You are on page 1of 8

Deregulating

Cellular Energetics
Cancer cells need a lot of energy to grow fast—to do so, they show abnormal metabolic pathways.
Most mammalian cells use glucose as a fuel source. Glucose is metabolized by glycolysis in a multistep set
of reactions, resulting in the creation of pyruvate. In typical cells under normal oxygen levels, much of this
pyruvate enters the mitochondria, where it is oxidized by the Krebs Cycle to generate ATP to meet the cell’s
energy demands.
However, in cancer cells or other highly proliferative cell types, much of the pyruvate from glycolysis is
directed away from the mitochondria to create lactate through the action of lactate dehydrogenase (LDH/
LDHA)—a process typically reserved for the low oxygen state. In contrast to mitochondrial glycolysis, lactate
production in the presence of oxygen is termed “aerobic glycolysis” or the Warburg Effect. Several signaling
pathways contribute to the Warburg Effect and other metabolic phenotypes of cancer cells. If you look at
the pathway, you might notice PI3K/Akt, mTOR, Erk1/2 MAPK, ULK1 (Autophagy), p53, HIF1α (Hypoxia –
Angiogenesis), and AMPK all represented.
Cancer cells frequently use glutamine as another fuel source, which enters the mitochondria and can
be used to replenish Krebs Cycle intermediates or to produce more pyruvate through the action of malic
enzyme, or to produce building blocks to help the increased cell growth.
Cancer cells can become addicted to glutamine, with glutamine itself promoting cell proliferation. For this
reason, glutamine metabolism is becoming a hot research area for cancer researchers. Because glycolysis
is an intricate process, influenced by so many factors, there are many pathways to research, such as the
Warburg Effect pathway, Energy Metabolism pathway, and Glutamine Signaling pathway.

References:
1. Hanahan D, Weinberg RA (January 2000). “The Hallmarks of Cancer”. Cell. 100 (1): 57–70. doi:10.1016/S0092-8674(00)81683-9
2. Hanahan D, Weinberg RA (March 2011). “Hallmarks of Cancer: the next generation”. Cell. 144 (5):646-74. doi: 10.1016/j.cell.2011.02.013

Warburg Effect Energy Metabolism Glutamine Signaling Glutamine Metabolism

Macropinocytosis and Insulin Receptor Glycolysis


other scavenging pathways Glycolysis Growth Factors Glutamine Glutamine Leucine
SNARE TNF GPNA
Glucose Glucose Complex Glucose
Glucose GLUT4
Transporters
p85
Gab1

FlotillinCav
TNFR1

PTEN PI3K ASCT2/SLC1A5 SLC7A5/LAT1


Ras
CAP

Synip p110 SOCS3


Cbl

IRS

Torin1
Shc

PI3K PIP3
APS

Hexokinase
Amino Acids NADPH NADP SHIP Akt2 SHP-2 PTP1B
and Lipids Ras
Crkll

c-Myc
Pentose 6-P- Glucose-6-P Akt GLUT4 EHD1 GRB2 Nck
Phosphate Translocation TC10 C3G Glucose
Gluconolactone IRS-1 Crkll c-Myc
6-P- Ras MEK1/2 EHBP1 Transporters
Shunt NADP Amino Acids CIP4/2 PKCλ/ζ Fyn Leucine
Gluconate TIGAR Fructose-6-P
SOS mTORC1
NADPH p53 LKB1 PDK1 Ras
SREBP Rac1 mTORC2 HK
PFK Erk1/2
Ribulose-5P AMPK
PKCθ GRB10
Jnk Glutamine AA/Protein
Fructose mTOR GLUT4 PKCλ/ζ IKK Akt Glucose-6-P
GFAT
Bisphosphate c-Myc Ras Synthesis
Nucleotide HIF-1α vesicle Cell Growth
Synthesis PP2A FFA Glucosamine-6-P γ-nitrogen
PHGDH
Serine 3-Phosphoglycerate LKB1 NO iNOS Fructose-6-P
Protein Synthesis Cell Proliferation 14-3-3
NADPH c-Myc LKB1 Glutamate CAD
GLUT4 AS160 Akt
NADP
PEP Exocytosis PFK UDP-GlcNac
Glycine ULK1 ROS c-Raf Pyrimidine
PKM2 TBC1D1 AMPK Glycosylation
HIF-1α Fatty Acid Oxidation PDK1 14-3-3 Cysteine Purine
NADP
Pyruvate Fatty Acids TSC2 PDE3B AMPK Fructose Glycine Nucleotide
c-Myc Autophagy
Malic Enzyme TSC1 Bisphosphate Mitochondrial Nonessential Synthesis
NADPH Bad SIK2
LDHA Dysfunction AA Synthesis
Pyruvate
PDHK GSK-3 PRAS40 Ras
Lactate Rheb [cAMP] MEK1/2
Dehydrogenase
Hypoxia Malate
Malate Pyruvate Acetyl-CoA SGK
Folate 3-Phosphoglycerate
Metabolism Oxaloacetate β-Oxidation Apoptosis SREBP CBP/p300
Acetate Torc2 Asp
mTORC1 PKA Ras
Akt p70 Erk1/2 HIF-1α
Bax

