ArticlePDF Available

Lactate in the tumor microenvironment: A rising star for targeted tumor therapy

Frontiers
Frontiers in Nutrition
Authors:

Abstract and Figures

Metabolic reprogramming is one of fourteen hallmarks of tumor cells, among which aerobic glycolysis, often known as the “Warburg effect,” is essential to the fast proliferation and aggressive metastasis of tumor cells. Lactate, on the other hand, as a ubiquitous molecule in the tumor microenvironment (TME), is generated primarily by tumor cells undergoing glycolysis. To prevent intracellular acidification, malignant cells often remove lactate along with H⁺, yet the acidification of TME is inevitable. Not only does the highly concentrated lactate within the TME serve as a substrate to supply energy to the malignant cells, but it also works as a signal to activate multiple pathways that enhance tumor metastasis and invasion, intratumoral angiogenesis, as well as immune escape. In this review, we aim to discuss the latest findings on lactate metabolism in tumor cells, particularly the capacity of extracellular lactate to influence cells in the tumor microenvironment. In addition, we examine current treatment techniques employing existing medications that target and interfere with lactate generation and transport in cancer therapy. New research shows that targeting lactate metabolism, lactate-regulated cells, and lactate action pathways are viable cancer therapy strategies.
This content is subject to copyright.
TYPE Review
PUBLISHED 16 February 2023
DOI 10.3389/fnut.2023.1113739
OPEN ACCESS
EDITED BY
Peng Liu,
Sun Yat-sen University, China
REVIEWED BY
Paolo Paoli,
University of Florence, Italy
María A. García,
University of Concepcion, Chile
*CORRESPONDENCE
Aihua Gong
ahg5@mail.ujs.edu.cn
These authors have contributed equally to this
work and share first authorship
SPECIALTY SECTION
This article was submitted to
Nutritional Immunology,
a section of the journal
Frontiers in Nutrition
RECEIVED 01 December 2022
ACCEPTED 30 January 2023
PUBLISHED 16 February 2023
CITATION
Li Z, Wang Q, Huang X, Yang M, Zhou S, Li Z,
Fang Z, Tang Y, Chen Q, Hou H, Li L, Fei F,
Wang Q, Wu Y and Gong A (2023) Lactate in the
tumor microenvironment: A rising star for
targeted tumor therapy.
Front. Nutr. 10:1113739.
doi: 10.3389/fnut.2023.1113739
COPYRIGHT
©2023 Li, Wang, Huang, Yang, Zhou, Li, Fang,
Tang, Chen, Hou, Li, Fei, Wang, Wu and Gong.
This is an open-access article distributed under
the terms of the Creative Commons Attribution
License (CC BY). The use, distribution or
reproduction in other forums is permitted,
provided the original author(s) and the
copyright owner(s) are credited and that the
original publication in this journal is cited, in
accordance with accepted academic practice.
No use, distribution or reproduction is
permitted which does not comply with these
terms.
Lactate in the tumor
microenvironment: A rising star for
targeted tumor therapy
Zhangzuo Li1,2†, Qi Wang3† , Xufeng Huang4†, Mengting Yang2†,
Shujing Zhou5, Zhengrui Li6,7,8, Zhengzou Fang2, Yidan Tang5,
Qian Chen2, Hanjin Hou2, Li Li2, Fei Fei2, Qiaowei Wang2, Yuqing Wu2
and Aihua Gong1,2*
1Hematological Disease Institute of Jiangsu University, Aliated Hospital of Jiangsu University, Jiangsu
University, Zhenjiang, China, 2Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang,
China, 3Department of Gastroenterology, Aliated Hospital of Jiangsu University, Jiangsu University,
Zhenjiang, China, 4Faculty of Dentistry, University of Debrecen, Debrecen, Hungary, 5Faculty of Medicine,
University of Debrecen, Debrecen, Hungary, 6School of Medicine, College of Stomatology, Shanghai Jiao
Tong University, Shanghai, China, 7National Center for Stomatology and National Clinical Research Center
for Oral Diseases, Shanghai, China, 8Shanghai Key Laboratory of Stomatology, Shanghai, China
Metabolic reprogramming is one of fourteen hallmarks of tumor cells, among which
aerobic glycolysis, often known as the “Warburg eect,” is essential to the fast
proliferation and aggressive metastasis of tumor cells. Lactate, on the other hand, as
a ubiquitous molecule in the tumor microenvironment (TME), is generated primarily
by tumor cells undergoing glycolysis. To prevent intracellular acidification, malignant
cells often remove lactate along with H+, yet the acidification of TME is inevitable.
Not only does the highly concentrated lactate within the TME serve as a substrate to
supply energy to the malignant cells, but it also works as a signal to activate multiple
pathways that enhance tumor metastasis and invasion, intratumoral angiogenesis,
as well as immune escape. In this review, we aim to discuss the latest findings on
lactate metabolism in tumor cells, particularly the capacity of extracellular lactate
to influence cells in the tumor microenvironment. In addition, we examine current
treatment techniques employing existing medications that target and interfere with
lactate generation and transport in cancer therapy. New research shows that targeting
lactate metabolism, lactate-regulated cells, and lactate action pathways are viable
cancer therapy strategies.
KEYWORDS
lactate, tumor microenvironment, metabolic, immunity, immune cells
1. Introduction
The cellular transformation includes uncontrolled cell proliferation, resistance to cell death,
immune evasion, and evasion of growth inhibitory activity, ultimately leading to cancer
formation (1,2). Furthermore, it has been observed that, as part of the tumor survival machinery,
tumor cells adapt to different survival challenges by altering their metabolism, a feature now
considered a hallmark of cancer (13). Most normal tissues obtain energy through aerobic
respiration in the presence of oxygen, while energy is provided only in the absence of oxygen
through glycolysis. However, in tumor tissues, tumor cells select a low adenosine triphosphate
(ATP)-generating mode of glycolysis to provide energy for their rapid growth and proliferation,
even under adequate oxygen conditions, a specific phenomenon now known as the Warburg
effect (4,5), also known as glycolysis. The reasons why tumor cells choose this seemingly
uneconomical method of metabolism include the fact that glycolysis allows tumor cells to better
adapt to fluctuations in oxygen partial pressure. The intermediates of glycolysis can be used by
Frontiers in Nutrition 01 frontiersin.org
Li et al. 10.3389/fnut.2023.1113739
tumor cells to synthesize macromolecules such as proteins, nucleic
acids, and lipids needed for cell construction, thus maintaining the
growth and proliferation of the tumor cells themselves, although
glycolysis produces less ATP than aerobic respiration, it produces
ATP at a higher rate to meet the energy demands of rapid tumor
growth, and using glycolysis as the primary mode of energy supply
reduces the mitochondrial of the electron transport chain (6), the
production of anaerobic glucose glycolysis reduces the production
of free radicals (ROS) and thus reduces the toxicity to tumor cells
(6). Cancer cells use large amounts of glucose as their energy source,
leading to the accumulation of extracellular lactate, which can alter
the metabolic patterns of multiple cells, including immune cells,
within the tumor microenvironment (TME) (7). In the early stages
of tumor growth, immune cells recruited and activated by tumor
cells can form a tumor-suppressive inflammatory microenvironment
that hinders tumor progression. However, as tumor cells continue to
proliferate and continue the immune activation response, the TME
undergoes dynamic changes: immune effector cells become depleted
or remodeled thus failing to function properly. An important cause
of these changes is the high concentration of lactate produced by
the Warburg effect, which affects the differentiation, metabolism, and
function of tumor-infiltrating immune cells through several pathways
(8). For many years lactate has been considered a waste metabolite,
but it is now clear that lactate plays an irreplaceable role in promoting
tumor cell survival, oncogene signaling (9), inflammation, metastasis
(10), tumor resistance (11), immunosuppression (12,13), and many
other oncogenic processes. In this review, we briefly discuss the role
of lactate in tumor progression, particularly its role in the metabolic
microenvironment, immune microenvironment, and metastasis. We
conclude that studies of targeted lactate therapeutic strategies and
transport and combination therapy with other agents offer a new
way of thinking to attack cancer and discuss the potential for clinical
translation of lactate therapy.
2. The role of lactate in promoting
tumor progression
2.1. The role of lactate in tumor cell
metabolism
It has been found that lactate can stabilize HIF-2αand activate c-
Myc, which results in an up-regulation in the expression of glutamine
transporter (ASCT2) and GLS1, and an enhancement in glutamine
uptake as well as catabolism (14). Damiani et al. found that tumor
cells can maximize the use of α-ketoglutaric acid (α-KG), a catabolite
of glutamine, into the TCA cycle, which is converted to pyruvate by
malase and finally to lactate by LDHA (15) (Figure 1). The above
preliminary clues suggest that glycolysis and glutamine catabolism
pathways may be interdependent and jointly maintain the interstitial
phenotype of tumor cells, but it is not clear whether there is a
regulatory relationship between key enzymes LDHA and GLS1.
As a “mitochondrial vent”, PYCR dissipates electron aggregation
by oxidizing NADH to NAD+, allowing the TCA cycle to proceed
independently of oxygen consumption (16). This is also necessary
for the oxidation of NADH produced by the glycolysis pathway (17).
In addition, early reports found that lactate could directly inhibit
the activity of proline oxidase (PRODH/POX) (18), and indirectly
negatively regulates the expression of PRODH/POX, while reducing
the degradation of Pro (1921). Lactate can activate c-Myc to up-
regulate the expression of PYCR, thus up-regulating the expression
of proline. The above results suggest that lactate plays a positive
regulatory role in the process of the Pro synthesis pathway.
2.2. Eect of lactate on tumor immunity
2.2.1. Lactate and innate immune cells in the TME
Through the activity of its stromal cells, TME is in a state
of continuous modification with the progress of the tumor. As
an important site for tumor cells to survive, TME has a complex
composition, including immune cells, cancer associated fibroblasts
(CAFs), vascular endothelial cells, and other types of cells, an
extracellular matrix, and a large number of active molecules.
On the one hand, innate immune cells present in TME, including
macrophages, neutrophils, dendritic cells, natural lymphoid cells,
invariant natural killer cells, and myeloid-derived suppressor cells, as
well as adaptive immune cells including T and B cells, are responsible
for detecting and eliminating cancer cells (22,23). On the other hand,
tumor cells can recruit immunosuppressive cell populations into
TME by secreting anti-inflammatory cytokines, where they directly
suppress immune responses (24) (Figure 2).
Invariant natural killer T (iNKT) cells are at the forefront
of the anti-tumor immune response. They can not only kill
target cells directly through antigen recognition but also enhance
anti-tumor immune responses by inhibiting tumor-associated
macrophages (TAMs) and promoting the activation of NK and
Cytotoxic T lymphocytes (CTLs) (2528). PPARγand BTB–zinc
finger transcriptional regulator PLZF synergically promote lipid
biosynthesis of iNKT cells after activation through enhancing
transcription of SERBP1. In the tumor microenvironment with high
concentrations of lactate, the expression of PPARγin intratumoral
iNKT cells will be inhibited, thus consequently reducing their
cholesterol synthesis. And cholesterol is necessary for iNKT cells to
produce the optimal IFN- γ(29).
Tumor-associated macrophages are the most numerous subsets
in the immune microenvironment, accounting for more than 50%
of the immune cells in the tumor microenvironment (30). There
are two main phenotypes of TAM: M1-TAM (tumor suppressor)
has the function of antigen presentation and can activate immune
factors, which is beneficial to anti-tumor immune response. M2-
TAM (tumor-promoting type) can inhibit the inflammatory reaction,
shield tumor immune surveillance, and promote tumor growth
and metastasis (31). Lactate can activate the MCT-HIF1αpathway
and promote macrophages to M2 polarization, which further
complements the regulatory mechanism of macrophage polarization
(32). The expression of IL-12 in M2-TAM decreased and the
expression of IL-10 increased, which could promote the occurrence
and growth of tumors. Experiments have shown that TAMs can
regulate cancer progression through a variety of mechanisms (33).
