SlideShare une entreprise Scribd logo
1  sur  22
Télécharger pour lire hors ligne
Dear Reviewer,
Please find the confined manuscript entitled “Cytokine Immunotherapy: A forthcoming
visible feature in cancer therapeutics”, which I would like to share with you all.
Cytokine are messengers protein produced by varieties of cells whose role in host responses
to cancer are remarkable. Different bioprocesses are interlinked with cytokine functions.
Preclinical research studies have shown the anti-cancerous property of cytokine which
suppress inflammatory and autoimmune microenvironment. Little evidence of rare patient
studies suggesting that the immune system can revoke again if stimulated appropriately. One
of the appropriate stimulation of immune system can be done with therapeutic manipulation
of cytokine balance.
However, at present status, mono-cytokine therapy doesn’t show promising features as
compare to combined cytokine therapy as combined cytokine therapy or cytokine therapy
with other traditional therapeutic module. It is expected that appropriate combination will
result with specific and reliable therapeutics for cancer treatment. It is time to focus on the
remaining checkpoints and else everything to make an option for the accurate efficacy of
treatment.
In this literature work, I have enlightened some therapeutic individuality along with
possibility of combination with other modalities. I think that this literature work will be
fruitful for those seekers who are looking for the appropriate combination of treatment to
overcome the drawbacks of traditional healing remedies.
Thank you for your consideration. I look forward to seeing your critics from you.
Sincerely,
Sachin K. S. Chauhan
New Delhi,
INDIA.
Cytokine Immunotherapy: A Forthcoming Visible Feature in Cancer
Therapeutics
Sachin K. S. Chauhan
Abstract: Cell-based cytokine therapy have prodigious promising role for treating a varieties
of diseases. Cytokines are intra- as well as intercellular messengers, which recruit the
immune effecter cells, thereby limiting the tumor growth; however, sometimes these
molecules promote tumor progression. Several studies have attempted the usage cytokines
alone as pharmaceutical targets to alter the immune response, which may eradicate solid
tumors. In the context of advancement in understanding of cytokine biology, there is a
definite need to develop combined cytokine therapy with the advent of gene technology to
improve efficacy of cancer treatment. This review focuses on understanding the factors of
combined cytokine therapy and that allows us to attain durable and long-lasting responses in
cancer therapy.
Introduction
Cancer is a natural wound healing related process, which includes cytokine secretion, tissue
remodeling, and oncogenes activation (1). Every cell has a unique type of characteristic
signature such as the abilities of genes or proteins to perform their function. These
characteristic features also alleviate molecular mechanisms of diseases and will give
prediction about individualization of therapies for suffering people. From many years, all
efforts to treat human malignancies have concentrated on the early detection and cure of
tumor. So these efforts have provided great opportunities for improving the management of
cancer patients by enhancing the efficacy of detection and treatment (2). Now it is well
known that the mutation, competition, and natural selection operating within the population
of somatic cells are basic requirements for initiations, progression, and survival of cancer
cells. These phenomena depend not only on the tumor cell, but on the complex interactions
between the tumor cells and its ‘host’ (3). Hence, it is required to focus on the strategies to
modulate the growth mechanism and host microenvironment, which offers a complementary
perspective to achieve a permanent cure.
Cancer cells are introduced by their heritable properties like they reproduce in defiance of the
normal restraints, invade, and colonize territories reserved for other cells. This renegade
coordination of malignancy has been creating lot of evidences that tumor cells elicit certain
hallmark characteristics which lead the somatic cells to micro-evolutionary developmental
process of malignant cancer (4). (Box 1)
This multistep process of tumor genesis could be explained by the requirement of incipient
cancer cells to adopt the traits of tumorigenic cells and ultimately become malignant (5). :
There are several therapeutic modalities have been developing on the basis of cancer
hallmarks individuality (Figure 1). It is mandatory to go through the overall metabolic
network and its acquired hallmarks which explain the majority of manipulated checkpoint for
the growth of tumor cells.
FOUNDATION OF CANCER BIOLOGY
New research applications of cancer biology are transforming our understanding about
cancer. The basic foundation of cancer biology explores these basic principles – cancer
excogitates by somatic selection, neoplasm ripens in complex ecosystems, natural selection
has designed effective cancer defenses (6). The limitations of these principles defenses have
the explanations which present foundation of cancer biology for understanding, preventing
and developing therapeutic modalities (7). The most fundamental traits of cancer cells
overcome cell growth signals and influence cell survival and energy metabolism (8). These
mutagenic signals are release via branched intracellular signaling through cell progression
which should be filtered in case of tumor growth. Alternatively, some tumor cells may be
able to overcome high level of oncogenic signaling by disabling their apoptosis-inducing
mechanism. If we enquire anomaly between level of complex network of proteases,
sulfatases, and other enzymes in normal and tumor cells, then it’ll be easy to find out that it
can switch the pathway in an alternate ways, hence it is necessary to seek a multi target
therapeutic approaches. In addition to two above aspect of fundamental biology of
insensitivity to antigrowth signaling and evasion of apoptosis, cancer cells riddance various
signaling pathways, especially apoptotic which allow them to seed, proliferate, and flourish
in other territories of body. They must also besiege dozens of powerful tumor suppressor
programs that control in various ways to limit the normal cell growth. For example, the
central effectors of apoptosis is caspases whose hypo-phosporylation makes impossible
antigrowth factors such as TGF-β, pRB and p53 from blocking the advancement of cell
growth (9).The other two factors are RB (Retinoblastoma) and TP53 have the function of
inhibition and execution of cell cycle growth in normal cells. Tumor cell with the defect in
these factor mechanisms exclude the check-point service of cell cycle whose absence permits
persistent growth. (10).
At the mechanistic level, several types of surface proteins are involved in the interactions of
metastatic cells through cell adhesions molecules (CAMs) and integrin to establish proper
invasive cell in distinct territories of body. These interactions play a vital role in allowing the
signal to enter into intracellular part of cell. (11). Alterations in expression of these CAMs in
Ig super family play a critical role in the invasion and metastasis processes like change in the
expression of integrin help to invade and proliferating cancer cells (12). E-cadherins are
Box 1
Hallmarks individuality of cancer:
1. The quality to prolong chronic proliferation by deregulating explicit of growth promoting signals.
2. Evading growth suppressors either by persecuting the effect of tumor suppressor genes or by annulling
cell-cell contact inhibition.
3. Resisting cell death and becoming tolerant to multiple apoptotic mechanisms.
4. Unlimited replicate potential by protecting against telomere erosion.
5. Inducing angiogenesis via production of vascular endothelial growth factors.
6. Activating invasion and metastasis by altering expression of cell-cell adhesion molecules.
Because of the proceeding impact of these hallmarks, they are usually mentioned as a starting point to study
for the anticancer strategies. Several progresses in these research studies has revealed two new emerging
hallmarks of cancer to this list – reprogramming of energy metabolism and evading immune destruction
(Douglas Hanahan, Hallmarks of Cancer: The Next Generation, 2011).
Box 2
Molecular and cellular pathways linking inflammation and cancer
Schematically, two pathways have emerged; the intrinsic and extrinsic. In the intrinsic pathways, activation
of different classes of oncogenes drives the expression of inflammation-related programs that guide the
construction of an inflammatory milieu. In the extrinsic pathways, inflammatory conditions promote cancer
development. The key factors of the inflammation-mediated tumor progression are transcription factors,
cytokines, chemokine and infiltrating leukocytes. (123).
widely known as a suppressor of invasion and metastasis by epithelial cells, which must be
altered by cancer cell to overcome to progress (13), (14).
As it is known that these CAM molecules are the basis of interaction between cells, so if put
a gate on the expression of these molecules in tumor cells then it’ll be easier to find a cure for
the responsible tumor type. For all malignancies, executing treatments is much harder to
come by. May be a after few years, it may be possible to sequence according to the soul of
tumor and offer up the diagnosis, not of colorectal cancer, but of an RB/TP53/APC/KRAS
malignancy like report. Then different combination system in the form of pills will be
recommended.
Over the past decade when viewed from all these perspective, tumors have been increasingly
recognized as abnormality whose complexity approaches and may even exceed that of normal
healthy tissues. It’s better to understand the biology of a tumor through studying the
specialized cell types within it as well as the ‘tumor Microenvironment’ that they are
interlinked.
These are the overall assessment about different recent aspects of foundation studies of
cancer biology. They were illustrated about cells involved in the proliferation, soluble factors
which enhance the progression, signaling molecules whose alteration promote the survival of
cancer cells, extracellular matrix and mechanical cues that can promote transformation,
invasion and protect the tumor from host immunity. These all aspects indicate toward the link
of cancer progression and tumor microenvironment.
Tumor microenvironment and Immunomodulation
Tumor is composed of stromal constituents like inflammatory cells, endothelial cells which
finally arrange tumor microenvironment to promote tumor growth, progression and survival
(15). In 1909, Paul Ehrlich originally hypothesized that the progression of neoplastic cells
could be constrained by host immune system. In the response to tissue injury induced by
tumor-host interaction, stromal cells represent a defense mechanism of the organism and
release cytokine, chemokine, growth factors, and lipid modulators that exert their effects. For
example, Transforming growth factor- beta (TGF-β) is a critical regulator of tumor
progression and a potent inhibitor of cell differentiation which is secreted by multiple cell
types within the tumor microenvironment (16). This defense mechanism suppressed by tumor
cells which effectively alter the growth inhibitory effect of stromal cells and harness the
growth promoting characteristics for their own beneficial (17). The purpose of studying
limitations of tumor microenvironment and immunomodulation is to identify the optimal
treatment for each individual patient to maximize the treatment benefit.
Tumor and immune cells both have been found to secrete inflammatory mediators which play
a dual role in tumor development. On the one side, they promote tumor survival whereas on
the other side they promote immune surveillance mechanisms against tumor cells. It is
during the equilibrium phase that the interplay between several components of the immune
system and the tumor will define the final outcome of the immune response. This immune
selection may be produced due to recognition of cytokine and chemokine in immune system.
The cytokines are endogenous substances behaving like hormones present within the immune
system, sending messages between immune cells mediated by receptors, stimulation of
immune cells showing some direct effect on tumor cells and infections, (18).
Several studies are presently going on chemokine and cytokine levels in tumor cells at
various stage of their growth phase to understand the levels of these factors causing
promotion or inhibition of tumor growth. They revealed that cancer cells amend only those
pathways which make those cells accessible to use them as proliferative agent without any
checkpoint. A new era of effectively harnessing these studies to treat and prevent cancer has
begun. There are several therapies are now approved for cancer treatment and others are on
clinical trials. These studies may lead to elevated interest and movement in developing novel
therapeutic strategies for oversetting the balance of host-tumor interaction toward tumor
growth inhibition. It is clearly practicing to prosper advanced therapies to re-engineer the
overall host ambience and tumor microenvironment to rattle pathways of suppression and
immune system tolerance.
Overall, Chemo, radiation and other traditional therapies are standard therapeutic modalities
for patients suffering from cancer. But the extensive use of antibiotics and chemicals in
human patients has shown several side effects with less efficacy of treatment (19). In another
case, William Coley (1891) attempted to demonstrate the immune system’s ability to cause
inflammation which further stimulates antibacterial phagocytes that might kill spectator
tumor cells also. Further this theory was discounted but it gave a channel towards Cancer
immunotherapy (20). Cytokine being natural messengers of immune response which on
administration eradicate solid tumor, thereby exhibiting bright side of cytokine therapy.
Presently, many researchers have tried the combination of Chemotherapy / radiotherapy with
different specific form of immunotherapy. Although, it is well known that chemotherapy /
radiation cause several lethal effect to rapidly progressive cells like hematopoietic cells,
epithelial lining in gut and sperms. So it is recommending that use chemotherapy /
radiotherapy to deplete tumor load and then introduce immunotherapy to final depletion of
tumor and prevent the re-growth (21).
Recent studies highlight the benefits of specificity of Immunotherapy for cancer which can
enable the induction of immune response via a variety of ways such as
Guidance of co-stimulatory signal molecules
During tumor progression, a tumor cell mutates the co-stimulatory genes and affects the
function of co-stimlatory molecules which are required for the interaction of antigen
presenting cells (APC) with immune cells. Like, B7 (a co-stimulatory molecule) on APC
interact with its receptor i.e. CTLA -4 and CD28 on T cells which provide signal for further
activation of T cells. The absence of expression of B7 molecules renders tumor growth
imperceptible to the immune system while the induced expression of B7 molecule prevent
tumor from effective cytolytic T lymphocyte (CTLs). The transfection of B7 molecule in
tumor cell micro-environment led to tumor regression due to induction of effective CTLs
response. The co-stimulation of T cell upto a balanced level may have valuable therapeutic
purpose for generating immunity against tumors (22), (23).
Augmentation of APC activity with monotherapy
This monotherapy attempts to utilize the expression of tumor-specific antigens in the
development of antigen-specific immunotherapy. In one approach, the result approved the
practicability to induce monocyte-derived DCs in the presence of GM-CSF and IL-4 by co-
culturing with melanoma tumor cells (24). This study highlights that tumor cells can be
genetically improvised to be able to identify tumor specific antigens (TSAs) through
activation of different cytokines and other molecules. If patients DCs co-culture with tumor
fragment from patients in the presence of recombinant GM-CSF, then it can induce the
activation of APCs, Th cells, and CTLs specific for TSAs. After decades of research, it is
shown that DCs are the central effecter cells of immune system. Thus, DCs highlighted as
beneficial target generate therapeutic modality against tumor.
Immunotoxins against tumor
Immunotoxins are bifunctional molecules that can induce intracellular toxin activity to kill
the target cells via binding of antibody with specific antigen (25). In an approach,
Immunotoxins as adjuvant which binds with binding domains of monoclonal antibodies,
designed for Cell-targeted treatment. In this design, Monoclonal antibodies were able to
target hematopoietic differentiation antigens whereas immunotoxin have been shown to be
highly cytotoxic toward malignant cells. These therapeutic designed will likely play a
promising role in treatment of leukemia (26). The incidence of antitoxin antibodies
production in bodies after multiple cycles of treatment causes immunogenicity. There are
several approaches are under trial to reduce immunogenicity for the treatment of solid tumor.
Monoclonal Antibodies
The monoclonal antibodies are a promising part of immune system which recognize and
binds with foreign antigens expressed on the cell surface. After binding, they highlight them
for further action of destruction of abnormal cells via antibody dependent cellular
cytotoxicity (ADCC). In addition to other therapy, they are designed to enhance body’s
immune responses which assist to act against cell growth factors. The induction of adaptive
anti-tumor immune responses continues to hold for immunization against cancer.
There is a practice developing for the manipulation of antibody Fcγ domains to further
improvement of efficacy and affinity for Fcγ receptor (s) (27). Still alots of things needs to be
exploring about monoclonal antibody as therapeutic agent like as adjuvant, combination with
other molecules which can activate co-stimulation, CTLs activation or some interleukin
which will give varieties of ways to target and treat tumors in patients.
There are several therapeutic monoclonal antibodies against cancer approved in clinical trial
like, Rituximab (for Non-Hodgkins lymphoma), Ipilimumab (for melanoma), Trastuzumab
(for Breast Cancer), Bevacizumab (For Colorectal Cancer), Cetuximab (For Colorectal, Head
and Neck Cancer), Panitumumab (For Metastatic colorectal carcinoma), Brentuximab
vedotin (For Hodgkins lymphoma), Alemtuzumab (For Fludaribine – refractory chronic
lymphocytic leukemia) (28).
In addition to these, several approaches are under trial to increase the ability of antibodies to
elicit an anti-tumor immune response.
Cytokine Therapy
Cytokines are the important regulator of innate immune system: natural killer (NK) cells,
neutrophills and macrophages also rectify adaptive immune system with T and B cell
immune responses. Usually cytokine functions in cascade, so individual cytokine therapy are
not a promising therapy in present. To overcome these drawbacks, there are several trials
going for the combination of different cytokine to target a specific mechanism directly. These
findings provide evidence for the unique therapeutic opportunity based on selective
interference with the action of pro-inflammatory and tumor-promoting cytokines. It has been
established that cancer promotion and exacerbation are linked with the cytokines produced by
activated innate immune cells. Here we provide an overview of the updated understanding of
the role of inflammation-induced cytokines in development of novel therapeutic strategies
(Table 1).
Cell-based Immunotherapies of Cytokine
Cytokine function as autocrine and paracrine, so they function locally or any distant part of
body to enhance or suppress immunity. In cytokine therapy for cancer, cytokine uses to
induce ant-tumor immunity to fight against cancer. Here we will examine cytokines which
are currently under evaluation for cancer therapy.
Tumor Necrosis Factor - alpha (TNF – α)
In 1975, TNF-α was extracted from the serum of mice treated with ‘Coley’s toxin’ and later
on showed the hemorrhagic necrosis of mice tumor (29) , (30). Tumor necrosis factor – α
(TNF- α) is a pleiotropic cytokines involved in regulating diverse functions like apoptosis,
cell survival, tumorigenesis, autoimmunity, inflammation, and immune response via two
receptors i.e. TNFR1 and TNFR2 (31).
The interaction of TNF- α molecule with TNFR1 or TNFR2 receptor leads to the activation
of various signaling pathways in cell microenvironment (32). In an experiment when TNF-α
is administered via ILP (Isolated limb perfusion) into tumor cells, then it was bound with
TNFR1 soluble receptors on the cell surface (33). It is well known that TNFR1 is less in
count on cell surface which inhibit the cause of cytotoxicity. The effect of binding of TNF - α
to TNFR1 results in hyper permeability of the neoplastic vessels and strong extravasations of
erythrocytes resulting into massive hemorrhage and necrosis in tumor (34). This excessive
hemorrhage is responsible for the inhibition of tumor associated vasculature and the growth
of malignant cells. Several clinical studies have also reported that low dose of TNF - α may
induce these antitumor effects (35), (36).
In case of tumor microenvironment, tumor or inflammatory cells induce macrophages, T
lymphocytes and natural killer (NK) cells, which further harvest TNF-α, which induces
gene-encoding NF-κB dependent anti-apoptotic molecules and also promote the tumor cell
survival (37), (38). TNF-α further stimulate the production of genotoxic molecule Nitric
oxide and reactive Oxygen Species that can lead to DNA damage and reduce the antigen-
induced cytotoxicity or enhance the tumor progression through suppression of T cell
responses and cytotoxic activity of activated macrophages (39). Oxidative stress caused by
ROS and NO is a contributor to senescence, which could be an option for developing therapy
for tumor treatment. These studies indicate vast therapeutic applications of TNF-α in tumor
cell depletion.
Another attribute of TNF-α induces hemorrhagic necrosis in certain set of tumor types in
which synergistic antitumor effect of the combination treatment with TNF - α and
chemotherapeutic drugs as compared to TNF-α alone. (40). TNF - α effect during solid
tumor treatment cannot be caused by a direct cytotoxic or cytostatic effect toward the tumor
cells (41). However, cell death induction by TNF - α plays only small role in necrosis, but by
combination with Fas ligand, activation of NF-κB pathway which transcript the inhibitory
proteins that interfere with death signaling in tumor cells, can be an alternative blueprint for
developing therapy (42). These strategies of developing combined therapy are very relevant
to show the antagonistic effects of TNF-alpha against tumor growth. The same set up also
revealed that TNF-α improved the accumulation of chemotherapeutic drugs in tumor.
TNF-α super family represents an active target for drug development. Some of the approved
therapeutics are primarily either a soluble receptor which binds the circulating TNF- α and
prevent the interaction between TNF- α and cell surface receptors, or antibodies against TNF-
α. Various antagonist against TNF family have been approved like Golimumab (a humanized
TNF- α monoclonal antibody) (43), Certolizumabpegol (a PEGylated Fab fragment of a
humanized TNF antibody) (44) and still more are in the process of approval.
As described, members of TNF- α are involved in mediation of chronic inflammation, there
is a need to regulate the members of TNF- α family members however the role of TNF-β is