PP1 GS PEP GSH


4E-BP1 S6K
AN

Krebs Lipin1
Malate Cycle Citrate Citrate ATP-citrate Degradation PKM-2 Redox
T

ACSS2 Asp
VDAC
Hexokinase ENaC lyase eIF4E HSL
ACL
Glycogen Homeostasis
IDH2 UCP2 Synthesis Protein Synthesis, Pyruvate
AMPK ME
AN

AC Acetyl-CoA Fatty Acid Growth, and SREBP-1


VD
Synthesis
T

α-Ketoglutarate Sodium Proliferation Lipolysis NADPH Glutamine


OMM Stability IDH1 ACC
Transport
Fatty Acid/
Inhibition Cytochrome C Release Glutaminase
NADP
Lipid Synthesis
NADPH m GLS
Inhibition of Apoptosis Epigenetic Cytoplas Erk1/2
Lactate miR23a/b c-Myc
p53 Glutamine Transporters Regulation Fatty Acid IDH2 HIF-1α
SGK Akt2 Acetyl-CoA Citrate
Glutamine and Cholesterol Disruption of
Nucleus Synthesis CBP/Torc2/CREB
Glutaminolysis NH4+ Autophagy
Complex Malate TA
FoxO3 FoxO4 FoxO1 Erk1/2
SREBP LXRα USF Gluconeogenesis Glutamate IDH1 α-KG
Transcription Apoptosis,
Oxaloacetate GDH SirT4 mTORC1
mTORC1 mTORC2 Autophagy, Lipin1 α-KG NADPH
Raptor Sin1 PRR5
Glucose and TCA Cycle
GβL
Rictor
Lipid Metabolism Growth
mTOR GβL Fatty Acid/
DEPTOR
mTOR Acetyl-CoA Lipid Synthesis
DEPTOR
Malate Citrate
Anaplerosis Oxaloacetate Amino Acid
Cataplerosis Synthesis

rev. 04/25/18

visit www.cellsignal.com/pathways to see all pathways 7


Inducing
Angiogenesis
Cancer cells stimulate the growth of blood vessels to supply nutrients to tumors. Angiogenesis is the formation
of new blood vessels from pre-existing blood vessels. This plays an important role in tumor growth.
Benign tumors can exist in a dormant state, which can be driven by inadequate access to sufficient blood
supply. However, the “angiogenic switch” occurs when angiogenesis is activated in a dormant tumor, and
growth factors are secreted to induce sprouting and chemotaxis of endothelial cells toward the tumor mass.
Within the hypoxic environment of the tumor mass, hypoxia inducible factor-1 (HIF-1) is stabilized and activates
the expression of multiple genes contributing to the angiogenic process, including vascular endothelial growth
factor (VEGF) and fibroblast growth factor (bFGF), or platelet-derived growth factor (PDGF).