Lactate can also inhibit the degradation of HIF2αby activating
mTORC1 in macrophages, thus promoting tumor development
(34). Lactate inhibits the activation of YAP and nuclear factor-
kB through a GPR81-mediated signal and reduces the production
of pro-inflammatory cytokines in macrophages, thus inhibiting
the pro-inflammatory response of macrophages to LPS stimulation
(35). Regulation of lactate levels can promote the transformation
Frontiers in Nutrition 02 frontiersin.org
Li et al. 10.3389/fnut.2023.1113739
FIGURE 1
Glycolysis and glutamine metabolism. Glucose is catabolized to pyruvate in the cytoplasm. Conversion of lactate from pyruvate stabilizes HIF-2a, which
then stabilizes c-Myc protein expression in the nucleus. c-Myc promotes glutamine transporter protein 2 (ASCT2) expression. Glutaminase 1 (GLS1)
catalyzes the breakdown of glutamine to glutamate in the mitochondria. In the mitochondrial matrix, glutamate is converted to a-KG and enters the
tricarboxylic acid cycle.
of macrophages from M1 to M2 and upregulate PD-L1 to help
tumor immune escape (36). In addition, studies have shown that
reducing the level of lactate in tumors can inhibit the polarization
of macrophages to M2, thus inhibiting the secretion of CCL17 and
finally inhibiting the invasion of pituitary adenomas (37).
Myeloid-derived suppressor cells (MDSCs) represent a group
of expanded, heterogeneous immature myeloid cells that can be
subdivided into monocytic MDSCs (M-MDSCs) and granulocyte
MDSCs (G-MDSCs). These two MDSC subsets are thought to
have a significant ability to block innate and adaptive immunity
(38). Several studies have shown that lactate in the acidic tumor
metabolic microenvironment induces increased HIFαin MDSCs,
leading to increased expression of programmed death ligand 1 (PD-
L1), which regulates the development of myeloid cells (39,40).
Notch-RBP-J signaling plays a key role in determining cell fate
and plasticity, and it is highly conserved (41). Recent studies have
shown that myeloid-specific activation of Notch/RBP-J signaling
could inhibit the transcription of the lactate transporter MCT2
through its downstream molecule Hes1, leading to a decrease in
intracellular lactate levels and inhibition of granulocyte MDSC (G-
MDSC) differentiation. Combining Notch activation and MCT2
inhibition in myeloid cells represses tumor growth (37).
DCs are the most functional specialized antigen-presenting cells
found in vivo and they play a crucial role in initiating specific
antitumor T-cell responses (42). One of the main functions of DCs
is to activate the immune response by processing and presenting
antigens through the MHC-II and MHC-I (43). The accumulation of
lactate in tumors prevents the differentiation of DCs and makes the
cells tolerant (44), limits the ability of DCs to recognize and present
antigens (45), inactivates cytokines released by DCs, and promotes
the production of an important immunosuppressive cytokine, IL-10,
by DCs and inhibits the secretion of the pro-inflammatory factor
IL-12 (46,47). The effect of lactate on DC cells was also reversible
when lactate was neutralized with NaOH to pH 7.4, showing that the
inhibitory effect of lactate on DC activation disappeared (48).
Neutrophils are derived from bone marrow hematopoietic stem
cells and have unique morphology because of their lobular nuclei, and
can be determined by the phenotypes expressed on the cell surface.
Neutrophils account for a significant proportion of the primary
tumor immune cell infiltration (49). Recent studies have shown that
neutrophils have both anti-tumor and tumor-promoting effects in
cancers (50). N1 neutrophils are thought to have the potential to kill
tumor cells due to the activation of immune factors such as TNF-α,
ICAM-1, and FAS, resulting in elevated levels of immune factors and
direct antibody-dependent cytotoxicity (51). N1 neutrophils can also
exert anti-tumor effects indirectly through the regulation of T-cell
function (52). It has been shown that N1 neutrophils exert antitumor
effects through the release of neutrophil elastase (53). Neutrophils
can undergo phenotypic and functional remodeling in response to
lactic acid, which is the effect of N2. N2 neutrophils release a variety
of pro-tumor factors and participate in promoting tumorigenesis
and progression through multiple mechanisms. N2 neutrophils can
release a variety of proteases that promote malignant proliferation
and metastasis of tumor cells. matrix metalloproteinases (MMPs)
mainly regulate tumor angiogenesis and metastasis. Blocking or
reducing the production of these proteases is expected to achieve the
purpose of inhibiting tumor progression (54).
2.2.2. Lactate and T cells in the TME
T cells have long been thought to be effective against tumor
cells and form lasting immunity, and their specificity to antigens
Frontiers in Nutrition 03 frontiersin.org
Li et al. 10.3389/fnut.2023.1113739
FIGURE 2
Eect of lactate in the regulation of the immune response. The lactate in the TME aects the dierentiation, metabolism, and function of innate and
adaptive tumor-infiltrating immune cells through multiple pathways and inhibits anti-tumor immune responses. The lactate secreted by tumor and
stromal cells acidifies the TME and promotes tumor cell proliferation and metastasis.
expressed by tumors is crucial (55), but other inherent characteristics
of T cells, such as persistence, longevity, and function, also play an
important role in determining the effectiveness of immunotherapy
(56). Tumor-derived lactate reduces CTL recruitment in TME and
further inhibits the function of infiltrating CTL in TME by impairing
its chemotactic and respiratory activities (57). Quinn et al. have
reported that the inhibitory effect of lactate on the proliferation of
effector T cells does not depend on acidity, which is achieved by the
transition from NAD+to NADH (lactate-induced reductive stress).
This impairs glycolysis and the production of glucose-derived serine,
which is necessary for effector T-cell proliferation (58). Tumor cells
often exhibit uncontrolled metabolic processes, leading to a tumor
microenvironment of metabolite depletion, hypoxia, and acidity,
which makes it difficult for effector T cells to exert their lethal
function. Recent studies have also found that lactate can promote
the stem cell-like characteristics of CD8+T cells, thus playing an
anti-tumor immune role in cancer treatment. This shows that lactate
has two sides in T cell anti-tumor immunity (59). Regulatory T
Cells (Tregs) also play an important role in immune homeostasis,
and unlike other immune cells, Tregs have increased activity and
recruitment in acidic TME, becoming a major barrier to anticancer
immunity (60). Lactate can be used as a carbon fuel source for Treg
to maintain its high inhibition ability. Inhibition of Monocarboxylate
transporter 1 (MCT1), direct targeting of lactate metabolism, or
inhibition of tumor acidity may break the metabolic symbiosis
between tumor cells and Treg cells, thus reducing the Treg barrier
of tumor immunity and enhancing the killing function of effector T
cells (61).
2.2.3. Lactate and CAFs in the TME
Cancer-associated fibroblasts (CAFs) are the predominant
stromal cells in TME (62,63). A vast number of studies have found
that CAFs play a multifaceted function in tumor growth (6366).
They could not only provide ATP to adjacent cancer cells, but also
regulate the tumor cells and tumor microenvironment by secreting
various growth factors, cytokines, and chemokines [48], and prevent
the deep infiltration of drugs and immune cells into tumor tissue
by shaping the tumor extracellular matrix and forming a drug or
therapeutic immune cell permeation barrier, thereby diminishing
the therapeutic effects of the anticancer drugs (67,68). In terms of
offering energetic sources for tumor cells, CAFs emit a substantial
quantity of lactate through glycolysis to the tumor cells which in turn
use it as a fuel to support their physiological activities (69). A recent
study demonstrated evidence in which the tumor-secreted lactate
downregulates p62 in stromal fibroblasts, which essentially induces
the CAF phenotype (70,71).
Frontiers in Nutrition 04 frontiersin.org
Li et al. 10.3389/fnut.2023.1113739
3. Mechanism of lactate inhibition of
antitumor response
3.1. Gene regulation by histone lactylation in
tumor cells
Lactate modification (lactylation) is a histone post-translational
modification reported by Zhang et al. for the first time, which plays
a role in gene transcriptional regulation (72). Follow-up studies
have further confirmed that protein lactylation is an important
way for lactate to exert its function and participate in cellular
life activities such as glycolysis-related cell function, macrophage
polarization, nervous system regulation, and so on (7375). The
discovery of histone lactylation has pointed out a new direction
for the research on the participation of tumor cell metabolites in
the tumor, immunity, and other fields. The researchers found that
lactylation of histone lysine residue, as an epigenetic modification,
directly promoted chromatin gene transcription. The researchers
identified 28 lactate sites on the core histones of human and mouse
cells. Hypoxia and bacterial stimulation induce the production of
lactate through glycolysis, and it acts as a precursor to promote
histone lactylation. Using M1 macrophages exposed to bacteria
as a model system, the researchers found that histone lactylation
and acetylation had different time dynamics. Stimulation of mouse
bone marrow-derived macrophages with lipopolysaccharide and
interferon γ(LPS+IFNγ) to simulate infection with Gram-negative
bacteria revealed that macrophages produced large amounts of
lactate and induced histone lactylation modifications at 16–24 h
of stimulation; while histone acetylation levels decreased at the
same time. In the late stage of M1 macrophage polarization, the
lactate modification of histones is enhanced, which induces steady-
state genes involved in wound healing, including Arg1. Overall,
these results suggest that the endogenous “Lactate clock” in M1
macrophages attacked by bacteria turns on gene expression to
promote balance in the body (67). Histone lactylation, therefore,
provides an opportunity to improve understanding of the function
of lactate and its role in various pathophysiological conditions,
including infection and cancer. A recent study has shown that histone
lactylation accelerates tumorigenesis by activating M6A interpreter
protein YTHDF2, which provides a new histone lactylation target for
the treatment of ocular melanoma (73). At the same time, it also links
histone modification with RNA modification, which provides a new
understanding of epigenetic regulation.
3.2. Lactate/GPR81 signaling in cancer cells
In recent years, the importance of lactate to the survival and
growth of cancer cells has been proven to be achieved in part by its
ability to activate the lactate receptor (HCAR1), which is also called
GPR81. GPR81 is widely distributed in human tissues. GPR81 has
been identified as a lactate receptor (75). It is expressed in a variety
of cell types, including adipocytes, brain cells, skeletal muscle cells,
and various cancer cells (76). Xie et al. clarified the transcriptional
mechanism of GPR81 expression regulation in cancer cells. Lactate
induces the transcription factor Snail/STAT3 pathway and up-
regulates GPR81 expression through autocrine regulation (77). A
study has emphasized the importance of GPR81 in tumor growth
and migration. GPR81 promotes cell proliferation and angiogenesis
in a PI3K/Akt/cAMP-dependent manner in breast cancer cells.
Silencing GPR81 and treating cells with PI3K inhibitors can reduce
angiogenesis in vitro, thus inhibiting tumor growth (78). Recent work
elucidates the up-regulation of PD-L1 in glucose-stimulated lung
cancer cells mediated by GPR81 through lactate dehydrogenase A
(LDHA). It is also proved that the activation of GPR81 reduces the
level of intracellular cAMP and inhibits the activity of protein kinase
A (PKA), resulting in the activation of transcriptional coactivator
TAZ. The interaction between TAZ and transcription factor TEAD
is the key to activating PD-L1 and inducing its expression (79).