still elusive. In several medical cohort studies in United States in which cytokine was
delivered at appropriate concentration to the tumor site, there are no evidences of
enhancement of cancer growth associated with Tumor necrosis factor alpha antagonists TNF
therapy in either analysis or follow-up after the patients discontinued therapy (45). So there is
much left yet to be learned about TNF-superfamily, and we expect this information to be
forthcoming.
Of note, there is a need to develop a strategy to identify the patient stratification criteria for
the development of individualization of therapy. Therefore, research in the future should be
focus on not only recognizing integrated TNFL/TNFR-based combined immunotherapy but
also on the individualization of appropriate patient treatment.
 Interleukin – 6
It is well established that innate and adaptive immunity released several pro-inflammatory
cytokines which contribute to tumor promotion and progression. It is a glycoprotein that
shows pleiotropic activity through binding with its transmembrane receptor (46).
Among these, the IL-6 receptor complex is a heterodimer structure which consist of IL-6Rα
and glycoprotein 130 (gp 130), that is reckon as key anti-inflammatory myokine, pro-
inflammatory cytokine, antiapoptotic and growth-promoting factor (47), (48). Experiments
based on link between inflammation and carcinogenesis entangled few constituents of
inflammatory avalanche such as IL-6 known for its key role in tumor growth and survival
(49). Other studies also revealed the multi-utility of IL-6 during chronic inflammation like it
induces Th17 cells and inhibits the differentiation of Tregs cells. During IL-6 signaling,
soluble IL-6 receptor (sIL-6R) secretion can induce IL-6 trans-signaling which further
promote the development of cancer cells. In addition, it has been suggested that shedding of
sIL-6R from carcinoma cell surface promotes the T cell survival and increase the production
of more IL-6 by T cells (50). Such theories suggested that opposing role of IL-6 might be
therapeutically useful for the treatment of colitis-associated colon cancer (CAC) (51).
The indefinite therapeutic methods have been developed for the treatment of disease
including anti-IL-6 or anti-IL-6R antibodies, soluble gp130Fc (sgp130Fc) and selective small
molecule JAK inhibitors which directly/indirectly inhibit the IL-6/STAT3 pathway that is
directly involved in sporadic and inflammation-associated CRC development (52) . The first
therapeutics targeting of IL-6/STAT3 pathway has been started in clinical studies through the
anti-IL-6 antibodies during the 1990s. However, these treatments only produced a limited
response. Another strategy to inhibit IL-6/STAT3 signaling is the inhibition of JAK
activation through ruxolitinib (53) and CEP-33779 i.e. JAK1 and JAK2 inhibitor could
successfully reduce tumor growth in clinical studies (54), (55). The studies of these arrays
can yield new insights on the effect of IL6/STAT3 deregulation.
All of the treatments mentioned above inhibit physiological functions of IL-6 through
inhibition of signaling which has been achieved by binding of sgp130Fc to IL-6 receptors
complexes (56).
It is mentioned in several studies that components of IL-6 signaling pathway such as IL-6, IL-
6R, JAK has been shown to promote tumor proliferation and anti-apoptotic effects in tumor
cells which can be proposed as promising target to achieve therapeutic effects (57), (58). IL-6
is not only involved in tumor growth is also related with the interaction within tumor
microenvironment which may also suggest a target for the development of novel IL-6
antibody based therapies like IL-6 inhibitors are being developed include human anti- IL-6R
Ab (Sarilumab/REGN88/SAR153191) (59), anti-IL-6R nanobody (ALX-0061), anti-IL-6
Abs such as sirukumab (CNTO 136), BMS – 945429 (ALD518), olokizumab (CDP6038),
and MEDI5117, and soluble gp130-Fc fusion protein (FE301) (60) which are in clinical
trials for different disease in human body.
In addition, Chronic inflammation causes DNA damage which has further shown affecting
several pathways including cell based homeostasis like cell-cycle regulation, apoptosis, DNA
repair (61). These conditions nourish the production of IL-6 in patients. However, attenuating
chronic inflammation with surgery, radiation and chemotherapy as well as combining these
therapies with IL-6 inhibition may be one of the ways to decrease inflammation and add
prognostic values to different methods (62).
Recent researches have concluded the potential for IL-6 as a prognostic factor. So there is
need of clinical studies to further evaluate potential which could help direct additional
treatment strategies in the future. However, additional studies and appropriate clinical trials
need to be done to make sure the effectiveness of anti-IL-6 therapeutic modalities in cancer
patients.
 Interleukin-17
Recent studies on the role of different T cell subset named Th17 identified by the production
of IL-17 (IL-17A) as well as related cytokine IL-17F cytokine, explain as implication of IL-
17 in inflammatory responses primarily as a proinflammatory factor (63). IL-17A expresses
on tumor cells and develops antitumor immunity in immunocompetent mice which
suppresses tumor progression whereas in some cases it also enhances the angiogenesis in
immune-deficient mice which promote tumor progression (64).
IL-17 is induced by IL-23 that responds to the invasion of immune system and induces
destruction of foreign antigen cellular matrix. IL-17 is the signature cytokine of a unique
lineage of CD4+ T cells termed as Th17 and also known as a potent mediator in delayed-type
reactions by enhancing chemokine production throughout body to recruit immune cells to the
site of chronic inflammation (65). The properties of IL-17 like foreign antigen destruction,
induction of chemokine to the site of inflammation, synergistics action with TNF and IL-17
are very promising features to target it as a strategy against tumor reduction.
IL-17 also acts as an angiogenesis factor whose up-regulation may results in blockade of
angiogenesis in tumor and provokes the host inflammatory response to tumor progression
(66). The up-regulation of IL-17F includes modulation of cytokines regulating IL-17F
production (67) whereas blocking of IL-17A signaling may inhibit tumor angiogenesis but
sustains the risk of diminishing host antitumor response. Tumor endothelial marker 8 (TEM8)
expressed on human tumor vasculature is the main factor in promotion of angiogenesis (68).
The anti-TEM8 antibody can be used to inhibit the expression of TEM8 and it is also helpful
to segment the other anti-angiogenesis agent’s activity. By controlling the diminishing of
antitumor response and increase in the blockade of tumor angiogenesis by using the
synergistic effect of different anti-angiogenesis agent, it is possible to inhibit the growth of
tumor.
It is clear that IL-17 is skillful of exerting multiple effects within tumor microenvironment
with bidirectional effects in tumor growth, so further investigation should be approached with
a definite knowledge of complex interaction of IL-17 within the tumor microenvironment.
Therefore, we propose that there should be some novel inhibitory molecule and
immunomodulators mixed with these cytokine based therapy to achieve the beneficiary effect
in treating malignant solid tumors. Several studies are under progress to investigate the
relation, functions and involvement of IL-17F and IL-17A in tumor progression which
provide an unusual strategy for any therapeutic application of these cytokine in therapy for
cancer.
 Interleukin-12 and Interleukin-23 (IL-12 cytokine family)
Both of these cytokine are the members of IL-12 cytokine family and can be released by
Antigen presenting cells (APC) like dendritic cells, macrophages, and human B-
lymphoblastoid cells into the inflammatory sites upon microbial infections (69). Recent
studies have shown that there are several similarities between IL-12 and IL-23 receptor
subunits and signaling but still they impel different immunological pathways and exhibit
different response towards tumor development. Research evidences has shown that IL-12
cytokine family members contain anti-proliferative and pro-apoptotic functions like IL-
12Rβ2 which could act as a tumor suppressor gene and IL-12R transfected B lymphoma cells
significantly deplete cell proliferation in animal models (70).
Although, it is well established that IL-23 enhance the production of TH17-mediated
response and induce IL-17 production which further promote end-stage inflammation (71),
whilst the proliferation of memory T cells and induction of IFN-γ are done by IL-12 which
play as a major role in exhibitions the antitumor immune response of IL-23 (72).
Interleukin-12 is a disulfide-linked heterodimer cytokine, composed of two subunits i.e. IL-
12p40 and IL-12p35 (73), (74), whereas another cytokine IL-23 is also an heterodimer
molecule composed of IL-12p40 and IL-23p19 subunits and both of these cytokines belongs
to the IL-12 family of proinflammatory cytokine (75). IL-12 binds to its heterodimer
receptors i.e. IL-12Rβ-IL-12Rβ2, whereas IL-23 binds an IL-12Rβ1-IL-23R heterodimer
(75). This heterodimer cytokine complex stimulate the expression of their receptors to
maintain the expression of several proteins involved in IL-12 based signaling pathways in
NK cells to enhance the cytotoxic activity.
IL-12 has been shown to exhibit antitumor activity through the immune-stimulatory effects
on certain T helper cells (Th1), as well as on cytotoxic T lymphocytes (CTL) and natural
killer (NK) cells (76), (77), (78). There are certain other cytokine like IL-1, IL-2, IL-15 and
tumor necrosis factor-α, which act synergistically with IL-12 to induce the secretion of IFN-γ
which exhibit direct toxic effect on tumor and antiangiogenic activity (77), (79), (80). This
type of enhanced functional response of IL-12 is demonstrated by IFN-γ production and
killing of target cells. As it is clear that IL-12 has the ability to induce anti-angiogenic
activity and immune system, there should be studies on using IL-12 as a possible anti-cancer
therapy.
Another synergistic property of IL-12 and IL-23 is that they both activate TYK2 and JAK2 as
well as STAT1, STAT3, STAT4 and STAT5, whilst IL-12 alone activates STAT4 efficiently
and IL-23 preferentially activates STAT3 (75). IL-23 has been known as a key factor
controlling inflammation in peripheral tissues which is mediated by induction of JAK/STAT
pathway following IL-23 binding.
As described above that IL-23 suppresses the innate immune response during tumor
progression and metastasis. Based on these hypotheses, IL-23 neutralization strategies may
promise to modulate immunosuppression in combination with immunostimulatory agents
(81). In an experimental study, α-IL-23 mAb has been given to wild-type mouse resulted in
fewer lung metastasis than in mice treated with control Ig (α-AGP3), which respond in the
form of NK cell-mediated antitumor immune response of α-IL-23 mAb (82). Collectively, the
use of α-IL-23 mAb in combination with IL-2 mAb treatment significantly inhibited
subcutaneous growth of established mammary carcinoma and suppressed established
experimental and spontaneous lung metastases.
In other trial studies, the anti-IL-23R treated mice displaced a significant reduction in the
number of tumor infiltrating Tregs compared with control antibody treated mice. These
studies exhibit another role of IL-23 driven STAT3 activation in tumor-infiltrating Tregs with
the addition of IL-23R dependent signaling pathway (83).
Since past 15 years, IL-12 cytokine family based therapies have acquired so much attention
and are apparently developing more effective therapy for the survival and treatment of
cancer-bearing patients (84). Of note, current studies postulated that over-expression of IL-
23R mainly suppress the expression of anti-apoptotic factor like Bcl-xL and induce the
intrinsic apoptotic pathways in 293ET and HeLa cell lines during trial (69). But tumor
immunosuppression still remains one of the major barriers in the development of novel
cancer therapies and vaccines. Several studies have been shown that initial stage of cancer
treatment, mAb neutralizing IL-23 alone and exerting a temporary effect. So there is a need
to target the combination of α-IL-23 mAb with other cytokine based antibodies to exert
potential effect in treatment of cancer.
 Interleukin-10
Interleukin-10, also known as human cytokine synthesis inhibitory factor (CSIF), having a
pleiotropic effects in inflammation and immune-regulation which directly affect the function
of antigen-presenting cells by inhibiting the expression of MHC and co-stimulatory molecule
(85), (86). In 1989, Mosmann and his coworkers postulated that T helper 2 (Th2) cell clones
produced a cytokine which inhibit the synthesis of interferon (IFN) in Th1 cell clones (87),
and inhibit the NF-κB activation through ill-defined mechanisms (88) (89), is known as
interleukin -10 (IL-10). It is also known for the production of proinflammatory cytokine like
TNF-α, IL-6, and IL-12 (90) and down-regulation of the expression of MHC class II antigens,
co-stimulatory molecules on macrophages.
Several transcription factor responsive elements promotes the major source of IL-10 i.e.
macrophages to modulate the production of IL-10 through these exogenous and endogenous
factors such as endotoxin via Toll-like receptor, tumor necrosis factor (TNF) , catecholamines
and cAMP-elevation drugs via protein kinase A (91), (92), (93), (94), (95), (96). In contrast
to these observations, IL-10 produced by B-cell lymphoma functions as an autocrine growth
factor (97). These previous mentioned reports concluded that IL-10 production within the
tumor microenvironment is a common phenomenon in cancer metastasis which might be the
major hurdle for anticancer immunotherapy such as cancer vaccines. In case of metastatic
melanoma patients, it was reported that high IL-10 production were used for synthesis of
homologous melanoma cell vaccine estimated fewer survival after vaccine treatment (98).
In another trial, anti-IL-10 antibody treatment has been administered in two mouse models of
acquired hepatitis (99). This study revealed that IL-10 plays a protective role down regulating
the expression of several other inflammations causing cytokine including IL-12, TNF-α and
INF-γ (100).
IL-10 is able of affecting the tumor cells and inhibiting the immune cells which may convert
tumor cells to a CTL resistant phenotype. Kiessling and his coworkers reported that IL-10
treatment results in 50% reduction of expression of MHC class-I in human melanoma cells
and 100% inhibition of CTL-mediated lysis of tumor cells (101). The consequence of IL-10
has been demonstrated in transgenic mice by uncontrolled growth of immunogenic tumors
because of the blockade of the antitumor immune reaction. The anti-IL-10 antibodies
treatment thus reinstates the anticancer response (102).
Several studies conducted on Systemic Lupus Erythematosus (SLE) patients concluded that
treatment of anti-IL-10 antibody to SLE-patients results in the lessening of autoantibody
production due to the depletion of IL-10 (103). Some other experimental models suggest that
IL-10 may retain antitumor response and immune stimulation (104), (105) like transfection of
mouse melanoma cell lines with IL-10, extorts induced immunogenicity following a strong
lymphocyte and antibody-dependent immune memory (106).
Additionally, IL-10 induces the production of Treg cells (107), (108). So rather than giving
anti-IL-10 therapy alone, it is suggested that a combination of anti-IL-10 antagonist with
other immune-stimulants such as cancer vaccines, other cytokine will be more synergistically
effective in inhibiting the tumor-specific immune tolerance and promoting antitumor immune
responses. It suggests that balance of IL-10 should be maintained to regulate the immune
response against any infection in body. These trials give new insights to the IL-10
immunobiology to become a new therapeutic target.
In contrast, complex effect of IL-10 may cause reduced potency of treatment depending upon
IL-10 rather than promising effect of other combined approaches. Given the overall outcomes
of systemic IL-10 therapies of disease, it has become apparent that future studies on the
sources of IL-10 and kinetics of IL-10 production may lead to the identification of new target
with better therapeutic efficiency and also facilitates the development of treatment strategies
aimed at regulating inflammation. These studies will have to understand the details of
circulating levels of cytokines and their gene variations inside the patients which will assist in
development of therapy with defined parameters of treatment side effects.
These above proclamation about IL-10 cytokine revealed the utility as an essential
immunoregulatory which favorably react toward different treatment, evaluating the assistance
of IL-10 for counteracting the inflammatory effects in the human body. The generalized
therapeutic effect of IL-10 in cancer treatment is hard to predict as both tumor-suppressive as
well as tumor-promoting effects have been demonstrated. Since, IL-10 is produced in
metastatic stage of cancer, it insinuate more equitable to clock the effects of IL-10, rather
than administrating IL-10 to cancer patients. These features can assist in the development of
recombinant therapy for several diseases.
 TGF-β
TGF-β is known as a secreted polypeptide skillful of inducing fibroblast growth and collagen
fructification which was further discovered as a pervasive and essential key regulator of
proliferation, differentiation, migration, cell survival, cell survival, angiogenesis and
immunosurveillance (109), (110), (111). Tumor progression is stimulated by TGF- β by two
mechanisms: firstly intrinsic activity and, another extrinsic activity (112).
During the growth phase of tumor, excessive amount of TGF-β is produced whereas in
normal epithelial cells, it is known as a potent growth inhibitor. Normally, TGF- β blocks the
G1 stage of normal cells to regulate the balance of growth in body but when a cell is
transformed into a malignant cell TGF- β signaling pathway is altered and it losses the
control on cell growth resulting into cancer and surrounding cells escalated the production
and increase in their own TGF- β which acts on microenvironment (113). It causes
immunosuppression and angiogenesis, which makes the malignant cells violent.
TGF-β has the dichotomous function in human cancers. First it is having the capability to act
as a tumor suppressor (via its effects on replication, proliferation, and apoptosis) and another
as a tumor promoter (via its effects on progression, metastasis, invasion and angiogenesis)
(114). So profiling of TGF-β ligand (in serum, urine, and tissue), TGF-β receptor levels and
TGF-β mRNA (in tissue) may serve as a diagnostic and therapeutic tools for cancer
treatment.
It is predicted that strategies such as inhibition of signaling pathway, monoclonal TGF-β-
neutralizing antibodies, large molecule ligand snare, lowering translational efficiency of
TGF-β ligand by antisense technology and estranging TGF-β receptor I/II kinase function by
inhibitor molecule which are the most conspicuous methods being investigate these days
(115), (116). For example, first is the utilization of soluble TGFIIR (human α2-
macroglobulin) plasma protein that binds TGF-β isoform, blockade the interaction between
cytokine and its receptor (117) and two other TGF-β1 and TGF-β2 specific humanized
monoclonal antibodies like CAT-192 and CAT-152 respectively, are going under clinical trial
for cure of fibrosis and their development will embolden the therapeutic application to anti-
cancer therapy (118), (119).
Antisense TGF-β treatment in a variety of cancer has also been delineating the response in
the form of reactivation of number of different therapeutic scope. In addition, it has been
shown that combined treatment with tumor cell vaccines and antisense TGF-β therapy or
monoclonal antibodies therapy respond synergistically and downsize the tumor volume and
escalate the survival benefit (120), (121), (122).
Now it is well known that increased quantity of TGF-β has superlative role in tumorigenesis
due to reprogramming of immune surveillance which makes it a very advisable target for
different therapeutic modalities like monoclonal TGF- β antibody, antisense oligonucleotide
or receptor kinase inhibitors. From different outcome of studies, it is revealed that continuous
suppression of TGF- β signaling pathway may lead to harmful off-target effect. So there is a
need to focus on combined therapy with these inhibitors for the therapeutic profit in future.
There are three leading therapeutic targeting of TGF- β in clinical trials: TGF-B antibodies,
antisense oligonucleotide, and receptor kinase inhibitors. These all strategies have their own
limitations. So it is necessary to reengineer the TGF- β inhibitors within a combinational
therapy.
Of note, it is known that increased level of TGF-β in tumor microenvironment induce the
tumorigenesis through various processes, thereby making it possible to target these for
therapeutic development. Conclusively, the precise mechanisms involved in transformation
of TGF-β during malignancies needs to be deeply understood only. After that, it will be
possible to prosper successful therapies as well as provide new therapeutic targets to fix the
normal TGF-β function.
Conclusion
We have sought here to revisit, refine, and extend the concept of therapeutic targeting of
cytokine, which has provided a useful conceptual framework for understanding the complex
foundation of cancer biology. Advancement of role of cytokines in immune and
inflammatory diseases has promoted the development of cytokine-based therapies. Cytokine
therapies have tremendous capabilities for treating a variety of diseases either by inhibiting or
restoring the activity of specific cytokines. As cancer is caused due to an alteration in cellular
programming, cytokine therapy is the one best choice to treat cancer. However, as cancer is a
part of the wound healing process, single cytokine therapy may not be effective. This might
increase the likelihood of cancer recurrence and development of alternate signaling pathway
to facilitate tumor cell growth. So it is necessary to work on dominant module in modern
immunologic research, which is the illustration of commutative effect of different cytokine
therapies. One of the approaches is known as Gene therapy as they enable therapy to restrict
to tumor cells only which will decrease the side-effect and increases the efficacy of therapy.
As prescribed above, several studies indicate the role of combined cytokine therapy like the
combination of TNF-α and IL-6, it is evident that IL-6 signaling embellished the protraction
of TNF-α, this interference restrict tumor progression and IL-6 production.
IL-23 induces IL-17 production which further endorses end-stage inflammation whilst the
rise of memory T cells and induction of IFN-γ are made by IL-12 which serves as a foremost
role to reveal the antitumor immune response of IL-2. As previously revealed that IL-12
based therapy is more valuable with smaller tumor burden, so there is a need to investigate
the role of combination of IL-10 with other therapy as an adjuvant to get more effective
therapeutic effects. The contribution of CD4 T cells, CD8 T cells, and IFN-γ and NK cells in
induction of antitumor effect of IL-12 therapy is still unclear and itmay give a better
opportunity to investigate the role of combined therapy in treatment.
Through the rapidly growing research of intricate signaling system of different cytokines,