References:
1. Hanahan D, Weinberg RA (January 2000). “The Hallmarks of Cancer”. Cell. 100 (1): 57–70. doi:10.1016/S0092-8674(00)81683-9
2. Hanahan D, Weinberg RA (March 2011). “Hallmarks of Cancer: the next generation”. Cell. 144 (5):646-74. doi: 10.1016/j.cell.2011.02.013

Tumor Angiogenesis
Basement
Pericyte Membrane
Endothelial
Precursor Platelet
Cell
Endothelial Cell

PDGFR

VEGFR2
Tie2 Tie2
FGFR MMPs

EPH Neuropilin • Growth Factors


• Cytokines
• ECM Proteases
ROBO3
Integrins Integrins
ROBO1/2 Ets2
Tip Cell HIF-1α Stat3
Target Genes

Tumor Associated
Macrophage (TAM)

OH

elF4E1 HIF-1α
Key

Angiopoietin 1
OH Normoxia
Angiopoietin 2
bFGF PHDs HIF-1α HIF-1β
Akt Erk1/2
Ephrin 4E- Hypoxia
BP1
PDGF
• Growth Factors
SLIT • Cytokines
• ECM Proteases CBP/p300
VEGF
MMPs HIF-1α HIF-1β
HRE Target Genes

Tumor Cell
Tumor Cell Nucleus

“Benign tumors can exist in a


dormant state, which can be driven
by inadequate access to sufficient
blood supply. ”
8
Sustaining
Proliferative
Signaling
Cancer cells stimulate their own growth, which means they become self-sufficient in growth signals and no longer
depend on external signals, such as epidermal growth factor (EGF/EGFR). Proliferation depends highly on these
three important pathways: Akt, MAPK/Erk, and mTOR.
Akt, also known as Protein Kinase B (PKB), represents a family of serine/threonine protein kinases, Akt1, Akt2,
and Akt3. Akt1 (v-Akt) was originally discovered as a proto-oncogene and plays a critical role in regulating diverse
cellular functions, which include metabolism, growth, proliferation, survival, transcription, and protein synthesis.
Akt is activated by various extracellular and intracellular signals, most involving the lipid kinase phosphoinositide
3-kinase (PI3K). It is also a major regulator of cell survival by inhibiting the apoptotic effects of signaling pathways
and proteins (like FoxO1).
Mitogen activated protein kinases (MAPKs) are the central component in numerous signaling cascades that
transmit growth, proliferation, and survival signals from the cell surface to the nucleus. Stimulation of receptor-
tyrosine kinases (RTKs) by growth factors, engagement of integrins, or changes in cellular homeostasis (eg,
stress) promote the activation of signaling pathways involving Erk (extracellular regulated kinase), p38 MAPK, or
Jnk (c-Jun N-terminal kinase). Each of these kinases, in turn, promotes activation of transcription factors such as
c-Jun, Ets, Alk, and ATF resulting in cellular growth, survival, repair, and proliferation.
mTOR (mammalian target of rapamycin) is a protein kinase that functions as an ATP and amino acid sensor to
balance nutrient and energy availability with cellular growth. mTOR can also be activated or inhibited by other
signaling pathways involving Akt, Erk, and AMPK. mTOR in turn regulates a series of metabolic enzymes and other
protein kinases that modulate lipid metabolism and biogenesis cellular growth and proliferation and autophagy.

References:
1. Hanahan D, Weinberg RA (January 2000). “The Hallmarks of Cancer”. Cell. 100 (1): 57–70. doi:10.1016/S0092-8674(00)81683-9
2. Hanahan D, Weinberg RA (March 2011). “Hallmarks of Cancer: the next generation”. Cell. 144 (5):646-74. doi: 10.1016/j.cell.2011.02.013.

PI3K/Akt MAPK/Erk mTOR


BCR Integrins
Cytokine RTKs Growth Factors, Glucose
Receptor Ag Hormones,
RTK CD19 Ion Channels RTKs AMP: ATP
[Ca2+] Cytokines, etc.
Wnt Stress AICAR
Integrin metformin
mIg α/β α/β GPCR Hypoxia
mIg Glucose,
SOS Ras SOS Tal Amino Acids
LRP
GRB2