Lactate helps to protect tumor cells from being attacked by T cells,
establishing the relationship between metabolic reprogramming of
tumor cells and tumor evasion of the immune response. Another
paper proved that immune cells are involved in the growth of GPR81-
dependent tumors. The antigen is GPR81 on the surface of DC
cells, and the activation of this receptor is related to the decrease
of cAMP, IL-6, and IL-12, and can down-regulate the expression
of MHCII on the cell surface (76). These findings suggest that
lactate from tumor cells activates GPR81 in dendritic cells and blocks
the expression of tumor-specific antigens to other immune cells.
This paracrine mechanism complements the autocrine mechanism
of PD-L1 induced by activating GPR81 in tumor cells in recent
years and provides an effective means for tumor cells to evade the
immune system. Therefore, blocking the GPR81 signal can promote
cancer immunotherapy. GPR81 is expressed in both tumor and the
surrounding immune cells at the same time, and the end result of
GPR81 activation is the promotion of angiogenesis, immune evasion,
and chemoresistance.
4. Anti-tumor metabolic therapeutic
strategies targeting lactate metabolism
4.1. Therapeutic strategies targeting lactate
metabolism
Tumor, composed of tumor cells and the TME, is currently a
major scientific challenge in the field of medicine. Traditional tumor
treatment methods include surgery, radiotherapy, chemotherapy,
and so on, but these methods may cause serious damage to the
surrounding normal tissue while killing tumor cells (8084).
The Warburg effect is prevalent in various tumors and is
characterized by a predominantly glycolytic energy metabolism
in cancer cells under adequate oxygen conditions. The glycolytic
product lactate can activate many essential signaling pathways
in cancer cells, promoting survival, invasion, immune escape,
metastasis, and angiogenesis. Combining aerobic glycolytic targeted
therapy with other therapeutic approaches, such as immunotherapy
and chemotherapy, is promising for cancer treatment.
Lactate oxidase (LOD) can catalyze the oxidation of lactate
to pyruvate and hydrogen peroxide. The use of lactate oxidase
can catalyze the change of lactate present in large amounts in
tumors to H2O2, which not only dismantles the tumor immune
microenvironment but H2O2can be converted to highly toxic
hydroxyl radicals (OH) bar catalyzed by other compounds, thus
killing tumor cells (8587). The depletion of lactic acid in the
tumor microenvironment by lactate oxidase can improve the
Frontiers in Nutrition 05 frontiersin.org
Li et al. 10.3389/fnut.2023.1113739
tumor immunosuppressive microenvironment and effectively inhibit
tumor growth.
LDH is mainly composed of different proportions of LDHA
and LDHB subunits, forming five different LDH tetramer isozymes
(LDH15). The main function of lactate dehydrogenase is to
convert pyruvate to lactate and NADH to NAD+. The up-regulated
expression of LDH can be detected in some tumor clinical samples
(8891), while the downregulation of LDH can inhibit the growth
and migration of cells in vitro and affect the occurrence of tumors
in vivo (92). Recently, lactate dehydrogenase-A (LDH-A) has been
found to protect the dryness of cancer and recruit tumor-related
macrophages to promote breast cancer progression (93). The method
targeting lactate dehydrogenase is used to inhibit the production of
lactate in TME.
In mouse cancer models, knockout of LDHA, LDHB, or knockout
alone can inhibit tumor growth, highlighting their key role in tumor
metabolism (9496). The R- (-) enantiomeric AT-101 of gossypol
acetate and its derivatives FX-11, Galloflavin, and N-hydroxyindolyl
compounds have been shown to give priority to the inhibition of
LDHA subtypes (21). In lymphoma and pancreatic cancer xenografts,
FX-11 can reduce cellular lactate production, induce oxidative stress,
and eventually lead to tumor cell apoptosis and inhibit tumor
progression (97). In prostate cancer, FX-11 as a single drug can also
effectively inhibit the glycolysis of tumor cells, and then inhibit the
growth of tumor cells (97). It is reported that Galloflavin can bind to
free LDHA and inhibit glycolysis in breast cancer cells, thus acting as
an anti-tumor agent (98).
The authors identified a new LDH inhibitor, NCI-006,
which overcomes many of the drawbacks of other inhibitors,
is highly targeted and is expected to have an effect in vivo
(92). N-hydroxyindole drugs have been shown to reduce
the growth of pancreatic and cervical cancer cells in vitro
(99). And when used in combination with gemcitabine, it
can increase the apoptosis rate of pancreatic cancer cell
lines (100).
MCTs is a proton-linked transporter responsible for transporting
several monocarboxylic acid metabolites, such as pyruvate, L-lactate,
and ketone bodies, across the plasma membrane with protons.
MCT1/4 subtypes are dominant in tumor metabolism (101). MCTs
are promising anticancer targets. The use of MCT1 inhibitors
or gene knockout can interfere with lactate-fueled respiration in
mitochondria (102). MCTs play an important role in the metabolic
homeostasis of the tumor microenvironment. MCT1 and MCT4 are
the most widely expressed MCT isoforms in cancer cells. MCT1
has a high affinity for lactate and is preferentially expressed in
respiratory cancer cells that take up lactate (101). In contrast,
MCT4 has a low affinity for lactate and is suitable for promoting
lactate export from glycolytic cancer cells, and its expression is
upregulated by hypoxia (103). The use of MCT1 inhibitors to
disrupt the communication between oxidative and glycolytic cancer
cells can inhibit the growth of breast cancer and promote the
death of myeloma cell lines (104106). MCTs inhibitors have been
shown to reduce the invasive and migratory capacity of glioma
cells (107), cervical squamous carcinoma cells (108), melanoma cells
(109), triple-negative breast cancer cells (110) and pancreatic ductal
adenocarcinoma cells (109). MCTs inhibitors could reduce glycolysis,
lactic acid and tumor growth and allow the immune response to
remain strong with increased tumor infiltration with CD8+T and
NK cells (53,111).
As the research on TME has intensified, many kinds of synergistic
therapies based on TME modulation have proliferated and achieved
better therapeutic effects in mice. Recently, Zhou et al. simultaneously
utilized lactate oxidase (LOD) and Fe3O4nanoparticles (NPs) to
treat tumors (86). the synergism between LOD and Fe3O4can
increase the consumption of lactate and produce more H2O2, and
concurrently hydrogen peroxide (H2O2) is subsequently converted
to highly toxic hydroxyl radicals (OH) catalyzed by Fe3O4NPs via
Fenton-like reactions to kill tumor cells. This ingenious strategy
showed an obvious inhibitory effect on tumor growth and resistance
to metastasis. Chen et al. effectively inhibited tumor growth and
resisted tumor metastasis by using metformin (Me) and fluvastatin
sodium (Flu) to interfere with lactate metabolism in tumor cells (112).
On the one hand, Me alters the gluconeogenesis pathway and inhibits
the tricarboxylic acid (TCA) cycle by inhibiting mitochondrial
respiration (113), leading to an increase in the conversion of
pyruvate to lactate. On the other hand, Flu inhibits lactate efflux,
leading to intracellular acidosis, which kills tumor cells (114).
Considering integrating cascaded enzymes and gene therapy, Tang
et al. creatively proposed a method that can effectively inhibit tumor
proliferation and angiogenesis even with the combined strategy
of lactate oxidase/catalase (LOD/CAT) and vascular endothelial
growth factor (VEGF) siRNA (SiVEGF) (115). The combination
of lactate depletion and VEGF silencing effectively inhibited the
migration of 4T1 cells in vitro and showed good anti-tumor and anti-
metastasis properties in vivo (116). A recent study has shown that
lactate degradation can be promoted with the assistance of lactate
oxidase (LOD) cationic polyethyleneimine (PEI) and nano-coated
with a certain amount of copper ion (PLNPCu). More importantly,
hydrogen peroxide (H2O2), a by-product of lactate degradation, can
be converted into anti-tumor ROS under the catalysis of copper
ions, which mediates immunogenic cell death (ICD) (117). With the
decrease of lactate in TME, the ICD process effectively promoted the
anti-tumor immune response of the 4T1 tumor model (the tumor
inhibition rate was 88%) (116). These strategies show a good anti-
tumor effect and verify the feasibility of endogenous lactate as one of
the key targets for tumor therapy. Although preclinical studies have
proved that glycolysis is effective in the targeted treatment of tumors,
their clinical transformation is still limited so far. The main reasons
that restrict the development of glycolysis targeting therapy include
metabolic heterogeneity and the damage of glycolysis targeting the
immune system.
4.2. Therapeutic strategies targeting immune
cells
Treatment of immune cells in the tumor microenvironment is
also a strategy. Although there are few reports of specific treatment
of immune cells, these reports have shown encouraging results in
different studies. Several methods have provided promising prospects
focused on targeting immune cells. INKT cells play an important
role in the clearance of tumor cells (2628). However, the tumor
microenvironment affects the metabolism of INKT cells and hinders
their anti-tumor functions. Fu et al. demonstrate that restoring lipid
synthesis via activating PPARγby using pioglitazone recovers αGC-
induced IFN-γproduction and significantly improves the efficacy
of iNKT cell-based immunotherapy against tumors. Importantly,
Frontiers in Nutrition 06 frontiersin.org
Li et al. 10.3389/fnut.2023.1113739
Pioglitazone has been already used in the treatment of type 2 diabetes,
which proves its potential application in clinical antineoplastic
therapy (29). This strategy of enhancing the antitumor efficacy of
iNKT cell-based immunotherapy by promoting lipid biosynthesis is
highly promising for clinical translation.
HIF1αcan directly regulate many M2-related genes, including
CD163 and ARG1 (118). Knockdown of HIF1αcan greatly impair
lactate-induced M2 polarization. By inhibiting the CCL17 released
by TAM, they can reduce the volume of prostate cancer, reduce the
invasiveness of the tumor, and the susceptibility to postoperative
recurrence (37). Tumor-derived lactate induces PD-L1+expression
on neutrophils via MCT1/NF-κB/COX-2 pathway, resulting in
inhibiting the efficacy of Lenvatinib. Thereby, it was believed that
the COX-2 inhibitor could reduce PD- L1+neutrophil and restore
T cell cytotoxicity (49). Lactate can impair T cell proliferation
by reducing stress independent of microenvironment acidification,
which depletes the GAPDH and PGDH responses of NAD+and
deprives proliferative T cells of glucose-derived serine. Manipulating
NAD redox metabolism may promote the differentiation and
activation of T cells and open a new way to selectively promote
immune regulation to enhance anti-tumor immunity (58).
GPR81 is expressed not only in cancer cells, but also in immune
cells such as dendritic cells and macrophages (119). Lactate can not
only affect cell proliferation, invasion, angiogenesis, and immune
tolerance by activating GPR81 on tumor cells but may also promote
tumor growth by activating GPR81 on non-tumor cells in the TME,
as there is a functional crosstalk between tumor cells and other
cells in the TME, making GPR81 a potential therapeutic target.
Targeting GPR81 could inhibit the growth of cancer cells and activate
the “enemy killing function” of the patient’s immune system to
fight the tumor. GPR81 was found closely related to tumorigenesis,
development, treatment, and prognosis in clinical trials, but still,
certain problems remain to be solved, such as the specific mechanism
of action is not fully understood, the safety and feasibility of
anti-GPR81 targeting therapy, and how to use it in clinical
trials. In vitro, silencing GPR81 can effectively eliminate GPR81-
induced angiogenesis (78). The discovery of small synthetic non-
metabolite ligands for structure-based metabolite receptors is rapidly
developing as many new GPCR structures are solved (120). With
the maturation of experimental techniques and further research,
it is believed that GPR81 will have good application prospects in
the future.
