0% found this document useful (0 votes)
12 views9 pages

nihms-1711679

The document discusses the tumor microenvironment (TME), highlighting its complex composition and the interactions between tumor cells and various cellular components, including immune cells and stromal cells. It emphasizes the dual roles of immune cells in either promoting or suppressing tumor growth and outlines the contributions of specific cell types, such as T-cells, B-cells, macrophages, and cancer-associated fibroblasts, to tumor progression and metastasis. The TME is portrayed as an active participant in cancer development, with emerging therapeutic targets identified within its structure.

Uploaded by

giangpear311
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
0% found this document useful (0 votes)
12 views9 pages

nihms-1711679

The document discusses the tumor microenvironment (TME), highlighting its complex composition and the interactions between tumor cells and various cellular components, including immune cells and stromal cells. It emphasizes the dual roles of immune cells in either promoting or suppressing tumor growth and outlines the contributions of specific cell types, such as T-cells, B-cells, macrophages, and cancer-associated fibroblasts, to tumor progression and metastasis. The TME is portrayed as an active participant in cancer development, with emerging therapeutic targets identified within its structure.

Uploaded by

giangpear311
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
You are on page 1/ 9

HHS Public Access

Author manuscript
Curr Biol. Author manuscript; available in PMC 2021 June 11.
Author Manuscript

Published in final edited form as:


Curr Biol. 2020 August 17; 30(16): R921–R925. doi:10.1016/j.cub.2020.06.081.

Tumor Microenvironment
Nicole M. Anderson1,2, M. Celeste Simon1,2,*
1.Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of
Medicine, Philadelphia, PA, USA.
2.Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA,
USA.
Author Manuscript

Tumor cells stimulate significant molecular, cellular and physical changes within their host
tissues. The emerging tumor microenvironment (TME) is a complex and continuously
evolving entity. The composition of the TME varies between tumor types, but hallmark
features include immune cells, stromal cells, blood vessels, and extracellular matrix (ECM).
It is believed that the “TME is not just a silent bystander, but rather an active promoter of
cancer progression”. Early in tumor growth, a dynamic and reciprocal relationship develops
between cancer cells and components of the TME to support cancer cell survival, local
invasion and metastatic dissemination. To overcome a hypoxic and acidic microenvironment,
the TME coordinate a program that promotes angiogenesis to restore oxygen/nutrient supply
and remove metabolic waste. Tumors become infiltrated with diverse adaptive and innate
immune cells that can perform both pro- and anti- tumorigenic effects (Figure 1). An
Author Manuscript

expanding literature on the TME has identified new targets within it for therapeutic
intervention.

Immune Cells
Immune cells are a critical component of the TME. Depending on the context, a dichotomy
exists in the relationship between immune cells and TMEs: immune cells can either suppress
tumor growth or promote it (Figure 1). Persistent inflammation due to chronic infection is a
common mechanism underlying tumor formation in several types of cancer, including:
colorectal, hepatocellular and cervical cancer. Broadly, immune cells fall into two
categories: adaptive immune cells and innate immune cells. Adaptive immunity is activated
by exposure to specific antigens and uses an immunological memory to “evaluate” the threat
and enhance immune responses. T-cells, B-cells and NK cells belong to the adaptive
Author Manuscript

immune response. Innate immunity is a non-specific defense mechanism that comes into
play within hours of a foreign antigen entering the body. Cells that carry out an innate
immune response include macrophages, neutrophils and dendritic cells.

T-cells
Each T-cell develops its own T cell receptor (TCR) that recognizes a specific antigen. Within
the TME there are several distinct populations of T-cells that influence tumorigenesis.

