Cancer Genetics and Biology

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

CHAPTER 4

Cancer Genetics and Biology


THERESA V. STRONG, PhD

OVERVIEW

Cancer development is a multistep process in which cells must acquire certain characteristics to circumvent the inherent
controls on abnormal cell proliferation and expansion. This chapter will review factors contributing to the initiation of
tumor development, as well as the biologic hallmarks necessary for cancer cells to reach a fully malignant state.

GENETICS OF TUMOR INITIATION AND Next-generation sequencing methods have allowed


PROGRESSION—DEFINING THE GENOMIC the numerous genetic changes that accompany transfor-
LANDSCAPE OF PEDIATRIC TUMORS mation to be delineated in greater detail. Point muta-
Cancer arises from a progressive accumulation of tions, copy number alterations (amplifications and
mutations and genomic alterations in normal tis- deletions), chromosomal rearrangements, loss or gain of
sue progenitor and stem cells, resulting in malig- entire chromosomes (aneuploidy), extrachromosomal
nant transformation. Both intrinsic and extrinsic DNA, and epigenetic changes can all contribute to the
factors contribute to the accumulation of mutations altered genetic makeup of cancer cells. Fully defining
and genomic changes. Mutations occur as a natural the molecular landscape of pediatric tumors is impor-
consequence of repeated cell division—although tant not only for advancing the understanding of cancer
the DNA is replicated with high fidelity, given the development and progression, but also for improving
size of the genome and the number of cell divisions the precision of diagnosis, prognosis, and treatment.
necessary for organ development and maintenance, The Cancer Genome Atlas (TCGA)3 is a multiyear col-
random errors are inevitable. The highly repetitive laborative project supported by the National Institutes
nature of many sequences in the human genome also of Health to understand the genomic changes in more
contributes to errors. “Slippage” during DNA synthe- than 30 types of cancer. This project has generated a tre-
sis may introduce small insertions and deletions, mendous amount of data and has revealed characteristic
and amplifications, deletions, and translocations can differences in genomic changes across tumor types and
be promoted by short stretches of homology found subclasses. Although some cancers found in the pediat-
across the genome. Normal cellular processes con- ric population are represented in this effort, most of the
tribute to the mutation load as well, because the cancers selected for TCGA analysis are predominantly
natural by-products of metabolism, such as reactive found in adult populations. To complement these efforts,
oxygen species, can cause DNA damage. Extrinsic a pediatric-specific genome project has been developed:
factors also play an important role in cancer devel- The St. Jude-Washington University Pediatric Cancer
opment and include exposure to radiation, carcino- Genome Project.4,5 This effort is not only defining the
gens, and other environmental factors that increase genomic landscape for common pediatric cancers, such
cancer risk over the lifetime of an individual. Finally, as lymphoblastic leukemias, medulloblastoma, Ewing
while the majority of cancers, including pediatric sarcoma, and neuroblastoma, but also investigating the
cancers, are the result of acquired mutations, patho- underlying genetic basis of typically adult tumors, such
genic germline mutations greatly increase cancer as melanoma, which sometime occur in children and
risk and are estimated to underlie ∼10% of pediatric adolescents. The last 5 years have generated a wealth of
cases, disproportionately contributing to adrenocor- knowledge in this regard, providing insight into the ori-
tical tumors, osteosarcoma, and acute lymphocytic gins, progression, and recurrence of both rare and com-
leukemia.1,2 mon pediatric cancers.4–6