5. Conclusions and perspectives
Lactate was neglected for a long time in the field of oncological
research, but studies conducted over the past decades reveal that
it plays a significant role in the development and progression
of cancer by increasing angiogenesis, cancer cell migration, and
metastasis. By modulating lactate in the tumor microenvironment,
it was found that the GPR81 receptor inhibits host immune cells,
resulting in an advanced immunological evasion by tumor cells.
According to a number of research, the suppression of the anti-
cancer effect of several immune cells raised by lactate deposited
in the TME is transient. If extracellular lactate is depleted and
the microenvironmental pH is restored, it is possible to boost the
therapeutic efficacy of PD-1/PD-L1 therapeutics. Given the crucial
role that lactate plays, anti-glycolytic agents such as LDHA inhibitors
are capable of greatly limiting tumorigenesis. In fact, numerous
other anti-tumor immune cells also rely on glycolysis to exert
their functionality. Therefore, accordingly, it is assumed that anti-
glycolytic inhibitors that inhibit the proliferation of tumor cells would
reduce the anti-tumor immunological action of the host, rendering
the treatment far less effective. Most current glycolysis inhibitors
are inefficient, require high doses, and have varying degrees of
damage to the immune system. Therefore, there is an urgent need
to find highly effective and specific glycolytic inhibitors. Further
understanding of the metabolic differences between tumor cells and
immune cells is needed to identify new targeted agents that are more
effective and can more appropriately modulate the immune response
in vivo. Therapeutic regimens that target metabolism can potentially
be viable and revolutionary in the fight against cancer by combining
them with other therapeutic approaches such as immunotherapy
and chemotherapy.
Author contributions
ZZL, QW, XFH, and MTY designed this manuscript. ZZL,
QW, XFH, MTY, ZRL, and SJZ prepared and drafted the
manuscript. QW, XFH, ZRL, and SJZ prepared the figure.
ZZF, QC, HJH, LL, FF, QWW, YQW, and YDT enhanced the
language and analyzed the literature. AHG edited and revised
manuscript. All authors contributed to the article and approved the
submitted version.
Funding
This study was supported by Jiangsu Provincial Science and
Technology Department (Social Development)—Clinical Forward
Technology Project, BE2022778.
Conflict of interest
The authors declare that the research was conducted in the
absence of any commercial or financial relationships that could be
construed as a potential conflict of interest.
Publisher’s note
All claims expressed in this article are solely those of the
authors and do not necessarily represent those of their affiliated
organizations, or those of the publisher, the editors and the reviewers.
Any product that may be evaluated in this article, or claim that may
be made by its manufacturer, is not guaranteed or endorsed by the
publisher.
Frontiers in Nutrition 07 frontiersin.org
Li et al. 10.3389/fnut.2023.1113739
References
1. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. (2011)
144:646–74. doi: 10.1016/j.cell.2011.02.013
2. Hanahan D. Hallmarks of cancer: new dimensions. Cancer Discov. (2022) 12:31–
46. doi: 10.1158/2159-8290.CD-21-1059
3. Liu P, Wang Z, Ou X, Wu P, Zhang Y, Wu S, et al. The FUS/circEZH2/KLF5/
feedback loop contributes to CXCR4-induced liver metastasis of breast
cancer by enhancing epithelial-mesenchymal transition. Mol Cancer. (2022)
21:198. doi: 10.1186/s12943-022-01653-2
4. Warburg O, Wind F, Negelein E. The metabolism of tumors in the body. J Gen
Physiol. (1927) 8:519–30. doi: 10.1085/jgp.8.6.519
5. Huang J, Deng X, Chen X, Chang Z, Lu Q, Tang A, et al. Circular RNA KIF4A
promotes liver metastasis of breast cancer by reprogramming glucose metabolism. J
Oncol. (2022) 2022:8035083. doi: 10.1155/2022/8035083
6. Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg
effect: the metabolic requirements of cell proliferation. Science. (2009) 324:1029–
33. doi: 10.1126/science.1160809
7. Lyssiotis CA, Kimmelman AC. Metabolic interactions in the tumor
microenvironment. Trends Cell Biol. (2017) 27:863–75. doi: 10.1016/j.tcb.2017.06.003
8. Certo M, Tsai CH, Pucino V, Ho PC, Mauro C. Lactate modulation of immune
responses in inflammatory vs. tumour microenvironments. Nat Rev Immunol. (2021)
21:151–61. doi: 10.1038/s41577-020-0406-2
9. Faubert B, Li KY, Cai L, Hensley CT, Kim J, Zacharias LG, et al. Lactate metabolism
in human lung tumors. Cell. (2017) 171:358–71. doi: 10.1016/j.cell.2017.09.019
10. Dhup S, Dadhich RK, Porporato PE, Sonveaux P. Multiple biological activities of
lactic acid in cancer: influences on tumor growth, angiogenesis, and metastasis. Curr
Pharm Des. (2012) 18:1319–30. doi: 10.2174/138161212799504902
11. San-Millán I, Brooks GA. Reexamining cancer metabolism: lactate production
for carcinogenesis could be the purpose and explanation of the Warburg Effect.
Carcinogenesis. (2017) 38:119–33. doi: 10.1093/carcin/bgw127
12. Elia I, Rowe JH, Johnson S, Joshi S, Notarangelo G, Kurmi K, et al.
Tumor cells dictate anti-tumor immune responses by altering pyruvate
utilization and succinate signaling in CD8(+) T cells. Cell Metab. (2022)
34:1137-50.e6. doi: 10.1016/j.cmet.2022.06.008
13. Zou Y, Ye F, Kong Y, Hu X, Deng X, Xie J, et al. The single-
cell landscape of intratumoral heterogeneity and the immunosuppressive
microenvironment in liver and brain metastases of breast cancer. Adv Sci. (2022)
5:e2203699. doi: 10.1002/advs.202203699
14. Pérez-Escuredo J, Dadhich RK, Dhup S, Cacace A, Van Hée VF, De Saedeleer CJ,
et al. Lactate promotes glutamine uptake and metabolism in oxidative cancer cells. Cell
Cycle. (2016) 15:72–83. doi: 10.1080/15384101.2015.1120930
15. Damiani C, Colombo R, Gaglio D, Mastroianni F, Pescini D, Westerhoff
HV, et al. A metabolic core model elucidates how enhanced utilization of
glucose and glutamine, with enhanced glutamine-dependent lactate production,
promotes cancer cell growth: the WarburQ effect. PLoS Comput Biol. (2017)
13:e1005758. doi: 10.1371/journal.pcbi.1005758
16. Burke L, Guterman I, Palacios Gallego R, Britton RG, Burschowsky D, Tufarelli
C, et al. The Janus-like role of proline metabolism in cancer. Cell Death Discov. (2020)
6:104. doi: 10.1038/s41420-020-00341-8
17. Luengo A, Li Z, Gui DY, Sullivan LB, Zagorulya M, Do BT, et al. Increased
demand for NAD(+) relative to ATP drives aerobic glycolysis. Mol Cell. (2021) 81:691–
707. doi: 10.1016/j.molcel.2020.12.012
18. Kowaloff EM, Phang JM, Granger AS, Downing SJ. Regulation of
proline oxidase activity by lactate. Proc Natl Acad Sci U S A. (1977)
74:5368–71. doi: 10.1073/pnas.74.12.5368
19. Oizel K, Yang C, Renoult O, Gautier F, Do QN, Joalland N, et al. Glutamine uptake
and utilization of human mesenchymal glioblastoma in orthotopic mouse model. Cancer
Metab. (2020) 8:9. doi: 10.1186/s40170-020-00215-8
20. Liu W, Le A, Hancock C, Lane AN, Dang CV, Fan TW, et al. Reprogramming
of proline and glutamine metabolism contributes to the proliferative and metabolic
responses regulated by oncogenic transcription factor c-MYC. Proc Natl Acad Sci U S
A. (2012) 109:8983–8. doi: 10.1073/pnas.1203244109
21. Ippolito L, Morandi A, Giannoni E, Chiarugi P. Lactate: a metabolic driver in the
tumour landscape. Trends Biochem Sci. (2019) 44:153–66. doi: 10.1016/j.tibs.2018.10.011
22. Liu C, Yang M, Zhang D, Chen M, Zhu D. Clinical cancer
immunotherapy: current progress and prospects. Front Immunol. (2022)
13:961805. doi: 10.3389/fimmu.2022.961805
23. Zou Y, Zheng S, Deng X, Yang A, Xie X, Tang H, et al. The role of circular
RNA CDR1as/ciRS-7 in regulating tumor microenvironment: a pan-cancer analysis.
Biomolecules. (2019) 9:429. doi: 10.3390/biom9090429
24. Russo V, Protti MP. Tumor-derived factors affecting immune cells.
Cytokine Growth Factor Rev. (2017) 36:79–87. doi: 10.1016/j.cytogfr.2017.
06.005
25. De Santo C, Arscott R, Booth S, Karydis I, Jones M, Asher R, et al. Invariant NKT
cells modulate the suppressive activity of IL-10-secreting neutrophils differentiated with
serum amyloid A. Nat Immunol. (2010) 11:1039–46. doi: 10.1038/ni.1942
26. Fujii S, Shimizu K, Smith C, Bonifaz L, Steinman RM. Activation of natural killer
T cells by alpha-galactosylceramide rapidly induces the full maturation of dendritic cells
in vivo and thereby acts as an adjuvant for combined CD4 and CD8 T cell immunity to a
coadministered protein. J Exp Med. (2003) 198:267–79. doi: 10.1084/jem.20030324
27. Moreno M, Molling JW, von Mensdorff-Pouilly S, Verheijen RH, Hooijberg E,
Kramer D, et al. IFN-gamma-producing human invariant NKT cells promote tumor-
associated antigen-specific cytotoxic T cell responses. J Immunol. (2008) 181:2446–
54. doi: 10.4049/jimmunol.181.4.2446
28. Silk JD, Hermans IF, Gileadi U, Chong TW, Shepherd D, Salio M, et al. Utilizing the
adjuvant properties of CD1d-dependent NK T cells in T cell-mediated immunotherapy. J
Clin Invest. (2004) 114:1800–11. doi: 10.1172/JCI200422046
29. Fu S, He K, Tian C, Sun H, Zhu C, Bai S, et al. Impaired lipid biosynthesis
hinders anti-tumor efficacy of intratumoral iNKT cells. Nat Commun. (2020)
11:438. doi: 10.1038/s41467-020-14332-x
30. Najafi M, Hashemi Goradel N, Farhood B, Salehi E, Nashtaei MS, Khanlarkhani
N, et al. Macrophage polarity in cancer: a review. J Cell Biochem. (2019) 120:2756–
65. doi: 10.1002/jcb.27646
31. Pascual-García M, Bonfill-Teixidor E, Planas-Rigol E, Rubio-Perez C, Iurlaro R,
Arias A, et al. LIF regulates CXCL9 in tumor-associated macrophages and prevents
CD8(+) T cell tumor-infiltration impairing anti-PD1 therapy. Nat Commun. (2019)
10:2416. doi: 10.1038/s41467-019-10369-9
32. Zhang L, Li S. Lactic acid promotes macrophage polarization
through MCT-HIF1αsignaling in gastric cancer. Exp Cell Res. (2020)
388:111846. doi: 10.1016/j.yexcr.2020.111846
33. Xu JY, Wang WS, Zhou J, Liu CY, Shi JL, Lu PH, et al. The importance of a conjoint
analysis of tumor-associated macrophages and immune checkpoints in pancreatic cancer.