*
Corresponding author: [email protected].
Anderson and Simon Page 2

Cytotoxic T-cells (CD8+) detect abnormal tumor antigens expressed on cancer cells and
Author Manuscript

target tumor cells for destruction. The presence of cytotoxic T-cells in the TME is often
associated with positive prognosis in cancer patients. Aside from killing tumor cells,
cytotoxic T-cells also suppress angiogenesis through the secretion of interferon gamma
(IFN-γ). CD4+ T-cells differentiate into a variety of subtypes and thus coordinate a wide
range of immune responses within the context of the TME. T helper 1 (Th-1) cells are
proinflammatory CD4+ T-cells that support CD8+ cells through the secretion of
interleukin-2 (IL-2) and IFN-γ. Increased levels of Th-1 cells within the TME are associated
with positive outcomes in many types of cancer. Regulatory T cells (Tregs) are normally
required to suppress inflammatory responses and control autoimmunity. In the context of the
TME, Tregs are ubiquitous and promote tumor development and progression by dampening
antitumor immune responses. For example, Tregs secrete IL-2 which modulates natural
killer (NK) cell homeostasis and function. Additionally, Tregs cells directly support the
Author Manuscript

survival of cancer cells through the secretion of growth factors, and indirectly through
interaction with stromal cells such as fibroblasts and endothelial cells. The immune
landscape within the TME falls into three main categories: immune infiltrated, immune
excluded, and immune silent. In an immune infiltrated tumor, immune cells (e.g. cytotoxic
T-cells) are homogenously distributed throughout the tumor indicating an active immune
response. Alternatively, some tumors are classified as immune excluded were T-cells are at
the periphery of tumors and have not infiltrated the TME. Finally, some tumors are
categorized as “immune silent” and completely lack immune cell infiltrate indicating no
immune response to the tumor.

B-cells
B-cells are specialized immune cells responsible for antibody production, antigen
Author Manuscript

presentation and secretion of cytokines. Typically, B-cells concentrate at the margin of


tumors and are commonly found in lymph nodes in close proximity to the TME. Compared
to T-cells, the TME has relatively few infiltrating B-cells; however, recent studies have
demonstrated that the presence and function of B-cells are important during tumorigenesis.
Tumor infiltrating B-cells are important in the formation of “tertiary lymphoid structures”,
which are ectopic lymphoid structures formed within the TME. Tertiary lymphoid structures
allow close association between T and B cells and are a positive prognostic marker in breast
cancer, melanoma and ovarian cancer. The anti-tumorigenic roles of B-cells, include
antigen-presentation to T-cells, anti-tumor antibody production and secretion of cytokines
that promote cytotoxic immune responses (e.g. IFN-γ). Alternatively, B-cells can have
protumor effects, and their presence in the TME can be predictive of poor outcome in
bladder cancer, prostate cancer, renal cell carcinoma. Regulatory B-cells promote tumor
Author Manuscript

aggression through production of cytokines (e.g. IL-10 and transforming growth factor-beta
(TGF-β)) that promote immune suppressive phenotypes in macrophages, neutrophils and
cytotoxic T cells.

Natural Killer Cells


Natural killer cells naturally patrol the blood stream and seek out virally infected host cells
or tumor cells. Functionally, NK cells can be broken down into two classes; they either
directly participate in cell-mediated killing of tumor cells or they secrete inflammatory

Curr Biol. Author manuscript; available in PMC 2021 June 11.


Anderson and Simon Page 3

cytokines. NK cells are highly efficient at killing tumor cells within the circulation and can
Author Manuscript

participate in blocking metastasis, but are less efficient at killing within the tumor
microenvironment.

Macrophages
Macrophages are a critical component of the innate immune system that modulate immune
responses through pathogen phagocytosis and antigen presentation. In addition,
macrophages are critical in wound healing and tissue repair. Monocyte derived macrophages
can be categorized into either inflammatory M1 macrophages that phagocytize and kill cells
or immune-suppressive M2 macrophages that participate in wound healing. While both
classes of macrophages can be found within a tumor, the TME promotes the M2 phenotype
through hypoxia and the secretion of cytokines (e.g. IL-4) to support tumor growth and
progression. Certain tumor types can be heavily infiltrated with macrophages and can make
Author Manuscript

up to 50% of a tumors mass. Typically, high macrophage infiltration is associated with poor
patient prognosis in many types of cancer, such as breast cancer, lung cancer and gastric
cancer. Often macrophages are found to surround blood vessels in the TME where they
secrete vascular endothelial growth factor (VEGF)-A and induce new blood vessel
formation.

