Vous êtes sur la page 1sur 9

[Downloaded free from http://www.jofs.in on Friday, January 5, 2018, IP: 157.48.122.

209]

Review Article

A Review on Tumor Immunology

Abstract Sri Lalitha Kaja,


The ability of immune system to detect and destroy the altered/abnormal cells may inhibit the S. V. N. Sasi Kiran,
development of various cancers. The immune system had been proposed as a tactic in sustaining Kiran Kumar
nonneoplastic state and also for the development of immunotherapy against cancer. Although the
immune system exerts a protective role, under certain circumstances, it could be damaging in terms Kattapagari,
of modulating the oncogenic process. The failure of host’s immunological responses against tumor Ravi Teja Chitturi,
growth and dissemination implicated that both immunologic and nonimmunologic factors may work S. Deepika
together to affect tumorigenesis. Hence, understanding the aspects pertaining to tumor immunology Chowdary,
which deals with the complex interactions between the host’s immune system and neoplasm Baddam Venkata
is essential. The current review focuses on the aspects concerned with tumor immunology, steps
involved, and cancer immunotherapy as a probable therapeutic tool. Ramana Reddy
Department of Oral Pathology
Keywords: Cancer, immune escape, immunosurveillance, immunoediting, immunology, tumor and Microbiology, SIBAR
Institute of Dental Sciences,
Takkellapadu, Guntur,
Introduction Tumors progress even in immunologically Andhra Pradesh, India
competent hosts, as they manage to escape
Tumor immunology is the “study of the
from the immune system. The escape process
complex interaction between a human host
is attributed to the inability of immune cells
and a neoplasm, which unless adequately
to develop an effective antitumor response or
treated causes the death of the host.”
to the inhibition of functions of the immune
Frequent failure of the immunologic
system by tumor‑derived factors.[3]
responses of the host to restrict tumor
growth and dissemination led to the The current review focuses on the aspects
realization that both immunologic and concerned with tumor immunology.
nonimmunologic factors may act in concert Tumor immunology includes:
to affect tumorigenesis. Numerous studies • Immunosurveillance
have focused on the immune system as • Immunoediting
a tactic in sustaining nonneoplastic state • Immune evasion.
and also for the possibility of developing
cancer immunotherapy. Although under Cancer Immunosurveillance
some circumstances, immune system exerts
The three primary roles of the immune
a protective role; under other conditions, it
system in the prevention of tumors are as
could be damaging or irrelevant in terms of
follows:
modulating the oncogenic process.[1]
• First, the immune system by eliminating Address for correspondence:
Paul Ehrlich was the first one to propose or suppressing viral infections, protects Dr. Sri Lalitha Kaja,
that “the immune system could repress the host from tumors induced by viruses Department of Oral Pathology
and Microbiology, SIBAR
a potentially overwhelming frequency of • Second, by the elimination of pathogens Institute of Dental Sciences,
carcinomas” which was found to be a key for and prompt resolution of inflammation, Takkellapadu, Guntur ‑ 522 509,
the cancer immunosurveillance hypothesis. it prevents the establishment of an Andhra Pradesh, India.
Despite tumor immunosurveillance, inflammatory environment that is E‑mail: srilalitha_kaza@yahoo.
com
development of tumors can occur even in conducive to tumorigenesis
the presence of functioning immune system, • Third, the immune system could
and hence an updated concept of tumor specifically identify and eliminate tumor Access this article online
immunoediting was proposed which gives cells by the expression of tumor‑specific
Website: www.jofs.in
a more complex explanation for the role of antigens or molecules that are induced
DOI: 10.4103/0975-8844.207949
immune system in tumor development.[2] by cellular stress.
Quick Response Code:

This is an open access article distributed under the terms of the


Creative Commons Attribution-NonCommercial-ShareAlike 3.0
License, which allows others to remix, tweak, and build upon the How to cite this article: Kaja SL, Kiran SS,
work non-commercially, as long as the author is credited and the Kattapagari KK, Chitturi RT, Chowdary SD,
new creations are licensed under the identical terms. Reddy BV. A review on tumor immunology. J Orofac
For reprints contact: reprints@medknow.com Sci 2017;9:7-15.

© 2017 Journal of Orofacial Sciences | Published by Wolters Kluwer - Medknow 7


[Downloaded free from http://www.jofs.in on Friday, January 5, 2018, IP: 157.48.122.209]