Pancreas. (2019) 48:904–12. doi: 10.1097/MPA.0000000000001364
34. Liu N, Luo J, Kuang D, Xu S, Duan Y, Xia Y, et al. Lactate inhibits ATP6V0d2
expression in tumor-associated macrophages to promote HIF-2α-mediated tumor
progression. J Clin Invest. (2019) 129:631–46. doi: 10.1172/JCI123027
35. Yang K, Xu J, Fan M, Tu F, Wang X, Ha T, et al. Lactate suppresses
macrophage pro-inflammatory response to LPS stimulation by inhibition of YAP
and NF-κB activation via GPR81-mediated signaling. Front Immunol. (2020)
11:587913. doi: 10.3389/fimmu.2020.587913
36. Shan T, Chen S, Chen X, Wu T, Yang Y, Li S, et al. M2-TAM subsets altered by
lactic acid promote T-cell apoptosis through the PD-L1/PD-1 pathway. Oncol Rep. (2020)
44:1885–94. doi: 10.3892/or.2020.7767
37. Zhang A, Xu Y, Xu H, Ren J, Meng T, Ni Y, et al. Lactate-induced M2 polarization of
tumor-associated macrophages promotes the invasion of pituitary adenoma by secreting
CCL17. Theranostics. (2021) 11:3839–52. doi: 10.7150/thno.53749
38. Bronte V, Brandau S, Chen SH, Colombo MP, Frey AB, Greten TF,
et al. Recommendations for myeloid-derived suppressor cell nomenclature and
characterization standards. Nat Commun. (2016) 7:12150. doi: 10.1038/ncomms12150
39. Biswas SK. Metabolic reprogramming of immune cells in cancer progression.
Immunity. (2015) 43:435–49. doi: 10.1016/j.immuni.2015.09.001
40. Condamine T, Ramachandran I, Youn JI, Gabrilovich DI. Regulation of tumor
metastasis by myeloid-derived suppressor cells. Annu Rev Med. (2015) 66:97–
110. doi: 10.1146/annurev-med-051013-052304
41. Kopan R, Ilagan MX. The canonical Notch signaling pathway: unfolding the
activation mechanism. Cell. (2009) 137:216–33. doi: 10.1016/j.cell.2009.03.045
42. Gupta S. Role of dendritic cells in innate and adaptive immune response in human
aging. Exp Gerontol. (2014) 54:47–52. doi: 10.1016/j.exger.2013.12.009
43. Steinman RM. The dendritic cell system and its role in immunogenicity. Annu Rev
Immunol. (1991) 9:271–96. doi: 10.1146/annurev.iy.09.040191.001415
44. Burgdorf S, Porubsky S, Marx A, Popovic ZV. Cancer acidity and hypertonicity
contribute to dysfunction of tumor-associated dendritic cells: potential impact on antigen
cross-presentation machinery. Cancers. (2020) 12:2403. doi: 10.3390/cancers12092403
45. Wang Y, Wang Y, Ren Y, Zhang Q, Yi P,Cheng C. Metabolic modulation of immune
checkpoints and novel therapeutic strategies in cancer. Semin Cancer Biol. (2022) 86(Pt
3):542–65. doi: 10.1016/j.semcancer.2022.02.010
46. Nasi A, Fekete T, Krishnamurthy A, Snowden S, Rajnavölgyi E, Catrina AI, et al.
Dendritic cell reprogramming by endogenously produced lactic acid. J Immunol. (2013)
191:3090–9. doi: 10.4049/jimmunol.1300772
47. Kelly B, O’Neill LA. Metabolic reprogramming in macrophages and dendritic cells
in innate immunity. Cell Res. (2015) 25:771–84. doi: 10.1038/cr.2015.68
48. Brandau S, Trellakis S, Bruderek K, Schmaltz D, Steller G, Elian M, et al. Myeloid-
derived suppressor cells in the peripheral blood of cancer patients contain a subset
of immature neutrophils with impaired migratory properties. J Leukoc Biol. (2011)
89:311–7. doi: 10.1189/jlb.0310162
Frontiers in Nutrition 08 frontiersin.org
Li et al. 10.3389/fnut.2023.1113739
49. Deng H, Kan A, Lyu N, He M, Huang X, Qiao S, et al. Tumor-derived
lactate inhibit the efficacy of lenvatinib through regulating PD-L1 expression
on neutrophil in hepatocellular carcinoma. J Immunother Cancer. (2021)
9:2305. doi: 10.1136/jitc-2020-002305
50. Zhang X, Zhang W, Yuan X, Fu M, Qian H, Xu W. Neutrophils in cancer
development and progression: roles, mechanisms, and implications (Review). Int J Oncol.
(2016) 49:857–67. doi: 10.3892/ijo.2016.3616
51. Shaul ME, Fridlender ZG. Tumour-associated neutrophils in patients with cancer.
Nat Rev Clin Oncol. (2019) 16:601–20. doi: 10.1038/s41571-019-0222-4
52. McNamara MJ, Hilgart-Martiszus I, Barragan Echenique DM, Linch SN, Kasiewicz
MJ, Redmond WL. Interferon-γproduction by peripheral lymphocytes predicts survival
of tumor-bearing mice receiving dual PD-1/CTLA-4 blockade. Cancer Immunol Res.
(2016) 4:650–7. doi: 10.1158/2326-6066.CIR-16-0022
53. Kornberg MD. The immunologic Warburg effect: evidence and therapeutic
opportunities in autoimmunity. Wiley Interdiscip Rev Syst Biol Med. (2020)
12:e1486. doi: 10.1002/wsbm.1486
54. Ardi VC, Kupriyanova TA,Deryugina EI, Quigley JP. Human neutrophils uniquely
release TIMP-free MMP-9 to provide a potent catalytic stimulator of angiogenesis. Proc
Natl Acad Sci U S A. (2007) 104:20262–7. doi: 10.1073/pnas.0706438104
55. Oh DY, Fong L, Newell EW, Turk MJ, Chi H, Chang HY, et al.
Toward a better understanding of T cells in cancer. Cancer Cell. (2021)
39:1549–52. doi: 10.1016/j.ccell.2021.11.010
56. Kishton RJ, Sukumar M, Restifo NP. Metabolic regulation of T
cell longevity and function in tumor immunotherapy. Cell Metab. (2017)
26:94–109. doi: 10.1016/j.cmet.2017.06.016
57. Dietl K, Renner K, Dettmer K, Timischl B, Eberhart K, Dorn C, et al. Lactic acid
and acidification inhibit TNF secretion and glycolysis of human monocytes. J Immunol.
(2010) 184:1200–9. doi: 10.4049/jimmunol.0902584
58. Quinn WJ. 3rd, Jiao J, TeSlaa T, Stadanlick J, Wang Z, Wang L, et al.
Lactate limits T cell proliferation via the NAD(H). Redox State Cell Rep. (2020)
33:108500. doi: 10.1016/j.celrep.2020.108500
59. Feng Q, Liu Z, Yu X, Huang T, Chen J, Wang J, et al. Lactate increases
stemness of CD8 +T cells to augment anti-tumor immunity. Nat Commun. (2022)
13:4981. doi: 10.1038/s41467-022-32521-8
60. Ohue Y, Nishikawa H. Regulatory T (Treg) cells in cancer: can Treg cells be a new
therapeutic target? Cancer Sci. (2019) 110:2080–9. doi: 10.1111/cas.14069
61. Watson MJ, Vignali PDA, Mullett SJ, Overacre-Delgoffe AE, Peralta RM,
Grebinoski S, et al. Metabolic support of tumour-infiltrating regulatory T cells by lactic
acid. Nature. (2021) 591:645–51. doi: 10.1038/s41586-020-03045-2
62. Kalluri R. The biology and function of fibroblasts in cancer. Nat Rev Cancer. (2016)
16:582–98. doi: 10.1038/nrc.2016.73
63. Xie J, Zheng S, Zou Y, Tang Y, Tian W, Wong CW, et al. Turning up a new pattern:
Identification of cancer-associated fibroblast-related clusters in TNBC. Front Immunol.
(2022) 13:1022147. doi: 10.3389/fimmu.2022.1022147
64. Fiori ME, Di Franco S, Villanova L, Bianca P, Stassi G, De Maria R. Cancer-
associated fibroblasts as abettors of tumor progression at the crossroads of EMT and
therapy resistance. Mol Cancer. (2019) 18:70. doi: 10.1186/s12943-019-0994-2
65. Hinshaw DC, Shevde LA. The tumor microenvironment innately modulates cancer
progression. Cancer Res. (2019) 79:4557–66. doi: 10.1158/0008-5472.CAN-18-3962
66. Joshi RS, Kanugula SS, Sudhir S, Pereira MP, Jain S, Aghi MK. The
role of cancer-associated fibroblasts in tumor progression. Cancers. (2021)
13:1399. doi: 10.3390/cancers13061399
67. Su S, Chen J, Yao H, Liu J, Yu S, Lao L, et al. CD10(+)GPR77(+) Cancer-
associated fibroblasts promote cancer formation and chemoresistance by sustaining
cancer stemness. Cell. (2018) 172:841–56. doi: 10.1016/j.cell.2018.01.009
68. Desbois M, Wang Y. Cancer-associated fibroblasts: key players in
shaping the tumor immune microenvironment. Immunol Rev. (2021)
302:241–58. doi: 10.1111/imr.12982
69. Bonuccelli G, Tsirigos A, Whitaker-Menezes D, Pavlides S, Pestell RG, Chiavarina
B, et al. Ketones and lactate “fuel” tumor growth and metastasis: evidence that
epithelial cancer cells use oxidative mitochondrial metabolism. Cell Cycle. (2010) 9:3506–
14. doi: 10.4161/cc.9.17.12731
70. Linares JF, Cid-Diaz T, Duran A, Osrodek M, Martinez-Ordoñez A, Reina-
Campos M, et al. The lactate-NAD(+) axis activates cancer-associated fibroblasts by
downregulating p62. Cell Rep. (2022) 39:110792. doi: 10.1016/j.celrep.2022.110792
71. Huang J, Duran A, Reina-Campos M, Valencia T, Castilla EA, Müller
TD, et al. Adipocyte p62/SQSTM1 suppresses tumorigenesis through opposite
regulations of metabolism in adipose tissue and tumor. Cancer Cell. (2018) 33:770–
84. doi: 10.1016/j.ccell.2018.03.001
72. Zhang D, Tang Z, Huang H, Zhou G, Cui C, Weng Y, et al. Metabolic
regulation of gene expression by histone lactylation. Nature. (2019) 574:575–
80. doi: 10.1038/s41586-019-1678-1
73. Yu J, Chai P, Xie M, Ge S, Ruan J, Fan X, et al. Histone lactylation drives oncogenesis
by facilitating m(6)A reader protein YTHDF2 expression in ocular melanoma. Genome
Biol. (2021) 22:85. doi: 10.1186/s13059-021-02308-z
74. Hagihara H, Shoji H, Otabi H, Toyoda A, Katoh K, Namihira M,
et al. Protein lactylation induced by neural excitation. Cell Rep. (2021)
37:109820. doi: 10.1016/j.celrep.2021.109820
75. Kuei C, Yu J, Zhu J, Wu J, Zhang L, Shih A, et al. Study of GPR81, the lactate
receptor, from distant species identifies residues and motifs critical for GPR81 functions.
Mol Pharmacol. (2011) 80:848–58. doi: 10.1124/mol.111.074500
76. Brown TP, Ganapathy V. Lactate/GPR81 signaling and proton motive force in
cancer: role in angiogenesis, immune escape, nutrition, and Warburg phenomenon.
