master Mix, 1 μL of cDNA template, 1 μL of
each forward and reverse primer (at a concen-
tration of 10 μM), and 7 μL of nuclease-free
water. The thermal cycling protocol consisted
of the following steps: an initial activation of
uracil-DNA glycosylase (UDG) at a tempera-
ture of 50°C for a duration of two minutes,
followed by activation of the dual-lock DNA
polymerase at 95°C. This was then followed
by 40 cycles of denaturation at 95°C for 15
seconds and annealing/extension at 60°C for
1 minute. The primer sequences for the target
genes (BAX, BCL-2, BCLX, FAS, and p27) and
the reference gene (ACTB) were designed uti-
lizing the Primer-BLAST tool provided by the
National Center for Biotechnology Informa-
tion (NCBI).
The analysis of the relative expression levels
of target genes was conducted according to
the method proposed by Livak and Schmitt-
gen [20]. The experiments were conducted
three times, and the data were reported as the
mean ± standard deviation (SD).
Statistical Analysis
The experiments were conducted in triplicate,
each experiment was performed independent-
ly. The resulting data were reported as the
mean value along with the standard deviation
(SD). The statistical method of one-way anal-
ysis of variance (ANOVA) was utilized, fol-
lowed by the application of Tukey’s post hoc
test, to identify signicant dierences in the
analyses. Analysis of ow cytometry data was
done by t-test. The gene expression level was
also determined as n-fold changes relative to
the calibrator. The statistical signicance of
the results was demonstrated by a P-value less
than 0.05.
Results
PMs Correlated with Cell Viability
The eect of PMs derived from LR and LGG
on acute lymphoblastic leukemia (Jurkat)
cells and PBMCs was evaluated using the
MTT assay. Cells were subjected to dierent
concentrations of the PMs (100%, 50%, and
25% (v/v)) for 24 hours and 48 hours (Fig-
ure-1).
Eects of LGG and LR PMs on Jurkat Cell
Viability
The viability of Jurkat cells, when exposed
to PMs of LGG at concentrations of 100%,
50%, and 25% (v/v), exhibited a signicant
decrease after 24 hours in comparison to the
control group (P<0.05), with mean viabilities
of 48.09%, 41.70%, and 45.39%, respectively.
Similarly, LR PMs at 100%, 50%, and 25%
(v/v) also signicantly reduced the viability
GMJ.2023;12:e3096
www.gmj.ir
5
Anticancer Eects of Postbiotic Mediators from Lactobacillus Species on Acute Lymphoblastic Leukemia Banakar M, et al.
of Jurkat cells compared to the control group
(P<0.05), with mean viabilities of 41.82%,
43.82%, and 51.00%, respectively. After a
duration of 48 hours, the viability of Jurkat
cells, when subjected to LGG and LR postbi-
otic mediators at various concentrations, did
not show any signicant dierence among the
concentrations investigated (P>0.05).
Eects of LGG and LR PMs on Normal Cell
Viability
The viability of PBMCs was assessed through
MTT assays following a 24-hour exposure to
LGG and LR PMs. Following 24-hours, there
was not any signicant dierence in cellular
viability between the experimental groups
subjected to LGG PMs and the control group
(P>0.05). The ndings of this study demon-
strate that the PMs derived from LGG and LR
did not exhibit a statistically signicant cyto-
toxic impact on PBMCs at the concentrations
examined.
Gene Expression
The gene expression levels of BAX, BCL-2,
BCLX, FAS, and p27 in Jurkat cells untreat-
ed and treated with PMs from IC50 (100v/v)
LGG and IC50 (25v/v) LR after 24 hours
were assessed by qRT-PCR (Figure-2). p27
expression increased notably in both groups
treated with PMs. Nevertheless, there was no
signicant dierence in BAX, BCL-2, BCLX,
FA S , and p27 gene expression levels between
the treated and untreated groups for either LR
or LGG (P>0.05).