Rap1 Frizzled
FRS2

CAS RACK1
Shc
IRS
GRB2

PLCγ Fyn IRS-1


CRK

DNA Damage Spry


BCAP Pax FAK C3G
PI3K

PI3K
PI3K

Jak1 Gαq/o Dvl DNA


IRS-1

PI3K
Gab1

Syk
PI3K

ATM/ATR DNA-PK Lyn EPAC Damage


FAK [Ca2+] GRB10 PIP3 PIP2
PI3K

Src mTORC2
Gab2
PI3K


Gβγ GTP Raptor mTOR
PYK2 PAK Rac [cAMP] PDK1 Ras GATOR2
Paxillin

Spred Signaling
p53
ILK AMPK Sin1 PRR5 PTEN
PTEN Akt mTORC2 PIP3 PTEN PP2A PHLPP1/2 CTMP c-Raf PKA rpS6 TCPβ
Mios WDR24
PIP3 Rictor GβL
PKC
S6 TRAF6 NEDD1-4 c-Raf B-Raf B-Raf IκBα WDR59 Sec13
Neutrophil eIF4B mTOR
TBK1 Membrane Respiratory Heterodimer 14-3-3 LKB1 DEPTOR Seh1L
p47phox
Protein Synthesis

Membrane PDK1 PDK1 Erk


Recruitment
Recruitment Burst PD98059 Filamin A
and Activation Tpl2/Cot1
and Activation
Lamin A
Organization MEK1 U0126 GATOR1
p70 S6K of Nuclear LAMTOR2 AZD6244 Sestrin-1/2
Proteins MP1 IMP PP1/ SGK1 PKCα Akt RSK DEPDC5 Nprl2
Other

Tpl2 Ion Channels, PP2At FMK


NF-κB Receptors C-TAK1 KSR
PDCD4 Pathway
SL0101
eEF2K GSK-3 TSC1 Nprl3
IKKα Erk1 1/2 BI-D1870
TSC2 cPLA2 SOS TSC2 GSK-3
Cardiovascular Torin1
4E-
BP1
mTORC1 TSC1 eNOS Homeostasis MEK1/2 Bim DAPK METTL1
PP242 PRAS40 TBC1D7
Inhibits V-ATPase
Autophagy Translation p90RSK KU63794 REDD1/2
Synaptic Late Endosome MNK1/2 Erk1/2 BAD nNos LKB1 LAMTOR
GABAAR Control WYE354 1/2/3/4/5
ATG13 Signaling
PRAS40 Akt p90RSK MKP-3 Rheb AMPK Ragulator
Blocks Erk1/2 TSC2 FLCN
science

XIAP Huntingtin Complex


Aggregation Rag A/B
Inhibits Apoptosis Cell Adhesion PEA-15 Cytoplasmic Anchoring Ca+2-regulated rapamycin
Promotes WAVE-2 PLD1 GTP FNIP1/2
MDM2 Ataxin-1 Neuroprotection Migration Exocytosis FKBP12 mTORC1 mTORC1
Neuro

FoxO1 Rag C/D


Aggregation Translocation GDP Autophagy/Lysosome
PFKFB2
GβL Raptor to Lysosome
Bad Neurodegeneration m Biogenesis
p53 GSK-3 Cytoplas TFEB
14-3-3 cdc25 PPARγ mTOR
Bim Glycolysis
DEPTOR Lipid
PIP5K AS160 Nucleus Metabolism
PPARα
Bcl-2 Hexokinase II p27 KIP1 p90RSK BUB1 VEGF/
MKP-1/2 Erk1/2 MSK1/2 Angiogenesis
Su

Skp2 SCF ATP-Citrate FIP200 4E- HIF-1


eIF4G
rv

Lyase Glucose p70S6K BP1/2 Mitochondrial


iva

Bax Palladin Atg13


GS Transport Metabolism
l

Girdin p27 Kip1 ULK PGC-1α


Myt1
sm

Apoptosis mRNA
Fatty Acid Adipogenesis
oli

ER Ets Elk-1 TIF1A FoxO3 CREB ATF1 ER81 SRF c-Fos ERα Nur77 MITF C/EBPβ Translation
Cytosolic
ab