41
42 Pediatric Cancer Genetics

Although a review of the findings to date of the subclones poses a challenging problem.8 Despite these
Pediatric Cancer Genome Project is beyond the scope challenges, however, genomics-driven precision medi-
of this chapter, it is apparent that the well-described cine has already taken a foothold in the management
mechanisms of adult cancer initiation (e.g., point of pediatric cancers and holds great promise for the
mutations in oncogenes and tumor suppressors, ampli- future.9 
fications and deletions, aneuploidy, rearrangements/
translocations, and epigenetic changes) are all at play
to a greater or lesser degree across the different pediatric BIOLOGIC HALLMARKS OF CANCER
cancers. Furthermore, the molecular characteristics and DEVELOPMENT
the underlying genomic alterations of pediatric cancers During progression, cancer cells must acquire certain
correlate with their cell of origin. Among the changes capabilities to ensure survival and continued expan-
found at the nucleotide level, point mutations in proto- sion. Six key “hallmarks” of cancer development have
oncogenes render normal genes capable of promoting been described by Hanahan and Weinberg.10,11 These
aberrant and continued proliferation, and mutations hallmarks include sustaining proliferative signal-
in known oncogenes such as NRAS, KRAS, and BRAF ing, evading growth suppressors, resisting cell death,
are among the most frequent changes found in pedi- enabling replicative immortality, inducing angiogen-
atric cancer. Similarly, mutations in tumor suppressor esis, and activating invasion and metastasis. Each of
genes, whose normal role is to suppress abnormal cel- these traits marks the evolution of cancer through a
lular growth, also constitute an important molecular complex, multistep process. These changes take place
alteration, and mutations in TP53, the most commonly in the presence of normal cells, which are recruited and
mutated gene in all cancers, is also the most common induced to actively support the acquisition of the hall-
mutation found in pediatric cancers. Overall, however, marks. Progress in understanding the contribution of
pediatric cancers have lower rates of point mutations each of these components can suggest new approaches
at diagnosis compared to adult cancers. Chromosome to cancer prevention and treatment.
rearrangements may be particularly relevant for pedi-
atric leukemia, renal cancer, and rhabdomyosarcoma. Sustaining Abnormal Proliferation
These rearrangements frequently result in the genera- A fundamental trait of cancer cells is the ability to
tion of an oncogenic fusion protein or overexpression maintain proliferation. Mutation of normal, growth-
of an oncogene, and they may provide rational tar- promoting protooncogenes represents the most
gets for therapeutic development. Epigenetic changes, straightforward way to achieve this goal. Mutations in
including chemical modification of DNA (e.g., methyl- protooncogenes may render the encoded protein per-
ation and hydroxymethylation) as well as modification manently activated, capable of ignoring the signals that
of the histone proteins around which DNA is packaged, would normally trigger inactivation. Protooncogenes
do not change the actual DNA sequence but do lead to relevant in pediatric cancers include those that play a
stable changes in gene expression that impact tumor role in signal transduction (NRAS, KRAS, BRAF) and
development and progression. Compared with adult transcription (MYC, RUNX1, ETV6), among others.
tumors, pediatric cancers demonstrate a high mutation In addition to activating the oncogenes mentioned
frequency in genes that encode epigenetic regulators, above, additional genetic alterations may enhance
and these changes may be particularly important in and sustain abnormal proliferation. For example, the
high-grade glioma, T cell lymphoblastic leukemia, and aberrant production and release of growth factors or
medulloblastoma.7 changes to growth factor receptor pathways to enhance
In the aggregate, studies of the cancer genome and responsiveness or completely deregulate signaling can
epigenome are providing a clearer picture of the initi­ promote proliferation. These changes may allow pro-
ating events and genetic drivers of pediatric cancers, liferation that is independent of the usual growth fac-
as well as defining the changes that allow progres- tor requirements. For example, acquisition of activating
sively more aggressive clones to develop and expand. mutations and/or amplification and overexpression of
Genome instability is a defining feature of cancer cells, growth factor receptor genes (such as those encoding
and it generates the intratumoral diversity that can the epidermal growth factor [EGF] receptor and fibro-
then expedite the development of features necessary for blast growth factor [FGF] receptor) allow the cell to
tumor progression. Genomic instability and tumor het- become hyperresponsive to otherwise limited growth
erogeneity also complicate the development of effective factor ligand concentrations or to proliferate in the
targeted therapies, as emergence of treatment-resistant absence of the ligand entirely. Modification of normal
CHAPTER 4  Cancer Genetics and Biology 43