Flow Cytometry
The apoptosis of Jurkat cells treated with
IC50 particulate matter PMs from LGG and
LR was assessed using ow cytometry (Fig-
ure-3). In the group of cells subjected to LGG
PMs treatment, a signicant increase was de-
tected in the occurrence of both early apop-
tosis (P=0.007) and late apoptosis (P=0.005)
when compared to the control group. Simi-
larly, signicant increases were noted in both
early apoptosis (P<0.001) and late apoptosis
(P=0.033) within the group subjected to LR
PMs treatment in comparison to the control
group.
Figure 1 A-C. PMs Correlated with Cell Viability. A: The impact of PMs on Jurkat cells after 24h. B: The
impact of PMs on Jurkat cells after 48h. C: The impact of PMs on peripheral blood monocular cells after
24h. (*P<0.05; **P<0.01).
6GMJ.2023;12:e3096
www.gmj.ir
Banakar M, et al. Anticancer Eects of Postbiotic Mediators from Lactobacillus Species on Acute Lymphoblastic Leukemia
Discussion
The primary objective of the present study was
to examine the potential anticancer properties
of PMs obtained from LGG and LR on Jur-
kat cells. The results of our study indicate that
the PMs derived from LGG and LR exhibited
a time-dependent decrease in Jurkat cell via-
bility. Importantly, these PMs showed limited
cytotoxicity towards normal peripheral blood
PBMCs. Furthermore, ow cytometry analy-
sis demonstrated a signicant increase in both
early and late apoptosis within Jurkat cells
subjected to PMs. This observation suggests
the potential of these mediators in inducing
cell death in leukemia cells.
The observed reduction in Jurkat cell viabil-
ity after treatment with LGG and LR PMs is
in line with previous research reporting the
anticancer potential of probiotics and their
derived products, specically in leukemia
[21-23]. Our results are consistent with other
studies reporting the anticancer eects of pro-
biotic bacteria and their metabolites on vari-
ous cancer cell lines, including colon, breast,
and esophageal cancer [22, 24, 25]. It should
also be noted that the cytotoxic impacts of
Lactobacillus strains on cancerous cells are
not limited to their live forms. Studies have
shown that heat-killed cells and cell-free L.
plantarum and LGG supernatants can reduce
cancerous cells’ growth rate [24]. Interesting-
ly, the current study demonstrated that LGG
and LR PMs had minimal cytotoxic eects
on normal PBMCs, indicating the potential
for selective cytotoxicity toward cancer cells
without harming healthy cells, consistent
with other studies [19, 23]. Although some
studies have shown that probiotic strains also
reduce the growth rate of normal cells [26],
the selective cytotoxic eect on cancer cells
is crucial for developing targeted therapies
that minimize damage to healthy cells, a ma-
jor challenge in cancer treatment [27]. Flow
cytometry analysis demonstrated a signicant
increase in early and late apoptosis in both
groups treated with LGG and LR, compared
to the control group. Our study’s results align
with other research demonstrating bacterial
strains’ apoptotic eects on cancer cells. For
instance, Lactobacillus brevis (LB) induced
time-dependent apoptosis in Jurkat cells s but
not normal human peripheral blood lympho-
cytes. LB more eciently induces apoptosis
in Jurkat cells than Streptococcus thermoph-
ilus [28].
Another study showed that cell-free superna-
tants postbiotics derived from Saccharomy-
ces cerevisiae var. boulardii have potential
antigenotoxic and cytotoxic eects on HT-29
human colon cancerous cells [29]. Despite
observing signicant eects on cell viabili-
ty and apoptosis, our study did not nd any
signicant changes in the expression levels of
pro-apoptotic (BAX and FA S ) and anti-apop-
totic (BCL-2 and BCLX) and cell cycle regu-
lator (p27) genes in Jurkat cells treated with
LGG and LR PMs. In contrast to our study,
when cells were treated with LR, the increased
expression of BAX and casp3 genes in HT29
cells and a decrease in Wnt signaling pathway
gene expression has been observed, which im-
pacts cell proliferation and dierentiation in
Kyse30 cells [24, 25]. Furthermore, a study
Figure 2. Expression of the genes in treated with PMs and untreated Jurkat cells
GMJ.2023;12:e3096
www.gmj.ir
7
Anticancer Eects of Postbiotic Mediators from Lactobacillus Species on Acute Lymphoblastic Leukemia Banakar M, et al.