Kaja, et al.: Tumor immunology

The third process by which the immune system identifies cancer.[2] Immunosurveillance could often fail due to the
cancer or precancer cells and eliminates them before they following factors:
can harm is known as “tumor immunosurveillance.”[4] • Inherited selective defects of the immune response that
are mediated directly by the genes or through various
In the 1950s, Medawar et al. implicated that cellular
mechanisms resulting in low threshold tolerance
components of the immune system have a critical role in
• Absence of tumor‑associated antigens (TAA)
allograft rejection. He demonstrated that immunized mice
• Shielding of TAA.[10]
against synergistic tumor transplants induced by viruses,
chemical carcinogens, or other means established the Cancer Immunoediting
presence of “tumor‑specific antigens.” This supports the
cancer immunosurveillance hypothesis.[2] Despite tumor immunosurveillance, tumors develop in the
presence of functioning immune system, and hence an
These emerging discoveries led to the formal hypothesis of updated concept of tumor immunoediting was proposed
cancer immunosurveillance by Sir Macfarlane Burnet and which gives a more complex explanation for the role of
Lewis Thomas in 1957. He stated that “it is by no means immune system in tumor development.[3] Recent works
inconceivable that small accumulations of tumor cells may suggest that immune system might promote the emergence
develop and because of their possession of new antigenic of primary tumors by reducing immunogenicity that are
potentialities provokes an effective immunological reaction with capable of escaping immune recognition and destruction.[11]
regression of the tumor and no clinical hint of its existence.”[5] Cancer immunoediting describes both the host protecting
Thomas suggested that primary function of cellular and tumor sculpting actions of the immune system that
immunity was not to promote allograft rejection but indeed not only prevents but also shapes the neoplastic disease.
to protect against neoplastic disease and maintenance of Cancer immunoediting can promote:
tissue homeostasis.[6] After these speculations, Burnet in • Complete elimination of some tumors
1970 redefined the immunosurveillance concept as “In • Generates a nonprotective immune state to some tumors
large, long‑lived animals, like most of the warm‑blooded • Favors the development of immunologic anergy,
vertebrates, inheritable genetic changes must be common tolerance, or indifference.[2]
in somatic cells and a proportion of these changes will Evidence for cancer immunoediting in humans was
represent a step towards malignancy. It is an evolutionary supported by the number of clinical observations such as:
necessity that there should be some mechanism for • Increased risk of developing tumors in
eliminating or inactivating such potentially dangerous immunosuppressed patients
mutant cells and it is postulated that this mechanism is of • Instances of spontaneous tumor regression
immunological character.” Burnet and Thomas postulated • Presence of tumor‑reactive T‑lymphocytes and
that lymphocytes acted as sentinels in recognition and B‑lymphocytes with respect to improved prognosis.[12]
elimination of continuously arising transformed cells.[7,8]
The ability of cancer immunoediting to control and shape
The immunosurveillance theory of Burnet was based on the cancer is the result of three phases which are called as the
capability of the immune system in distinguishing between three E’s of cancer immunoediting. The phases of cancer
self and nonself. Tumors induced by virally encoded immunoediting are:[13]
proteins are foreign. They are potentially immunogenic • Elimination
targets for the immune system that can readily identify • Equilibrium
them as “nonself.” However, whether the cancer cells of • Escape.
nonviral origin are recognized as self or nonself by the
immune system was questionable. It was thought that Elimination represents the concept of immunosurveillance.
transformation into a cancer cell would change from self to Equilibrium refers to the period of immune‑mediated
nonself that is recognizable by the immune system.[8] latency after an incomplete abolition of tumor in the
elimination phase. Escape is the terminal outgrowth of
The concept of immunosurveillance had a dominating tumors which have escaped the immunological restraints of
position in the past, but currently, it is restricted only the equilibrium phase.[14]
to the virus‑induced tumors. The immune system can
Elimination
protect the host against outgrowth of cells transformed by
human papillomaviruses, Epstein–Barr virus, and human If the tumors are successfully eradicated in this phase, it
herpesvirus‑8 carrying Kaposi sarcoma cells. However, represents the complete immunoediting process without
they may grow into full‑fledged tumors in congenitally progressing to further phases. Elimination phase works out
or iatrogenically immunodefective individuals or in in four steps.
HIV‑infected persons with immunodeficiency.[9]
Initiation of antitumor responses occurs when the local
Cancer immunosurveillance represents only one dimension tissue disruption that occurs as a result of the stromal
of the complex interaction between the immune system and remodeling processes at the growing tumor alerts the cells

8 Journal of Orofacial Sciences | Volume 9 | Issue 1 | January-June 2017


[Downloaded free from http://www.jofs.in on Friday, January 5, 2018, IP: 157.48.122.209]