Pharmacol Ther. (2020) 206:107451. doi: 10.1016/j.pharmthera.2019.107451
77. Xie Q, Zhu Z, He Y, Zhang Z, Zhang Y, Wang Y, et al. A lactate-induced Snail/STAT3
pathway drives GPR81 expression in lung cancer cells. Biochim Biophys Acta Mol Basis
Dis. (2020) 1866:165576. doi: 10.1016/j.bbadis.2019.165576
78. Lee YJ, Shin KJ, Park SA, Park KS, Park S, Heo K, et al. G-protein-coupled receptor
81 promotes a malignant phenotype in breast cancer through angiogenic factor secretion.
Oncotarget. (2016) 7:70898–911. doi: 10.18632/oncotarget.12286
79. Feng J, Yang H, Zhang Y, Wei H, Zhu Z, Zhu B, et al. Tumor cell-derived lactate
induces TAZ-dependent upregulation of PD-L1 through GPR81 in human lung cancer
cells. Oncogene. (2017) 36:5829–39. doi: 10.1038/onc.2017.188
80. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J Clin.
(2021) 71:7–33. doi: 10.3322/caac.21654
81. Li Y, Duy Le TM, Nam Bui Q, Yang HY, Lee DS. Tumor acidity and CD44 dual
targeting hyaluronic acid-coated gold nanorods for combined chemo- and photothermal
cancer therapy. Carbohydr Polym. (2019) 226:115281. doi: 10.1016/j.carbpol.2019.115281
82. Wen M, Ouyang J, Wei C, Li H, Chen W, Liu YN. Artificial enzyme catalyzed
cascade reactions: antitumor immunotherapy reinforced by NIR-II light. Angew Chem
Int Ed Engl. (2019) 58:17425–32. doi: 10.1002/anie.201909729
83. Gao Z, Li Y, Zhang Y, Cheng K, An P, Chen F, et al. Biomimetic platinum
nanozyme immobilized on 2D metal-organic frameworks for mitochondrion-targeting
and oxygen self-supply photodynamic therapy. ACS Appl Mater Interfaces. (2020)
12:1963–72. doi: 10.1021/acsami.9b14958
84. Song G, Chen Y, Liang C, Yi X, Liu J, Sun X, et al. Catalase-loaded TaOx
nanoshells as bio-nanoreactors combining high-Z element and enzyme delivery
for enhancing radiotherapy. Adv Mater. (2016) 28:7143–8. doi: 10.1002/adma.2016
02111
85. Tian Z, Yang K, Yao T, Li X, Ma Y, Qu C, et al. Catalytically selective
chemotherapy from tumor-metabolic generated lactic acid. Small. (2019)
15:e1903746. doi: 10.1002/smll.201903746
86. Zhou X, Zhao W, Wang M, Zhang S, Li Y, Hu W, et al. Dual-modal therapeutic
role of the lactate oxidase-embedded hierarchical porous zeolitic imidazolate framework
as a nanocatalyst for effective tumor suppression. ACS Appl Mater Interfaces. (2020)
12:32278–88. doi: 10.1021/acsami.0c05783
87. Tang J, Meka AK, Theivendran S, Wang Y, Yang Y, Song H, et al.
Openwork@dendritic mesoporous silica nanoparticles for lactate depletion and
tumor microenvironment regulation. Angew Chem Int Ed Engl. (2020) 59:22054–
62. doi: 10.1002/anie.202001469
88. Quon E, Hart ML, Sullivan LB. Redox debt leads to metabolic bankruptcy in
tumors. Trends Cancer. (2020) 6:359–61. doi: 10.1016/j.trecan.2020.02.012
89. Shi M, Cui J, Du J, Wei D, Jia Z, Zhang J, et al. A novel KLF4/LDHA signaling
pathway regulates aerobic glycolysis in and progression of pancreatic cancer. Clin Cancer
Res. (2014) 20:4370–80. doi: 10.1158/1078-0432.CCR-14-0186
90. Cai H, Li J, Zhang Y, Liao Y, Zhu Y, Wang C, et al. LDHA promotes oral squamous
cell carcinoma progression through facilitating glycolysis and epithelial-mesenchymal
transition. Front Oncol. (2019) 9:1446. doi: 10.3389/fonc.2019.01446
91. Huang X, Li X, Xie X, Ye F, Chen B, Song C, et al. High expressions of
LDHA and AMPK as prognostic biomarkers for breast cancer. Breast. (2016) 30:39–
46. doi: 10.1016/j.breast.2016.08.014
92. Oshima N, Ishida R, Kishimoto S, Beebe K, Brender JR, Yamamoto
K, et al. Dynamic imaging of LDH inhibition in tumors reveals
rapid in vivo metabolic rewiring and vulnerability to combination
therapy. Cell Rep. (2020) 30:1798–810. doi: 10.1016/j.celrep.2020.
01.039
93. Wang S, Ma L, Wang Z, He H, Chen H, Duan Z, et al. Lactate dehydrogenase-
A (LDH-A) preserves cancer stemness and recruitment of tumor-associated
macrophages to promote breast cancer progression. Front Oncol. (2021)
11:654452. doi: 10.3389/fonc.2021.654452
94. Xie H, Hanai J, Ren JG, Kats L, Burgess K, Bhargava P, et al. Targeting
lactate dehydrogenase—A inhibits tumorigenesis and tumor progression in mouse
models of lung cancer and impacts tumor-initiating cells. Cell Metab. (2014) 19:795–
809. doi: 10.1016/j.cmet.2014.03.003
95. Brisson L, Ba´
nski P, Sboarina M, Dethier C, Danhier P, Fontenille MJ, et al. Lactate
dehydrogenase B controls lysosome activity and autophagy in cancer. Cancer Cell. (2016)
30:418–31. doi: 10.1016/j.ccell.2016.08.005
96. Ždralevi´
c M, Brand A, Di Ianni L, Dettmer K, Reinders J, Singer K, et al. Double
genetic disruption of lactate dehydrogenases A and B is required to ablate the “Warburg
effect” restricting tumor growth to oxidative metabolism. J Biol Chem. (2018) 293:15947–
61. doi: 10.1074/jbc.RA118.004180
Frontiers in Nutrition 09 frontiersin.org
Li et al. 10.3389/fnut.2023.1113739
97. Le A, Cooper CR, Gouw AM, Dinavahi R, Maitra A, Deck LM, et al. Inhibition
of lactate dehydrogenase A induces oxidative stress and inhibits tumor progression. Proc
Natl Acad Sci U S A. (2010) 107:2037–42. doi: 10.1073/pnas.0914433107
98. Farabegoli F, Vettraino M, Manerba M, Fiume L, Roberti M, Di Stefano G.
Galloflavin, a new lactate dehydrogenase inhibitor, induces the death of human breast
cancer cells with different glycolytic attitude by affecting distinct signaling pathways. Eur
J Pharm Sci. (2012) 47:729–38. doi: 10.1016/j.ejps.2012.08.012
99. Granchi C, Roy S, Giacomelli C, Macchia M, Tuccinardi T, Martinelli A, et al.
Discovery of N-hydroxyindole-based inhibitors of human lactate dehydrogenase isoform
A (LDH-A) as starvation agents against cancer cells. J Med Chem. (2011) 54:1599–
612. doi: 10.1021/jm101007q
100. Maftouh M, Avan A, Sciarrillo R, Granchi C, Leon LG, Rani R, et al. Synergistic
interaction of novel lactate dehydrogenase inhibitors with gemcitabine against pancreatic
cancer cells in hypoxia. Br J Cancer. (2014) 110:172–82. doi: 10.1038/bjc.2013.681
101. Payen VL, Mina E, Van Hée VF, Porporato PE, Sonveaux P. Monocarboxylate
transporters in cancer. Mol Metab. (2020) 33:48–66. doi: 10.1016/j.molmet.2019.07.006
102. Ma Y, Kepp O, Ghiringhelli F, Apetoh L, Aymeric L, Locher C, et al.
Chemotherapy and radiotherapy: cryptic anticancer vaccines. Semin Immunol. (2010)
22:113–24. doi: 10.1016/j.smim.2010.03.001
103. Pérez de Heredia F, Wood IS, Trayhurn P. Hypoxia stimulates lactate release
and modulates monocarboxylate transporter (MCT1, MCT2, and MCT4) expression in
human adipocytes. Pflugers Arch. (2010) 459:509–18. doi: 10.1007/s00424-009-0750-3
104. Morais-Santos F, Miranda-Gonçalves V, Pinheiro S, Vieira AF, Paredes J, Schmitt
FC, et al. Differential sensitivities to lactate transport inhibitors of breast cancer cell lines.
Endocr Relat Cancer. (2014) 21:27–38. doi: 10.1530/ERC-13-0132
105. Hanson DJ, Nakamura S, Amachi R, Hiasa M, Oda A, Tsuji D, et al. Effective
impairment of myeloma cells and their progenitors by blockade of monocarboxylate
transportation. Oncotarget. (2015) 6:33568–86. doi: 10.18632/oncotarget.5598
106. Hong CS, Graham NA, Gu W, Espindola Camacho C, Mah V, Maresh EL, et al.
MCT1 modulates cancer cell pyruvate export and growth of tumors that co-express MCT1
and MCT4. Cell Rep. (2016) 14:1590–601. doi: 10.1016/j.celrep.2016.01.057
107. Miranda-Gonçalves V, Honavar M, Pinheiro C, Martinho O, Pires MM,
Pinheiro C, et al. Monocarboxylate transporters (MCTs) in gliomas: expression and
exploitation as therapeutic targets. Neuro Oncol. (2013) 15:172–88. doi: 10.1093/neuonc/n
os298
108. Payen VL, Hsu MY, Rädecke KS, Wyart E, Vazeille T,
Bouzin C, et al. Monocarboxylate transporter MCT1 promotes tumor
metastasis independently of its activity as a lactate transporter.
Cancer Res. (2017) 77:5591–601. doi: 10.1158/0008-5472.CAN-1
7-0764
109. Kong SC, Nøhr-Nielsen A, Zeeberg K, Reshkin SJ, Hoffmann EK, Novak
I, et al. Monocarboxylate transporters MCT1 and MCT4 regulate migration and
invasion of pancreatic ductal adenocarcinoma cells. Pancreas. (2016) 45:1036–
47. doi: 10.1097/MPA.0000000000000571
110. Zou Y, Xie J, Zheng S, Liu W, Tang Y, Tian W, et al. Leveraging diverse cell-death
patterns to predict the prognosis and drug sensitivity of triple-negative breast cancer
patients after surgery. Int J Surg. (2022) 107:106936. doi: 10.1016/j.ijsu.2022.106936
111. Cascone T, McKenzie JA, Mbofung RM, Punt S, Wang Z, Xu C, et al. Increased
tumor glycolysis characterizes immune resistance to adoptive T cell therapy. Cell Metab.
(2018) 27:977–87. doi: 10.1016/j.cmet.2018.02.024
112. Chen ZX, Liu MD, Guo DK, Zou MZ, Wang SB, Cheng H, et al. A MSN-
based tumor-targeted nanoplatform to interfere with lactate metabolism to induce tumor
cell acidosis for tumor suppression and anti-metastasis. Nanoscale. (2020) 12:2966–
72. doi: 10.1039/C9NR10344A
113. Madiraju AK, Erion DM, Rahimi Y, Zhang XM, Braddock DT, Albright RA, et al.
Metformin suppresses gluconeogenesis by inhibiting mitochondrial glycerophosphate
dehydrogenase. Nature. (2014) 510:542–6. doi: 10.1038/nature13270
114. Kobayashi M, Otsuka Y, Itagaki S, Hirano T, Iseki K. Inhibitory effects
of statins on human monocarboxylate transporter 4. Int J Pharm. (2006) 317:19–
25. doi: 10.1016/j.ijpharm.2006.02.043
115. Tang Y, Jia C, Wang Y, Wan W, Li H, Huang G, et al. Lactate
consumption via cascaded enzymes combined VEGF siRNA for synergistic anti-
proliferation and anti-angiogenesis therapy of tumors. Adv Healthc Mater. (2021)
10:e2100799. doi: 10.1002/adhm.202100799
116. He R, Zang J, Zhao Y, Liu Y, Ruan S, Zheng X, et al. Nanofactory for metabolic
and chemodynamic therapy: pro-tumor lactate trapping and anti-tumor ROS transition.