on the cytotoxicity of probiotic Lactobacil-
lus spp. found that the probiotic supernatants
were cytotoxic to HT-29 and HCT-116 colon
cancer cell lines and signicantly upregu-
lated cfos and cjun transcripts in these cells
[30]. These ndings suggest that these PMs’
apoptosis-inducing eects might be mediated
through a dierent molecular mechanism or
may be related to post-transcriptional modi-
cations [31]. Further investigation is required
to clarify the molecular mechanisms by which
LGG and LR PMs exert their anticancer prop-
erties.
Our study demonstrated the potential antican-
cer impacts of PMs derived from LGG and LR
on ALL cells, but some limitations should be
considered. The study focused on one cancer
cell line and one normal cell type, and future
research should investigate additional cancer
cell lines and normal cell types. Moreover, in
vitro experiments may not fully represent the
complexities of the in vivo tumor microenvi-
ronment, making animal models necessary
for further investigation. The specic active
components in LGG and LR PMs were not
identied, and gene expression analysis was
limited to a single time point. Despite these
limitations, our study provides preliminary
evidence for the potential use of LGG and LR
PMs in leukemia treatment. It warrants further
research to explore their broader applicability
in cancer therapy.
Conclusion
Findings demonstrate the potential anticancer
activity of PMs derived from LGG and LR on
ALL cells. The observed selective cytotoxic-
ity towards leukemia cells and induction of
apoptosis indicates that these PMs may hold
promise as novel natural therapeutic agents in
leukemia treatment.
Future research should prioritize investigat-
ing the molecular mechanisms contributing to
the anticancer eects of PMs and exploring
their potential in combination with conven-
tional chemotherapeutic agents to enhance
therapeutic outcomes. Additionally, the po-
tential of LGG and LR PMs in other cancer
types should be investigated to determine
their broader applicability in cancer therapy.
It would also be valuable to assess these PMs’
long-term safety and ecacy in preclinical
and clinical settings. Identifying the specic
active components responsible for their anti-
cancer eects may lead to the development
of targeted therapies for leukemia and other
cancers, ultimately enhancing outcomes and
patients’ quality of life.
Acknowledgments
This study was supported by grants from the
National Institute of Medical Sciences Re-
search, Tehran, Iran (4001382). The funding
Figure 3. The e󰀨ects of LR and LGG PMs on Jurkat cells apoptosis
8GMJ.2023;12:e3096
www.gmj.ir
Banakar M, et al. Anticancer Eects of Postbiotic Mediators from Lactobacillus Species on Acute Lymphoblastic Leukemia
1. Sung H, Ferlay J, Siegel R, Laversanne M,
Soerjomataram I, Jemal A, et al. Global Can-
cer Statistics 2020: GLOBOCAN Estimates
of Incidence and Mortality Worldwide for 36
Cancers in 185 Countries. CA Cancer J Clin.
2021; 71: 209-49.
2. Pui C-H, Yang JJ, Hunger SP, Pieters R,
Schrappe M, Biondi A, et al. Childhood
Acute Lymphoblastic Leukemia: Prog-
ress Through Collaboration. J Clin Oncol.
2015;33(27):2938-48.
3. Wu C, Li W. Genomics and pharmacogenom-
ics of pediatric acute lymphoblastic leuke-
mia. Crit Rev Oncol Hematol. 2018;126:100-
11.
4. Hunger SP, Mullighan CG. Acute Lympho-
blastic Leukemia in Children. N Engl J Med.
2015;373(16):1541-52.