new effective therapies have become available to treat oncology patient like blocking or
neutralizing cytokine action with monoclonal antibodies and one of the drugs that inhibit
release of inflammatory cytokines, such as tumor necrosis factor-α (TNF- α). It is thus
predictable that new combined therapies of cytokine which are based on innovative
technologies will progressively reach the efficacy of new cancer treatment strategies. In some
cases, the cure afforded of by these cell based cytokine therapies may prove to be even more
effective than chemotherapies and other traditional treatment methods.
In my vision, I can foresee the limitative single cytokine therapy being overcome by the help
of combined cytokine therapy along with gene therapy.
What do we expect in the future?
Combined immunotherapy may be the next great hope in the treatment of cancer. Several of
these trials have shown impressive clinical results signaling that we are in the entry and
exciting phase of cancer treatment. It is likely our responsibility to combat cancer from all
fronts. Additional studies required to define the mechanism of cytokine cascade in body
which will help to reduce the opposite effect of therapy and make it specific for every cancer
type. One of the hurdles have been identified, i.e. immunogenicity, which may be resolved by
depleting or suppressing T and B cell epitopes. At present, nonspecific and specific
immunotherapy combination showing promise for another potent strategy for treatment.
Even, the effect of any aforementioned therapy module in combination with some traditional
cancer treatment method is another avenue, as it is come out in trial in terms of duration /
remission period with cytokine and chemotherapy.
All through with these concerted efforts, our decisive ambition may be a long-lasting anti-
tumor immune response that can be maintained throughout patient’s lifespan.
…………………………………..
References
1. Meng X, Zhong J, Liu S, Murray M, Gonzalez – Angulo AM. A new hypothesis for the cancer
mechanism. Cancer Metastasis Rev. 2012 Jun 31: 247-68.
2. Bhatt A N, Mathur R, Farooque A, Verma A, Dwarakanath B S. Cancer Biomarkers – current
perspectives. Indian J. Med. Res. 2010: 132: 129-149.
3. Kinzler K.W., Vogelstein B.. Lessons from hereditary colorectal cancer. Cell. 1996; 87: 159-170.
4. Laubenbacher R., Hower V., Abdul Jarrah A., Torti SV, Shulaev V, Mendes P. et al., A Systems
Biology View of Cancer. Elsevier. 2009 Dec 2; 1796: 129-139.
5. Hanahan D., Weinberg RA. Hallmarks of Cancer: The Next Generation. Elsevier. 2011 March 4;
144: 646-674.
6. C Athena Aktipis CA, Nesse RM. Evolutionary foundations for cancer biology.Evo app. 2013; 6:
144-159.
7. Cavallo F, Giovanni CD, Nanni P , Forni G , Lollini PL. The immune hallmarks of cancer. Cancer
Immunol Immunother. 2011 Mar; 60(3): 319-326.
8. Lemmon, M.A., and Schlessinger. Cell signaling by receptor tyrosine. Cell. 2010; 141: 1117-34.
9. Matsubara H, Suzuki K. Recent Advances in p53 Research and Cancer Treatment. Journal of
Biomedicine and Biotechnology. 2011; 1-7.
10. Burkhart D.L., and Sage J. Cellular mechanisms of tumour suppressionn by the retinoblastoma
gene. Nat. Rev. Cancer. 2008; 8: 671-682.
11. Berx G, Roy FV. Involvement of members of the cadherin superfamily in cancer. Cold Spring
Harb Perspect Biol. 2009 Dec; 1(6).
12. Johnson, J. P. Cell adhesion molecules of the immunoglobulin supergene family and their role in
malignant transformation and progression to metastatic disease. Cancer Metastasis Rev. 1991; 10:
11-22.
13. Cavallaro U, Christofori G. Cell adhesion and signalling by cadherins and Ig-CAMs in cancer.
Nat. Rev. Cancer. 2004; 4: 118-132.
14. Coussens LM, Werb Z. Matrix metalloproteinases and the develoment of cancer . Chem. Biol.
1996; 3: 895-904.
15. Liub, Jing Zhanga and Jinsong. Tumor stroma as targets for cancer therapy. Pharmacol Ther.
2013; 137: 200-215.
16. Witz I P, Levy-Nissenbaum O. The tumor microenvironment in the post-PAGET. Cancer Lett.
2006; 242: 1-10.
17. Marie-Aude Le Bitoux, Ivan Stamenkovic. Tumor-host interactions: the role of inXammation
Histochem Cell Biol. 2008; 130: 1079-1090.
18. Balkwill F. Cancer and chemokine network. Nat. Rev. Cancer. 2004; 4: 540-550.
19. Shiao SL and Coussens LM. The Tumor-Immune Microenvironment and Response to Radiation
Therapy. J Mammary Gland Biol Neoplasia. 2010; 15: 411-421.
20. Ira Mellman, George Coukos and Glenn Dranoff. Cancer immunotherapy comes of age. Nature.
2011; 480: 480-489.
21. Gabrilovich DI Combination of chemotherapy and immunotherapy for cancer: a paradigm
revisited. The Lancet Oncology. 2007 jan 1; 8: 2-3.
22. Chen L, Ashe S, Brady WA, Hellstrom I, Hellstrom KE, Ledbetter JA et al. Costimulation of
antitumor immunity by the B7 counterreceptor for the T lymphocyte molecules CD28 and CTLA-
4. CELL. 1992 Dec 7; 71: 1093-1102.
23. Zang X and Allison JP.. The B7 Family and Cancer Therapy: Costimulation and Coinhibition.
clinical cancer research. 2007; 13: 5271-5279.
24. Kim YT, Hersh EM, Trevor KT. Feasibility to generate monocyte-derived dendritic cell from
coculture with melanoma tumor cells in the presence of granulocyte/macrophage colony-
stimulating factor (GM-CSF) and interleukin-4. Am J Reprod Immunol. 2003; 49: 230-8.
25. Antignani A and FitzGerald D. Immunotoxins: The Role of the Toxin. Toxins. 2013; 5: 1486-
1502.
26. Wayne AS, FitzGerald DJ, Kreitman RJ, Pastan I. Immunotoxins for leukemia. Blood. 2014;
123: 2470-2477.
27. Weiner LM, Dhodapkar MV, and Ferrone S. Monoclonal Antibodies for Cancer Immunotherapy.
Lancet. 2009; 373: 1033-1040.
28. Adler MJ and Dimitrov DS. Therapeutic antibodies against cancer. Hematol Oncol Clin North
Am. 2012; 26: 447-481.
29. Carswell EA, Old LJ, Kassel RL. An endotoxin-induced serum factor that causes necrosis of
tumors. Proc Natl Acad Sci USA. 1975 Sep ; 72(9): 3666-3670.
30. Balkwill F. Tumour necrosis factor and cancer. Nature Reviews Cancer. 2009 May; 9: 361-371.
31. Faustman DL and Davis M. TNF receptor 2 and disease: autoimmunity and regenerative
medicine. Frontiers in immunobiology. 2013; 4: 1-8.
32. Croft M, Benedict CA, and Ware CF. Clinical targeting of the TNF and TNFR superfamilies. Nat
Rev Drug Discov. 2013; 12: 147-168.
33. A M Eggermont AM, H Schraffordt Koops HS, J M Klausner JM, B B Kroon BB, P M Schlag
PM, D Liénard D et al. Isolated limb perfusion with tumor necrosis factor and melphalan for limb
salvage in 186 patients with locally advanced soft tissue extremity sarcomas. The cumulative
multicenter European experience. Ann Surg. 1996 Dec; 224: 756-765.
34. Bradley JR, Thiru S, Pober JS. Disparate localization of 55-kD and 75-kD tumor necrosis factor
receptors in human endothelial cells. Am. J. Pathol. 1995; 146: 27-32.
35. Hill S, Fawcett WJ, Sheldon J. Low-dose tumor necrosis factor alpha and melphalan in
hyperthermic isolated limb perfusion. Br. J. Surg. 1993; 80: 995-997.
36. Bonvalot S, Laplanche A, Lejeune F. Limb salvage with isolated perfusion for soft tissue
sarcoma: could less TNF-alpha be better? Ann. Oncol. 2005; 16: 1061-1068.
37. Luo JL , Maeda S, Hsu LC, Yagita H, and Karin M. Inhibition of NF-kB in cancer cells converts
inflammation-induced tumor growth mediated by TNF-alpha to TRAIL-mediated tumor
regression. Cancer cell. 2004; 6: 297-305.
38. van Horssen R, Ten Hagen TL, Eggermont AM. TNF-α in Cancer Treatment:Molecular Insights,
Antitumor Effects, and Clinical Utility. The Oncologist. 2006; 11: 397-408.
39. Lin WW, Karin M. A cytokine-mediated link between innate immunity, inflammation, and
cancer. J. Clin. Investigation. 2007; 117: 1175-1183.
40. Manusama ER, Nooijen PT, Stavast J. Synergistic antitumor effect of recombinant human tumor
necrosis factor alpha with melphalan in isolated limb perfusion in the rat. Br. J. Surg. 1996; 83:
551-555.
41. Brouckaert P, Takahashi N, Van Tiel ST. Tumor necrosis factor-alpha augmented tumor response
in B16BL6 melanoma-bearing mice treated with stealth liposomal doxorubicin. Int J. Cancer.
2004; 109: 442-448.
42. Özören N, El-Deiry WS. Cell surface Death Receptor signaling in normal and cancer cells.
Elsevier. 2003; 13: 135-147.
43. Hirohata S. Fully human anti TNF-alpha monoclonal antibodies (adalimumab, golimumab).
Nihon Rinsho. 2007 Jul 7; 65: 1202-08.
44. Goel N and Stephens S. Certolizumab pegol. Landes Biosciences. 2010; 2: 137-147.
45. Haynes K, Beukelman T, Curtis JR, Newcomb C, Herrinton LJ, Graham DJ et al. Tumor
Necrosis Factor Alpha Inhibitor Therapy and Cancer Risk In Chronic Immune Mediated Diseases.
Arthritis Rheum. 2013 Jan 1; 65: 1-22.
46. Yoshida Y and Toshio T. Interleukin 6 and Rheumatoid Arthritis. BioMed Research International.
2014; 2014: 01-12.
47. Ishihara K, Hirano T. IL-6 in autoimmune disease and chronic inflammatory proliferative disease.
Cytokine growth factor Rev. 2002; 13: 357-368.
48. Hodge, D.R., Hurt, E.M., and Farrar, W. L. The role of IL-6 and STAT3 in inflammation and
cancer . Eur. J. Cancer. 2005; 41: 2502-2512.
49. Mantovani A. Role of inflammatory cells and mediators in tumor invasion and metastasis. Cancer
Metastasis Rev. 2010; 29: 241.
50. Rose-John S, Scheller J, Elson G, and Jones SA. Interleukin-6 biology is coordinated by
membrane-bound and soluble receptors: role in inflammation and cancer. J. Leukoc. Biol. 2006;
80: 227-236.
51. Becker C. TGF-beta suppresses tumor progression in colon cancer by inhibition of IL-6 trans-
signalling. Immunity. 2004; 21: 491-501.
52. Jones SA, Scheller J, Rose-John S. Therapeutic strategies for the clinical blockade of IL-6/gp130
signaling. J. Clin. Invest. 2011; 121: 3375-83.
53. Hobson SA, PharmD, Waddell JA, Solimando DA Jr. Enzalutamide and Ruxolitinib. Hosp
Pharm. 2013; 48: 104-107.
54. Eghtedar A, Verstovsek S, Estrov Z, Burger J, Cortes J, Bivins C. Phase II study of the JAK
kinase inhibitor ruxolitinib in patients with refractory leukemias, including post
myeloproliferative neoplasms (MPN) acute myeloid leukemia (AML). Blood. 2012; 119: 4614-8.
55. Seavey MM, Lu LD, Stump KL, Wallace NH, Hockeimer W, O'Kane TM. Therapeutic efficacy
of CEP-33779, a novel selective JAK2 inhibitor, in a mouse model of colitis-induced colorectal
cancer . Mol. Cancer Ther. 2012; 11: 984-93.
56. Becker C, Fantini MC, Schramm C, Lehr HA, Wirtz S, Nikolaev A. TGF-beta suppress tumor
progression in colon cancer by inhibition of IL-6 trans-signaling . Immunity. 2004; 21: 491-501.
57. Taga T, Hibi M, Hirata Y, Yamasaki K, Yasukawa K, Matsuda T et al. Interleukin-6 triggers the
association of its receptor with a possible signal transducer, gp130. Cell. 1989; 58: 573-581.
58. N. Stahl N, T. G. Boulton, T. Farruggella. Association and activation of Jak-Tyk kinases by
CNTF-LIF-OSM-IL-6 ? receptor components. Science. 1994; 263: 92-95.
59. Huizinga TW , Fleischmann RM, Jasson M, Radin AR, Adelsberg JV, Fiore S et al. Sarilumab, a
fully human monoclonal antibody against IL-6Rα in patients with rheumatoid arthritis and an
inadequate response to methotrexate: efficacy and safety results from the randomised SARIL-RA-
MOBILITY Part A trial. Clinical and epidemiological research. 2014 Sep;73(9): 1-9.
60. Tanaka T, Ogata A, and Kishimoto T. Targeting of interleukin-6 for the treatment of rheumatoid
arthritis: a review and update. Rheumatology: Current Research. 2013 June 17; 3:1-14.
61. Hussain SP, Harris CC. Inflammation and cancer: an ancient link with novel potentials. Int J
Cancer. 2007; 121: 2372-2380.
62. Duffy SA, Teknos T, Taylor JM, Fowler KE, Islam M, Wolf GT et al. Health Behaviors Predict
Higher Interleukin-6 levels Among Patients Newly Diagnosed with Head and Neck Squamous
Cell Carcinoma. Cancer Epidemiol Biomarkers. 2013; 22: 374-381.
63. Fujino S, Andoh A, Bamba S. Increased expression of interleukin 17 in inflammatory bowel
disease. Gut. 2003 Jan; 52(1): 65-70.
64. Wang M, Wang L, Ren T, Xu L, Wen Z. IL-17A/IL-17RA interaction promoted metastasis of
osteosarcoma cells. Cancer Biology and Therapy. 2013; 14: 155-163.
65. Qiu Z, Dillen C, Hu J, Verbeke H, Struyf S, Van Damme J, Opdenakker G. Interleukin-17
regulates chemokine and gelatinase B expression in fibroblasts to recruit both neutrophils and
monocytes. J.of Immunol. 2009; 214: 835-842.
66. Yan H, Tong Z. Interleukin-17F: A Promising Candidate of Cancer Therapy. Immuno-
Gastroenterology. 2012; 1: 100-103.
67. Chang SH and Dong C . IL-17F: Regulation, signaling and function in inflammation. Elsvier.
2009; 46: 7-11.
68. Chaudhary A, St Croix B. Selective blockade of tumor angiogenesis. Cell Cycle; 2012 Jun 15; 11:
2253-9.
69. Wei-Ye Shi, Chang-Yan Che and Li Liu. Human Interleukin 23 Receptor Induces Cell Apoptosis
in Mammalian Cells by Intrinsic Mitochondrial Pathway Associated with the Down-Regulation of
RAS/Mitogen-Activated Protein Kinase and Signal Transducers and Activators of Transcription
factor 3 Signaling. Int J of Mol Sci. 2013 Dec 18; 14(12): 24656-24669.
70. Airoldi I, Carlo ED, Banelli B, Moserle L, Cocco C, Pezzolo A et al. The IL-12Rβ2 gene
functions as a tumor suppressor in human B cell malignancies. J. Clin. Invest. 2004; 113(11):
1651-59.
71. Cho ML, Kang JW, Moon YM, Nam HJ, Jhun JY, Heo SB et al. STAT3 and NF-kappa B signal
pathways is required for IL-23-mediated IL-17 production in spontaneous arthritis animal model
IL-1 receptor antagonist-defecient mice . J. Immunol. 2006 May; 176(9): 5652-5661.
72. Hao JS and Shan BE. Immune enhancement and anti-tumor activity of IL-23. Cancer Immunol.
Immunother. 2006 Nov; 55(11): 1426-1431.
73. Kobayashi M, Fitz L, Ryan M, Hewick RM, Clark SC, Chan S. Identification and purification of
natural killer cell stimulatory factor (NKSF), acytokine with multiple biologic effects on human
lymphocytes. J Exp Med. 1989 Sep 1; 170(3): 827-45.
74. Stern AS, Podlaski FJ, Hulmes JD. Purification to homogeneity and partial characterization of
cytotoxic lymphocyte maturation factor from human B-lymphoblastoid cells. Proc Natl Acad Sci
USA. 1990 Sep; 87(17): 6808-12.
75. Watford WT, Hissong BD, Bream JH, Kanno Y, Muul L, OShea JJ. Signaling by IL-12 and IL-23
and the immunoregulatory roles of STAT4. Immunol Rev. 2004 Dec; 202: 139-156.
76. Trinchieri, G. Interleukin-12 and the regulation of innate resistance and adaptive immunity . Nat.
Rev. 2003 Feb; 3(2): 133-46.
77. Chan SH, Perussia B, Gupta JW, Kobayashi M, Pospisil M, Young HA et al. Induction of
interferon γ production by natural killer cell stimulatory factor: characterization of responder cells
and synergy with other inducers. J Exp Med. 1991 Apr 1; 173(4): 869-79.
78. Brunda MJ, Luistro L, Warrier RR, Wright RB, Hubbard BR, Murphy M et al. Antitumor and
antimetastatic activity of interleukin 12 against marine tumors. JEM. 1993 Oct 1; 178: 1223-1230.
79. Carson WE, Ross ME, Baiocchi RA, Marien MJ, Boiani N, Grabstain K.. Endogenous production
of interleukin 15 by activated human monocytes is critical for optimal production of interferon-γ
by natural killer cells in vitro. J Clin Invest. 1995 Dec; 96(6): 2578-82.
80. Gazzinelli RT, Hieny S, Wynn TA, Wolf S, Sher A. Interleukin 12 is required for the T-
lymphocyte-independent induction of interferon γ by an intracellular parasite and induces
resistance in T-cell-deficient hosts . Proc Natl Acad Sci USA. 1993 Jul 1; 90(13): 6115-6119.
81. Teng MW, Scheidt BV, Duret H, Towne JE, and Smyth MJ. Anti-IL-23 Monoclonal Antibody
Synergizes in Combination with Targeted Therapies or IL-2 to Suppress Tumor Growth and
Metastases. Cancer Research. 2011 Mar; 71(6): 2077-2086.
82. Michele W.L. Teng, Bianca von Scheidt, Helene Duret, Jennifer E. Towne, and Mark J. Smyth.
Anti-IL 23 Monoclonal Antibody Synergies in Combination with Targeted Therapies or IL – 2 to
suppress Tumor Growth and Metastases. Cancer Research. 2011 Mar 15; 71: 2077-2086.
83. Kortylewski M, Xin H, Kujawski M, Lee H, Liu Y, Harris T. Regulation of the IL-23 and Il-12
balance by Stat3 signaling in the tumor microenvironment. Cancer cell. 2009 Feb 3; 15(2): 114-
23.
84. Witold Lasek W, Zagozdzon R, Jakobisiak M. Interleukin 12: still a promising candidate for
tumor immunotherapy? Cancer Immunol Immunother. 2014; 63: 419-435.
85. Pestka S, Krause CD, Sarkar D, Walter MR, Shi Y, Fisher PB. Interleukin-10 and related
cytokines and receptors. Annu. Rev. Immunol. 2004; 22: 929-979.
86. Sato T, Sato MT, Terai M, Tamura Y, Alexeev V, Mastrangelo MJ et al. Interleukin 10 in the
tumor microenvironment: a target for anticancer immunotherapy. Immunol Res. 2011 Dec; 51(2):
172-80.
87. Fiorentino DF, Bond MW, and Mosmann TR. Two types of mouse T-helper cell. IV. Th2 clones
secrete a factor that inhibits cytokine production by Th1 clones. J. Exp Med. 1989 Dec 1; 170(6):
2081-95.
88. Schottelius AJ, Mayo MW, Sartor RB, and Baldwin AS Jr. Interleukin-10 signaling blocks
inhibitor of kappa-B kinase activity and nuclear factor kappaB DNA binding. J. Biol. Chem. 1999
Nov 5; 274(45): 31868-31874.
89. Hoentjen F, Sartor RB, Ozaki M, and Jobin C. STAT3 regulates NF-kappaB recruitment to the
IL-12p40 promoter in dendritic cells . Blood. 2005 Jan 15; 105(2): 689-696.
90. Moore KW, De Waal MR, Coffman RL, O’Garra A. Interleukin-10 and the interleukin-10
receptor. Annu. Rev. Immunol. 2001; 19: 683-765.
91. Platzer C, Meisel C, Vogt K, Platzer M, and Volk HD. Up-regulation of monocytic IL-10 by
tumor necrosis factor-alpha and cAMP elevating drugs . Int. Immunol. 1995 Apr; 7(4): 517–523.
92. Platzer C, Fritsch E, Elstner T, Lehmann MH, Volk HD, and Prosch S. Cyclic adenosine
monophosphate-responsive elements are involved in the transcriptional activation of the human
IL-10 gene in monocytic cells. Eur J Immunol. 1999 Oct; 29(10): 3098-104.
93. Platzer C, Docke WD, Volk HD, and Prosch S. Catecholamines trigger IL-10 release in acute
systemic stress reaction by direct stimulation of its promoter/ enhancer activity in monocytic cells.
J Neuroimmunol. 2000 Jun 1; 105(1): 31–38.
94. Meisel C, Vogt K, Platzer C, Randow F, Liebenthal C, and Volk HD. Differential regulation of
monocytic tumor necrosis factor-alpha and interleukin-10 expression. Eur J Immunol. 1996 Jul;
26(7): 1580–1586.
95. Woiciechowsky C, Asadullah K, Nestler D, Eberhardt B, Platzer C, Schoning B et al.
Sympathetic activation triggers systemic interleukin-10 release in immunodepression induced by
brain injury. Nat. Med. 1998; 4: 808–813.
96. Riese U, Brenner S, Docke WD, Prosch S, Reinke P, Oppert M et al. Catecholamines induce IL-
10 release in patients suffering from acute myocardial infarction by transactivating its promoter in
monocytic but not in T cells. Mol Cell Biochem. 2000 Sep; 212 (1-2): 45-50.
97. Masood R, Zhang Y, Bond MW, Scadden DT, Moudgil T, Law RE et al. Interleukin-10 is an
autocrine growth factor for acquired immunodeficiency syndrome-related B-cell lymphoma.
Blood. 1995 jul 15; 85(12): 3423–30.
98. Mahipal A, Terai M, Berd D, Chervoneva I, Patel K, Mastrangelo MJ et al. Tumor-derived
interleukin-10 as a prognostic factor in stage III patients undergoing adjuvant treatment with an
autologous melanoma cell vaccine. Cancer Immunol Immunother. 2011 Jul; 60(7); 1039–45.
99. Di Marco R, Xiang M, Zaccone P, Leonardi C, Franco S, Meroni P, Nicoletti F. Concanavalin A
induced hepatitis in mice is prevented by interleukin (IL)-10 and exacerbated by endogenous IL-
10 deficiency. Autoimmunity. 1999 Oct; 31(2): 75–83.
100. Park YB, Lee SK, Kim DS, Lee J, Lee CH, Song CH. Elevated interleukin-10 levels
correlated with disease activity in systemic lupus erythematosus. Clin Exp Rheumatol. 1998 May-
Jun; 16(3): 283–88.
101. Matsuda M, Salazar F, Petersson M, Masucci G, Hansson J, Pisa P et al. Interleukin 10
pretreatment protects target cells from tumor- and allo-specific cytotoxic T cells and
downregulates HLA class I expression. J Exp Med. 1994 Dec 1; 180(6): 2371–2376.
102. Hagenbaugh A, Sharma S, Dubinett SM, Wei SH, Aranda R, Cheroutre H et al. Altered
immune responses in interleukin 10 transgenic mice. J Exp Med; 1997 Jun 16; 185(12): 2101-
2110.
103. Tournoy KG, Kips JC, Pauwels RA. Endogenous interleukin-10 suppresses allergen-induced
airway inflammation and nonspecific airway responsiveness. Clin Exp Allergy. 2000 jun; 30(6):
775–83.
104. Mocellin S, Wang E, Marincola FM. Cytokines and immune response in the tumor
microenvironment. J Immunother. 2001 Sep-Oct; 24(5): 392-407.
105. Mocellin S, Marincola FM, Young HA. Interleukin-10 and the immune response against
cancer: a counterpoint. J Leukoc Biol. 2005 Nov; 78(5):1043-51.
106. Gerard CM, Bruyns C, Delvaux A, Baudson N, Dargent JL, Goldman M et al. Loss of
tumorigenicity and increased immunogenicity induced by interleukin-10 gene transfer in B16
melanoma cells. Hum Gene Ther. 1996 Jan; 7(1): 23-31.
107. Roncarolo MG, Bacchetta R, Bordignon C, Narula S, Levings MK. Type 1 T regulatory
cells. Immunol Rev. 2001 Aug; 182: 68-79.
108. Levings MK, Sangregorio R, Roncarolo MG. Human cd25(+) cd4(+) t regulatory cells
suppress naive and memory T cell proliferation and can be expanded in vitro without loss of
function. J Exp Med. 2001 Jun 4; 193(11): 1295-302.
109. Roberts AB, Sporn MB, Assoian RK, Smith JM, Roche NS, Wakefield LM et al.
Transforming growth factor type beta: rapid induction of fibrosis and angiogenesis in vivo and
stimulation of collagen formation in vitro. Proc. Natl. Acad. Sci. USA. 1986 Jun; 83(12): 4167–
4171.
110. Tucker RF, Shipley GD, Moses HL, Holley RW. Growth inhibitor from BSC-1 cells closely
related to platelet type β transforming growth factor. Science. 1984 Nov 9; 226(4675): 705–707.
111. Roberts AB, Anzano MA, Wakefield LM, Roche NS, Stern DF, Sporn MB. Type beta
transforming growth factor: a bifunctional regulator of cellular growth. Proc Natl Acad Sci USA.
1985 Jan; 82(1): 119–123.
112. Giannelli G, Villa E and Lahn M. Transforming Growth Factor-b as a Therapeutic Target in
Hepatocellular Carcinoma. Cancer Res. 2014 Apr 1; 74(7): 1890-4.
113. Blobe GC, Schiemann WP, Lodish HF. Role of transforming growth factor beta in human
disease. N. Engl. J. Med. 2000 May 4; 342(18): 1350-8.
114. Blobe, Rebecca L. Elliott and Gerard C. Role of Transforming Growth Factor Beta in Human
Cancer. J. Clin Oncol. 2005 Mar 20; 23(9): 2078-2093.
115. Korpal M, Kang Y. Targeting the transforming growth factor-beta signalling pathway in
metastatic cancer. Eur J Cancer. 2010 Mar; 46(7): 1232-40.
116. Lampropoulos P, Zizi-Sermpetzoglou A, Rizos S, Kostakis A, Nikiteas N, Papavassiliou
AG. TGF-beta signalling in colon carcinogenesis. Cancer Lett. 2012 Jan 1; 314(1): 1-7.
117. Won J, Kim H, Park EJ, Hong Y, Kim SJ, Yun Y. Tumorigenicity of mouse thymoma is
suppressed by soluble Type II transforming growth factor b receptor therapy. Cancer Res. 1999
Mar 15; 59(6): 1273-1277.
118. Benigni A, Zoja C, Corna D, Zatelli C, Conti S, Campana M et al. Add-on anti-TGF-b
antibody to ACE inhibitor arrests progressive diabetic nephropathy in the rat. J Am Soc Nephrol;
1999 Mar 15: 59(6): 1816-1824.
119. Mead AL, Wong TT, Cordeiro MF, Anderson IK, Khaw PT. Evaluation of anti-TGF-b2
antibody as a new postoperative anti-scarring agent in glaucoma surgery. Invest Ophthalmol Vis
Sci. 2003 Aug; 44(8):3394-3401.
120. Nemunaitis J, Dillman RO, Schwarzenberger PO, Senzer N, Cunningham C, Cutler J et al.
Phase II study of belagenpumatucel-L, a transforming growth factor beta-2 antisense gene-
modified allogeneic tumor cell vaccine in non-small-cell lung cancer. J Clin Oncol. 2006 Oct 10;
24(29): 4721–4730.
121. Fakhrai H, Mantil JC, Liu L, Nicholson GL, Murphy-Satter CS, Ruppert J, Shawler DL.
Phase I clinical trial of a TGF-beta antisense-modified tumor cell vaccine in patients with
advanced glioma. Cancer Gene Ther. 2006 Dec ; 13(12): 1052–1060.
122. Gadir N, Jackson DN, Lee E, Foster DA. Defective TGF-beta signaling sensitizes human
cancer cells to rapamycin. Oncogene. 2008 Feb 14; 27(8): 1055-62.
123. Candido J, Hagemann T. Cancer-related inflammation. J Clin Immunol. 2013 Jan;33(1):79-
84.
Cytokine Immunotherapy: A Forthcoming Visible Feature in Cancer Therapeutics