Neutrophils
Neutrophils make up to 70% of circulating leukocytes and provide the first line of defense
against many pathogens. In the context of cancer, neutrophils can act to either suppress or
promote tumor growth, depending on tumor type and stage of development. As a tumor
begins to grow neutrophils are recruited to the TME and promote inflammation through
release of cytokines and reactive oxygen species to promote tumor cell apoptosis. Later in
Author Manuscript

tumor development neutrophils promote tumor growth through modification of the


extracellular matrix, releasing VEGF and producing matrix metalloprotease (MMP) −9 to
stimulate angiogenesis and ultimately tumor progression and local invasion.

Dendritic Cells
Dendritic cells (DCs) play a critical role in the immune system as antigen presenting cells;
they recognize, capture and present antigens to T-cells (antigen presenting cells, APC) at
secondary lymphoid organs (e.g. lymph nodes). Ultimately, DCs bridge a gap between
adaptive and innate immunity to initiate pathogen-specific T-cell responses. The tumor
microenvironment shapes the fate of DCs by providing environmental cues that either create
an immune response to tumor cells or accept them. Cytokines secreted from the TME trigger
DCs to tolerate the presence of tumor cells and block the induction of an immune response.
Author Manuscript

Stromal Cells
Cancer cells recruit supporting cells from nearby endogenous tissue stroma to promote
critical steps in tumor formation. Stromal cell composition can vary significantly between
tumors types and include: vascular endothelial cells, fibroblasts, adipocytes and stellate
cells. Once recruited to the TME, stromal cells secrete many factors that influence
angiogenesis, proliferation, invasion, and metastasis (Figure 2).

Curr Biol. Author manuscript; available in PMC 2021 June 11.


Anderson and Simon Page 4

Endothelial Cells
Author Manuscript

Vascular endothelium is a thin monolayer of endothelial cells (ECs) and help to orchestrate
the formation of blood vessels. Not only does vascular endothelium separate circulating
blood from tissues, it also delivers water and nutrients, maintains metabolic homeostasis,
carries immune cells and participates in the formation of new blood vessels. During the
initial stages of tumor development cancer cells rely on passive diffusion for gas exchange
and the transportation of nutrients. Once tumors reach 1–2mm3 in volume, insufficient
oxygen and a build-up metabolic waste results in the TME becoming hypoxic and acidic. To
overcome hypoxia and acidification, tumors must develop their own blood supply. A
hypoxic TME leads to the activation hypoxia inducible factors (HIFs), transcription factors
critical in coordinating cellular responses to low O2. Vessel sprouting is a common
mechanism used by tumors to co-opt existing blood vessels and induce the growth of new
vessels. Specifically, HIFs initiate vessel sprouting by instructing ECs to secrete
Author Manuscript

proangiogenic factors such as, platelet derived growth factor (PDGF), epidermal growth
factor (EGF) and VEGF. In an autocrine and paracrine fashion, VEGF stimulates migration
of ECs to form new blood vessel lumens. Next ECs secrete proteins to form new basement
membranes. Blood vessels in the TME often fail to achieve the final stages of maturation,
resulting in leaky vasculature.

Beyond angiogenesis, ECs are critical in promoting cancer cell migration, invasion and
metastasis. ECs are highly plastic in nature and can change cell fate. During tumor
progression, ECs undergo endothelial-mesenchymal transition to become cancer associated
fibroblasts (CAFs). The transition from an EC to a CAF is organized by TGF-β and bone
morphogenetic protein (BMP) and leads to loss of cell-to-cell connections, detachment and
elongation, enhanced migration and loss of endothelial properties. CAF are critical in
stimulating migration and invasion of tumor cells (see below). Metastasis is a multistep
Author Manuscript

process that involves translocation of cancer cells from the primary TME to distant
locations. Tumor cells must first escape the primary tumor site and enter the vasculature in a
process known as intravasation. During intravasation, tumor cells adhere to ECs and this
interaction changes the endothelial barrier and allows tumor cells to migrate between two
ECs. Blood vessels formed in the TME are usually immature and lack proper cell-to-cell
connections, enabling cancer cells to extravasate.