negative feedback loops to render them less effective through the cell cycle. The p53 protein has been termed
is another method by which proliferation can be pro- the “guardian of the genome” because it confers sta-
moted and sustained. bility by sensing cellular stress and DNA damage and
Reprogramming cellular metabolism is another halting cell cycle progression until conditions improve.
strategy the cancer cells use to sustain proliferation.12 Alternatively, if faced with extensive genomic damage
This adaptation is necessary to allow cancer cells to or physiologic stress, p53 normally functions to trigger
continue to propagate in a nutrient-poor environ- apoptosis and eliminate the defective cell. Loss or inac-
ment. Tumors markedly increase glucose consumption tivation of p53 represents the most common genetic
in comparison to normal, nonproliferating tissue and alteration in cancer and can impact tumor initiation,
exhibit enhanced uptake of amino acids. Several major progression, and metastasis.
signaling pathways, frequently activated in cancer, In addition to well-characterized tumor suppres-
contribute to this process. Activating mutations in the sors genes, there are several pathways that serve to con-
phosphoinositide 3-kinase (PI3K)/Akt signaling path- trol proliferation and to maintain tissue homeostasis
way serve as a master regulator of glucose uptake and within confined spaces, and the genes involved in these
glycolysis, while activation of MYC, mutation of RAS pathways can serve as noncanonical tumor suppressors.
and SRC alleles, or deletion of the tumor suppressor Chief among these are the signals that maintain cells in
RB1 can enhance amino acid uptake. Beyond changes a quiescent state when they are in contact with other
in nutrient acquisition, tumor cells often decouple cells, known as contact-mediated growth inhibition.
glycolysis from oxidative phosphorylation, switching In differentiated tissues, cells interact and communi-
largely to a glycolytic pathway for carbon utilization. cate with neighboring cells through junctions formed
Although at first pass it may seem counterintuitive by cell surface adhesion molecules such as E-cadherin,
that rapidly dividing malignant cells use a less effi- and this network provides a powerful means of control-
cient pathway for ATP generation, in fact, the glycolytic ling growth and maintaining tissue integrity. Cellular
intermediates generated by this metabolic shift provide polarization is also important for maintaining tissue
a necessary pool of precursors for the diverse biosyn- integrity and attenuating proliferation. These three-
thetic reactions required by proliferating cancer cells.  dimensional cues can override intrinsic cell prolifera-
tion signals and restrict the growth of mutated cells,
Circumventing Growth Suppressors while loss of the molecules that mediate cell-to-cell
In addition to acquiring the genetic changes that serve contact and polarization allows unfettered expansion
to activate and sustain abnormal proliferation, cancer of transformed cells and disruption of normal tissue
cells must be able to evade the signals that normally architecture. 
serve to limit excess cell division. Tumor suppressor
genes encode proteins that act in a variety of ways to Resisting Cell Death
restrict cell growth, allowing progression through the Apoptosis, or programmed cell death, is a normal
cell cycle only when conditions are favorable. Two developmental process to eliminate damaged or
major tumor suppressor genes disrupted in a number unnecessary cells in a controlled manner. A key part of
of tumor types, including pediatric cancers, are the organ development, tissue homeostasis, and immunity,
retinoblastoma (RB1) and TP53 (tumor protein p53) apoptosis serves to eliminate potentially tumorigenic
genes, encoding the RB and p53 proteins, respectively. cells as well. Cells experiencing extensive DNA dam-
RB1 was the first tumor suppressor gene identified age or physiologic stress will trigger the apoptotic pro-
by studying a rare, inherited form of retinoblastoma. cess through a p53-dependent process. Alternatively,
Alfred Knudson noted that inheriting one defective extracellular signals, such as the Fas ligand, can trigger
copy of RB1 led to a vastly increased risk of developing apoptosis in Fas receptor positive cells. To survive and
retinoblastoma. Despite inheriting the defective allele continue to proliferate, cancer cells use a variety of strat-
in all cells, however, only a small number of cells actu- egies to resist or evade apoptosis. The apoptotic process
ally initiated tumors. This observation led Knudson to consists of a cascade of signaling events that culminates
propose that retinoblastoma development required the in the activation of the proteolytic caspase enzymes,
loss of both copies of RB1 in a single cell, with the sec- which advance the controlled demolition of the cell.
ond “hit” coming as a somatic mutation in the normal Dysregulation or disruption of the apoptotic pathway
RB1 gene. The normal function of the RB1 protein is can be achieved through loss of p53, inhibition of pro-
to integrate cellular signals from internal and exter- apoptotic signals, or overexpression of antiapoptotic
nal sources and to assess whether to advance the cell molecules. The BCL-2 family of regulatory proteins
44 Pediatric Cancer Genetics