Wee1 p21 Synthesis PPARγ


et

Sequestration
M

Waf1/Cip1 Glycogen e c-Myc/


Histone
os Stat1/3 N-Myc Pax6 c-Fos HMGN1 ETV1 TIF1A ATF4 Myt1 Mad1 YB1 Ribosome Lipogenesis
Synthesis uc H3
Cell Growth Autophagy Proliferation
mTORC1 Cyclin D1 Gl mTORC2 Biogenesis SREBP-1
Migration CDK2 Cyclin A UBF Lipin 1
Raptor Sin1 PRR5 Transcription
GβL
Mig SKAR
mTOR Rictor GβL
rati
DEPTOR on Cell Cycle mTOR Transcription Lipid Synthesis mRNA Splicing
DEPTOR

Proliferation

visit www.cellsignal.com/pathways to see all pathways 9


Enabling
Replicative
Immortality
Cancer cells can revert to a pre-differentiated, stem-cell-like phenotype, allowing uninhibited cellular division and other
metabolic adaptations that enable survival in adverse conditions.
While there are multiple signaling pathways involved in these changes, two key components enable replicative immortality,
Hippo and WNT. There are multiple pathways involved in this characteristic of cancer cells. Below, we’ll focus on two of
these key pathways: Hippo signaling and Wnt signaling.
Hippo signaling is an evolutionarily conserved pathway that controls organ size by regulating cell proliferation, apoptosis,
and stem cell self-renewal. In addition, dysregulation of the Hippo pathway contributes to cancer development. Important
targets include:
• Y AP and TAZ are key mediators to Hippo signaling. YAP acts as a transcriptional co-activator, can be phosphorylated
at multiple sites and translocates from the nucleus to the cytoplasm
• W
 hen the Hippo pathway is turned off, YAP is phosphorylated, translocates to the nucleus and is associated with
various transcription factors, including the TEAD YAP/TEAD complexes regulate the expression of genes involved in
cell proliferation and apoptosis.
The Wnt/β-Catenin pathway is another evolutionarily conserved mechanism that contributed to cancer’s ability to replicate
indefinitely. This pathway regulates stem cell pluripotency and cell fate decisions during development. Wnt signaling has
also been shown to promote nuclear accumulation of transcriptional regulators implicated in cancer, such as TAZ. Key
regulators include:
• β -Catenin is a key downstream effector in the Wnt signaling pathway. It is implicated in two major biological
processes in vertebrates: early embryonic development and tumorigenesis. It can translocate to the nucleus.
• L EF1 and TCF bind to Wnt response elements to provide docking sites for β-catenin, which translocates to the
nucleus to promote the transcription of target genes upon activation of Wnt signaling.

References:
1. Hanahan D, Weinberg RA (January 2000). “The Hallmarks of Cancer”. Cell. 100 (1): 57–70. doi:10.1016/S0092-8674(00)81683-9
2. 2 Hanahan D, Weinberg RA (March 2011). “Hallmarks of Cancer: the next generation”. Cell. 144 (5):646-74. doi: 10.1016/j.cell.2011.02.013.

Hippo Signaling Wnt Signaling


GPCR Signaling Mechanical Signaling OFF-State ON-State

Porc WIF R-spondins


CD44

FAT4

?
Crb

WLS SFRP LGR5/6


DKKs SFRP
Tight Junction

Wnt
Cadherin
RNF43
? ?
LRP5/6

ZO-2 YAP/TAZ
LRP5/6

Frizzled ZNRF3
Frizzled
AMOT Gα12/13 Gαs p120
FRMD KIBRA
YAP/TAZ Merlin CK1 GSK-3
β-catenin Dvl
Axin Naked
Gαq/11 α-catenin

Gαi/o Tankyrase ub
TAOK-1-3 Dvl
CYLD
RASSF
SIK1-3
Adherens Junction

PP2A

PP2A PAR-1
α-catenin Mst1/2 LKB1 Axin
SAV1 Skp APC GSK-3 β-catenin ub TAB2
14-3-3 MARK4
WTX β-catenin TAK1
β-TrCP
CK1 TAB1
YAP Merlin Rho

Ajuba
Rac1
MOB1 F-actin
ub β-catenin NLK
AMOT LATS1/2
Proteasomal
Degradation
ITCH