Cancer Associated Fibroblasts


Cancer associated fibroblasts (CAFs) are a major component of the tumor stroma and play a
critical role in facilitating crosstalk between cancer cells and TME. While diverse in origin,
CAFs often arise from tissue resident fibroblasts. CAFS are also formed from adipocytes,
Author Manuscript

ECs, pericytes, stellate cells and bone marrow derive mesenchymal stem cells. Upon injury,
fibroblasts that normally reside within tissues can become reversibly induced to form
myofibroblasts, which actively participate in wound healing. Myofibroblasts are activated by
TGF-β signaling and develop characteristics important in wound healing, such as
proliferation, contractile properties, secretory phenotypes and extracellular matrix formation.
Tumors have been aptly termed “wounds that never heal”. In the TME, cancer cells and
other stroma cells secrete transforming growth factors to convert fibroblasts into CAFs, such
as TGF-β, platelet derived growth factor (PGDF) and FGF2. A build-up of CAFs within the

Curr Biol. Author manuscript; available in PMC 2021 June 11.


Anderson and Simon Page 5

tumor microenvironment is often associated with poor prognosis in many cancer types. For
Author Manuscript

example, in colorectal cancer the presence of CAFs is strongly associated with disease
reoccurrence. Despite an association with poor prognosis, CAFs have been shown to both
promote and restrain tumorigenesis. Alternatively, some cancer types, such as breast cancer
and lung cancer, have improved prognosis and overall survival when they are highly
desmoplastic.

Within the TME, CAFs produce the majority of extracellular components, including growth
factors, cytokines and extracellular matrix. CAFs shape the TME in four main ways: tumor
proliferation and metastasis, neoangiogenesis, ECM remodeling and immunosuppression. In
tumors of epithelial origin, EMT or epithelial-mesenchymal transition is a critical step in
metastasis were epithelial cells lose cell polarity and cell-to-cell adhesions and gain
migratory and invasive phenotypes. One way CAFs control metastasis is through the
secretion of TGF-β, a growth factor required for EMT and angiogenesis. To facilitate
Author Manuscript

migration of cancer cells through the TME, CAFs secrete MMP-3 which degrades E-
cadherin to promote cancer cell invasion. The ECM is also an important source of VEGF,
which can be released by MMP-13 to promote angiogenesis. In general, CAFs promote an
immunosuppressive phenotype through the production of immune modulatory chemokines
and cytokines.

Adipocytes
Adipocytes are specialized cells within the body that regulate energy balance and are
responsible for storing excess energy as fat. Adipocytes exert their effects on the TME
through secretion of metabolites, enzymes, hormones, growth factors and cytokines. Within
the context of the TME, adipocytes are in a dynamic and reciprocal relationship with tumor
cells to support tumor progression. Breast tissue is largely composed of white adipose tissue
Author Manuscript

(WAT); therefore, adipocytes are a critical player in the breast cancer TME. Breast cancer
cells can stimulate adipocytes to undergo lipidlysis, which breaks down lipid stores making
free fatty acids available uptake by the cancer cells. Breast cancer cells use free fatty acids
for energy production, cell membrane formation, lipid bioactive molecules and exosomes.
Leptin is an important hormone produced by adipocytes and promotes tumor progression
directly by influencing breast cancer cell proliferation and indirectly by activation of
macrophages. Adipocytes also play an important role in modifying ECM through secretion
of metalloproteases, such as MMP-1, MMP-7, MMP-10, MMP-11 and MMP-14. More than
40% of cancer patients are overweight, making obesity is a major risk factor for many types
of cancer, including: breast, pancreatic and ovarian. White adipose tissue is an endocrine
organ and can promote breast cancer cells to metastasis to the liver and lungs through
paracrine signaling.
Author Manuscript

Stellate Cells
Stellate cells are quiescent stromal cells of mesenchymal origin located within the liver and
pancreas. Upon tissue injury, stellate cells become activated, enter into cell cycle and are
induced to transform into myofibroblasts. A characteristic feature of stellate cells is the
deposition of vitamin A in lipid droplets. In the liver, hepatic stellate cells (HSC) are
normally located within perisinusoidal and portal areas and can constitute as much as 15%

Curr Biol. Author manuscript; available in PMC 2021 June 11.