acts at several points along the apoptotic pathway to Inducing Angiogenesis


either promote or inhibit apoptosis, and it is the inter- The growth of solid tumors, like any tissue mass,
action of BCL-2 proteins with each other and the mito- requires that cells have adequate access to oxygen and
chondria that determines the threshold for induction nutrients and that they are able to clear waste and car-
of apoptosis.13 Overexpression of prosurvival BCL-2 bon dioxide. For tumors to grow beyond approximately
accelerates tumorigenesis and promotes resistance to 1–2 mm3 in size, a blood supply must be developed to
therapy, while inhibition or loss of proapoptotic fam- support metabolic demands. Angiogenesis, the induc-
ily members also enables tumor progression. Targeting tion of new, tumor-associated vasculature, is thus
the regulation or function of the BCL-2 proteins has critical to tumor development. Tumors must be able
emerged as a strategy to enhance the efficacy of conven- to induce an “angiogenic switch” to coax the prolif-
tional cytotoxic agents. eration of normally quiescent vasculature endothelial
Autophagy is another normal cellular process cells. Tumor neovasculature development is a complex
designed to recycle cellular organelles and nutrients process that involves a variety of cell types and signal-
to sustain cellular metabolism and homeostasis. Cells ing pathways. Direct effects on vascular endothelial
maintain a low level of autophagy as a means to remove cells or their bone-marrow derived precursors are key
damaged proteins and organelles and efficiently recycle to the process, and vascular endothelial growth fac-
the nutrients. Initially thought to play primarily a tumor tor A (VEGF-A) is a critical molecule orchestrating the
suppressor role, the process of autophagy can also be development of new blood vessels through its actions
hijacked in cancer cells to promote survival in stressful on these cells. VEGF is upregulated under conditions
conditions and facilitate continued aggressive growth of hypoxia, inflammation, and oncogenic signaling,
where it might not be possible otherwise.14 Autophagy which are commonly found in tumors. Additional pro-
can promote tumor dormancy and may allow cancer angiogenic and antiangiogenic factors secreted by the
cells to survive during treatment, setting the stage for tumor, as well as cancer-associated fibroblasts (CAFs)
an eventual recurrence. A better understanding of how and immune cells, include angiopoietins, EGF, trans-
autophagy serves both tumor-suppressing and tumor- forming growth factors, platelet-derived growth factor,
promoting roles is needed to fully exploit this process to and thrombospondin-1. Under hypoxic conditions,
control tumor initiation, progression, and recurrence.  the balance of proangiogenic and antiangiogenic fac-
tors switches in favor of angiogenesis, and the signals
Enabling Replicative Immortality coordinate to encourage the recruitment, proliferation,
To achieve unlimited replication, cancer cells must and differentiation of endothelial cells into functional
overcome the normal attrition of telomere length vessels. However, vascular development in tumors is
that accompanies division of nonimmortalized cells. far from the precise process in normal development;
Telomeres are composed of a six-base pair nucleotide tumor vasculature is typically disorganized and exhib-
sequence, tandemly repeated hundreds to thousands its poor structural integrity with increased permeabil-
of times, which protects the ends of chromosomes. ity. The density and pattern of neovascularization also
Because of the nature of DNA replication, wherein the shows diversity, with some tumor types characterized
DNA polymerase cannot replicate the very end of the by hypovascularization, while others exhibit dense
chromosomal DNA on the lagging strand, telomeres vascularization. The timing of induction of angiogen-
shorten with every cell division. Shortened telomeres esis can vary in tumor development, with some early,
mediate end-to-end fusions between chromosomes, premalignant lesions already exhibiting robust blood
which reduce cell viability, trigger crisis, and induce supply. Regardless of the density, integrity, and timing,
senescence. The cellular enzyme telomerase uses an the new vasculature provides a venue to deliver the
RNA template to add on the telomere repeat DNA, necessary oxygen and nutrients to cancer cells and also
lengthening telomeres. Although inactive in most nor- provides a means for the next step in the clinical pro-
mal differentiated cells, telomerase is reexpressed in the gression of tumors—metastasis. 
vast majority of tumor cells, where it restores telomere
length to the point where senescence and/or apoptosis Activating Invasion and Metastasis
are averted. Residual end-to-end chromosomal fusions, A number of steps are needed for solid tumors to
persisting from a period before telomerase activation, invade and disseminate beyond the primary tumor
may be carried in neoplastic cells and may evolve into mass.15 Cancer cells from solid tumors must be able to
chromosome deletions or amplifications as cancer survive without cell-to-cell contact, acquire migratory
development progresses.  ability, break through the basement membrane, enter
CHAPTER 4  Cancer Genetics and Biology 45