CK1 AMOT
14-3-3
YAP/TAZ
YAP PP1
oplasm
Cyt
ASPP2 ICAT
β-TrCP
leus Chibby
Nuc
HDAC Pygo
BCL9 Brg1 NLK TAZ Snail1
Groucho
β-catenin
OFF CBP Target Genes: Migration
Cy TCF3 Myc, Cyclin D1,
to pla LEF1/TCF1
WBP2 s m TCF-1, PPAR-δ, Adhesion
Nu MMP-7, Axin-2,
VGL4 YAP/TAZ YAP/TAZ YAP/TAZ YAP/TAZ cle
CD44, etc.
us HDAC

TEAD1-4 TEAD1-4 Smad p73 RUNX Transcription Groucho β-catenin β-catenin


Hesx1 Pit1
FoxO FoxO

10
Evading Growth
Suppressors
Cancer cells resist inhibitory signals that might otherwise stop their growth. The major pathways involved are Autophagy
and Death Receptor Signaling (Apoptosis), both of which can ultimately lead to cell death, and reduction in tumor growth.
Autophagy is a dynamic cellular recycling system that results in the degradation of cytoplasmic contents, abnormal protein
aggregates, and excess or damaged organelles so that the building blocks (e.g., amino acids) can be used to create new
cellular components. Therefore, autophagy is a survival-promoting process. Cancers can upregulate autophagy to survive
microenvironmental stress and to increase growth and aggressiveness. The autophagy process involves the formation of
an autophagosome, which then fuses with lysosomes to form an autophagolysosome. The process is regulated by mTOR/
AMPK/PI3K/MAPK pathways.
Proteins of Interest:
• A tg16L1 plays a key role in helping to form a huge complex that then is needed to form the autophagosomes, the
structures that deliver cytoplasmic components to the lysosomes
• S equestosome 1 (SQSTM1, p62) is a ubiquitin binding protein that interacts with ubiquitin and provides a scaffold
for several signaling proteins. It triggers degradation of proteins through the proteasome or lysosome
• A utophagy marker Light Chain 3 (LC3) becomes associated with autophagic vesicles. The presence of LC3 in
autophagosomes and the conversion of LC3 to LC3-II have been used as indicators of autophagy
• Atg1/ULK1 can act as a convergence point for multiple signals that control autophagy
Apoptosis (programmed cell death) can be induced through the activation of death receptors.
Proteins of Interest:
• C
 aspase-8: Apoptosis is induced through several receptors which activate caspase-8 and lead to the release of the
caspase-8 active fragments, which then cleave and activate downstream caspases.
• R IP kinases are important regulators of cellular stress that trigger pro-survival and inflammatory responses through
the activation of NF-κB, as well as pro-apoptotic pathways
• B cl-2 exerts a survival function in response to a wide range of apoptotic stimuli through inhibition of mitochondrial
cytochrome c release

References:
1. Hanahan D, Weinberg RA (January 2000). “The Hallmarks of Cancer”. Cell. 100 (1): 57–70. doi:10.1016/S0092-8674(00)81683-9
2. 2 Hanahan D, Weinberg RA (March 2011). “Hallmarks of Cancer: the next generation”. Cell. 144 (5):646-74. doi: 10.1016/j.cell.2011.02.013.