Anderson and Simon Page 6

of liver mass. Hepatocellular carcinoma (HCC) is the predominant form of liver cancer and
Author Manuscript

HSCs function to promote crosstalk within the TME. A key signaling molecule, TGF-β is
produced by HCC and triggers HSCs to become activated. Once activated HSCs modify the
ECM and produce proangiogenic factors such as VEGF-A and MMP-2. Lipid droplets are
critical structures in HSCs used to produce new ECM and remodel it through the production
of MMPs. Pancreatic ductal adenocarcinoma is the most common form of pancreatic cancer
(95%), characterized by dense fibrotic tissue or desmoplasia. When pancreatic stellate cells
(PSCs) are quiescent, they contribute to ECM modification through the production of ECM
proteins (e.g. desmin, vimentin) and degradation enzymes. Vitamin A depletion results in
PSC activation, leading to the secretion of cytokines and chemokines, enhanced migration
and proliferation potential. Activated PSCs play a critical role in promoting the desmoplastic
phenotype of PDAC tumors and their hypoxic microenvironments.
Author Manuscript

Non-cellular components of the TME


ECM
The extracellular matrix (ECM) has important functions within the TME; not only does it
provide a physical scaffold for cells; it also is a key factor in promoting tumor cell
dissemination. ECM is an important molecular component of the TME and composed of
collagen, fibronectin, elastin, and laminin. Solid tumors contain large ECM deposits and
constitute up to 60% of tumors mass. Large collagen deposits together with a high
percentage of fibroblast infiltration result in desmoplasia, which is strongly linked to poor
patient prognosis. Many cells within the TME secrete components of the ECM, although
CAFs are the predominant source of ECM. MMPs are proteases that breakdown ECM
proteins and are critical in remodeling ECM to promote tumor progression and metastasis.
The ECM is a depot for cytokines and growth factors, which are released by proteases (e.g.
Author Manuscript

MMPs). For example, the ECM can be a deposit for proangiogenic factors, like VEGF, FGF,
PDGFB, TGFB.

Exosomes
Exosomes are microvesicles that range in size from 30nm to 200nm. Their contents reflect
the original cell, including protein, RNA, DNA and lipids. Within the TME exosomes play a
critical role in facilitating cross talk between cancer cells and stromal cells. Functionally,
exosomes have been shown to promote inflammation, tumor progression, angiogenesis, and
metastasis within the TME. Conditions of hypoxia appear to exacerbate exosome production
by cancer cells and promote the transition of stromal cells into CAFs.

Therapeutically Targeting the TME


Author Manuscript

Over the last decade cancer treatment has undergone a revolution. Traditionally, drugs
targeted tumors more broadly (e.g. chemotherapy) and now new therapeutic strategies target
specific cells within the TME. Immune check-point blockade (ICB) was the first generation
of antibody-based therapies to target immune cells in the TME. ICB works by blocking
receptor/ligand interactions (e.g. CTLA4 and PD1) dulling T-cell activation and function.
Patients who respond to ICB have significant clinical benefit, but the majority of patients do

Curr Biol. Author manuscript; available in PMC 2021 June 11.


Anderson and Simon Page 7

not respond to ICB. The identification of relevant biomarkers is required to recognize


Author Manuscript

patients who are expected to benefit from immune check-point blockade therapy.
Therapeutically targeting DC activation through the use of DC vaccination has been
successfully used in the treatment of prostate cancer. The “Provenge” protocol involves
harvesting monocytes from prostate cancer patients, differentiation into DC, activation with
PAP antigen, and then re-introducing them back into patients. Provenge therapy can result in
significant reduction in tumor burden of prostate cancer patients.