Contenu connexe

Tendances

Tendances (20)

Immunotherapy
ImmunotherapyImmunotherapy
Immunotherapy
 
Immunotherapeutic approaches in cancer
Immunotherapeutic approaches in cancerImmunotherapeutic approaches in cancer
Immunotherapeutic approaches in cancer
 
Basic principles of cancer immunotherapy
Basic principles of cancer immunotherapyBasic principles of cancer immunotherapy
Basic principles of cancer immunotherapy
 
Theory of Immune surveillance
Theory of  Immune surveillance Theory of  Immune surveillance
Theory of Immune surveillance
 
Cancer Immunotherapy
Cancer ImmunotherapyCancer Immunotherapy
Cancer Immunotherapy
 
Tumor Immunology and Cancer Immunotherapy
Tumor Immunology and Cancer ImmunotherapyTumor Immunology and Cancer Immunotherapy
Tumor Immunology and Cancer Immunotherapy
 
Cancer vaccines
Cancer vaccinesCancer vaccines
Cancer vaccines
 
Immunotherapy Update in Oncology
Immunotherapy Update in OncologyImmunotherapy Update in Oncology
Immunotherapy Update in Oncology
 
cancer immunotherapy
cancer immunotherapycancer immunotherapy
cancer immunotherapy
 
Global cancer immunotherapy market outlook 2020
Global cancer immunotherapy market outlook 2020Global cancer immunotherapy market outlook 2020
Global cancer immunotherapy market outlook 2020
 
Cancer Immunotherapy
Cancer ImmunotherapyCancer Immunotherapy
Cancer Immunotherapy
 
Cancer Immunotherapy
Cancer ImmunotherapyCancer Immunotherapy
Cancer Immunotherapy
 
Immunotherapy for cancer
Immunotherapy for cancerImmunotherapy for cancer
Immunotherapy for cancer
 
Cancer therapy via vaccine
Cancer therapy via vaccineCancer therapy via vaccine
Cancer therapy via vaccine
 
Tumor Immunology presentation by Sharmista
Tumor Immunology presentation by SharmistaTumor Immunology presentation by Sharmista
Tumor Immunology presentation by Sharmista
 
Cancer immunology
Cancer immunologyCancer immunology
Cancer immunology
 
Cancer immunotherapy ppt
Cancer immunotherapy pptCancer immunotherapy ppt
Cancer immunotherapy ppt
 
Immunobiology of cancer
Immunobiology of cancerImmunobiology of cancer
Immunobiology of cancer
 
Immunotherapeutics
Immunotherapeutics Immunotherapeutics
Immunotherapeutics
 
Immunotherapy and gene therapy
Immunotherapy and gene therapyImmunotherapy and gene therapy
Immunotherapy and gene therapy
 

En vedette (6)

Cytokines
CytokinesCytokines
Cytokines
 
Biologic Therapy
Biologic TherapyBiologic Therapy
Biologic Therapy
 
Cytokines by Dr Rahul , Physiology SMS MEDICAL JAIPUR MOBILE NO-8764324067
Cytokines by Dr Rahul , Physiology  SMS MEDICAL JAIPUR MOBILE NO-8764324067Cytokines by Dr Rahul , Physiology  SMS MEDICAL JAIPUR MOBILE NO-8764324067
Cytokines by Dr Rahul , Physiology SMS MEDICAL JAIPUR MOBILE NO-8764324067
 
Cytokinins
CytokininsCytokinins
Cytokinins
 
Cytokines ppt
Cytokines pptCytokines ppt
Cytokines ppt
 
Cytokines
CytokinesCytokines
Cytokines
 

Similaire à Cytokine Immunotherapy: A Forthcoming Visible Feature in Cancer Therapeutics

Immuotherapy 2
Immuotherapy 2Immuotherapy 2
Immuotherapy 2
drmcbansal
 
REVIEWCancer stem cells a new framework for the designo.docx
REVIEWCancer stem cells a new framework for the designo.docxREVIEWCancer stem cells a new framework for the designo.docx
REVIEWCancer stem cells a new framework for the designo.docx
joellemurphey
 
In vitro and in vivo models of cancer.docx
In vitro and in vivo models of cancer.docxIn vitro and in vivo models of cancer.docx
In vitro and in vivo models of cancer.docx
write4
 
Clinical oncology basic fundamental For undergraduate studies part I .pdf
Clinical oncology basic fundamental For undergraduate studies  part I .pdfClinical oncology basic fundamental For undergraduate studies  part I .pdf
Clinical oncology basic fundamental For undergraduate studies part I .pdf
Mona Quenawy
 
Citologia relatório de aula prática
Citologia   relatório de aula práticaCitologia   relatório de aula prática
Citologia relatório de aula prática
André Rangel
 

Similaire à Cytokine Immunotherapy: A Forthcoming Visible Feature in Cancer Therapeutics (20)

Immuotherapy 2
Immuotherapy 2Immuotherapy 2
Immuotherapy 2
 
Immuotherapy
ImmuotherapyImmuotherapy
Immuotherapy
 
Vol 87-no2-article-3
Vol 87-no2-article-3Vol 87-no2-article-3
Vol 87-no2-article-3
 
SCT60103 Group 2 Presentation
SCT60103 Group 2 PresentationSCT60103 Group 2 Presentation
SCT60103 Group 2 Presentation
 
التاثير الدوائي على الخلايا السرطانية في
التاثير الدوائي على الخلايا السرطانية فيالتاثير الدوائي على الخلايا السرطانية في
التاثير الدوائي على الخلايا السرطانية في
 
REVIEWCancer stem cells a new framework for the designo.docx
REVIEWCancer stem cells a new framework for the designo.docxREVIEWCancer stem cells a new framework for the designo.docx
REVIEWCancer stem cells a new framework for the designo.docx
 
Intracellular Events in Antigen Processing.pptx
Intracellular Events in Antigen Processing.pptxIntracellular Events in Antigen Processing.pptx
Intracellular Events in Antigen Processing.pptx
 
Hallmarks of Cancer
Hallmarks of Cancer Hallmarks of Cancer
Hallmarks of Cancer
 
13-chap-24-lecture.ppt
13-chap-24-lecture.ppt13-chap-24-lecture.ppt
13-chap-24-lecture.ppt
 
13-chap-24-lecture.ppt
13-chap-24-lecture.ppt13-chap-24-lecture.ppt
13-chap-24-lecture.ppt
 
13-chap-24-lecture.ppt
13-chap-24-lecture.ppt13-chap-24-lecture.ppt
13-chap-24-lecture.ppt
 
Anticancer.ppt
Anticancer.pptAnticancer.ppt
Anticancer.ppt
 
13-chap-24-lecture.ppt
13-chap-24-lecture.ppt13-chap-24-lecture.ppt
13-chap-24-lecture.ppt
 
بدر الشهاري.ppt
بدر الشهاري.pptبدر الشهاري.ppt
بدر الشهاري.ppt
 
In vitro and in vivo models of cancer.docx
In vitro and in vivo models of cancer.docxIn vitro and in vivo models of cancer.docx
In vitro and in vivo models of cancer.docx
 
Clinical oncology basic fundamental For undergraduate studies part I .pdf
Clinical oncology basic fundamental For undergraduate studies  part I .pdfClinical oncology basic fundamental For undergraduate studies  part I .pdf
Clinical oncology basic fundamental For undergraduate studies part I .pdf
 
Citologia relatório de aula prática
Citologia   relatório de aula práticaCitologia   relatório de aula prática
Citologia relatório de aula prática
 
640917.pptx
640917.pptx640917.pptx
640917.pptx
 
Immune surveillance of tumors.
Immune surveillance of tumors.Immune surveillance of tumors.
Immune surveillance of tumors.
 