visit www.cellsignal.com/pathways to see all pathways 11


Avoiding Immune
Destruction
Some cancer cells adapt mechanisms to evade detection and destruction by the host’s immune system. One way
cells do this is by hijacking normal mechanisms of immune checkpoint control and modulation of the innate immune
response via STING.
Immune checkpoints refer to the built-in control mechanisms of the immune system that maintain self-
tolerance and help to avoid collateral damage during a physiological immune response. It is now evident that
tumors engineer microenvironments to evade immune surveillance and attack, particularly by modulating
certain immune-checkpoint pathways.
Tumor-specific T cells must discriminate between destruction of the tumor cell and survival of the target cell. Important
for discrimination are proteins on both the T-cell and the target cell:
• C
 D8 is a T cell and the coreceptor for the T cell receptor (TCR). These two distinct structures recognize the
Antigen–Major Histocompatibility Complex (MHC). In tumors, T cells are also known as tumor infiltrating
lymphocytes (TIL), present in high abundance at the tumor site to influence overall survival. During cancer
therapy, TILs are sometimes removed from a patient’s tumor and are then treated with substances that activate
the lymphocytes to help them better kill the patient’s cancer cells.
• P D-L1 and PD-L2 (programmed cell death proteins) are transmembrane proteins that suppress the adaptive arm
of the immune system during particular events such as pregnancy, tissue allografts, autoimmune disease and
other disease states like hepatitis. PD-1 on the T-cell is activated by the cell surface ligands on the tumor cell.
Upregulation of PD-L1 may allow cancers to evade the host immune system.
• T IM-3 is an inhibitory molecule that is induced following T cell activation. As a negative regulatory immune
checkpoint, it is detected in different types of immune cells, including T cells, regulatory T cells (Tregs), dendritic
cells (DCs), B cells, macrophages, nature killer (NK) cells, and mast cells. TIM-3 inhibits antitumor immunity by
mediating T-cell exhaustion.
STING (stimulator of interferon genes) is a key mediator of innate immunity, and the STING pathway has been shown
to be involved in the induction of an anti-tumor immune response. STING is responsible for regulation of type-I
interferon production and cellular defense against intracellular pathogens (like bacteria or viruses). Key regulators of
the STING pathway are:
• Interferon regulatory factors (IRFs) comprise a family of transcription factors that function within the Jak/Stat
pathway to regulate interferon (IFN) and IFN-inducible gene expression in response to viral infection
• T ING is a signaling molecule associated with the endoplasmic reticulum (ER) and is essential for controlling the
transcription of numerous host defense genes (including type I interferons (IFNs) and pro-inflammatory cytokines)
following the recognition of aberrant DNA species or cyclic dinucleotides in the cytosol of the cell. Sting can
translocate out of the ER upon activation.

12
Learn more about the pathways and proteins involved in Avoiding Immune Destruction:
• Immune Checkpoints

• STING

References:
1. Hanahan D, Weinberg RA (January 2000). “The Hallmarks of Cancer”. Cell. 100 (1): 57–70. doi:10.1016/S0092-8674(00)81683-9
2. Hanahan D, Weinberg RA (March 2011). “Hallmarks of Cancer: the next generation”. Cell. 144 (5):646-74. doi: 10.1016/j.cell.2011.02.013.
3. Yayi He, et al. “TIM-3, a promising target for cancer immunotherapy”. Onco Targets Ther. 2018; 11: 7005–7009.
4. Barber, Glen N., “STING: infection, inflammation and cancer”. Nat Rev Immunol. 2015 Dec; 15(12): 760–770.

Immune Checkpoints STING


Cell Intrinsic Innate Immunity Signaling

Growth Factor
Receptor Activation
Akt RNA Virus Extracellular Space
TGF-β DNA Pathogen ds DNA
Replicative Damage Host ds DNA
Senescence
FoxO1/3
Cytoplasm
Differentiation ATM/
ATR RNA
Pol III
Hormones
Smad3
OAS ds DNA TREX1
BMI1 Myc ds RNA
Smad4 p53
ATP
Ultra Violet Replicative ds RNA
Senescence LGP2 MDA-5 RIG-I RNaseL
Stress Response
LGP2 RNase L 2-5A Bacterial
p21 Cip1 Chk1/2 MAVS CDNs
p19 INK4D p18 INK4C p16 INK4A p15 INK4B TRIM25
Ub
MAVS RNAseL
Myc Degradation RNase L C-di
Growth p27 Kip1 Ubiquitination Products
Ub
GMP
Ub TRIM21
Factor
Withdrawal TRAF3 GTP cGAS C-di
and ATP AMP
DDX41
Myc Myc Ub
cdc25A TRAF3 Mitochondria
GSK-3β MAVS Ub
DAI IFI16
DUBA TANK C-
SCF NAP1 SINTBAD MAVS GAMP
DDX41

SCF MAVS
CDK4/6 CDK2
Ubiquitination Cyclin D Cyclin E Ubiquitination
NLRX1
R RNF5
SE S-PH
PH A
Ub
ASE TRAF2/5/6
G1 -
NEMO TANK Ub
Ub
ULK1
IRF3 TBK1 TBK1 TRAF2/5/6
Ub STING TRIM32