the bloodstream, transit through blood and lymphatic stimulate normal, resident cells to release growth fac-
vessels, adhere to vessel walls at a distant site, penetrate tors that, in turn, stimulate cancer cell growth, recruit
through the vessel in a process termed “extravasation,” immunosuppressive cells, and promote angiogenesis.
and finally, begin to divide and colonize the distant CAFs, for example, are an important source of inflam-
site, establishing a secondary tumor. To successfully matory cytokines that drive angiogenesis and immune
perform this series of steps, cancer cells undergo a cell recruitment to the tumor. The composition of the
major shift in gene expression toward a developmental ECM also impacts this process by influencing the bio-
regulatory program, termed “epithelial to mesenchy- availability of growth factors. These factors are normally
mal transition” (EMT). Activation of an EMT transcrip- sequestered in pericellular spaces and the ECM, but an
tional program through a set of transcriptional factors, array of proteases, sulfatastes, and other enzymes can
including Snail, Slug, and Twist, results in loss of E-cad- liberate and activate the growth factors, promoting pro-
herin expression to allow the tumor cell to detach from liferation and invasion. Tumor cells themselves secrete
its neighbors and the surrounding extracellular matrix matrix-degrading proteases, but these can also be sup-
(ECM). The trascriptional changes also direct changes plied by stromal cells and infiltrating immune cells
in morphology to a more spindle-like morphology, and such as tumor-associated macrophages (TAMs).
increased motility to facilitate transit. Upregulation of Metabolic changes in the tumor contribute to the
expression of alternative adherens molecules, such as perturbations in the microenvironment that encour-
N-cadherin, is consistent with an increased migratory age tumor growth, promote angiogenesis, and suppress
capacity, reminiscent of embryonic development. Addi- the action of immune cells. Increased extracellular lac-
tionally, cells undergoing EMT secrete proteases that tate and subsequent acidification of extracellular space
degrade the basement membrane and allow access to attenuates the function of antigen presenting dendritic
the bloodstream. Once residing at the distant metastatic cells (DCs) and cytotoxic T cells, while polarizing
site, tumor cells may revert back to a less invasive state infiltrating macrophages to a tumor-promoting, M2
through the reverse, mesenchymal to epithelial transi- phenotype. Overexpression of tryptophan-degrading
tion. Although the EMT reprogramming is perhaps the dioxygenases indoleamine2,3-dioxygenase (IDO1 and
most well-described method of achieving metastatic 2) by cancer cells depletes tryptophan from the micro-
spread, other programs of invasion are apparent in environment and promotes apoptosis of effector T
nonepithelial tumor types. For example, gliomas rarely cells. Even the way cancer cells die can impact the TME.
escape the brain but have a highly invasive phenotype, Cell death by necrosis is common as tumors develop,
invading normal brain tissue by following vascular and and it contributes to continued tumor growth by releas-
nerve tracks. These cells commandeer chloride and ing proinflammatory signals into the TME, recruiting
potassium ion channels to drastically alter their cell the inflammatory cells and setting up an environment
volume and navigate the narrow spaces of the brain. It that fosters angiogenesis and cancer cell invasion.
is likely that additional, alternative strategies of tumor A critical and dynamic component of the TME is
spreading are routinely used by cancer cells, and a bet- cells of the immune system. The interaction of can-
ter understanding of all of the possible routes of metas- cer cells with the host immune system is a complex,
tasis will suggest new approaches to intervention.  ever-evolving process. Tumors are infiltrated by a broad
array of immune cells, including TAMs, monocytes,
myeloid-derived suppressor cells, DCs, neutrophils,
THE ROLE OF THE TUMOR and lymphocytes such as T-helper (Th) (CD4+) cells,
MICROENVIRONMENT cytotoxic T cells (CD8+), and T regulatory (Treg) cells.
As has been noted, tumors are not composed purely Although some of these cells serve to eliminate tumor
of cancerous cells but rather represent a complex tissue cells (e.g., DC, Th, CD8+ T cells), overall these cells are
that incorporates noncancerous stromal and immune protumorigenic and may release inflammatory cyto-
cells, which play important supporting and tumor-pro- kines (TAMs) or immunosuppressive cytokines (Treg)
moting roles. The tumor microenvironment (TME) is that impede the induction of effective cytotoxic T cell
composed of CAFs, vascular endothelial cells, pericytes, responses. Infiltrating immune cells (neutrophils,
and innate and adaptive immune cells, as well as their TAMs) also induce inflammation, promote angiogen-
associated molecules. The TME can act as a critical regu- esis, and support the expansion of stem cell pools.
lator of cancer progression and metastasis, and cancer Tumors actively evade immune destruction through a
cells actively reprogram their microenvironment to number of strategies, and a “successful” tumor needs
support these processes.16 Tumors release signals that to tip the balance of the immune system in favor of
46 Pediatric Cancer Genetics