Kaja, et al.: Tumor immunology

of the innate immune system. The stromal remodeling the draining lymph nodes and induce the activation of
produces pro‑inflammatory molecules which along with naïve tumor‑specific Th1 CD4+  T‑cells. Th1  cells through
the chemokines produced by the tumor cells themselves cross‑presentation of antigenic tumor peptides on DC major
comprise the cells of the innate system that are recruited histocompatibility (MHC) Class I molecules facilitate the
to the site of danger. Once recruited to the tumor site, development of tumor‑specific CD8+ cytotoxic T‑cells.[2]
these innate cells such as the natural killer (NK) cells,
In the fourth step, the host is provided with a capacity to
γδ T‑lymphocytes, and macrophages recognize molecules
completely eliminate the tumor development by means of
such as ligands for NK Group 2 D (NKG2D) ligand
tumor‑specific adaptive immune responses. Tumor‑specific
which have been induced on the tumor cells either by CD4+  and CD8+  T‑cells are primed to the site of tumor
inflammation or by the cellular transformation process where they participate in the killing of antigen‑positive
itself. γδ T‑lymphocytes and NK cells may recognize tumor cells. IL‑2 produced by the CD4+  T‑cells together
developing tumors through T‑cell receptor interaction with IL‑15 produced by the host cells helps to maintain
with either NKG2D ligands or glycolipid‑CD1 complexes the function and viability of tumor‑specific cytotoxic
expressed on the tumor cells, respectively. This precise CD8+  T‑cells. CD8+  T‑cells induce tumor cell death
mechanism of recognition leads to the progression directly and also indirectly by producing IFN‑γ which in
of antitumor immune response, i.e., production of turn activates IFN‑γ‑dependent mechanisms.[14]
interferon‑γ (IFN‑γ).[14]
The elimination phase of cancer immunoediting is a
In the second step, the effects of innate immune continuous process and must be repeated each time
recognition of tumor are amplified. IFN‑γ, released at antigenically distinct neoplastic cells arise. Hence, cancer
tumor site, induces the production of chemokines such is more prevalent in aged individuals where the functioning
as CXCL‑9, 10, and 11 from the tumor cells as well as of immune system declines.
from surrounding normal host tissue which further recruits
more cells of the innate immune system to the tumor. Equilibrium
Tumor‑infiltrating macrophages are induced to produce The host immune system and the tumor cell variant that
low levels of interleukin‑12 (IL‑12) by the products that have survived the elimination phase enter into equilibrium.
are generated during the remodeling of extracellular Here, the lymphocytes and IFN‑γ exert potent selection
matrix. IL‑12 stimulates NK cells to produce low amounts pressure on tumor cells which could try to extinguish
of IFN‑γ, which further activates macrophages to produce a tumor bed containing many genetically unstable and
more amounts of IL‑12 thereby leading to increased mutating cells. Equilibrium phase of cancer immunoediting
production of IFN‑γ by the NK cells. These work in a is the longest of all the three phases and occurs over a
positive feedback mechanism. NK cells also produce period of many years in humans. This is said to have the
more IFN‑γ production upon stimulation by binding of Darwinian selection; where many of the original tumor
NK cell receptors to their cognate ligands on tumor cells, cell escape variants are destroyed, but new variants arise
i.e., NKG2D ligands. IFN‑γ can now activate a number carrying different mutations. Different mutations in the
of IFN‑γ dependent processes which enhance the killing new variants provide increased resistance to attack by the
of a proportion of the tumor. The processes activated are immune system. Thereby, the end result of equilibrium
antiproliferative, proapoptotic, and angiostatic.[2] IFN‑γ phase is a new population of tumor clones with reduced
activates macrophages that express reactive oxygen and immunogenicity.[2,16]
nitrogen metabolites which are tumoricidal products.
NK cells activated by IFN‑γ or by engagement of their Clinical evidence supporting the equilibrium phase of
cancer immunoediting is provided by a number of findings.
receptors can kill tumor cells either by tumor necrosis
• The existence of an immune response to premalignant
factor alpha‑inducing ligand or perforin‑dependent
monoclonal gammopathy of undetermined significant
mechanisms, respectively.[14]
cells that eventually progress to multiple myeloma,
The processes in the second step provide a source of tumor in which the immune system is controlling, but not
antigens from dead tumor cells and thereby activating eliminating the premalignant cells that eventually
tumor‑specific adaptive immune responses in the third step. evolve and progress to malignancy
Dendritic cells (DC) recruited to the tumor site get activated • Passive immunization with a specific antibody and in
by exposure to the cytokines generated by innate immune conjunction with cytokine therapy or chemotherapy
responses or by interacting with the tumor‑infiltrating NK is known to induce remission in few patients having
cells.[15] The activated DC can acquire tumor antigens by low‑grade B‑cell lymphoma. However, the tumor cells
either direct or indirect mechanisms. Direct mechanism are not completely eliminated, and they can be detected
is by the ingestion of tumor cell debris. Indirect in the blood/bone marrow for up to 8 years following
mechanisms involve transferring of tumor cell‑derived clinical remission
heat shock protein or tumor antigen complexes to DC. • Pediatric acute myeloid leukemia patients on
These activated antigen‑bearing mature DCs migrate to chemotherapy or chemotherapy combined with
Journal of Orofacial Sciences | Volume 9 | Issue 1 | January-June 2017 9
[Downloaded free from http://www.jofs.in on Friday, January 5, 2018, IP: 157.48.122.209]