Targeted Therapy
Targeted TherapyTargeted Therapy
Targeted Therapy
 

Dernier

👉 Amritsar Call Girls 👉📞 8725944379 👉📞 Just📲 Call Ruhi Call Girl Near Me Amri...
👉 Amritsar Call Girls 👉📞 8725944379 👉📞 Just📲 Call Ruhi Call Girl Near Me Amri...👉 Amritsar Call Girls 👉📞 8725944379 👉📞 Just📲 Call Ruhi Call Girl Near Me Amri...
👉 Amritsar Call Girls 👉📞 8725944379 👉📞 Just📲 Call Ruhi Call Girl Near Me Amri...
Sheetaleventcompany
 
Electrocardiogram (ECG) physiological basis .pdf
Electrocardiogram (ECG) physiological basis .pdfElectrocardiogram (ECG) physiological basis .pdf
Electrocardiogram (ECG) physiological basis .pdf
MedicoseAcademics
 
Pune Call Girl Service 📞9xx000xx09📞Just Call Divya📲 Call Girl In Pune No💰Adva...
Pune Call Girl Service 📞9xx000xx09📞Just Call Divya📲 Call Girl In Pune No💰Adva...Pune Call Girl Service 📞9xx000xx09📞Just Call Divya📲 Call Girl In Pune No💰Adva...
Pune Call Girl Service 📞9xx000xx09📞Just Call Divya📲 Call Girl In Pune No💰Adva...
Sheetaleventcompany
 
Dehradun Call Girl Service ❤️🍑 8854095900 👄🫦Independent Escort Service Dehradun
Dehradun Call Girl Service ❤️🍑 8854095900 👄🫦Independent Escort Service DehradunDehradun Call Girl Service ❤️🍑 8854095900 👄🫦Independent Escort Service Dehradun
Dehradun Call Girl Service ❤️🍑 8854095900 👄🫦Independent Escort Service Dehradun
Sheetaleventcompany
 
Jual Obat Aborsi Di Dubai UAE Wa 0838-4800-7379 Obat Penggugur Kandungan Cytotec
Jual Obat Aborsi Di Dubai UAE Wa 0838-4800-7379 Obat Penggugur Kandungan CytotecJual Obat Aborsi Di Dubai UAE Wa 0838-4800-7379 Obat Penggugur Kandungan Cytotec
Jual Obat Aborsi Di Dubai UAE Wa 0838-4800-7379 Obat Penggugur Kandungan Cytotec
jualobat34
 
Control of Local Blood Flow: acute and chronic
Control of Local Blood Flow: acute and chronicControl of Local Blood Flow: acute and chronic
Control of Local Blood Flow: acute and chronic
MedicoseAcademics
 
Cara Menggugurkan Kandungan Dengan Cepat Selesai Dalam 24 Jam Secara Alami Bu...
Cara Menggugurkan Kandungan Dengan Cepat Selesai Dalam 24 Jam Secara Alami Bu...Cara Menggugurkan Kandungan Dengan Cepat Selesai Dalam 24 Jam Secara Alami Bu...
Cara Menggugurkan Kandungan Dengan Cepat Selesai Dalam 24 Jam Secara Alami Bu...
Cara Menggugurkan Kandungan 087776558899
 
Jaipur Call Girl Service 📞9xx000xx09📞Just Call Divya📲 Call Girl In Jaipur No💰...
Jaipur Call Girl Service 📞9xx000xx09📞Just Call Divya📲 Call Girl In Jaipur No💰...Jaipur Call Girl Service 📞9xx000xx09📞Just Call Divya📲 Call Girl In Jaipur No💰...
Jaipur Call Girl Service 📞9xx000xx09📞Just Call Divya📲 Call Girl In Jaipur No💰...
Sheetaleventcompany
 

Dernier (20)

Call Girls Shahdol Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Shahdol Just Call 8250077686 Top Class Call Girl Service AvailableCall Girls Shahdol Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Shahdol Just Call 8250077686 Top Class Call Girl Service Available
 
👉 Amritsar Call Girls 👉📞 8725944379 👉📞 Just📲 Call Ruhi Call Girl Near Me Amri...
👉 Amritsar Call Girls 👉📞 8725944379 👉📞 Just📲 Call Ruhi Call Girl Near Me Amri...👉 Amritsar Call Girls 👉📞 8725944379 👉📞 Just📲 Call Ruhi Call Girl Near Me Amri...
👉 Amritsar Call Girls 👉📞 8725944379 👉📞 Just📲 Call Ruhi Call Girl Near Me Amri...
 
Shazia Iqbal 2024 - Bioorganic Chemistry.pdf
Shazia Iqbal 2024 - Bioorganic Chemistry.pdfShazia Iqbal 2024 - Bioorganic Chemistry.pdf
Shazia Iqbal 2024 - Bioorganic Chemistry.pdf
 
ANATOMY AND PHYSIOLOGY OF RESPIRATORY SYSTEM.pptx
ANATOMY AND PHYSIOLOGY OF RESPIRATORY SYSTEM.pptxANATOMY AND PHYSIOLOGY OF RESPIRATORY SYSTEM.pptx
ANATOMY AND PHYSIOLOGY OF RESPIRATORY SYSTEM.pptx
 
Electrocardiogram (ECG) physiological basis .pdf
Electrocardiogram (ECG) physiological basis .pdfElectrocardiogram (ECG) physiological basis .pdf
Electrocardiogram (ECG) physiological basis .pdf
 
❤️Call Girl Service In Chandigarh☎️9814379184☎️ Call Girl in Chandigarh☎️ Cha...
❤️Call Girl Service In Chandigarh☎️9814379184☎️ Call Girl in Chandigarh☎️ Cha...❤️Call Girl Service In Chandigarh☎️9814379184☎️ Call Girl in Chandigarh☎️ Cha...
❤️Call Girl Service In Chandigarh☎️9814379184☎️ Call Girl in Chandigarh☎️ Cha...
 
Genuine Call Girls Hyderabad 9630942363 Book High Profile Call Girl in Hydera...
Genuine Call Girls Hyderabad 9630942363 Book High Profile Call Girl in Hydera...Genuine Call Girls Hyderabad 9630942363 Book High Profile Call Girl in Hydera...
Genuine Call Girls Hyderabad 9630942363 Book High Profile Call Girl in Hydera...
 
Chandigarh Call Girls Service ❤️🍑 9809698092 👄🫦Independent Escort Service Cha...
Chandigarh Call Girls Service ❤️🍑 9809698092 👄🫦Independent Escort Service Cha...Chandigarh Call Girls Service ❤️🍑 9809698092 👄🫦Independent Escort Service Cha...
Chandigarh Call Girls Service ❤️🍑 9809698092 👄🫦Independent Escort Service Cha...
 
Pune Call Girl Service 📞9xx000xx09📞Just Call Divya📲 Call Girl In Pune No💰Adva...
Pune Call Girl Service 📞9xx000xx09📞Just Call Divya📲 Call Girl In Pune No💰Adva...Pune Call Girl Service 📞9xx000xx09📞Just Call Divya📲 Call Girl In Pune No💰Adva...
Pune Call Girl Service 📞9xx000xx09📞Just Call Divya📲 Call Girl In Pune No💰Adva...
 
Dehradun Call Girl Service ❤️🍑 8854095900 👄🫦Independent Escort Service Dehradun
Dehradun Call Girl Service ❤️🍑 8854095900 👄🫦Independent Escort Service DehradunDehradun Call Girl Service ❤️🍑 8854095900 👄🫦Independent Escort Service Dehradun
Dehradun Call Girl Service ❤️🍑 8854095900 👄🫦Independent Escort Service Dehradun
 
7 steps How to prevent Thalassemia : Dr Sharda Jain & Vandana Gupta
7 steps How to prevent Thalassemia : Dr Sharda Jain & Vandana Gupta7 steps How to prevent Thalassemia : Dr Sharda Jain & Vandana Gupta
7 steps How to prevent Thalassemia : Dr Sharda Jain & Vandana Gupta
 
Circulatory Shock, types and stages, compensatory mechanisms
Circulatory Shock, types and stages, compensatory mechanismsCirculatory Shock, types and stages, compensatory mechanisms
Circulatory Shock, types and stages, compensatory mechanisms
 
Jual Obat Aborsi Di Dubai UAE Wa 0838-4800-7379 Obat Penggugur Kandungan Cytotec
Jual Obat Aborsi Di Dubai UAE Wa 0838-4800-7379 Obat Penggugur Kandungan CytotecJual Obat Aborsi Di Dubai UAE Wa 0838-4800-7379 Obat Penggugur Kandungan Cytotec
Jual Obat Aborsi Di Dubai UAE Wa 0838-4800-7379 Obat Penggugur Kandungan Cytotec
 
Most Beautiful Call Girl in Chennai 7427069034 Contact on WhatsApp
Most Beautiful Call Girl in Chennai 7427069034 Contact on WhatsAppMost Beautiful Call Girl in Chennai 7427069034 Contact on WhatsApp
Most Beautiful Call Girl in Chennai 7427069034 Contact on WhatsApp
 
Kolkata Call Girls Shobhabazar 💯Call Us 🔝 8005736733 🔝 💃 Top Class Call Gir...
Kolkata Call Girls Shobhabazar  💯Call Us 🔝 8005736733 🔝 💃  Top Class Call Gir...Kolkata Call Girls Shobhabazar  💯Call Us 🔝 8005736733 🔝 💃  Top Class Call Gir...
Kolkata Call Girls Shobhabazar 💯Call Us 🔝 8005736733 🔝 💃 Top Class Call Gir...
 
Bhawanipatna Call Girls 📞9332606886 Call Girls in Bhawanipatna Escorts servic...
Bhawanipatna Call Girls 📞9332606886 Call Girls in Bhawanipatna Escorts servic...Bhawanipatna Call Girls 📞9332606886 Call Girls in Bhawanipatna Escorts servic...
Bhawanipatna Call Girls 📞9332606886 Call Girls in Bhawanipatna Escorts servic...
 
Control of Local Blood Flow: acute and chronic
Control of Local Blood Flow: acute and chronicControl of Local Blood Flow: acute and chronic
Control of Local Blood Flow: acute and chronic
 
Bandra East [ best call girls in Mumbai Get 50% Off On VIP Escorts Service 90...
Bandra East [ best call girls in Mumbai Get 50% Off On VIP Escorts Service 90...Bandra East [ best call girls in Mumbai Get 50% Off On VIP Escorts Service 90...
Bandra East [ best call girls in Mumbai Get 50% Off On VIP Escorts Service 90...
 
Cara Menggugurkan Kandungan Dengan Cepat Selesai Dalam 24 Jam Secara Alami Bu...
Cara Menggugurkan Kandungan Dengan Cepat Selesai Dalam 24 Jam Secara Alami Bu...Cara Menggugurkan Kandungan Dengan Cepat Selesai Dalam 24 Jam Secara Alami Bu...
Cara Menggugurkan Kandungan Dengan Cepat Selesai Dalam 24 Jam Secara Alami Bu...
 
Jaipur Call Girl Service 📞9xx000xx09📞Just Call Divya📲 Call Girl In Jaipur No💰...
Jaipur Call Girl Service 📞9xx000xx09📞Just Call Divya📲 Call Girl In Jaipur No💰...Jaipur Call Girl Service 📞9xx000xx09📞Just Call Divya📲 Call Girl In Jaipur No💰...
Jaipur Call Girl Service 📞9xx000xx09📞Just Call Divya📲 Call Girl In Jaipur No💰...
 