a permissive environment. Tumor cells secrete fac- knowledge gained will allow the selection of tailored,
tors that enhance the function of tumor-promoting targeted treatments based on genomic profile, rather
innate immune cells (e.g., stimulation of macrophages than tissue of origin, toward the goal of treating cancer
through secretion of colony stimulating factor 1) and with reduced toxicity and improved survival and qual-
suppress the function of antigen presenting cells (DC) ity of life.
and effector T cells (e.g., by secretion of the cytokine
IL-10). As cancer cells acquire more mutations and
express neoantigens that may be recognized by T cells, REFERENCES
a process of “immunoediting” occurs, whereby cells 1. Parsons DW, Roy A, Yang Y, et al. Diagnostic yield of clini-
selectively downregulate expression of immunogenic cal tumor and germline whole-exome sequencing for chil-
proteins. Tumor cells can also evade destruction by dren with solid tumors. JAMA Oncol. 2016;2:616–624.
2. Zhang J, Walsh MF, Wu G, et al. Germline mutations in
activated T cells by downregulating expression of anti-
predisposition genes in pediatric cancer. N Engl J Med.
gen presenting HLA-1 molecules, effectively “hiding”
2015;373:2336–2346.
from these effector cells. Alternatively, or in addition, 3. The Cancer Genome Atlas. https://cancergenome.nih.gov.
cancer cells may express inhibitory ligands, such as pro- 4. Downing JR, Wilson RK, Zhang J, et al. The Pediatric Can-
grammed cell death ligand 1 (PD-L1), which engages T cer Genome Project. Nat Genet. 2012;44:619–622.
cells through the PD-1 receptor to inhibit T cell prolif- 5. The Pediatric Cancer Genome Project. https://www.stjude.
eration, survival, and effector functions. org/research/pediatric-cancer-genome-project.html.
As complex as the interactions are, the TME offers 6. Childhood Cancer Genomics (PDQ®) Health Profession-
new opportunities for therapeutic target development al Version PDQ Pediatric Treatment Editorial Board. https:
that are being exploited. For example, after years of //www.ncbi.nlm.nih.gov/books/NBK374260/.
7. Huether R, Dong L, Chen X, et al. The landscape of so-
investigating the fine details of the cancer-immune
matic mutations in epigenetic regulators across 1,000 pae-
cells interaction, strategies to modulation of the effec-
diatric cancer genomes. Nat Commun. 2014;5:3630.