J Nanobiotechnol. (2021) 19:426. doi: 10.1186/s12951-021-01169-9
117. Xie J, Luo X, Deng X, Tang Y, Tian W, Cheng H, et al. Advances in
artificial intelligence to predict cancer immunotherapy efficacy. Front Immunol. (2022)
13:1076883. doi: 10.3389/fimmu.2022.1076883
118. Colegio OR, Chu NQ, Szabo AL, Chu T,R hebergenAM, Jairam V, et al. Functional
polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature.
(2014) 513:559–63. doi: 10.1038/nature13490
119. Ranganathan P, Shanmugam A, Swafford D, Suryawanshi A, Bhattacharjee P,
Hussein MS, et al. GPR81, a cell-surface receptor for lactate, regulates intestinal
homeostasis and protects mice from experimental colitis. J Immunol. (2018) 200:1781–
9. doi: 10.4049/jimmunol.1700604
120. Lückmann M, Trauelsen M, Frimurer TM, Schwartz TW. Structural basis for
GPCR signaling by small polar versus large lipid metabolites-discovery of non-metabolite
ligands. Curr Opin Cell Biol. (2020) 63:38–48. doi: 10.1016/j.ceb.2019.12.005
Frontiers in Nutrition 10 frontiersin.org
... In atherosclerosis, impaired efferocytosis exacerbates inflammation and increased plaque destabilization, whereas improved efferocytosis in atherosclerosis regression reversed these changes. Although the pro-tolerogenic function of myeloid cell efferocytosis and its associated downstream signals, e.g., secreted lactate or kynurenine, are beneficial in the context of chronic inflammation and atherosclerosis progression, this process can also contribute to immunosuppression in cancer, leading to cancer cell evasion of the immune system (142)(143)(144)(145). Thus, conceptualizing methods to target the crosstalk between efferocytosing myeloid cells and T-cells for the treatment of atherosclerosis must also consider that excessive immunosuppression may result in impaired anti-tumor immunity. ...
Article
Full-text available
The interplay between myeloid cells and T-lymphocytes is critical to the regulation of host defense and inflammation resolution. Dysregulation of this interaction can contribute to the development of chronic inflammatory diseases. Important among these diseases is atherosclerosis, which refers to focal lesions in the arterial intima driven by elevated apolipoprotein B-containing lipoproteins, notably low-density lipoprotein (LDL), and characterized by the formation of a plaque composed of inflammatory immune cells, a collection of dead cells and lipids called the necrotic core, and a fibrous cap. As the disease progresses, the necrotic core expands, and the fibrous cap becomes thin, which increases the risk of plaque rupture or erosion. Plaque rupture leads to a rapid thrombotic response that can give rise to heart attack, stroke, or sudden death. With marked lowering of circulating LDL, however, plaques become more stable and cardiac risk is lowered—a process known as atherosclerosis regression. A critical aspect of both atherosclerosis progression and regression is the crosstalk between innate (myeloid cells) and adaptive (T-lymphocytes) immune cells. Myeloid cells are specialized at clearing apoptotic cells by a process called efferocytosis, which is necessary for inflammation resolution. In advanced disease, efferocytosis is impaired, leading to secondary necrosis of apoptotic cells, inflammation, and, most importantly, defective tissue resolution. In regression, efferocytosis is reawakened aiding in inflammation resolution and plaque stabilization. Here, we will explore how efferocytosing myeloid cells could affect T-cell function and vice versa through antigen presentation, secreted factors, and cell-cell contacts and how this cellular crosstalk may contribute to the progression or regression of atherosclerosis.
... While the Warburg effect continues, the lactate produced is not a useless waste product. Lactate is not only important for tumor invasion, metastasis, and angiogenesis [24,25], but also has immunosuppressive functions, such as inducing and recruiting immunosuppressive cells and molecules, thereby promoting tumor development and escape [26,27]. Cancer cell-derived lactate can induce the expression of vascular endothelial growth factor (VEGF) and arginase 1 (Arg1) through HIF-1α signaling pathway, which promotes the polarization of tumor-associated macrophages (TAMs) toward the M2 phenotype, enabling TAMs to promote tumor growth [28]. ...
Article
Full-text available
Metabolic reprogramming is an emerging hallmark of cancer cells, enabling them to meet increased nutrient and energy demands while withstanding the challenging microenvironment. Cancer cells can switch their metabolic pathways, allowing them to adapt to different microenvironments and therapeutic interventions. This refers to metabolic heterogeneity, in which different cell populations use different metabolic pathways to sustain their survival and proliferation and impact their response to conventional cancer therapies. Thus, targeting cancer metabolic heterogeneity represents an innovative therapeutic avenue with the potential to overcome treatment resistance and improve therapeutic outcomes. This review discusses the metabolic patterns of different cancer cell populations and developmental stages, summarizes the molecular mechanisms involved in the intricate interactions within cancer metabolism, and highlights the clinical potential of targeting metabolic vulnerabilities as a promising therapeutic regimen. We aim to unravel the complex of metabolic characteristics and develop personalized treatment approaches to address distinct metabolic traits, ultimately enhancing patient outcomes.
Article
Full-text available
Protein therapeutics offer high therapeutic potency and specificity; the broader adoptions and development of protein therapeutics, however, have been constricted by their intrinsic limitations such as inadequate stability, immunogenicity, suboptimal pharmacokinetics and biodistribution, and off‐target effects. This review describes a platform technology that formulates individual protein molecules with a thin formulation layer of crosslinked polymers, which confers the protein therapeutics with high activity, enhanced stability, controlled release capability, reduced immunogenicity, improved pharmacokinetics and biodistribution, and ability to cross the blood brain barriers. Based on currently approved protein therapeutics, this formulating platform affords the development of a vast family of superior protein therapeutics with improved efficacy and broadened indications at significantly reduced cost.
Article
Full-text available
This review briefly introduces readers to an area where glycomics meets modern oncodiagnostics with a focus on the analysis of sialic acid (Neu5Ac)-terminated structures. We present the biochemical perspective of aberrant sialylation during tumourigenesis and its significance, as well as an analytical perspective on the detection of these structures using different approaches for diagnostic and therapeutic purposes. We also provide a comparison to other established liquid biopsy approaches, and we mathematically define an early-stage cancer based on the overall prognosis and effect of these approaches on the patient’s quality of life. Finally, some barriers including regulations and quality of clinical validations data are discussed, and a perspective and major challenges in this area are summarised.
Article
Lymphotoxin beta receptor (LTBR) is a positive T cell proliferation regulator gene. It is closely associated with the tumor immune microenvironment. However, its role in cancer and immunotherapy is unclear. Firstly, the expression level and prognostic value of LTBR were analyzed. Secondly, the expression of LTBR in clinical stages, immune subtypes, and molecular subtypes was analyzed. The correlation between LTBR and immune regulatory genes, immune checkpoint genes, and RNA modification genes was then analyzed. Correlations between LTBR and immune cells, scores, cancer-related functional status, tumor stemness index, mismatch repair (MMR) genes, and DNA methyltransferase were also analyzed. In addition, we analyzed the role of LTBR in DNA methylation, mutational status, tumor mutation burden (TMB), and microsatellite instability (MSI). Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA) were used to explore the role of LTBR in pan-cancer. Finally, the drugs associated with LTBR were analyzed. The expression of LTBR was confirmed using quantitative real-time PCR and Western blot. LTBR is significantly overexpressed in most cancers and is associated with low patient survival. In addition, LTBR expression was strongly correlated with immune cells, score, cancer-related functional status, tumor stemness index, MMR genes, DNA methyltransferase, DNA methylation, mutational status, TMB, and MSI. Enrichment analysis revealed that LTBR was associated with apoptosis, necroptosis, and immune-related pathways. Finally, multiple drugs targeting LTBR were identified. LTBR is overexpressed in several tumors and is associated with a poor prognosis. It is related to immune-related genes and immune cell infiltration.
Article
Full-text available
Background: Colorectal cancer is one of the most common malignant tumors worldwide. A various of neurotransmitter receptors have been found to be expressed in tumor cells, and the activation of these receptors may promote tumor growth and metastasis. This study aimed to construct a novel neurotransmitter receptor-related genes signature to predict the survival, immune microenvironment, and treatment response of colorectal cancer patients. Methods: RNA-seq and clinical data of colorectal cancer from The Cancer Genome Atlas database and Gene Expression Omnibus were downloaded. Neurotransmitter receptor-related gene were collected from publicly available data sources. The Weighted Gene Coexpression Network Analysis (WGCNA), Least Absolute Shrinkage and Selection Operator (LASSO) logistic regression, Support Vector Machine-Recursive Feature Elimination (SVM-RFE), and Random Forest (RF) algorithms were employed to construct the Neurotransmitter receptor-related gene prognostic signature. Further analyses, functional enrichment, CIBERSORTx, The Tumor Immune Single Cell Center (TISCH), survival analysis, and CellMiner, were performed to analyze immune status and treatment responses. Quantitative real-time polymerase chain reaction (qRT-PCR) assays were carried out to confirm the expression levels of prognostic genes. Results: By combining machine learning algorithm and WGCNA, we identified CHRNA3, GABRD, GRIK3, and GRIK5 as Neurotransmitter receptor-related prognostic genes signature. Functional enrichment analyses showed that these genes were enriched with cellular metabolic-related pathways, such as organic acid, inorganic acid, and lipid metabolism. CIBERSORTx and Single cell analysis showed that the high expression of genes were positively correlated with immunosuppressive cells infiltration, and the genes were mainly expressed in cancer-associated fibroblasts and endothelial cells. A nomogram was further built to predict overall survival (OS). The expression of CHRNA3, GABRD, GRIK3, and GRIK5 in cancer cells significantly impacted their response to chemotherapy. Conclusion: A neurotransmitter receptor-related prognostic gene signature was developed and validated in the current study, giving novel sights of neurotransmitter in predicting the prognostic and improving the treatment of CRC.
Article
Full-text available
Among all malignant tumors, lung cancer has the highest mortality and morbidity rates. The non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC) are the most common histological subtypes. Although there are a number of internationally recognized lung cancer therapy regimens, their therapeutic effects remain inadequate. The outlook for individuals with lung carcinoma has ameliorated partly thanks to the intensive study of the tumor microenvironment and immune checkpoint inhibitors. Numerous cancers have been effectively treated with immunotherapy, which has had positive therapeutic results. Global clinical trials have validated that PD-1/PD-L1 inhibitors are effective and safe for treating lung cancer either independently or in combination, and they are gradually being recommended as systemic treatment medications by numerous guidelines. However, the immunotherapy resistance restricts the immunotherapy efficacy due to the formation of tumor immunosuppressive microenvironment and tumor mutations, and immunotherapy is only effective for a small percentage of lung cancer patients. To summarize, while tumor immunotherapy is benefiting an increasing number of lung cancer patients, most of them still develop natural or acquired resistance during immunotherapy. Consequently, a crucial and urgent topic is understanding and tackling drug resistance triggered by immunotherapy in lung cancer treatment. This review will outline the presently recognized mechanisms of immunotherapy resistance and reversal strategies in lung cancer.