Cytokine Immunotherapy: A Forthcoming Visible Feature in Cancer Therapeutics

  • 1. Dear Reviewer, Please find the confined manuscript entitled “Cytokine Immunotherapy: A forthcoming visible feature in cancer therapeutics”, which I would like to share with you all. Cytokine are messengers protein produced by varieties of cells whose role in host responses to cancer are remarkable. Different bioprocesses are interlinked with cytokine functions. Preclinical research studies have shown the anti-cancerous property of cytokine which suppress inflammatory and autoimmune microenvironment. Little evidence of rare patient studies suggesting that the immune system can revoke again if stimulated appropriately. One of the appropriate stimulation of immune system can be done with therapeutic manipulation of cytokine balance. However, at present status, mono-cytokine therapy doesn’t show promising features as compare to combined cytokine therapy as combined cytokine therapy or cytokine therapy with other traditional therapeutic module. It is expected that appropriate combination will result with specific and reliable therapeutics for cancer treatment. It is time to focus on the remaining checkpoints and else everything to make an option for the accurate efficacy of treatment. In this literature work, I have enlightened some therapeutic individuality along with possibility of combination with other modalities. I think that this literature work will be fruitful for those seekers who are looking for the appropriate combination of treatment to overcome the drawbacks of traditional healing remedies. Thank you for your consideration. I look forward to seeing your critics from you. Sincerely, Sachin K. S. Chauhan New Delhi, INDIA.
  • 2. Cytokine Immunotherapy: A Forthcoming Visible Feature in Cancer Therapeutics Sachin K. S. Chauhan Abstract: Cell-based cytokine therapy have prodigious promising role for treating a varieties of diseases. Cytokines are intra- as well as intercellular messengers, which recruit the immune effecter cells, thereby limiting the tumor growth; however, sometimes these molecules promote tumor progression. Several studies have attempted the usage cytokines alone as pharmaceutical targets to alter the immune response, which may eradicate solid tumors. In the context of advancement in understanding of cytokine biology, there is a definite need to develop combined cytokine therapy with the advent of gene technology to improve efficacy of cancer treatment. This review focuses on understanding the factors of combined cytokine therapy and that allows us to attain durable and long-lasting responses in cancer therapy. Introduction Cancer is a natural wound healing related process, which includes cytokine secretion, tissue remodeling, and oncogenes activation (1). Every cell has a unique type of characteristic signature such as the abilities of genes or proteins to perform their function. These characteristic features also alleviate molecular mechanisms of diseases and will give prediction about individualization of therapies for suffering people. From many years, all efforts to treat human malignancies have concentrated on the early detection and cure of tumor. So these efforts have provided great opportunities for improving the management of cancer patients by enhancing the efficacy of detection and treatment (2). Now it is well known that the mutation, competition, and natural selection operating within the population of somatic cells are basic requirements for initiations, progression, and survival of cancer cells. These phenomena depend not only on the tumor cell, but on the complex interactions between the tumor cells and its ‘host’ (3). Hence, it is required to focus on the strategies to modulate the growth mechanism and host microenvironment, which offers a complementary perspective to achieve a permanent cure. Cancer cells are introduced by their heritable properties like they reproduce in defiance of the normal restraints, invade, and colonize territories reserved for other cells. This renegade coordination of malignancy has been creating lot of evidences that tumor cells elicit certain hallmark characteristics which lead the somatic cells to micro-evolutionary developmental process of malignant cancer (4). (Box 1) This multistep process of tumor genesis could be explained by the requirement of incipient cancer cells to adopt the traits of tumorigenic cells and ultimately become malignant (5). :
  • 3. There are several therapeutic modalities have been developing on the basis of cancer hallmarks individuality (Figure 1). It is mandatory to go through the overall metabolic network and its acquired hallmarks which explain the majority of manipulated checkpoint for the growth of tumor cells. FOUNDATION OF CANCER BIOLOGY New research applications of cancer biology are transforming our understanding about cancer. The basic foundation of cancer biology explores these basic principles – cancer excogitates by somatic selection, neoplasm ripens in complex ecosystems, natural selection has designed effective cancer defenses (6). The limitations of these principles defenses have the explanations which present foundation of cancer biology for understanding, preventing and developing therapeutic modalities (7). The most fundamental traits of cancer cells overcome cell growth signals and influence cell survival and energy metabolism (8). These mutagenic signals are release via branched intracellular signaling through cell progression which should be filtered in case of tumor growth. Alternatively, some tumor cells may be able to overcome high level of oncogenic signaling by disabling their apoptosis-inducing mechanism. If we enquire anomaly between level of complex network of proteases, sulfatases, and other enzymes in normal and tumor cells, then it’ll be easy to find out that it can switch the pathway in an alternate ways, hence it is necessary to seek a multi target therapeutic approaches. In addition to two above aspect of fundamental biology of insensitivity to antigrowth signaling and evasion of apoptosis, cancer cells riddance various signaling pathways, especially apoptotic which allow them to seed, proliferate, and flourish in other territories of body. They must also besiege dozens of powerful tumor suppressor programs that control in various ways to limit the normal cell growth. For example, the central effectors of apoptosis is caspases whose hypo-phosporylation makes impossible antigrowth factors such as TGF-β, pRB and p53 from blocking the advancement of cell growth (9).The other two factors are RB (Retinoblastoma) and TP53 have the function of inhibition and execution of cell cycle growth in normal cells. Tumor cell with the defect in these factor mechanisms exclude the check-point service of cell cycle whose absence permits persistent growth. (10). At the mechanistic level, several types of surface proteins are involved in the interactions of metastatic cells through cell adhesions molecules (CAMs) and integrin to establish proper invasive cell in distinct territories of body. These interactions play a vital role in allowing the signal to enter into intracellular part of cell. (11). Alterations in expression of these CAMs in Ig super family play a critical role in the invasion and metastasis processes like change in the expression of integrin help to invade and proliferating cancer cells (12). E-cadherins are Box 1 Hallmarks individuality of cancer: 1. The quality to prolong chronic proliferation by deregulating explicit of growth promoting signals. 2. Evading growth suppressors either by persecuting the effect of tumor suppressor genes or by annulling cell-cell contact inhibition. 3. Resisting cell death and becoming tolerant to multiple apoptotic mechanisms. 4. Unlimited replicate potential by protecting against telomere erosion. 5. Inducing angiogenesis via production of vascular endothelial growth factors. 6. Activating invasion and metastasis by altering expression of cell-cell adhesion molecules. Because of the proceeding impact of these hallmarks, they are usually mentioned as a starting point to study for the anticancer strategies. Several progresses in these research studies has revealed two new emerging hallmarks of cancer to this list – reprogramming of energy metabolism and evading immune destruction (Douglas Hanahan, Hallmarks of Cancer: The Next Generation, 2011).
  • 4. Box 2 Molecular and cellular pathways linking inflammation and cancer Schematically, two pathways have emerged; the intrinsic and extrinsic. In the intrinsic pathways, activation of different classes of oncogenes drives the expression of inflammation-related programs that guide the construction of an inflammatory milieu. In the extrinsic pathways, inflammatory conditions promote cancer development. The key factors of the inflammation-mediated tumor progression are transcription factors, cytokines, chemokine and infiltrating leukocytes. (123). widely known as a suppressor of invasion and metastasis by epithelial cells, which must be altered by cancer cell to overcome to progress (13), (14). As it is known that these CAM molecules are the basis of interaction between cells, so if put a gate on the expression of these molecules in tumor cells then it’ll be easier to find a cure for the responsible tumor type. For all malignancies, executing treatments is much harder to come by. May be a after few years, it may be possible to sequence according to the soul of tumor and offer up the diagnosis, not of colorectal cancer, but of an RB/TP53/APC/KRAS malignancy like report. Then different combination system in the form of pills will be recommended. Over the past decade when viewed from all these perspective, tumors have been increasingly recognized as abnormality whose complexity approaches and may even exceed that of normal healthy tissues. It’s better to understand the biology of a tumor through studying the specialized cell types within it as well as the ‘tumor Microenvironment’ that they are interlinked. These are the overall assessment about different recent aspects of foundation studies of cancer biology. They were illustrated about cells involved in the proliferation, soluble factors which enhance the progression, signaling molecules whose alteration promote the survival of cancer cells, extracellular matrix and mechanical cues that can promote transformation, invasion and protect the tumor from host immunity. These all aspects indicate toward the link of cancer progression and tumor microenvironment. Tumor microenvironment and Immunomodulation Tumor is composed of stromal constituents like inflammatory cells, endothelial cells which finally arrange tumor microenvironment to promote tumor growth, progression and survival (15). In 1909, Paul Ehrlich originally hypothesized that the progression of neoplastic cells could be constrained by host immune system. In the response to tissue injury induced by tumor-host interaction, stromal cells represent a defense mechanism of the organism and release cytokine, chemokine, growth factors, and lipid modulators that exert their effects. For example, Transforming growth factor- beta (TGF-β) is a critical regulator of tumor progression and a potent inhibitor of cell differentiation which is secreted by multiple cell types within the tumor microenvironment (16). This defense mechanism suppressed by tumor cells which effectively alter the growth inhibitory effect of stromal cells and harness the growth promoting characteristics for their own beneficial (17). The purpose of studying limitations of tumor microenvironment and immunomodulation is to identify the optimal treatment for each individual patient to maximize the treatment benefit.
  • 5. Tumor and immune cells both have been found to secrete inflammatory mediators which play a dual role in tumor development. On the one side, they promote tumor survival whereas on the other side they promote immune surveillance mechanisms against tumor cells. It is during the equilibrium phase that the interplay between several components of the immune system and the tumor will define the final outcome of the immune response. This immune selection may be produced due to recognition of cytokine and chemokine in immune system. The cytokines are endogenous substances behaving like hormones present within the immune system, sending messages between immune cells mediated by receptors, stimulation of immune cells showing some direct effect on tumor cells and infections, (18). Several studies are presently going on chemokine and cytokine levels in tumor cells at various stage of their growth phase to understand the levels of these factors causing promotion or inhibition of tumor growth. They revealed that cancer cells amend only those pathways which make those cells accessible to use them as proliferative agent without any checkpoint. A new era of effectively harnessing these studies to treat and prevent cancer has begun. There are several therapies are now approved for cancer treatment and others are on clinical trials. These studies may lead to elevated interest and movement in developing novel therapeutic strategies for oversetting the balance of host-tumor interaction toward tumor growth inhibition. It is clearly practicing to prosper advanced therapies to re-engineer the overall host ambience and tumor microenvironment to rattle pathways of suppression and immune system tolerance. Overall, Chemo, radiation and other traditional therapies are standard therapeutic modalities for patients suffering from cancer. But the extensive use of antibiotics and chemicals in human patients has shown several side effects with less efficacy of treatment (19). In another case, William Coley (1891) attempted to demonstrate the immune system’s ability to cause inflammation which further stimulates antibacterial phagocytes that might kill spectator tumor cells also. Further this theory was discounted but it gave a channel towards Cancer immunotherapy (20). Cytokine being natural messengers of immune response which on administration eradicate solid tumor, thereby exhibiting bright side of cytokine therapy. Presently, many researchers have tried the combination of Chemotherapy / radiotherapy with different specific form of immunotherapy. Although, it is well known that chemotherapy / radiation cause several lethal effect to rapidly progressive cells like hematopoietic cells, epithelial lining in gut and sperms. So it is recommending that use chemotherapy / radiotherapy to deplete tumor load and then introduce immunotherapy to final depletion of tumor and prevent the re-growth (21). Recent studies highlight the benefits of specificity of Immunotherapy for cancer which can enable the induction of immune response via a variety of ways such as Guidance of co-stimulatory signal molecules During tumor progression, a tumor cell mutates the co-stimulatory genes and affects the function of co-stimlatory molecules which are required for the interaction of antigen presenting cells (APC) with immune cells. Like, B7 (a co-stimulatory molecule) on APC interact with its receptor i.e. CTLA -4 and CD28 on T cells which provide signal for further activation of T cells. The absence of expression of B7 molecules renders tumor growth imperceptible to the immune system while the induced expression of B7 molecule prevent
  • 6. tumor from effective cytolytic T lymphocyte (CTLs). The transfection of B7 molecule in tumor cell micro-environment led to tumor regression due to induction of effective CTLs response. The co-stimulation of T cell upto a balanced level may have valuable therapeutic purpose for generating immunity against tumors (22), (23). Augmentation of APC activity with monotherapy This monotherapy attempts to utilize the expression of tumor-specific antigens in the development of antigen-specific immunotherapy. In one approach, the result approved the practicability to induce monocyte-derived DCs in the presence of GM-CSF and IL-4 by co- culturing with melanoma tumor cells (24). This study highlights that tumor cells can be genetically improvised to be able to identify tumor specific antigens (TSAs) through activation of different cytokines and other molecules. If patients DCs co-culture with tumor fragment from patients in the presence of recombinant GM-CSF, then it can induce the activation of APCs, Th cells, and CTLs specific for TSAs. After decades of research, it is shown that DCs are the central effecter cells of immune system. Thus, DCs highlighted as beneficial target generate therapeutic modality against tumor. Immunotoxins against tumor Immunotoxins are bifunctional molecules that can induce intracellular toxin activity to kill the target cells via binding of antibody with specific antigen (25). In an approach, Immunotoxins as adjuvant which binds with binding domains of monoclonal antibodies, designed for Cell-targeted treatment. In this design, Monoclonal antibodies were able to target hematopoietic differentiation antigens whereas immunotoxin have been shown to be highly cytotoxic toward malignant cells. These therapeutic designed will likely play a promising role in treatment of leukemia (26). The incidence of antitoxin antibodies production in bodies after multiple cycles of treatment causes immunogenicity. There are several approaches are under trial to reduce immunogenicity for the treatment of solid tumor. Monoclonal Antibodies The monoclonal antibodies are a promising part of immune system which recognize and binds with foreign antigens expressed on the cell surface. After binding, they highlight them for further action of destruction of abnormal cells via antibody dependent cellular cytotoxicity (ADCC). In addition to other therapy, they are designed to enhance body’s immune responses which assist to act against cell growth factors. The induction of adaptive anti-tumor immune responses continues to hold for immunization against cancer. There is a practice developing for the manipulation of antibody Fcγ domains to further improvement of efficacy and affinity for Fcγ receptor (s) (27). Still alots of things needs to be exploring about monoclonal antibody as therapeutic agent like as adjuvant, combination with other molecules which can activate co-stimulation, CTLs activation or some interleukin which will give varieties of ways to target and treat tumors in patients. There are several therapeutic monoclonal antibodies against cancer approved in clinical trial like, Rituximab (for Non-Hodgkins lymphoma), Ipilimumab (for melanoma), Trastuzumab (for Breast Cancer), Bevacizumab (For Colorectal Cancer), Cetuximab (For Colorectal, Head and Neck Cancer), Panitumumab (For Metastatic colorectal carcinoma), Brentuximab vedotin (For Hodgkins lymphoma), Alemtuzumab (For Fludaribine – refractory chronic lymphocytic leukemia) (28). In addition to these, several approaches are under trial to increase the ability of antibodies to elicit an anti-tumor immune response. Cytokine Therapy
  • 7. Cytokines are the important regulator of innate immune system: natural killer (NK) cells, neutrophills and macrophages also rectify adaptive immune system with T and B cell immune responses. Usually cytokine functions in cascade, so individual cytokine therapy are not a promising therapy in present. To overcome these drawbacks, there are several trials going for the combination of different cytokine to target a specific mechanism directly. These findings provide evidence for the unique therapeutic opportunity based on selective interference with the action of pro-inflammatory and tumor-promoting cytokines. It has been established that cancer promotion and exacerbation are linked with the cytokines produced by activated innate immune cells. Here we provide an overview of the updated understanding of the role of inflammation-induced cytokines in development of novel therapeutic strategies (Table 1). Cell-based Immunotherapies of Cytokine Cytokine function as autocrine and paracrine, so they function locally or any distant part of body to enhance or suppress immunity. In cytokine therapy for cancer, cytokine uses to induce ant-tumor immunity to fight against cancer. Here we will examine cytokines which are currently under evaluation for cancer therapy. Tumor Necrosis Factor - alpha (TNF – α) In 1975, TNF-α was extracted from the serum of mice treated with ‘Coley’s toxin’ and later on showed the hemorrhagic necrosis of mice tumor (29) , (30). Tumor necrosis factor – α (TNF- α) is a pleiotropic cytokines involved in regulating diverse functions like apoptosis, cell survival, tumorigenesis, autoimmunity, inflammation, and immune response via two receptors i.e. TNFR1 and TNFR2 (31). The interaction of TNF- α molecule with TNFR1 or TNFR2 receptor leads to the activation of various signaling pathways in cell microenvironment (32). In an experiment when TNF-α is administered via ILP (Isolated limb perfusion) into tumor cells, then it was bound with TNFR1 soluble receptors on the cell surface (33). It is well known that TNFR1 is less in count on cell surface which inhibit the cause of cytotoxicity. The effect of binding of TNF - α to TNFR1 results in hyper permeability of the neoplastic vessels and strong extravasations of erythrocytes resulting into massive hemorrhage and necrosis in tumor (34). This excessive hemorrhage is responsible for the inhibition of tumor associated vasculature and the growth of malignant cells. Several clinical studies have also reported that low dose of TNF - α may induce these antitumor effects (35), (36). In case of tumor microenvironment, tumor or inflammatory cells induce macrophages, T lymphocytes and natural killer (NK) cells, which further harvest TNF-α, which induces gene-encoding NF-κB dependent anti-apoptotic molecules and also promote the tumor cell survival (37), (38). TNF-α further stimulate the production of genotoxic molecule Nitric oxide and reactive Oxygen Species that can lead to DNA damage and reduce the antigen- induced cytotoxicity or enhance the tumor progression through suppression of T cell responses and cytotoxic activity of activated macrophages (39). Oxidative stress caused by ROS and NO is a contributor to senescence, which could be an option for developing therapy for tumor treatment. These studies indicate vast therapeutic applications of TNF-α in tumor cell depletion.