Suv39H1 P
NEMO STING TRIM56
Abl HDAC Rb E2F/DP Target Genes: TBK1 STING
Rb Cyclin E/A, E2F-1/2/3,
OTUB1/2 27 Ub
Ub 63
IKKß
FoxO1 Bim cdc2, c-Myc, p107,
IKKα IRF3 Atg9α Endoplasmic
E2F E2F Reticulum
FasL RanGAP, TK, DHFR,
DBE Apoptosis DP-1 DP-1
TRAIL OFF ON PCNA, H2A, etc. Ub AMFR INSIG1
IκB STING
NF-κB P P
IRF3 IRF3
P P NF-κB
IRF3 IRF3

Nucleus

P P

IRF3 TYPE 1 IRF3


IFN NF-κBIRF3 TYPE 1
IFN
ISRE ISRE

rev. 06/13/18

“It is now evident that tumors


engineer microenvironments
to evade immune surveillance
and attack....”

visit www.cellsignal.com/pathways to see all pathways 13


Genome Instability
and Mutation
Not all cancer cells are equal, they evolve in response to selective pressure driven by accumulation of mutations.
Cancer cells have to out-compete nearby cells for nutrients and other resources, avoid immune cell attack, and
suppress apoptotic self-destruction.
Due to the aberrant proliferation associated with cancer cells, there is an increased tendency of genomic changes
and mutations that contribute to the damage of multiple genes regulating cell division and tumor suppression. This
is known as genomic instability. Genomic instability has the tendency to compound in cancer cells, since survival-
enhancing mutations increase the probability that those mutations will propagate in future cells.
A mutation is an alteration of an organism’s DNA sequence. The nucleotides that compose our DNA can be added,
replaced, or deleted, and single- or double-stranded breaks can occur within the DNA strand. Complete sections of
DNA can also swap positions, be inadvertently replicated, or deleted. Most of these mutations are not cancer-related.
They can either be spontaneous or the result of environmental insults like chemicals and radiation. Despite the high
probability that such mutations can occur, our DNA is maintained relatively error-free. Our genome surveillance and
maintenance systems, mitotic checkpoints and DNA repair mechanisms are always working to mitigate common
daily factors that attempt to mutate our genetic code. A defect in any of these systems can increase the DNA’s
susceptibility to mutations, resulting in genomic instability and an increased risk of malignancy.
 ne such mechanism is the G2/M DNA damage checkpoint, which serves to prevent the cell from entering mitosis
O
(M-phase) with genomic DNA damage, facilitating genome surveillance and DNA repair. There are several key
proteins involved:
• D
 NA-dependent protein kinase (DNA-PK), a serine/threonine kinase complex composed of a heterodimer of
Ku proteins (Ku70/Ku80) and the catalytic subunit DNA-PKcs, is deployed to the site of double-stranded DNA
breaks almost instantly to initiate repair via non-homologous end joining.
• B RCA1 and BRCA2, two tumor suppressors that are found in breast and other tissue, contribute to DNA repair,
chromosomal stability and transcriptional regulation in response to DNA damage. Studies have shown that,
in response to DNA damage, BRCA1 is hyperphosphorylated and translocated to specific sites within the
replication fork. BRCA1 has also been shown to regulate the expression levels of several genes activated in
response to DNA damage. In addition, BRCA1 is required for the S-phase and G2/M-phase mitotic checkpoints.
BRCA2 plays a slightly different role than BRCA1 and is predominantly active in maintaining chromosomal
stability and mitotic recombination. Both BRCA1 and BRCA2 have been shown to repair double-stranded DNA
breaks via homologous recombination.
• C
 hk1 and Chk2 are key signaling transducers that are part of a complex network of gene integrity checkpoints,
damage detectors and tumor suppressors.
• p 53 is also known as the “guardian of the genome” for its role in conserving genomic stability. p53 plays a
central role in in a pathway that recognizes and mitigates oncogenic stress by halting proliferation and inducing
apoptosis/senescence in an attempt to allay accumulating DNA damage that could lead to malignancy. (Fun
fact, elephants have over 20 copies of the p53 gene to protect them from mutations.)
14

You might also like