5. Motevaseli E, Dianatpour A, Ghafouri-Fard
S. The role of probiotics in cancer treat-
ment: emphasis on their in vivo and in vitro
anti-metastatic eects. Int J Mol Cell Med.
2017;6(2):66.
6. Galdeano CM, Cazorla SI, Dumit JML,
Vélez E, Perdigón G. Benecial eects of
probiotic consumption on the immune sys-
tem. Ann Nutr Metab. 2019;74(2):115-24.
7. Alam Z, Shang X, Eat K, Kanwal F, He X,
Li Y, et al. The potential role of prebiotics,
probiotics, and synbiotics in adjuvant cancer
therapy especially colorectal cancer. J Food
Biochem. 2022;46(10):e14302.
8. Ghorbani E, Avan A, Ryzhikov M, Ferns
G, Khazaei M, Soleimanpour S. Role of
Lactobacillus strains in the management
of colorectal cancer An overview of recent
advances. Nutrition. 2022:111828.
9. Thomas CM, Hong T, Van Pijkeren JP,
Hemarajata P, Trinh DV, Hu W, et al. Hista-
mine derived from probiotic Lactobacillus
reuteri suppresses TNF via modulation
of PKA and ERK signaling. PLoS One.
2012;7(2):e31951.
10. Lozenov S, Krastev B, Nikolaev G, Peshevs-
ka-Sekulovska M, Peruhova M, Velikova
T. Gut Microbiome Composition and Its
Metabolites Are a Key Regulating Factor
for Malignant Transformation, Metastasis
and Antitumor Immunity. Int J Mol Sci.
2023;24(6):5978.
11. Thorakkattu P, Khanashyam AC, Shah
K, Babu KS, Mundanat AS, Deliephan
A, et al. Postbiotics: Current trends in
food and Pharmaceutical industry. Foods.
2022;11(19):3094.
12. Aguilar-Toalá JE, Garcia-Varela R, Garcia
HS, Mata-Haro V, González-Córdova AF,
Vallejo-Cordoba B, et al. Postbiotics: An
evolving term within the functional foods
eld. Trends Food Sci Technol. 2018;75:105-
14.
13. Ambesh P, Stroud S, Franzova E, Gotes-
man J, Sharma K, Wolf L, et al. Recurrent
Lactobacillus Bacteremia in a Patient With
Leukemia. J Investig Med High Impact Case
Rep. 2017;5(4):2324709617744233.
14. Zitvogel L, Daillère R, Roberti MP, Routy B,
Kroemer G. Anticancer eects of the micro-
biome and its products. Nat Rev Microbiol.
2017;15(8):465-78.
15. Sharma M, Chandel D, Shukla G. Antigeno-
toxicity and Cytotoxic Potentials of Met-
abiotics Extracted from Isolated Probiotic,
Lactobacillus rhamnosus MD 14 on Caco-2
and HT-29 Human Colon Cancer Cells. Nutr
Cancer. 2020;72(1):110-9.
16. Schaefer L, Auchtunger F, Mallett G, Foer-
ster F, Porter A, Malvezzi M, et al. Global
trends in colorectal cancer mortality: projec-
tions to the year 2035. Int J Cancer. 2018;
142(12): 2489-96.
17. Zahran WE, Elsonbaty SM, Moawed FSM.
Lactobacillus rhamnosus ATCC 7469 exo-
polysaccharides synergizes with low level
ionizing radiation to modulate signaling mo-
lecular targets in colorectal carcinogenesis in
rats. Biomed Pharmacother. 2017;92:384-93.
18. Banakar M, Pourhajibagher M, Ete-
mad-Moghadam S, Mehran M, Yazdi MH,
Haghgoo R, et al. Antimicrobial Eects of
Postbiotic Mediators Derived from Lacto-
bacillus rhamnosus GG and Lactobacillus
reuteri on Streptococcus mutans. FBL.
2023;28(5):88.