Kaja, et al.: Tumor immunology

autologous bone marrow transplantation suggested a Cancer Immune Evasion


role for the immune system in establishing long‑term
Immune evasion is a mechanism used by pathogenic
remission
organisms and tumors to evade host’s immune response
• Tumor transmission from an organ donor to recipient
and to maximize their capability of transmission to a fresh
has also provided evidence for the equilibrium phase of
host or to continue in growth, respectively.[19] Although
cancer immunoediting. It was shown that the tumor was
tumors possess tumor rejection antigens, they are capable
being held under control by an immunologic mechanism
of escaping destruction by host immunity which indicates
in the donor and the transplantation of organ into an
that some form of immune evasion occurs.[20] The escape
immunosuppressed host allowed tumor outgrowth.[4]
process is attributed to the inability of immune cells to
Escape develop an effective antitumor response or to the inhibition
The escape phase represents the failure of the immune of functions of the immune system by tumor‑derived
system to eliminate or control transformed cells, factors.
which further allows tumor cell variants to grow in Tumors can interfere with all the components of an immune
an immunologically unrestricted manner. Genetic and system and could also affect all the stages of immune
epigenetic changes in the tumor cell confer resistance response, but tumor‑induced immunosuppression is not
to immune detection and/or elimination, allowing the an immunodeficiency in a classical sense. It is selective
expansion of tumors and they being clinically detectable. and aggressively inhibits the functions of immune cells
To achieve progressive growth, the tumor cells have to responsible for antitumor responses such as T‑lymphocytes,
circumvent either one or both the arms of the immune B‑lymphocytes, macrophages, NK cells, DC, granulocytes,
system, i.e., innate and adaptive immune responses in and mast cells. The immune cell infiltrates may not always
the cancer immunosurveillance network. To circumvent favor antitumor immune responses but may benefit the
these responses, several distinct immunologically tumor, particularly when the tumor is aggressive. In
driven tumor sculpting events should occur before the developing an immunosuppressive environment, the tumor
immunogenic phenotype of a malignant cell is ultimately utilizes various strategies to:
established.[17] • Regulate the cytokine/chemokine network
• Controls the regulatory cells recruitment
Tumors can impede the development of antitumor immune
• Modify the expression of receptors for tumor
responses either directly or indirectly which target the
growth‑promoting factors or release of vesicular bodies
immune system to achieve tumor escape. It could be by:
that carry tumor‑derived “molecular messages” in the
• Expression of immunosuppressive cytokines such as
host.[21]
transforming growth factor‑β and IL‑10 or
• Mechanisms involving T‑lymphocytes with Tumors elaborate assembly of tricks to fool the immune
immunosuppressive activities, i.e., T‑regulatory (TRegs) system in general to avoid recognition.
cells.[18] i. They either hide from immune cells thus avoiding
recognition
Tumor escape could also result from the changes that occur
ii. They proceed to disable or eliminate immune cells.[22]
directly at the level of tumor. These include:
• Alterations that affect the recognition of tumor by Tumors can evade the host immune response either by
immune effector cells such as loss of antigen expression, being (a) poor stimulators of T‑lymphocytes or (b) poor
loss of MHC components, shedding of NKG2D ligands, targets for tumor‑specific T‑lymphocytes. Tumors skillfully
and further development of IFN‑γ insensitivity shed their surface antigens thereby being poor stimulators
• Tumor mechanisms to escape immune destruction for T‑lymphocytes or downregulates the expression
such as defects in death receptor signaling pathways of molecules needed for interaction with immune
or antiapoptotic signal expressions such as those cells and hence being poor targets for tumor‑specific
induced by active signal transducer and activation of T‑lymphocytes.[22]
transcription 3.
In tumor cells, the expression of molecules such as TAA,
Of this dysregulation of MHC Class I processing and human leukocyte antigen (HLA) Class I molecules, or
presentation and development of IFN‑γ insensitivity in antigen‑processing machinery components (APM) is often
tumor cells, allows tumors to escape from the events in downregulated or altered. Abnormalities in the APM
the elimination phase of the cancer immunoediting process. components such as downregulation, absence, or mutation
The unresponsive state in these tumors may be caused due do not generate peptides from TAA, or they are generated
to the absence/abnormal function of components in the in a different form that does not allow for the formation of
IFN‑γ receptor signaling. Tumor escape strategies finally HLA Class I peptide complexes by the T‑cells.[23] Tumors
result in a clinically detectable malignant disease, when if frequently misprocessed with TAA, where the immunogenic
left unchecked results in the death of the host.[14] peptides cannot be made. These immunogenic peptides

10 Journal of Orofacial Sciences | Volume 9 | Issue 1 | January-June 2017


[Downloaded free from http://www.jofs.in on Friday, January 5, 2018, IP: 157.48.122.209]