Growing tumors require the formation of new blood vessels to relieve oxygen deprivation
and accumulating metabolic waste, therefore targeting angiogenesis was an attractive
strategy. Antiangiogenic (AA) therapy has focused on targeting the VEGF/VEGFR signaling
axis and has included: a neutralizing antibody to VEGF-A (Bevacizumab); decoy receptor
for VEGF-A or B (Aflibercept); tyrosine kinase inhibitor (Sorafenib); and antibody that
blocks VEGF binding its receptor (Ramucirumab). As a single agent most patients either do
Author Manuscript

not respond to AA therapy or develop resistance. Successful integration of AA therapy into


the clinic will likely require combination with other agents. For example, Bevacizumab in
combination with PDL-1 has shown some success for the treatment of HCC and renal
cancer.

While therapeutically targeting the TME is an attractive strategy for the treatment of cancer,
existing FDA approved treatments have limited efficacy. As we continue to understand how
the TME contributes to tumorigenesis, new therapeutic targets and strategies will be
identified. Promising preclinical studies have shown potential for the use of CAR-NK cells,
liver stellate cells and fibroblasts.

Further Reading:
Author Manuscript

Bussard KM, Mutkus L, Stumpf K, Gomez-Manzano C, and Marini FC (2016). Tumor-associated


stromal cells as key contributors to the tumor microenvironment. Breast Cancer Res. 18.
Henke E, Nandigama R, and Ergün S (2020). Extracellular Matrix in the Tumor Microenvironment and
Its Impact on Cancer Therapy. Front. Mol. Biosci 6, 160. [PubMed: 32118030]
Hinshaw DC, and Shevde LA (2019). The tumor microenvironment innately modulates cancer
progression. Cancer Res. 79, 4557–4567. [PubMed: 31350295]
Maimela NR, Liu S, and Zhang Y (2019). Fates of CD8+ T cells in Tumor Microenvironment.
Comput. Struct. Biotechnol. J 17, 1–13. [PubMed: 30581539]
Plitas G, and Rudensky AY (2020). Regulatory T Cells in Cancer. Annu. Rev. Cancer Biol 4, 459–477.
Sharonov GV, Serebrovskaya EO, Yuzhakova DV, Britanova OV, and Chudakov DM (2020). B cells,
plasma cells and antibody repertoires in the tumour microenvironment. Nat. Rev. Immunol 20, 294–
307. [PubMed: 31988391]
Turley SJ, Cremasco V, and Astarita JL (2015). Immunological hallmarks of stromal cells in the
tumour microenvironment. Nat. Rev. Immunol 15, 669–682. [PubMed: 26471778]
Author Manuscript

Xie H, and Simon MC (2017). Oxygen availability and metabolic reprogramming in cancer. J. Biol.
Chem 292, 16825–16832. [PubMed: 28842498]

Curr Biol. Author manuscript; available in PMC 2021 June 11.


Anderson and Simon Page 8
Author Manuscript
Author Manuscript

Figure 1. Impact of immune cells within the TME.


The role of immune cells in the TME can be to either suppress tumor formation (anti-tumor
microenvironment) or promote tumorigenesis (immune suppressive microenvironment).
Depending on context and tumor type immune cells can both be pro or anti-tumorigenic.
Author Manuscript
Author Manuscript

Curr Biol. Author manuscript; available in PMC 2021 June 11.


Anderson and Simon Page 9
Author Manuscript
Author Manuscript

Figure 2. Defining the role of stromal cells in promoting cancer progression.


Cancer cells recruit stromal cells from neighboring tissue during tumorigenesis. In the TME,
Author Manuscript

stromal cells and cancer are in a dynamic relationship promoting the cancer progression.
The stromal cell composition varies between tumor types but includes endothelial cells,
fibroblasts, adipocytes and stellate cells. The TME orchestrates angiogenesis, proliferation,
invasion and metastasis through the secretion of growth factors and cytokines.
Author Manuscript

Curr Biol. Author manuscript; available in PMC 2021 June 11.

You might also like