  • 8. Another attribute of TNF-α induces hemorrhagic necrosis in certain set of tumor types in which synergistic antitumor effect of the combination treatment with TNF - α and chemotherapeutic drugs as compared to TNF-α alone. (40). TNF - α effect during solid tumor treatment cannot be caused by a direct cytotoxic or cytostatic effect toward the tumor cells (41). However, cell death induction by TNF - α plays only small role in necrosis, but by combination with Fas ligand, activation of NF-κB pathway which transcript the inhibitory proteins that interfere with death signaling in tumor cells, can be an alternative blueprint for developing therapy (42). These strategies of developing combined therapy are very relevant to show the antagonistic effects of TNF-alpha against tumor growth. The same set up also revealed that TNF-α improved the accumulation of chemotherapeutic drugs in tumor. TNF-α super family represents an active target for drug development. Some of the approved therapeutics are primarily either a soluble receptor which binds the circulating TNF- α and prevent the interaction between TNF- α and cell surface receptors, or antibodies against TNF- α. Various antagonist against TNF family have been approved like Golimumab (a humanized TNF- α monoclonal antibody) (43), Certolizumabpegol (a PEGylated Fab fragment of a humanized TNF antibody) (44) and still more are in the process of approval. As described, members of TNF- α are involved in mediation of chronic inflammation, there is a need to regulate the members of TNF- α family members however the role of TNF-β is still elusive. In several medical cohort studies in United States in which cytokine was delivered at appropriate concentration to the tumor site, there are no evidences of enhancement of cancer growth associated with Tumor necrosis factor alpha antagonists TNF therapy in either analysis or follow-up after the patients discontinued therapy (45). So there is much left yet to be learned about TNF-superfamily, and we expect this information to be forthcoming. Of note, there is a need to develop a strategy to identify the patient stratification criteria for the development of individualization of therapy. Therefore, research in the future should be focus on not only recognizing integrated TNFL/TNFR-based combined immunotherapy but also on the individualization of appropriate patient treatment.  Interleukin – 6 It is well established that innate and adaptive immunity released several pro-inflammatory cytokines which contribute to tumor promotion and progression. It is a glycoprotein that shows pleiotropic activity through binding with its transmembrane receptor (46). Among these, the IL-6 receptor complex is a heterodimer structure which consist of IL-6Rα and glycoprotein 130 (gp 130), that is reckon as key anti-inflammatory myokine, pro- inflammatory cytokine, antiapoptotic and growth-promoting factor (47), (48). Experiments based on link between inflammation and carcinogenesis entangled few constituents of inflammatory avalanche such as IL-6 known for its key role in tumor growth and survival (49). Other studies also revealed the multi-utility of IL-6 during chronic inflammation like it induces Th17 cells and inhibits the differentiation of Tregs cells. During IL-6 signaling, soluble IL-6 receptor (sIL-6R) secretion can induce IL-6 trans-signaling which further promote the development of cancer cells. In addition, it has been suggested that shedding of sIL-6R from carcinoma cell surface promotes the T cell survival and increase the production
  • 9. of more IL-6 by T cells (50). Such theories suggested that opposing role of IL-6 might be therapeutically useful for the treatment of colitis-associated colon cancer (CAC) (51). The indefinite therapeutic methods have been developed for the treatment of disease including anti-IL-6 or anti-IL-6R antibodies, soluble gp130Fc (sgp130Fc) and selective small molecule JAK inhibitors which directly/indirectly inhibit the IL-6/STAT3 pathway that is directly involved in sporadic and inflammation-associated CRC development (52) . The first therapeutics targeting of IL-6/STAT3 pathway has been started in clinical studies through the anti-IL-6 antibodies during the 1990s. However, these treatments only produced a limited response. Another strategy to inhibit IL-6/STAT3 signaling is the inhibition of JAK activation through ruxolitinib (53) and CEP-33779 i.e. JAK1 and JAK2 inhibitor could successfully reduce tumor growth in clinical studies (54), (55). The studies of these arrays can yield new insights on the effect of IL6/STAT3 deregulation. All of the treatments mentioned above inhibit physiological functions of IL-6 through inhibition of signaling which has been achieved by binding of sgp130Fc to IL-6 receptors complexes (56). It is mentioned in several studies that components of IL-6 signaling pathway such as IL-6, IL- 6R, JAK has been shown to promote tumor proliferation and anti-apoptotic effects in tumor cells which can be proposed as promising target to achieve therapeutic effects (57), (58). IL-6 is not only involved in tumor growth is also related with the interaction within tumor microenvironment which may also suggest a target for the development of novel IL-6 antibody based therapies like IL-6 inhibitors are being developed include human anti- IL-6R Ab (Sarilumab/REGN88/SAR153191) (59), anti-IL-6R nanobody (ALX-0061), anti-IL-6 Abs such as sirukumab (CNTO 136), BMS – 945429 (ALD518), olokizumab (CDP6038), and MEDI5117, and soluble gp130-Fc fusion protein (FE301) (60) which are in clinical trials for different disease in human body. In addition, Chronic inflammation causes DNA damage which has further shown affecting several pathways including cell based homeostasis like cell-cycle regulation, apoptosis, DNA repair (61). These conditions nourish the production of IL-6 in patients. However, attenuating chronic inflammation with surgery, radiation and chemotherapy as well as combining these therapies with IL-6 inhibition may be one of the ways to decrease inflammation and add prognostic values to different methods (62). Recent researches have concluded the potential for IL-6 as a prognostic factor. So there is need of clinical studies to further evaluate potential which could help direct additional treatment strategies in the future. However, additional studies and appropriate clinical trials need to be done to make sure the effectiveness of anti-IL-6 therapeutic modalities in cancer patients.  Interleukin-17 Recent studies on the role of different T cell subset named Th17 identified by the production of IL-17 (IL-17A) as well as related cytokine IL-17F cytokine, explain as implication of IL- 17 in inflammatory responses primarily as a proinflammatory factor (63). IL-17A expresses on tumor cells and develops antitumor immunity in immunocompetent mice which suppresses tumor progression whereas in some cases it also enhances the angiogenesis in immune-deficient mice which promote tumor progression (64). IL-17 is induced by IL-23 that responds to the invasion of immune system and induces destruction of foreign antigen cellular matrix. IL-17 is the signature cytokine of a unique lineage of CD4+ T cells termed as Th17 and also known as a potent mediator in delayed-type
  • 10. reactions by enhancing chemokine production throughout body to recruit immune cells to the site of chronic inflammation (65). The properties of IL-17 like foreign antigen destruction, induction of chemokine to the site of inflammation, synergistics action with TNF and IL-17 are very promising features to target it as a strategy against tumor reduction. IL-17 also acts as an angiogenesis factor whose up-regulation may results in blockade of angiogenesis in tumor and provokes the host inflammatory response to tumor progression (66). The up-regulation of IL-17F includes modulation of cytokines regulating IL-17F production (67) whereas blocking of IL-17A signaling may inhibit tumor angiogenesis but sustains the risk of diminishing host antitumor response. Tumor endothelial marker 8 (TEM8) expressed on human tumor vasculature is the main factor in promotion of angiogenesis (68). The anti-TEM8 antibody can be used to inhibit the expression of TEM8 and it is also helpful to segment the other anti-angiogenesis agent’s activity. By controlling the diminishing of antitumor response and increase in the blockade of tumor angiogenesis by using the synergistic effect of different anti-angiogenesis agent, it is possible to inhibit the growth of tumor. It is clear that IL-17 is skillful of exerting multiple effects within tumor microenvironment with bidirectional effects in tumor growth, so further investigation should be approached with a definite knowledge of complex interaction of IL-17 within the tumor microenvironment. Therefore, we propose that there should be some novel inhibitory molecule and immunomodulators mixed with these cytokine based therapy to achieve the beneficiary effect in treating malignant solid tumors. Several studies are under progress to investigate the relation, functions and involvement of IL-17F and IL-17A in tumor progression which provide an unusual strategy for any therapeutic application of these cytokine in therapy for cancer.  Interleukin-12 and Interleukin-23 (IL-12 cytokine family) Both of these cytokine are the members of IL-12 cytokine family and can be released by Antigen presenting cells (APC) like dendritic cells, macrophages, and human B- lymphoblastoid cells into the inflammatory sites upon microbial infections (69). Recent studies have shown that there are several similarities between IL-12 and IL-23 receptor subunits and signaling but still they impel different immunological pathways and exhibit different response towards tumor development. Research evidences has shown that IL-12 cytokine family members contain anti-proliferative and pro-apoptotic functions like IL- 12Rβ2 which could act as a tumor suppressor gene and IL-12R transfected B lymphoma cells significantly deplete cell proliferation in animal models (70). Although, it is well established that IL-23 enhance the production of TH17-mediated response and induce IL-17 production which further promote end-stage inflammation (71), whilst the proliferation of memory T cells and induction of IFN-γ are done by IL-12 which play as a major role in exhibitions the antitumor immune response of IL-23 (72). Interleukin-12 is a disulfide-linked heterodimer cytokine, composed of two subunits i.e. IL- 12p40 and IL-12p35 (73), (74), whereas another cytokine IL-23 is also an heterodimer molecule composed of IL-12p40 and IL-23p19 subunits and both of these cytokines belongs to the IL-12 family of proinflammatory cytokine (75). IL-12 binds to its heterodimer receptors i.e. IL-12Rβ-IL-12Rβ2, whereas IL-23 binds an IL-12Rβ1-IL-23R heterodimer (75). This heterodimer cytokine complex stimulate the expression of their receptors to maintain the expression of several proteins involved in IL-12 based signaling pathways in NK cells to enhance the cytotoxic activity.
  • 11. IL-12 has been shown to exhibit antitumor activity through the immune-stimulatory effects on certain T helper cells (Th1), as well as on cytotoxic T lymphocytes (CTL) and natural killer (NK) cells (76), (77), (78). There are certain other cytokine like IL-1, IL-2, IL-15 and tumor necrosis factor-α, which act synergistically with IL-12 to induce the secretion of IFN-γ which exhibit direct toxic effect on tumor and antiangiogenic activity (77), (79), (80). This type of enhanced functional response of IL-12 is demonstrated by IFN-γ production and killing of target cells. As it is clear that IL-12 has the ability to induce anti-angiogenic activity and immune system, there should be studies on using IL-12 as a possible anti-cancer therapy. Another synergistic property of IL-12 and IL-23 is that they both activate TYK2 and JAK2 as well as STAT1, STAT3, STAT4 and STAT5, whilst IL-12 alone activates STAT4 efficiently and IL-23 preferentially activates STAT3 (75). IL-23 has been known as a key factor controlling inflammation in peripheral tissues which is mediated by induction of JAK/STAT pathway following IL-23 binding. As described above that IL-23 suppresses the innate immune response during tumor progression and metastasis. Based on these hypotheses, IL-23 neutralization strategies may promise to modulate immunosuppression in combination with immunostimulatory agents (81). In an experimental study, α-IL-23 mAb has been given to wild-type mouse resulted in fewer lung metastasis than in mice treated with control Ig (α-AGP3), which respond in the form of NK cell-mediated antitumor immune response of α-IL-23 mAb (82). Collectively, the use of α-IL-23 mAb in combination with IL-2 mAb treatment significantly inhibited subcutaneous growth of established mammary carcinoma and suppressed established experimental and spontaneous lung metastases. In other trial studies, the anti-IL-23R treated mice displaced a significant reduction in the number of tumor infiltrating Tregs compared with control antibody treated mice. These studies exhibit another role of IL-23 driven STAT3 activation in tumor-infiltrating Tregs with the addition of IL-23R dependent signaling pathway (83). Since past 15 years, IL-12 cytokine family based therapies have acquired so much attention and are apparently developing more effective therapy for the survival and treatment of cancer-bearing patients (84). Of note, current studies postulated that over-expression of IL- 23R mainly suppress the expression of anti-apoptotic factor like Bcl-xL and induce the intrinsic apoptotic pathways in 293ET and HeLa cell lines during trial (69). But tumor immunosuppression still remains one of the major barriers in the development of novel cancer therapies and vaccines. Several studies have been shown that initial stage of cancer treatment, mAb neutralizing IL-23 alone and exerting a temporary effect. So there is a need to target the combination of α-IL-23 mAb with other cytokine based antibodies to exert potential effect in treatment of cancer.  Interleukin-10 Interleukin-10, also known as human cytokine synthesis inhibitory factor (CSIF), having a pleiotropic effects in inflammation and immune-regulation which directly affect the function of antigen-presenting cells by inhibiting the expression of MHC and co-stimulatory molecule (85), (86). In 1989, Mosmann and his coworkers postulated that T helper 2 (Th2) cell clones produced a cytokine which inhibit the synthesis of interferon (IFN) in Th1 cell clones (87), and inhibit the NF-κB activation through ill-defined mechanisms (88) (89), is known as interleukin -10 (IL-10). It is also known for the production of proinflammatory cytokine like
  • 12. TNF-α, IL-6, and IL-12 (90) and down-regulation of the expression of MHC class II antigens, co-stimulatory molecules on macrophages. Several transcription factor responsive elements promotes the major source of IL-10 i.e. macrophages to modulate the production of IL-10 through these exogenous and endogenous factors such as endotoxin via Toll-like receptor, tumor necrosis factor (TNF) , catecholamines and cAMP-elevation drugs via protein kinase A (91), (92), (93), (94), (95), (96). In contrast to these observations, IL-10 produced by B-cell lymphoma functions as an autocrine growth factor (97). These previous mentioned reports concluded that IL-10 production within the tumor microenvironment is a common phenomenon in cancer metastasis which might be the major hurdle for anticancer immunotherapy such as cancer vaccines. In case of metastatic melanoma patients, it was reported that high IL-10 production were used for synthesis of homologous melanoma cell vaccine estimated fewer survival after vaccine treatment (98). In another trial, anti-IL-10 antibody treatment has been administered in two mouse models of acquired hepatitis (99). This study revealed that IL-10 plays a protective role down regulating the expression of several other inflammations causing cytokine including IL-12, TNF-α and INF-γ (100). IL-10 is able of affecting the tumor cells and inhibiting the immune cells which may convert tumor cells to a CTL resistant phenotype. Kiessling and his coworkers reported that IL-10 treatment results in 50% reduction of expression of MHC class-I in human melanoma cells and 100% inhibition of CTL-mediated lysis of tumor cells (101). The consequence of IL-10 has been demonstrated in transgenic mice by uncontrolled growth of immunogenic tumors because of the blockade of the antitumor immune reaction. The anti-IL-10 antibodies treatment thus reinstates the anticancer response (102). Several studies conducted on Systemic Lupus Erythematosus (SLE) patients concluded that treatment of anti-IL-10 antibody to SLE-patients results in the lessening of autoantibody production due to the depletion of IL-10 (103). Some other experimental models suggest that IL-10 may retain antitumor response and immune stimulation (104), (105) like transfection of mouse melanoma cell lines with IL-10, extorts induced immunogenicity following a strong lymphocyte and antibody-dependent immune memory (106). Additionally, IL-10 induces the production of Treg cells (107), (108). So rather than giving anti-IL-10 therapy alone, it is suggested that a combination of anti-IL-10 antagonist with other immune-stimulants such as cancer vaccines, other cytokine will be more synergistically effective in inhibiting the tumor-specific immune tolerance and promoting antitumor immune responses. It suggests that balance of IL-10 should be maintained to regulate the immune response against any infection in body. These trials give new insights to the IL-10 immunobiology to become a new therapeutic target. In contrast, complex effect of IL-10 may cause reduced potency of treatment depending upon IL-10 rather than promising effect of other combined approaches. Given the overall outcomes of systemic IL-10 therapies of disease, it has become apparent that future studies on the sources of IL-10 and kinetics of IL-10 production may lead to the identification of new target with better therapeutic efficiency and also facilitates the development of treatment strategies aimed at regulating inflammation. These studies will have to understand the details of circulating levels of cytokines and their gene variations inside the patients which will assist in development of therapy with defined parameters of treatment side effects. These above proclamation about IL-10 cytokine revealed the utility as an essential immunoregulatory which favorably react toward different treatment, evaluating the assistance
  • 13. of IL-10 for counteracting the inflammatory effects in the human body. The generalized therapeutic effect of IL-10 in cancer treatment is hard to predict as both tumor-suppressive as well as tumor-promoting effects have been demonstrated. Since, IL-10 is produced in metastatic stage of cancer, it insinuate more equitable to clock the effects of IL-10, rather than administrating IL-10 to cancer patients. These features can assist in the development of recombinant therapy for several diseases.  TGF-β TGF-β is known as a secreted polypeptide skillful of inducing fibroblast growth and collagen fructification which was further discovered as a pervasive and essential key regulator of proliferation, differentiation, migration, cell survival, cell survival, angiogenesis and immunosurveillance (109), (110), (111). Tumor progression is stimulated by TGF- β by two mechanisms: firstly intrinsic activity and, another extrinsic activity (112). During the growth phase of tumor, excessive amount of TGF-β is produced whereas in normal epithelial cells, it is known as a potent growth inhibitor. Normally, TGF- β blocks the G1 stage of normal cells to regulate the balance of growth in body but when a cell is transformed into a malignant cell TGF- β signaling pathway is altered and it losses the control on cell growth resulting into cancer and surrounding cells escalated the production and increase in their own TGF- β which acts on microenvironment (113). It causes immunosuppression and angiogenesis, which makes the malignant cells violent. TGF-β has the dichotomous function in human cancers. First it is having the capability to act as a tumor suppressor (via its effects on replication, proliferation, and apoptosis) and another as a tumor promoter (via its effects on progression, metastasis, invasion and angiogenesis) (114). So profiling of TGF-β ligand (in serum, urine, and tissue), TGF-β receptor levels and TGF-β mRNA (in tissue) may serve as a diagnostic and therapeutic tools for cancer treatment. It is predicted that strategies such as inhibition of signaling pathway, monoclonal TGF-β- neutralizing antibodies, large molecule ligand snare, lowering translational efficiency of TGF-β ligand by antisense technology and estranging TGF-β receptor I/II kinase function by inhibitor molecule which are the most conspicuous methods being investigate these days (115), (116). For example, first is the utilization of soluble TGFIIR (human α2- macroglobulin) plasma protein that binds TGF-β isoform, blockade the interaction between cytokine and its receptor (117) and two other TGF-β1 and TGF-β2 specific humanized monoclonal antibodies like CAT-192 and CAT-152 respectively, are going under clinical trial for cure of fibrosis and their development will embolden the therapeutic application to anti- cancer therapy (118), (119). Antisense TGF-β treatment in a variety of cancer has also been delineating the response in the form of reactivation of number of different therapeutic scope. In addition, it has been shown that combined treatment with tumor cell vaccines and antisense TGF-β therapy or monoclonal antibodies therapy respond synergistically and downsize the tumor volume and escalate the survival benefit (120), (121), (122). Now it is well known that increased quantity of TGF-β has superlative role in tumorigenesis due to reprogramming of immune surveillance which makes it a very advisable target for different therapeutic modalities like monoclonal TGF- β antibody, antisense oligonucleotide or receptor kinase inhibitors. From different outcome of studies, it is revealed that continuous
  • 14. suppression of TGF- β signaling pathway may lead to harmful off-target effect. So there is a need to focus on combined therapy with these inhibitors for the therapeutic profit in future. There are three leading therapeutic targeting of TGF- β in clinical trials: TGF-B antibodies, antisense oligonucleotide, and receptor kinase inhibitors. These all strategies have their own limitations. So it is necessary to reengineer the TGF- β inhibitors within a combinational therapy. Of note, it is known that increased level of TGF-β in tumor microenvironment induce the tumorigenesis through various processes, thereby making it possible to target these for therapeutic development. Conclusively, the precise mechanisms involved in transformation of TGF-β during malignancies needs to be deeply understood only. After that, it will be possible to prosper successful therapies as well as provide new therapeutic targets to fix the normal TGF-β function. Conclusion We have sought here to revisit, refine, and extend the concept of therapeutic targeting of cytokine, which has provided a useful conceptual framework for understanding the complex foundation of cancer biology. Advancement of role of cytokines in immune and inflammatory diseases has promoted the development of cytokine-based therapies. Cytokine therapies have tremendous capabilities for treating a variety of diseases either by inhibiting or restoring the activity of specific cytokines. As cancer is caused due to an alteration in cellular programming, cytokine therapy is the one best choice to treat cancer. However, as cancer is a part of the wound healing process, single cytokine therapy may not be effective. This might increase the likelihood of cancer recurrence and development of alternate signaling pathway to facilitate tumor cell growth. So it is necessary to work on dominant module in modern immunologic research, which is the illustration of commutative effect of different cytokine therapies. One of the approaches is known as Gene therapy as they enable therapy to restrict to tumor cells only which will decrease the side-effect and increases the efficacy of therapy. As prescribed above, several studies indicate the role of combined cytokine therapy like the combination of TNF-α and IL-6, it is evident that IL-6 signaling embellished the protraction of TNF-α, this interference restrict tumor progression and IL-6 production. IL-23 induces IL-17 production which further endorses end-stage inflammation whilst the rise of memory T cells and induction of IFN-γ are made by IL-12 which serves as a foremost role to reveal the antitumor immune response of IL-2. As previously revealed that IL-12 based therapy is more valuable with smaller tumor burden, so there is a need to investigate the role of combination of IL-10 with other therapy as an adjuvant to get more effective therapeutic effects. The contribution of CD4 T cells, CD8 T cells, and IFN-γ and NK cells in induction of antitumor effect of IL-12 therapy is still unclear and itmay give a better opportunity to investigate the role of combined therapy in treatment. Through the rapidly growing research of intricate signaling system of different cytokines, new effective therapies have become available to treat oncology patient like blocking or neutralizing cytokine action with monoclonal antibodies and one of the drugs that inhibit release of inflammatory cytokines, such as tumor necrosis factor-α (TNF- α). It is thus predictable that new combined therapies of cytokine which are based on innovative technologies will progressively reach the efficacy of new cancer treatment strategies. In some cases, the cure afforded of by these cell based cytokine therapies may prove to be even more effective than chemotherapies and other traditional treatment methods.