Kaja, et al.: Tumor immunology

cannot include in Human leukocytic antigen class I. In Table 1: Tumor factors modulating the immune cells
such cases, trimolecular HLA class I‑chain‑β2m‑peptide Factor-Modulatory mechanism
is absent from the tumor cell surface. The peptide‑HLA The TNF family ligands:
molecule complex thus formed cannot be recognized by the Induce leukocyte apoptosis
cytotoxic T‑lymphocytes.[24] via the TNF family receptors
FasL Fas
Another aberration in tumor cells involves the
TRAIL TRAIL‑R
downregulated expression of costimulatory molecules on
TNF TNFR1
the cell surface that are essential for producing interactions
Small molecules
with T‑lymphocytes. Decreased expression of costimulatory
Prostaglandin E2 Inhibits leukocyte functions
molecules such as B7 family on tumor cells could lead to
through increased cAMP
unresponsiveness based on MHC Class I restricted antigen
Histamine Inhibits leukocyte functions
presentation without transmission of the costimulatory through increased cAMP
signals that are critical for the activation of leukocytes.[25] Epinephrine Inhibits leukocyte functions
Tumors either produce or induce factors which in turn through increased cAMP
modulate the functions of immune cells or induce apoptosis INOS Promotes or inhibits Fas‑mediated
apoptosis by regulation of no
of the immune cells. These factors include a broad range of
levels
biological effector molecules such as [Table 1]:[22]
H 2O 2 Has pro‑oxidant activity, increases
• Several distinct receptor–ligand systems cAMP levels, causes apoptosis in
• Small molecular species NK cells, inhibits tumor‑specific
• Cellular enzymes CTL
• Soluble cell components Enzymes
• Cytokines/chemokines. Indoleamine Suppresses T‑cell responses
2,3‑dioxygenase
This diversity indicates that tumors are incredible in their
Arginase I Impairs T‑cell functions, decreases
ability to defend or debilitate the host immune responses.
chain expression
Mechanisms of tumor immune escape Cytokines
TGF‑β Inhibits perforin and granzyme
Mechanisms responsible for immune cell dysfunction in mRNA expression; inhibits
cancer are numerous. The escape from tumor immunity can lymphocyte proliferation
be achieved in three principally distinct ways [Table 2]. IL‑10 Inhibits production of IL‑1, IFN‑γ,
They are by: IL‑12 and TNF‑α
• Lack of recognition by means of loss or alteration of GM‑CSF Promotes expansion
molecules which are important for the recognition and of immunosuppressive
activation of the immune system tumor‑associated macrophages
• Lack of susceptibility, i.e.,  escape from the effector Tumor‑associated Inhibit IL‑2 dependent lymphocyte
mechanisms of cytotoxic lymphocytes gangliosides proliferation or induce apoptotic
• Induction of immune dysfunction.[26] signals
TNF: Tumor necrosis factor, TRAIL‑R: Tumor necrosis
These mechanisms render the tumor cells less recognizable factor alpha‑inducing ligand‑receptor, TNFR1: Tumor
by NK cells and T‑cells and also make them less susceptible necrosis factor receptor 1, TGF: Transforming growth factor,
to the effector molecules of the immune system. There GM‑CSF: Granulocyte‑macrophage colony‑stimulating factor,
is a continuous shaping of the tumor cell clones by both IFN: Interferon, IL: Interleukin, CTL: Cytolytic T lymphocytes,
nonimmune and immunosurveillance mechanisms. This NK: Natural killer, INOS: Inducible nitric oxide synthase,
sculpting process often extends for longer periods, probably cAMP: Cyclic adenosine monophosphate
many years and perhaps for decades.
and relies on glycolysis to produce energy which is called
Immune escape mechanisms related to tumor metabolic
the Warburg effect. Warburg in 1920 found that even in
changes
the presence of ample oxygen, the tumor cells prefer to
Tumors confront the adverse environment in two contexts. metabolize glucose by glycolysis, a metabolic pathway
First, the developing tumors must acquire nutrients to that converts glucose into pyruvate.[41] Pyruvate is reduced
ensure their rapid growth, and second, they must escape to lactic acid by fermentation. Although it is less efficient
from the attack by host immune system. Recent studies for producing adenosine triphosphate, it is quicker than
suggest that tumor cell metabolism plays a pivotal role in OXPHOS and confers a selective advantage to rapidly
tumor immune escape. growing tumor cells. This was related to low oxygen
Tumor cell metabolism avoids the activities of concentration in the inner region of nascent tumors that
mitochondria and oxidative phosphorylation (OXPHOS) progressively become distanced from the vasculature
Journal of Orofacial Sciences | Volume 9 | Issue 1 | January-June 2017 11
[Downloaded free from http://www.jofs.in on Friday, January 5, 2018, IP: 157.48.122.209]

Kaja, et al.: Tumor immunology

Table 2: Mechanisms responsible for immune escape by tumors


Mechanism-Pathway for Immune escape
Lack of recognition[26]
Alterations in the antigen processing Mutations and/or deletions of one or several genes encoding components of the antigen
and presentation pathway[27,28] processing machinery
Complete loss of HLA molecules
Absence of peptide and β2 microglobulin, needed for MHC class I molecule
Mutations of proteasomal subunits such as LMP‑2 and LMP‑7 as well as of the peptide
transporters such as TAP‑1 and TAP‑2
Loss of antigenic variants[29] Simultaneous loss of antigen and HLA class I
Decreased activation of NK cells Structural alterations ‑ total, allelic, or locus‑specific loss of MHC class I heavy chain;
and T‑cells by downregulating B2‑microglobulin deletions, rearrangements, or mutations; transporter associated with antigen
MHC[30,31] processing mutations
Dysregulation‑methylation and acetylation; transcriptional downregulation; posttranscriptional
downregulation; deficient IFN‑γ signal transduction
Enhanced negative signaling and Ectopic expression of nonclassical HLA molecule such as HLA‑G inhibits NK cell function by
counter attack[26,32‑34] binding to inhibitory receptors including the LILRB1
HLA‑G also shuts down the production of IFN‑γ and Fcγ receptor‑mediated activation of LIR‑1
expressing macrophages
Via a Fas–FasL‑dependent mechanism, HLA‑G1 were known to induce apoptosis of CD8+
T‑cells. Nonclassical HLA‑E and HLA‑F molecules, as well as alleles of HLA‑C, were shown
to induce apoptosis of NK cells. Tumor cells also secrete FasL‑bearing vesicles that induce
lymphocyte apoptosis known as “Fas‑counterattack”
Suppression of antitumor responses Cell‑to‑cell contact‑independent soluble factors, such as immunoinhibitory cytokines
by TRegs cells[21,35] (IL‑10 and TGF‑β) or adenosine
The cell contact‑dependent accumulation of cAMP in target cells or target cell death mediated by
perforin/granzyme B or death receptor‑ligand interactions
The competition for IL‑2, between TRegs and responders leading to death by starvation of
responder cells or for CTLA‑4/B7 co‑stimulation between TRegs and antigen‑presenting cells,
depriving responder cells of co‑stimulatory signaling necessary for proliferation
Lack of susceptibility
Induction and regulation of Tumor cells overexpress antiapoptotic molecules such as bcl‑2 thereby resists apoptosis and
apoptosis[26,36] hence escape from both nonimmune and immune surveillance mechanisms
Escaping death receptor Tumors escape death receptor signaling by overexpression of flip and also by release of soluble
signaling[26,37,38] factors such as Fas receptors, decoy receptors, and FasL
Presence of decoy death receptors (TRAIL‑R3 and TRAIL‑R4) lacking the intracellular domain
Missense mutations in the Fas gene leading to decreased expression or expression of
nonfunctional receptors
Metalloproteases such as matrix metalloprotease‑7 by cleaving Fas thereby decreasing the
surface expression of Fas and rendering the tumor resistant to FasL‑mediated apoptosis
Alterations in the FADD and of caspase ten by inactivation mutations
Escape from perforin and granzyme Tumors overexpress the serine PI‑9 that inhibits Granzyme B
B‑induced apoptosis[26] Cathepsin B, a cysteine proteinase is that inactivates perforin is overexpressed by tumors
Failure of IFN‑γ to induce the platelet activating factor receptor could confer tumor cell escape
from perforin‑mediated killing
Tumor‑induced immune suppression
Chronic inflammation[39,40] Tumors gain a growth advantage by secreting immunosuppressive cytokines or specifically
recruiting TRegs and MSC that dampen immune reactivity
Decreased Th1 cytokine production and poor cytolytic capacity of tumor infiltrating lymphocytes
and peripheral blood lymphocytes. This is associated with decreased levels of signaling
molecules such as p56, p59, ZAP70, JAK3, STAT‑5 and block of nuclear factor‑κβ which helps
in immune escape of the tumors
TRAIL: Tumor necrosis factor alpha‑inducing ligand, TGF: Transforming growth factor, IFN: Interferon, IL: Interleukin, NK: Natural killer,
TRegs: T regulatory, HLA: Human leukocyte antigen, TAP: Transporter associated with antigen presentation, MHC: Major histocompatibility,
Th: T‑helper cells, LMP: Low molecular mass polypeptides, CTLA‑4: Cytotoxic T‑lymphocyte‑associated antigen‑4, FADD: Fas‑associated
death domain, PI: Protease inhibitor, MSC: Myeloid suppressor cells, LILRB1: Leukocyte immunoglobulin‑like receptor B1, cAMP: Cyclic
adenosine monophosphate, LIR: Leukocyte immunoglobulin‑like receptor
12 Journal of Orofacial Sciences | Volume 9 | Issue 1 | January-June 2017
[Downloaded free from http://www.jofs.in on Friday, January 5, 2018, IP: 157.48.122.209]