tor arm of the immune system are showing some 8. Morrissy AS, Garzia L, Shih DJ, et al. Divergent clonal
encouraging effects and are entering the forefront of selection dominates medulloblastoma at recurrence. Na-
cancer therapies.17 These approaches include treat- ture. 2016;529:351–357.
ment with molecules targeting immune checkpoints, 9. Mody RJ, Presner JR, Evertt J, et al. Precision medicine in
which normally restrict effector cell proliferation and pediatric oncology: lessons learned and next steps. Pediatr
function. For example, inhibition of the PD-1/PD-L1 Blood Cancer. 2017;64:e26288.
interaction allows propagation of T cell stimulation, 10. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell.
resulting in a robust T cell activation and expan- 2000;100:57–70.
11. Hanahan D, Weinberg RA. Hallmarks of cancer: the next
sion, and more effective tumor cell killing. Addition-
generation. Cell. 2011;144:646–674.
ally, autologous T cells genetically modified to target
12. Pavlova NN, Thompson CB. The emerging hallmarks of
tumor-associated antigens (chimeric antigen receptor cancer metabolism. Cell Metab. 2016;23:27–47.
T (CAR-T) cells) have shown impressive results in 13. Czabotar PE, Lessene G, Strasser A, Adams JM. Con-
early stage clinical trials and suggest the potential of trol of apoptosis by the BCL-2 protein family: implica-
immune-based therapies for dramatically impacting tions for physiology and therapy. Nat Rev Mol Cell Biol.
patient survival.  2014;15:49–63.
14. White E. The role of autophagy in cancer. J Clin Invest.
2015;125:42–46.
FUTURE DIRECTIONS 15. Talmadge JE, Fidler IJ. AACR centennial series: the biol-
ogy of cancer metastasis: historical perspective. Cancer Res.
As genome sequencing and other “omics” technologies
2010;70:5649–5669.
continue to mature and be broadly applied to pediat-
16. Hanahan D, Coussens LM. Accessories to the crime: func-
ric cancers, the underlying genetic features and path- tions of cells recruited to the tumor microenvironment.
way alterations driving pediatric cancer development Cancer Cell. 2012;21:309–322.
will be fully appreciated. These insights will continue 17. Whiteside TL, Demaria S, Rodriguez-Ruiz ME, et al.
to refine our understanding of the complex processes Emerging opportunities and challenges in cancer immu-
that comprise the hallmarks of cancer. Ultimately, the notherapy. Clin Cancer Res. 2016;22:1845–1855.

You might also like