Article
Full-text available
Immune checkpoint therapy via PD-1 antibodies has shown exciting clinical value and robust therapeutic potential in clinical practice. It can significantly improve progression-free survival and overall survival. Following surgery, radiotherapy, chemotherapy, and targeted therapy, cancer treatment has now entered the age of immunotherapy. Although cancer immunotherapy has shown remarkable efficacy, it also suffers from limitations such as irAEs, cytokine storm, low response rate, etc. In this review, we discuss the basic classification, research progress, and limitations of cancer immunotherapy. Besides, by combining cancer immunotherapy resistance mechanism with analysis of combination therapy, we give our insights into the development of new anticancer immunotherapy strategies.
Article
Full-text available
Background Metastasis of breast cancer have caused the majority of cancer-related death worldwide. The circRNAs are associated with tumorigenesis and metastasis in breast cancer according to recent research. However, the biological mechanism of circRNAs in liver metastatic breast cancer remains ambiguous yet. Methods Microarray analysis of three pairs of primary BC tissues and matched hepatic metastatic specimens identified circEZH2. We used RT-qPCR and FISH assays to confirm circEZH2 existence, characteristics, and expression. Both in vivo and in vitro, circEZH2 played an oncogenic role which promoted metastasis as well. A range of bioinformatic analysis, Western blot, RNA pull-down, RIP, ChIP, and animal experiments were used to define the feedback loop involving FUS, circEZH2, miR-217-5p, KLF5, FUS, CXCR4 as well as epithelial and mesenchymal transition. Results In our research, circEZH2 was proved to be upregulated in liver metastases in BC and predicted the worse prognosis in breast cancer patients. Overexpression of circEZH2 notably accentuated the vitality and invasion of BC cells, whereas knockdown of circEZH2 elicited the literally opposite effects. Besides, overexpressed circEZH2 promoted tumorigenesis and liver metastasis in vivo. Moreover, circEZH2 could adsorb miR-217-5p to upregulate KLF5 thus leading to activate FUS transcription which would facilitate the back-splicing program of circEZH2. Meanwhile, KLF5 could upregulated CXCR4 transcriptionally to accelerate epithelial and mesenchymal transition of breast cancer. Conclusions Consequently, a novel feedback loop FUS/circEZH2/KLF5/CXCR4 was established while circEZH2 could be novel biomarker and potential target for BC patients’ therapy.
Article
Full-text available
Growing evidence indicates a connection between cancer-associated fibroblasts (CAFs) and tumor microenvironment (TME) remodeling and tumor progression. Nevertheless, how patterns of CAFs impact TME and immunotherapy responsiveness in triple-negative breast cancer (TNBC) remains unclear. Here, we systematically investigate the relationship between TNBC progression and patterns of CAFs. By using unsupervised clustering methods in the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) dataset, we identified two distinct CAF-associated clusters that were related to clinical features, characteristics of TME, and prognosis of patients. Then, we established a CAF-related prognosis index (CPI) by the least absolute shrinkage and selection operator (LASSO)-Cox regression method. CPI showed prognostic accuracy in both training and validation cohorts (METABRIC, GSE96058, and GSE21653). Consequently, we constructed a nomogram with great predictive performance. Moreover, the CPI was verified to be correlated with the responsiveness of immunotherapy in three independent cohorts (GSE91061, GSE165252, and GSE173839). Taken together, the CPI might help us improve our recognition of the TME of TNBC, predict the prognosis of TNBC patients, and offer more immunotherapy strategies in the future.
Article
Full-text available
Lactate is a key metabolite produced from glycolytic metabolism of glucose molecules, yet it also serves as a primary carbon fuel source for many cell types. In the tumor-immune microenvironment, effect of lactate on cancer and immune cells can be highly complex and hard to decipher, which is further confounded by acidic protons, a co-product of glycolysis. Here we show that lactate is able to increase stemness of CD8 ⁺ T cells and augments anti-tumor immunity. Subcutaneous administration of sodium lactate but not glucose to mice bearing transplanted MC38 tumors results in CD8 ⁺ T cell-dependent tumor growth inhibition. Single cell transcriptomics analysis reveals increased proportion of stem-like TCF-1-expressing CD8 ⁺ T cells among intra-tumoral CD3 ⁺ cells, a phenotype validated by in vitro lactate treatment of T cells. Mechanistically, lactate inhibits histone deacetylase activity, which results in increased acetylation at H3K27 of the Tcf7 super enhancer locus, leading to increased Tcf7 gene expression. CD8 ⁺ T cells in vitro pre-treated with lactate efficiently inhibit tumor growth upon adoptive transfer to tumor-bearing mice. Our results provide evidence for an intrinsic role of lactate in anti-tumor immunity independent of the pH-dependent effect of lactic acid, and might advance cancer immune therapy.
Article
Full-text available
Background: Circular RNAs (circRNAs) regulate complex functional processes and play crucial roles in cancer development and progression. It was reported that circKIF4 regulates the progression of triple-negative breast cancer (TNBC). This study evaluates the role of circKIF4 in breast cancer distant metastasis and metabolic reprogramming. Methods: RT-qPCR was performed to verify the expression of circKIF4A in breast cancer, liver metastatic tissues, and cell lines. The function of circKIF4A in metastasis was evaluated both in vitro and in vivo through a series of experiments, including cell migration and glucose intake experiments. Additionally, we conducted molecular experiments to clarify the regulatory role of circKIF4A. We then conducted a Luciferase reporter assay and an RNA immunoprecipitation assay to identify the molecular interactions between circKIF4A and miRNA. Results: circKIF4A was overexpressed in breast cancer cell lines and tissues, inhibiting its expression and suppressing breast cancer growth and metastasis. Interestingly, we observed that circKIF4A reprogrammed the glucose metabolism of breast cancer, and silencing circKIF4A greatly affected glucose uptake and lactate production in breast cancer cells. miR-335 can be sponged by circKIF4A, which affected the expression of ALDOA/OCT4 protein and regulated HK2/PKM2 expression. Conclusions: This study demonstrated that the circKIF4A-miR-335-OCT4/ALDOA-HK2/PKM2 axis is critical to breast cancer metabolic reprogramming, indicating that this axis could be a novel therapeutic target for the treatment of liver metastasis of breast cancer.
Article
Full-text available
Lactate has diverse roles in the brain at the molecular and behavioral levels under physiological and pathophysiological conditions. This study investigates whether lysine lactylation (Kla), a lactate-derived post-translational modification in macrophages, occurs in brain cells and if it does, whether Kla is induced by the stimuli that accompany changes in lactate levels. Here, we show that Kla in brain cells is regulated by neural excitation and social stress, with parallel changes in lactate levels. These stimuli increase Kla, which is associated with the expression of the neuronal activity marker c-Fos, as well as with decreased social behavior and increased anxiety-like behavior in the stress model. In addition, we identify 63 candidate lysine-lactylated proteins and find that stress preferentially increases histone H1 Kla. This study may open an avenue for the exploration of a role of neuronal activity-induced lactate mediated by protein lactylation in the brain.
Article
Full-text available
Background Neutrophils play a controversial role in tumor development. The function of programmed cell death-1 ligand (PD-L1 ⁺ ) neutrophils, however, may inhibit the cytotoxicity of anti-tumor immunity. In this study, we elucidate the stimulators of PD-L1 ⁺ neutrophils in tumor microenvironment (TME) and explore the optimal combination to enhance the effect of lenvatinib by inhibiting PD-L1 ⁺ neutrophils in hepatocellular carcinoma. Methods Neutrophil infiltration after lenvatinib treatment was examined with RNA sequencing and multicolor flow cytometry analysis in patient samples, subcutaneous and orthotopic mouse models. Neutrophils and T cells were isolated from peripheral blood and tumor tissues and purified with magnetic beads for cytotoxicity assay. Metabolites and cytokines were detected by a biochemical analyzer manufactured by Yellow Springs Instrument (YSI) and proteome profiler cytokines array. In vitro screening of pathway inhibitors was used to identify possible candidates that could reduce PD-L1 ⁺ neutrophil infiltration. Further in vivo assays were used for verification. Results Lenvatinib increased neutrophil recruitment by inducing CXCL2 and CXCL5 secretion in TME. After entering TME, neutrophils polarized toward N2 phenotype. PD-L1 expression was simultaneously upregulated. Thus, lenvatinib efficacy on tumor cells hindered. The increasing PD-L1 ⁺ neutrophils positively corelated with a suppressive T cell phenotype. Further investigation indicated that JAK/STAT1 pathway activated by immune-cell-derived interferon γ and MCT1/NF-kB/COX-2 pathway activated by high concentrations of tumor-derived lactate could induce PD-L1 ⁺ neutrophils. The latter could be significantly inhibited by COX-2 inhibitor celecoxib. Further in vivo assays verified that Celecoxib decreased the survival of lactate-stimulated PD-L1 ⁺ neutrophil and promoted the antitumor effect of lenvatinib. Conclusions PD-L1 ⁺ neutrophils decrease T cell cytotoxicity. Tumor-derived lactate induces PD-L1 expression on neutrophils via MCT1/NF-κB/COX-2 pathway. Thus, COX-2 inhibitor could reduce PD-L1 ⁺ neutrophil and restore T cell cytotoxicity. This may provide a potent addition to lenvatinib.
Article
The tumor microenvironment (TME) is a unique metabolic niche that can inhibit T cell metabolism and cytotoxicity. To dissect the metabolic interplay between tumors and T cells, we establish an in vitro system that recapitulates the metabolic niche of the TME and allows us to define cell-specific metabolism. We identify tumor-derived lactate as an inhibitor of CD8⁺ T cell cytotoxicity, revealing an unexpected metabolic shunt in the TCA cycle. Metabolically fit cytotoxic T cells shunt succinate out of the TCA cycle to promote autocrine signaling via the succinate receptor (SUCNR1). Cytotoxic T cells are reliant on pyruvate carboxylase (PC) to replenish TCA cycle intermediates. By contrast, lactate reduces PC-mediated anaplerosis. The inhibition of pyruvate dehydrogenase (PDH) is sufficient to restore PC activity, succinate secretion, and the activation of SUCNR1. These studies identify PDH as a potential drug target to allow CD8⁺ T cells to retain cytotoxicity and overcome a lactate-enriched TME.
Article
The hallmarks of cancer conceptualization is a heuristic tool for distilling the vast complexity of cancer phenotypes and genotypes into a provisional set of underlying principles. As knowledge of cancer mechanisms has progressed, other facets of the disease have emerged as potential refinements. Herein, the prospect is raised that phenotypic plasticity and disrupted differentiation is a discrete hallmark capability, and that nonmutational epigenetic reprogramming and polymorphic microbiomes both constitute distinctive enabling characteristics that facilitate the acquisition of hallmark capabilities. Additionally, senescent cells, of varying origins, may be added to the roster of functionally important cell types in the tumor microenvironment. Significance Cancer is daunting in the breadth and scope of its diversity, spanning genetics, cell and tissue biology, pathology, and response to therapy. Ever more powerful experimental and computational tools and technologies are providing an avalanche of “big data” about the myriad manifestations of the diseases that cancer encompasses. The integrative concept embodied in the hallmarks of cancer is helping to distill this complexity into an increasingly logical science, and the provisional new dimensions presented in this perspective may add value to that endeavor, to more fully understand mechanisms of cancer development and malignant progression, and apply that knowledge to cancer medicine.
Article
T cells mediate anti-tumor immune responses and are the key target of immune checkpoint therapy, but they can also promote immune tolerance. A clear understanding of the specific contributions and biology of different T cell subsets is required to fully harness the curative potential of immunotherapies. Experts discuss the state of the field and key challenges for moving forward.