Kaja, et al.: Tumor immunology

thereby resulting in a metabolic shift from OXPHOS to molecules are tyrosinase derived from the endosomes
fermentation.[42] in melanosomes and melanoma‑associated antigen‑3.
However, the pathway by which these peptides reach the
A new hypothesis proposes that the metabolism of tumor
Class II molecules is unclear. Recent data suggest that
cells can be changed to adjust to their different needs by
autophagy could be involved in the presentation of tumor
alterations in gene regulation, originated by deregulated
antigens to MHC Class II molecule. It was revealed that
expression of oncogenes. Tumor metabolic shift could
large portion of MHC Class II bound peptides is derived
be due to several processes such as overexpression of
from intracellular proteins, and this proportion appears to
glycolytic enzymes and metabolite transporters, defective
be correlated with autophagy.[44]
cellular respiration, and oncogenic alterations.[43] Apart
from the growth advantage given by the tumor metabolic Cancer Immunotherapy
shift, it also facilitates the immune escape of the tumor
cells by a process of Darwinian selection of the clones that “Immunotherapy is a treatment that uses certain parts of a
are able to perform the appropriate metabolic changes. person’s immune system to fight diseases such as cancer.”
Specific mechanisms of the immune system through
Processing and Presentation of Tumor Antigens antigen recognition played a key role in the possibility of
The antigenic peptides expressed by the tumor cells immune‑based therapy for cancer.
are presented by MHC molecules at the cell surface The main modes of tumor immunotherapy have been
to T‑lymphocytes. The presentation of peptides at the categorized as.[46]
surface of tumor cells is through the classical pathways
Stimulation of active host immune responses
of antigen processing. Presentation of peptides to
cytotoxic T‑lymphocytes by MHC Class I is derived from • Vaccination using tumor cells and antigens:
intracellular proteins degraded by the proteasome. Peptides o Immunization of tumor‑bearing individuals with
presented to CD4+  T‑lymphocytes by MHC Class  II are killed tumor cells or tumor antigens may result in
derived from proteins that have been transported to an enhanced immune responses against the tumor
endocytic compartment. • Augmentation of host immunity with cytokines and
costimulators against tumors:
Major histocompatibility Class I presentation
o Cell‑mediated immunity to tumors may be enhanced
Majority of peptides presented by MHC Class I molecules by expressing costimulators and cytokines in tumor
are produced by the proteasome and some appear to be cells. Cytokines stimulate the proliferation and
from degradation of newly synthesized proteins, which differentiation of T‑lymphocytes and NK cells
are either misfolded or fallacious and have been called as • Nonspecific stimulation of immune system:
“defective ribosomal products.” All the class I restricted o Immune responses to tumors may be stimulated
tumor antigenic peptides are derived from endogenous by the local administration of inflammatory
proteins that may be either cytosolic or nuclear proteins. substances/by systemic treatment with agents that
These peptides gain immediate access to the proteasome act as polyclonal activators of lymphocytes such as
and return to the cytosolic proteasome through the anti‑CD3 antibodies.
endoplasmic reticulum‑associated degradation pathway
that eliminates misfolded or unassembled proteins from the Passive immunotherapy with T‑cells and antibodies
endoplasmic reticulum.[44] • Adoptive cellular therapy:
Some peptides follow a different presenting pathway. For o It is the transfer of cultured immune cells having
example, peptide presented by melanoma cells derived from antitumor reactivity into a tumor‑bearing host
secreted matrix metalloproteinase‑2 (MMP2). The lone • Therapy with antitumor antibodies:
expression of MMP2 is not sufficient for the presentation of o Tumor‑specific monoclonal antibodies
the peptide by Class I molecules. However, its presentation o Cancer vaccines.
is dependent on the secretion of MMP2 in the extracellular
Conclusion
space and subsequent uptake of exogenous MMP2 by
endocytosis mediated through integrin αVβ3 that functions A large body of evidence implicates the potential of
as a receptor of MMP2. It could be due to the blockade of the immune system to control tumor promotion and the
the presentation of the peptide by proteasome inhibition.[45] various modalities of immunotherapy that can induce
strong immune responses for fighting against cancer.
Major histocompatibility Class II presentation
Such knowledge has stimulated the invention of novel
Tumors expressing Class II molecules present peptides to immunotherapeutic modalities including therapeutic
CD4+  T‑lymphocytes. These peptides are derived from antibodies, vaccines, and cell‑based treatments. Moreover,
proteins present either at the cell surface or in intracellular improvements in our understanding of antiapoptotic
organelles. Some peptides presented by the MHC Class II and immune escape mechanisms have resulted in the