19. Chuah L-O, Foo HL, Loh TC, Mohammed
Alitheen NB, Yeap SK, Abdul Mutalib NE, et
al. Postbiotic metabolites produced by Lac-
tobacillus plantarum strains exert selective
References
bodies of the study did not play any role in
its design, collection, analysis, data interpre-
tation, and manuscript writing.
Conict of Interest
The authors declare no conict of interest.
cytotoxicity eects on cancer cells. BMC
Complement Altern Med. 2019;19(1):114.
20. Livak KJ, Schmittgen TD. Analysis of
relative gene expression data using real-time
quantitative PCR and the 2− ΔΔCT method.
Methods. 2001;25(4):402-8.
21. Aghebati-Maleki L, Hasannezhad P, Abbasi
A, Khani N. Antibacterial, antiviral, antioxi-
dant, and anticancer activities of postbiotics:
a review of mechanisms and therapeutic
perspectives. Biointerface Res Appl Chem.
2021;12:2629-45.
22. Hadad SE, Baik H, Alhebshi AM, Aburas
M, Alrahimi J, Hassoubah S. Lactobacillus
acidophilus’ post biotic extracts combined
with methotrexate regulated the levels of
the apoptotic genes’ expressions in the acute
lymphoblastic leukemia cell line. Med Sci.
2022;26(122):1.
23. Maftuni K, Zare P. Eects of Lactobacillus
casei culture supernatant on dierentiation of
K562 cell line. Med Lab J. 2017;11(5):16-21.
24. ZibasazTalebi A, Ahmadizadh C. The Study
of Expression of Casp3/Bax Genes in HT29
Colon Cancer Cell Line with Lactobacillus
rhamnosus Co-Culturing. J Ilam Univ Med
Sci. 2020;28(4):25-35.
25. Hashemi-Khah MS, Arbab-Soleimani N,
Forghanifard MM, Gholami O, Taheri S,
Amoueian S. An In Vivo Study of Lacto-
bacillus rhamnosus (PTCC 1637) as a New
Therapeutic Candidate in Esophageal Cancer.
Biomed Res Int. 2022;2022:7607470.
26. Sadeghi-Aliabadi H, Mohammadi F, Faze-
li H, Mirlohi M. Eects of Lactobacillus
plantarum A7 with probiotic potential on
colon cancer and normal cells proliferation in
comparison with a commercial strain. Iran J
Basic Med Sci. 2014;17(10):815-9.
27. Zhong L, Li Y, Xiong L, Wang W, Wu M,
Yuan T, et al. Small molecules in targeted
cancer therapy: Advances, challenges, and
future perspectives. Signal Transduct Target
Ther. 2021;6(1):201.
28. Afrin S, Giampieri F, Gasparrini M,
Forbes-Hernández TY, Cianciosi D, Rebore-
do-Rodriguez P, et al. Dietary phytochemi-
cals in colorectal cancer prevention and treat-
ment: A focus on the molecular mechanisms
involved. Biotechnol Adv. 2020;38:107322.
29. Abbasi A, Rad AH, Maleki LA, Kal HS,
Baghbanzadeh A. Antigenotoxicity and Cy-
totoxic Potentials of Cell-Free Supernatants
Derived from Saccharomyces cerevisiae var
boulardii on HT-29 Human Colon Cancer
Cell Lines. Probiotics Antimicrob Proteins.
2023:1-3.
30. Shyu PT, Oyong GG, Cabrera EC. Cytotox-
icity of probiotics from Philippine commer-
cial dairy products on cancer cells and the
eect on expression of cfos and cjun early
apoptotic-promoting genes and Interleukin-1
β and Tumor Necrosis Factor-α proinam-
matory cytokine genes. Biomed Res Int.
2014;2014:491740.
31. Pfeer CM, Singh AT. Apoptosis: a target
for anticancer therapy. IJMS. 2018 Feb
2;19(2):448.
GMJ.2023;12:e3096
www.gmj.ir
9
Anticancer Eects of Postbiotic Mediators from Lactobacillus Species on Acute Lymphoblastic Leukemia Banakar M, et al.