Journal of Orofacial Sciences | Volume 9 | Issue 1 | January-June 2017 13


[Downloaded free from http://www.jofs.in on Friday, January 5, 2018, IP: 157.48.122.209]

Kaja, et al.: Tumor immunology

discovery of numerous biologically active drugs that Garaude J. From tumor cell metabolism to tumor immune
can specifically target and alter such mechanisms. In escape. Int J Biochem Cell Biol 2013;45:106-13.
the near future, the progress in cancer immunology will 20. Mitra R, Singh S, Khar A. Antitumour immune responses. Expert
Rev Mol Med 2003;5:1‑19.
introduce elegant combinational treatments involving novel
21. Whiteside TL. Tricks tumors use to escape from immune control.
immunotherapeutic strategies aiming at immunostimulation Oral Oncol 2009;45:e119‑23.
with simultaneous elimination of tumor‑induced suppressive 22. Whiteside TL. Immune suppression in cancer: Effects on
mechanisms. These new therapies in cancer are expected to immune cells, mechanisms and future therapeutic intervention.
induce improved clinical responses and eventually cancer Semin Cancer Biol 2006;16:3‑15.
prevention. 23. Meissner M, Reichert TE, Kunkel M, Gooding W, Whiteside TL,
Ferrone S, et al. Defects in the human leukocyte antigen class I
Financial support and sponsorship antigen processing machinery in head and neck squamous cell
carcinoma: Association with clinical outcome. Clin Cancer Res
Nil.
2005;11:2552‑60.
Conflicts of interest 24. Hoffmann TK, Nakano K, Elder EM, Dworacki G,
Finkelstein SD, Appella E, et al. Generation of T cells specific
There are no conflicts of interest. for the wild‑type sequence p53(264‑272) peptide in cancer
patients: Implications for immunoselection of epitope loss
References variants. J Immunol 2000;165:5938‑44.
1. Siegel BV. Tumor immunity. An overview. Am J Pathol 25. Wang S, Chen L. Co‑signaling molecules of the B7‑CD28 family
1978;93:515‑24. in positive and negative regulation of T lymphocyte responses.
2. Dunn GP, Bruce AT, Ikeda H, Old LJ, Schreiber RD. Cancer Microbes Infect 2004;6:759‑66.
immunoediting: From immunosurveillance to tumor escape. Nat 26. Malmberg KJ, Ljunggren HG. Escape from immune‑ and
Immunol 2002;3:991‑8. nonimmune‑mediated tumor surveillance. Semin Cancer Biol
3. Finn OJ. Human tumor antigens, immunosurveillance, and 2006;16:16‑31.
cancer vaccines. Immunol Res 2006;36:73‑82. 27. Seliger B, Cabrera T, Garrido F, Ferrone S. HLA class I antigen
4. Swann JB, Smyth MJ. Immune surveillance of tumors. J Clin abnormalities and immune escape by malignant cells. Semin
Invest 2007;117:1137‑46. Cancer Biol 2002;12:3‑13.
28. Seliger B. Strategies of tumor immune evasion. BioDrugs
5. Burnet FM. Cancer; a biological approach. I. The processes of
2005;19:347‑54.
control. Br Med J 1957;1:841‑7.
29. Khong  HT, Wang  QJ, Rosenberg  SA. Identification of multiple
6. Thomas L. On immunosurveillance in human cancer. Yale J Biol
antigens recognized by tumor‑infiltrating lymphocytes from a
Med 1982;55:329‑33.
single patient: Tumor escape by antigen loss and loss of MHC
7. Burnet FM. The concept of immunological surveillance. Prog
expression. J Immunother 2004;27:184‑90.
Exp Tumor Res 1970;13:1‑27.
30. Wu JD, Higgins LM, Steinle A, Cosman D, Haugk K,
8. Burnet M. Immunological factors in the process of
Plymate SR. Prevalent expression of the immunostimulatory
carcinogenesis. Br Med Bull 1964;20:154‑8.
MHC class I chain‑related molecule is counteracted by shedding
9. Klein G, Klein E. Surveillance against tumors – Is it mainly in prostate cancer. J Clin Invest 2004;114:560‑8.
immunological? Immunol Lett 2005;100:29‑33.
31. Doubrovina ES, Doubrovin MM, Vider E, Sisson RB,
10. Laroye  GJ. How efficient is immunological surveillance against O’Reilly RJ, Dupont B, et al. Evasion from NK cell immunity
cancer and why does it fail? Lancet 1974;1:1097‑100. by MHC class I chain‑related molecules expressing colon
11. Dunn GP, Old LJ, Schreiber RD. The immunobiology of adenocarcinoma. J Immunol 2003;171:6891‑9.
cancer immunosurveillance and immunoediting. Immunity 32. Spaggiari GM, Contini P, Dondero A, Carosio R, Puppo F,
2004;21:137‑48. Indiveri F, et al. Soluble HLA class I induces NK cell apoptosis
12. Penn I. Tumors of the immunocompromised patient. Annu Rev upon the engagement of killer‑activating HLA class I receptors
Med 1988;39:63‑73. through FasL‑Fas interaction. Blood 2002;100:4098‑107.
13. Sengupta N, MacFie TS, MacDonald TT, Pennington D, 33. Poggi A, Massaro AM, Negrini S, Contini P, Zocchi MR.
Silver AR. Cancer immunoediting and spontaneous tumor Tumor‑induced apoptosis of human IL‑2‑activated NK cells: Role
regression. Pathol Res Pract 2010;206:1‑8. of natural cytotoxicity receptors. J Immunol 2005;174:2653‑60.
14. Dunn GP, Old LJ, Schreiber RD. The three Es of cancer 34. Restifo NP. Not so Fas: Re‑evaluating the mechanisms of
immunoediting. Annu Rev Immunol 2004;22:329‑60. immune privilege and tumor escape. Nat Med 2000;6:493‑5.
15. Gerosa F, Baldani‑Guerra B, Nisii C, Marchesini V, Carra G, 35. Roncarolo MG, Bacchetta R, Bordignon C, Narula S, Levings MK.
Trinchieri G. Reciprocal activating interaction between natural Type 1 T regulatory cells. Immunol Rev 2001;182:68‑79.
killer cells and dendritic cells. J Exp Med 2002;195:327‑33. 36. Campos L, Rouault JP, Sabido O, Oriol P, Roubi N, Vasselon C,
16. Lengauer C, Kinzler KW, Vogelstein B. Genetic instabilities in et al. High expression of bcl‑2 protein in acute myeloid leukemia
human cancers. Nature 1998;396:643‑9. cells is associated with poor response to chemotherapy. Blood
17. Vesely MD, Kershaw MH, Schreiber RD, Smyth MJ. Natural 1993;81:3091‑6.
innate and adaptive immunity to cancer. Annu Rev Immunol 37. Strand S, Vollmer P, van den Abeelen L, Gottfried D, Alla V,
2011;29:235‑71. Heid H, et al. Cleavage of CD95 by matrix metalloproteinase‑7
18. Khong HT, Restifo NP. Natural selection of tumor variants induces apoptosis resistance in tumour cells. Oncogene
in the generation of tumor escape phenotypes. Nat Immunol 2004;23:3732‑6.
2002;3:999‑1005. 38. Hallermalm K, De Geer A, Kiessling R, Levitsky V, Levitskaya J.
19. Villalba M, Rathore MG, Lopez- Royuela N, Krzywinska E, Autocrine secretion of Fas ligand shields tumor cells from

14 Journal of Orofacial Sciences | Volume 9 | Issue 1 | January-June 2017


[Downloaded free from http://www.jofs.in on Friday, January 5, 2018, IP: 157.48.122.209]

Kaja, et al.: Tumor immunology

Fas‑mediated killing by cytotoxic lymphocytes. Cancer Res 43. Smolková K, Plecitá‑Hlavatá L, Bellance N, Benard G,
2004;64:6775‑82. Rossignol  R, Ježek P. Waves of gene regulation suppress and
39. Malmberg KJ. Effective immunotherapy against cancer: then restore oxidative phosphorylation in cancer cells. Int J
A question of overcoming immune suppression and immune Biochem Cell Biol 2011;43:950‑68.
escape. Cancer Immunol Immunother 2004;53:879‑92. 44. van der Bruggen P, Van den Eynde BJ. Processing and
40. Kiessling R, Wasserman K, Horiguchi S, Kono K, Sjöberg J, presentation of tumor antigens and vaccination strategies. Curr
Pisa P, et al. Tumor‑induced immune dysfunction. Cancer Opin Immunol 2006;18:98‑104.
Immunol Immunother 1999;48:353‑62. 45. Godefroy E, Moreau‑Aubry A, Diez E, Dreno B, Jotereau F,
41. Warburg O. On respiratory impairment in cancer cells. Science Guilloux Y. αVβ3‑dependent cross‑presentation of matrix
1956;124:269‑70. metalloproteinase‑2 by melanoma cells gives rise to a new tumor
42. Samudio I, Fiegl M, Andreeff M. Mitochondrial uncoupling and antigen. J Exp Med 2005;202:61‑72.
the Warburg effect: Molecular basis for the reprogramming of 46. Abbas AK, Lichtman AH. Cellular and Molecular Immunology.
cancer cell metabolism. Cancer Res 2009;69:2163‑6. 5th ed. Philadelphia: Elsevier Saunders; 2005. p. 391‑410.

Journal of Orofacial Sciences | Volume 9 | Issue 1 | January-June 2017 15

Vous aimerez peut-être aussi