  • 15. In my vision, I can foresee the limitative single cytokine therapy being overcome by the help of combined cytokine therapy along with gene therapy. What do we expect in the future? Combined immunotherapy may be the next great hope in the treatment of cancer. Several of these trials have shown impressive clinical results signaling that we are in the entry and exciting phase of cancer treatment. It is likely our responsibility to combat cancer from all fronts. Additional studies required to define the mechanism of cytokine cascade in body which will help to reduce the opposite effect of therapy and make it specific for every cancer type. One of the hurdles have been identified, i.e. immunogenicity, which may be resolved by depleting or suppressing T and B cell epitopes. At present, nonspecific and specific immunotherapy combination showing promise for another potent strategy for treatment. Even, the effect of any aforementioned therapy module in combination with some traditional cancer treatment method is another avenue, as it is come out in trial in terms of duration / remission period with cytokine and chemotherapy. All through with these concerted efforts, our decisive ambition may be a long-lasting anti- tumor immune response that can be maintained throughout patient’s lifespan. ………………………………….. References 1. Meng X, Zhong J, Liu S, Murray M, Gonzalez – Angulo AM. A new hypothesis for the cancer mechanism. Cancer Metastasis Rev. 2012 Jun 31: 247-68. 2. Bhatt A N, Mathur R, Farooque A, Verma A, Dwarakanath B S. Cancer Biomarkers – current perspectives. Indian J. Med. Res. 2010: 132: 129-149. 3. Kinzler K.W., Vogelstein B.. Lessons from hereditary colorectal cancer. Cell. 1996; 87: 159-170. 4. Laubenbacher R., Hower V., Abdul Jarrah A., Torti SV, Shulaev V, Mendes P. et al., A Systems Biology View of Cancer. Elsevier. 2009 Dec 2; 1796: 129-139. 5. Hanahan D., Weinberg RA. Hallmarks of Cancer: The Next Generation. Elsevier. 2011 March 4; 144: 646-674. 6. C Athena Aktipis CA, Nesse RM. Evolutionary foundations for cancer biology.Evo app. 2013; 6: 144-159. 7. Cavallo F, Giovanni CD, Nanni P , Forni G , Lollini PL. The immune hallmarks of cancer. Cancer Immunol Immunother. 2011 Mar; 60(3): 319-326. 8. Lemmon, M.A., and Schlessinger. Cell signaling by receptor tyrosine. Cell. 2010; 141: 1117-34. 9. Matsubara H, Suzuki K. Recent Advances in p53 Research and Cancer Treatment. Journal of Biomedicine and Biotechnology. 2011; 1-7. 10. Burkhart D.L., and Sage J. Cellular mechanisms of tumour suppressionn by the retinoblastoma gene. Nat. Rev. Cancer. 2008; 8: 671-682. 11. Berx G, Roy FV. Involvement of members of the cadherin superfamily in cancer. Cold Spring Harb Perspect Biol. 2009 Dec; 1(6).
  • 16. 12. Johnson, J. P. Cell adhesion molecules of the immunoglobulin supergene family and their role in malignant transformation and progression to metastatic disease. Cancer Metastasis Rev. 1991; 10: 11-22. 13. Cavallaro U, Christofori G. Cell adhesion and signalling by cadherins and Ig-CAMs in cancer. Nat. Rev. Cancer. 2004; 4: 118-132. 14. Coussens LM, Werb Z. Matrix metalloproteinases and the develoment of cancer . Chem. Biol. 1996; 3: 895-904. 15. Liub, Jing Zhanga and Jinsong. Tumor stroma as targets for cancer therapy. Pharmacol Ther. 2013; 137: 200-215. 16. Witz I P, Levy-Nissenbaum O. The tumor microenvironment in the post-PAGET. Cancer Lett. 2006; 242: 1-10. 17. Marie-Aude Le Bitoux, Ivan Stamenkovic. Tumor-host interactions: the role of inXammation Histochem Cell Biol. 2008; 130: 1079-1090. 18. Balkwill F. Cancer and chemokine network. Nat. Rev. Cancer. 2004; 4: 540-550. 19. Shiao SL and Coussens LM. The Tumor-Immune Microenvironment and Response to Radiation Therapy. J Mammary Gland Biol Neoplasia. 2010; 15: 411-421. 20. Ira Mellman, George Coukos and Glenn Dranoff. Cancer immunotherapy comes of age. Nature. 2011; 480: 480-489. 21. Gabrilovich DI Combination of chemotherapy and immunotherapy for cancer: a paradigm revisited. The Lancet Oncology. 2007 jan 1; 8: 2-3. 22. Chen L, Ashe S, Brady WA, Hellstrom I, Hellstrom KE, Ledbetter JA et al. Costimulation of antitumor immunity by the B7 counterreceptor for the T lymphocyte molecules CD28 and CTLA- 4. CELL. 1992 Dec 7; 71: 1093-1102. 23. Zang X and Allison JP.. The B7 Family and Cancer Therapy: Costimulation and Coinhibition. clinical cancer research. 2007; 13: 5271-5279. 24. Kim YT, Hersh EM, Trevor KT. Feasibility to generate monocyte-derived dendritic cell from coculture with melanoma tumor cells in the presence of granulocyte/macrophage colony- stimulating factor (GM-CSF) and interleukin-4. Am J Reprod Immunol. 2003; 49: 230-8. 25. Antignani A and FitzGerald D. Immunotoxins: The Role of the Toxin. Toxins. 2013; 5: 1486- 1502. 26. Wayne AS, FitzGerald DJ, Kreitman RJ, Pastan I. Immunotoxins for leukemia. Blood. 2014; 123: 2470-2477. 27. Weiner LM, Dhodapkar MV, and Ferrone S. Monoclonal Antibodies for Cancer Immunotherapy. Lancet. 2009; 373: 1033-1040. 28. Adler MJ and Dimitrov DS. Therapeutic antibodies against cancer. Hematol Oncol Clin North Am. 2012; 26: 447-481. 29. Carswell EA, Old LJ, Kassel RL. An endotoxin-induced serum factor that causes necrosis of tumors. Proc Natl Acad Sci USA. 1975 Sep ; 72(9): 3666-3670. 30. Balkwill F. Tumour necrosis factor and cancer. Nature Reviews Cancer. 2009 May; 9: 361-371. 31. Faustman DL and Davis M. TNF receptor 2 and disease: autoimmunity and regenerative medicine. Frontiers in immunobiology. 2013; 4: 1-8. 32. Croft M, Benedict CA, and Ware CF. Clinical targeting of the TNF and TNFR superfamilies. Nat Rev Drug Discov. 2013; 12: 147-168. 33. A M Eggermont AM, H Schraffordt Koops HS, J M Klausner JM, B B Kroon BB, P M Schlag PM, D Liénard D et al. Isolated limb perfusion with tumor necrosis factor and melphalan for limb salvage in 186 patients with locally advanced soft tissue extremity sarcomas. The cumulative multicenter European experience. Ann Surg. 1996 Dec; 224: 756-765. 34. Bradley JR, Thiru S, Pober JS. Disparate localization of 55-kD and 75-kD tumor necrosis factor receptors in human endothelial cells. Am. J. Pathol. 1995; 146: 27-32.
  • 17. 35. Hill S, Fawcett WJ, Sheldon J. Low-dose tumor necrosis factor alpha and melphalan in hyperthermic isolated limb perfusion. Br. J. Surg. 1993; 80: 995-997. 36. Bonvalot S, Laplanche A, Lejeune F. Limb salvage with isolated perfusion for soft tissue sarcoma: could less TNF-alpha be better? Ann. Oncol. 2005; 16: 1061-1068. 37. Luo JL , Maeda S, Hsu LC, Yagita H, and Karin M. Inhibition of NF-kB in cancer cells converts inflammation-induced tumor growth mediated by TNF-alpha to TRAIL-mediated tumor regression. Cancer cell. 2004; 6: 297-305. 38. van Horssen R, Ten Hagen TL, Eggermont AM. TNF-α in Cancer Treatment:Molecular Insights, Antitumor Effects, and Clinical Utility. The Oncologist. 2006; 11: 397-408. 39. Lin WW, Karin M. A cytokine-mediated link between innate immunity, inflammation, and cancer. J. Clin. Investigation. 2007; 117: 1175-1183. 40. Manusama ER, Nooijen PT, Stavast J. Synergistic antitumor effect of recombinant human tumor necrosis factor alpha with melphalan in isolated limb perfusion in the rat. Br. J. Surg. 1996; 83: 551-555. 41. Brouckaert P, Takahashi N, Van Tiel ST. Tumor necrosis factor-alpha augmented tumor response in B16BL6 melanoma-bearing mice treated with stealth liposomal doxorubicin. Int J. Cancer. 2004; 109: 442-448. 42. Özören N, El-Deiry WS. Cell surface Death Receptor signaling in normal and cancer cells. Elsevier. 2003; 13: 135-147. 43. Hirohata S. Fully human anti TNF-alpha monoclonal antibodies (adalimumab, golimumab). Nihon Rinsho. 2007 Jul 7; 65: 1202-08. 44. Goel N and Stephens S. Certolizumab pegol. Landes Biosciences. 2010; 2: 137-147. 45. Haynes K, Beukelman T, Curtis JR, Newcomb C, Herrinton LJ, Graham DJ et al. Tumor Necrosis Factor Alpha Inhibitor Therapy and Cancer Risk In Chronic Immune Mediated Diseases. Arthritis Rheum. 2013 Jan 1; 65: 1-22. 46. Yoshida Y and Toshio T. Interleukin 6 and Rheumatoid Arthritis. BioMed Research International. 2014; 2014: 01-12. 47. Ishihara K, Hirano T. IL-6 in autoimmune disease and chronic inflammatory proliferative disease. Cytokine growth factor Rev. 2002; 13: 357-368. 48. Hodge, D.R., Hurt, E.M., and Farrar, W. L. The role of IL-6 and STAT3 in inflammation and cancer . Eur. J. Cancer. 2005; 41: 2502-2512. 49. Mantovani A. Role of inflammatory cells and mediators in tumor invasion and metastasis. Cancer Metastasis Rev. 2010; 29: 241. 50. Rose-John S, Scheller J, Elson G, and Jones SA. Interleukin-6 biology is coordinated by membrane-bound and soluble receptors: role in inflammation and cancer. J. Leukoc. Biol. 2006; 80: 227-236. 51. Becker C. TGF-beta suppresses tumor progression in colon cancer by inhibition of IL-6 trans- signalling. Immunity. 2004; 21: 491-501. 52. Jones SA, Scheller J, Rose-John S. Therapeutic strategies for the clinical blockade of IL-6/gp130 signaling. J. Clin. Invest. 2011; 121: 3375-83. 53. Hobson SA, PharmD, Waddell JA, Solimando DA Jr. Enzalutamide and Ruxolitinib. Hosp Pharm. 2013; 48: 104-107. 54. Eghtedar A, Verstovsek S, Estrov Z, Burger J, Cortes J, Bivins C. Phase II study of the JAK kinase inhibitor ruxolitinib in patients with refractory leukemias, including post myeloproliferative neoplasms (MPN) acute myeloid leukemia (AML). Blood. 2012; 119: 4614-8. 55. Seavey MM, Lu LD, Stump KL, Wallace NH, Hockeimer W, O'Kane TM. Therapeutic efficacy of CEP-33779, a novel selective JAK2 inhibitor, in a mouse model of colitis-induced colorectal cancer . Mol. Cancer Ther. 2012; 11: 984-93. 56. Becker C, Fantini MC, Schramm C, Lehr HA, Wirtz S, Nikolaev A. TGF-beta suppress tumor progression in colon cancer by inhibition of IL-6 trans-signaling . Immunity. 2004; 21: 491-501.
  • 18. 57. Taga T, Hibi M, Hirata Y, Yamasaki K, Yasukawa K, Matsuda T et al. Interleukin-6 triggers the association of its receptor with a possible signal transducer, gp130. Cell. 1989; 58: 573-581. 58. N. Stahl N, T. G. Boulton, T. Farruggella. Association and activation of Jak-Tyk kinases by CNTF-LIF-OSM-IL-6 ? receptor components. Science. 1994; 263: 92-95. 59. Huizinga TW , Fleischmann RM, Jasson M, Radin AR, Adelsberg JV, Fiore S et al. Sarilumab, a fully human monoclonal antibody against IL-6Rα in patients with rheumatoid arthritis and an inadequate response to methotrexate: efficacy and safety results from the randomised SARIL-RA- MOBILITY Part A trial. Clinical and epidemiological research. 2014 Sep;73(9): 1-9. 60. Tanaka T, Ogata A, and Kishimoto T. Targeting of interleukin-6 for the treatment of rheumatoid arthritis: a review and update. Rheumatology: Current Research. 2013 June 17; 3:1-14. 61. Hussain SP, Harris CC. Inflammation and cancer: an ancient link with novel potentials. Int J Cancer. 2007; 121: 2372-2380. 62. Duffy SA, Teknos T, Taylor JM, Fowler KE, Islam M, Wolf GT et al. Health Behaviors Predict Higher Interleukin-6 levels Among Patients Newly Diagnosed with Head and Neck Squamous Cell Carcinoma. Cancer Epidemiol Biomarkers. 2013; 22: 374-381. 63. Fujino S, Andoh A, Bamba S. Increased expression of interleukin 17 in inflammatory bowel disease. Gut. 2003 Jan; 52(1): 65-70. 64. Wang M, Wang L, Ren T, Xu L, Wen Z. IL-17A/IL-17RA interaction promoted metastasis of osteosarcoma cells. Cancer Biology and Therapy. 2013; 14: 155-163. 65. Qiu Z, Dillen C, Hu J, Verbeke H, Struyf S, Van Damme J, Opdenakker G. Interleukin-17 regulates chemokine and gelatinase B expression in fibroblasts to recruit both neutrophils and monocytes. J.of Immunol. 2009; 214: 835-842. 66. Yan H, Tong Z. Interleukin-17F: A Promising Candidate of Cancer Therapy. Immuno- Gastroenterology. 2012; 1: 100-103. 67. Chang SH and Dong C . IL-17F: Regulation, signaling and function in inflammation. Elsvier. 2009; 46: 7-11. 68. Chaudhary A, St Croix B. Selective blockade of tumor angiogenesis. Cell Cycle; 2012 Jun 15; 11: 2253-9. 69. Wei-Ye Shi, Chang-Yan Che and Li Liu. Human Interleukin 23 Receptor Induces Cell Apoptosis in Mammalian Cells by Intrinsic Mitochondrial Pathway Associated with the Down-Regulation of RAS/Mitogen-Activated Protein Kinase and Signal Transducers and Activators of Transcription factor 3 Signaling. Int J of Mol Sci. 2013 Dec 18; 14(12): 24656-24669. 70. Airoldi I, Carlo ED, Banelli B, Moserle L, Cocco C, Pezzolo A et al. The IL-12Rβ2 gene functions as a tumor suppressor in human B cell malignancies. J. Clin. Invest. 2004; 113(11): 1651-59. 71. Cho ML, Kang JW, Moon YM, Nam HJ, Jhun JY, Heo SB et al. STAT3 and NF-kappa B signal pathways is required for IL-23-mediated IL-17 production in spontaneous arthritis animal model IL-1 receptor antagonist-defecient mice . J. Immunol. 2006 May; 176(9): 5652-5661. 72. Hao JS and Shan BE. Immune enhancement and anti-tumor activity of IL-23. Cancer Immunol. Immunother. 2006 Nov; 55(11): 1426-1431. 73. Kobayashi M, Fitz L, Ryan M, Hewick RM, Clark SC, Chan S. Identification and purification of natural killer cell stimulatory factor (NKSF), acytokine with multiple biologic effects on human lymphocytes. J Exp Med. 1989 Sep 1; 170(3): 827-45. 74. Stern AS, Podlaski FJ, Hulmes JD. Purification to homogeneity and partial characterization of cytotoxic lymphocyte maturation factor from human B-lymphoblastoid cells. Proc Natl Acad Sci USA. 1990 Sep; 87(17): 6808-12. 75. Watford WT, Hissong BD, Bream JH, Kanno Y, Muul L, OShea JJ. Signaling by IL-12 and IL-23 and the immunoregulatory roles of STAT4. Immunol Rev. 2004 Dec; 202: 139-156. 76. Trinchieri, G. Interleukin-12 and the regulation of innate resistance and adaptive immunity . Nat. Rev. 2003 Feb; 3(2): 133-46.
  • 19. 77. Chan SH, Perussia B, Gupta JW, Kobayashi M, Pospisil M, Young HA et al. Induction of interferon γ production by natural killer cell stimulatory factor: characterization of responder cells and synergy with other inducers. J Exp Med. 1991 Apr 1; 173(4): 869-79. 78. Brunda MJ, Luistro L, Warrier RR, Wright RB, Hubbard BR, Murphy M et al. Antitumor and antimetastatic activity of interleukin 12 against marine tumors. JEM. 1993 Oct 1; 178: 1223-1230. 79. Carson WE, Ross ME, Baiocchi RA, Marien MJ, Boiani N, Grabstain K.. Endogenous production of interleukin 15 by activated human monocytes is critical for optimal production of interferon-γ by natural killer cells in vitro. J Clin Invest. 1995 Dec; 96(6): 2578-82. 80. Gazzinelli RT, Hieny S, Wynn TA, Wolf S, Sher A. Interleukin 12 is required for the T- lymphocyte-independent induction of interferon γ by an intracellular parasite and induces resistance in T-cell-deficient hosts . Proc Natl Acad Sci USA. 1993 Jul 1; 90(13): 6115-6119. 81. Teng MW, Scheidt BV, Duret H, Towne JE, and Smyth MJ. Anti-IL-23 Monoclonal Antibody Synergizes in Combination with Targeted Therapies or IL-2 to Suppress Tumor Growth and Metastases. Cancer Research. 2011 Mar; 71(6): 2077-2086. 82. Michele W.L. Teng, Bianca von Scheidt, Helene Duret, Jennifer E. Towne, and Mark J. Smyth. Anti-IL 23 Monoclonal Antibody Synergies in Combination with Targeted Therapies or IL – 2 to suppress Tumor Growth and Metastases. Cancer Research. 2011 Mar 15; 71: 2077-2086. 83. Kortylewski M, Xin H, Kujawski M, Lee H, Liu Y, Harris T. Regulation of the IL-23 and Il-12 balance by Stat3 signaling in the tumor microenvironment. Cancer cell. 2009 Feb 3; 15(2): 114- 23. 84. Witold Lasek W, Zagozdzon R, Jakobisiak M. Interleukin 12: still a promising candidate for tumor immunotherapy? Cancer Immunol Immunother. 2014; 63: 419-435. 85. Pestka S, Krause CD, Sarkar D, Walter MR, Shi Y, Fisher PB. Interleukin-10 and related cytokines and receptors. Annu. Rev. Immunol. 2004; 22: 929-979. 86. Sato T, Sato MT, Terai M, Tamura Y, Alexeev V, Mastrangelo MJ et al. Interleukin 10 in the tumor microenvironment: a target for anticancer immunotherapy. Immunol Res. 2011 Dec; 51(2): 172-80. 87. Fiorentino DF, Bond MW, and Mosmann TR. Two types of mouse T-helper cell. IV. Th2 clones secrete a factor that inhibits cytokine production by Th1 clones. J. Exp Med. 1989 Dec 1; 170(6): 2081-95. 88. Schottelius AJ, Mayo MW, Sartor RB, and Baldwin AS Jr. Interleukin-10 signaling blocks inhibitor of kappa-B kinase activity and nuclear factor kappaB DNA binding. J. Biol. Chem. 1999 Nov 5; 274(45): 31868-31874. 89. Hoentjen F, Sartor RB, Ozaki M, and Jobin C. STAT3 regulates NF-kappaB recruitment to the IL-12p40 promoter in dendritic cells . Blood. 2005 Jan 15; 105(2): 689-696. 90. Moore KW, De Waal MR, Coffman RL, O’Garra A. Interleukin-10 and the interleukin-10 receptor. Annu. Rev. Immunol. 2001; 19: 683-765. 91. Platzer C, Meisel C, Vogt K, Platzer M, and Volk HD. Up-regulation of monocytic IL-10 by tumor necrosis factor-alpha and cAMP elevating drugs . Int. Immunol. 1995 Apr; 7(4): 517–523. 92. Platzer C, Fritsch E, Elstner T, Lehmann MH, Volk HD, and Prosch S. Cyclic adenosine monophosphate-responsive elements are involved in the transcriptional activation of the human IL-10 gene in monocytic cells. Eur J Immunol. 1999 Oct; 29(10): 3098-104. 93. Platzer C, Docke WD, Volk HD, and Prosch S. Catecholamines trigger IL-10 release in acute systemic stress reaction by direct stimulation of its promoter/ enhancer activity in monocytic cells. J Neuroimmunol. 2000 Jun 1; 105(1): 31–38. 94. Meisel C, Vogt K, Platzer C, Randow F, Liebenthal C, and Volk HD. Differential regulation of monocytic tumor necrosis factor-alpha and interleukin-10 expression. Eur J Immunol. 1996 Jul; 26(7): 1580–1586.
  • 20. 95. Woiciechowsky C, Asadullah K, Nestler D, Eberhardt B, Platzer C, Schoning B et al. Sympathetic activation triggers systemic interleukin-10 release in immunodepression induced by brain injury. Nat. Med. 1998; 4: 808–813. 96. Riese U, Brenner S, Docke WD, Prosch S, Reinke P, Oppert M et al. Catecholamines induce IL- 10 release in patients suffering from acute myocardial infarction by transactivating its promoter in monocytic but not in T cells. Mol Cell Biochem. 2000 Sep; 212 (1-2): 45-50. 97. Masood R, Zhang Y, Bond MW, Scadden DT, Moudgil T, Law RE et al. Interleukin-10 is an autocrine growth factor for acquired immunodeficiency syndrome-related B-cell lymphoma. Blood. 1995 jul 15; 85(12): 3423–30. 98. Mahipal A, Terai M, Berd D, Chervoneva I, Patel K, Mastrangelo MJ et al. Tumor-derived interleukin-10 as a prognostic factor in stage III patients undergoing adjuvant treatment with an autologous melanoma cell vaccine. Cancer Immunol Immunother. 2011 Jul; 60(7); 1039–45. 99. Di Marco R, Xiang M, Zaccone P, Leonardi C, Franco S, Meroni P, Nicoletti F. Concanavalin A induced hepatitis in mice is prevented by interleukin (IL)-10 and exacerbated by endogenous IL- 10 deficiency. Autoimmunity. 1999 Oct; 31(2): 75–83. 100. Park YB, Lee SK, Kim DS, Lee J, Lee CH, Song CH. Elevated interleukin-10 levels correlated with disease activity in systemic lupus erythematosus. Clin Exp Rheumatol. 1998 May- Jun; 16(3): 283–88. 101. Matsuda M, Salazar F, Petersson M, Masucci G, Hansson J, Pisa P et al. Interleukin 10 pretreatment protects target cells from tumor- and allo-specific cytotoxic T cells and downregulates HLA class I expression. J Exp Med. 1994 Dec 1; 180(6): 2371–2376. 102. Hagenbaugh A, Sharma S, Dubinett SM, Wei SH, Aranda R, Cheroutre H et al. Altered immune responses in interleukin 10 transgenic mice. J Exp Med; 1997 Jun 16; 185(12): 2101- 2110. 103. Tournoy KG, Kips JC, Pauwels RA. Endogenous interleukin-10 suppresses allergen-induced airway inflammation and nonspecific airway responsiveness. Clin Exp Allergy. 2000 jun; 30(6): 775–83. 104. Mocellin S, Wang E, Marincola FM. Cytokines and immune response in the tumor microenvironment. J Immunother. 2001 Sep-Oct; 24(5): 392-407. 105. Mocellin S, Marincola FM, Young HA. Interleukin-10 and the immune response against cancer: a counterpoint. J Leukoc Biol. 2005 Nov; 78(5):1043-51. 106. Gerard CM, Bruyns C, Delvaux A, Baudson N, Dargent JL, Goldman M et al. Loss of tumorigenicity and increased immunogenicity induced by interleukin-10 gene transfer in B16 melanoma cells. Hum Gene Ther. 1996 Jan; 7(1): 23-31. 107. Roncarolo MG, Bacchetta R, Bordignon C, Narula S, Levings MK. Type 1 T regulatory cells. Immunol Rev. 2001 Aug; 182: 68-79. 108. Levings MK, Sangregorio R, Roncarolo MG. Human cd25(+) cd4(+) t regulatory cells suppress naive and memory T cell proliferation and can be expanded in vitro without loss of function. J Exp Med. 2001 Jun 4; 193(11): 1295-302. 109. Roberts AB, Sporn MB, Assoian RK, Smith JM, Roche NS, Wakefield LM et al. Transforming growth factor type beta: rapid induction of fibrosis and angiogenesis in vivo and stimulation of collagen formation in vitro. Proc. Natl. Acad. Sci. USA. 1986 Jun; 83(12): 4167– 4171. 110. Tucker RF, Shipley GD, Moses HL, Holley RW. Growth inhibitor from BSC-1 cells closely related to platelet type β transforming growth factor. Science. 1984 Nov 9; 226(4675): 705–707. 111. Roberts AB, Anzano MA, Wakefield LM, Roche NS, Stern DF, Sporn MB. Type beta transforming growth factor: a bifunctional regulator of cellular growth. Proc Natl Acad Sci USA. 1985 Jan; 82(1): 119–123. 112. Giannelli G, Villa E and Lahn M. Transforming Growth Factor-b as a Therapeutic Target in Hepatocellular Carcinoma. Cancer Res. 2014 Apr 1; 74(7): 1890-4.
  • 21. 113. Blobe GC, Schiemann WP, Lodish HF. Role of transforming growth factor beta in human disease. N. Engl. J. Med. 2000 May 4; 342(18): 1350-8. 114. Blobe, Rebecca L. Elliott and Gerard C. Role of Transforming Growth Factor Beta in Human Cancer. J. Clin Oncol. 2005 Mar 20; 23(9): 2078-2093. 115. Korpal M, Kang Y. Targeting the transforming growth factor-beta signalling pathway in metastatic cancer. Eur J Cancer. 2010 Mar; 46(7): 1232-40. 116. Lampropoulos P, Zizi-Sermpetzoglou A, Rizos S, Kostakis A, Nikiteas N, Papavassiliou AG. TGF-beta signalling in colon carcinogenesis. Cancer Lett. 2012 Jan 1; 314(1): 1-7. 117. Won J, Kim H, Park EJ, Hong Y, Kim SJ, Yun Y. Tumorigenicity of mouse thymoma is suppressed by soluble Type II transforming growth factor b receptor therapy. Cancer Res. 1999 Mar 15; 59(6): 1273-1277. 118. Benigni A, Zoja C, Corna D, Zatelli C, Conti S, Campana M et al. Add-on anti-TGF-b antibody to ACE inhibitor arrests progressive diabetic nephropathy in the rat. J Am Soc Nephrol; 1999 Mar 15: 59(6): 1816-1824. 119. Mead AL, Wong TT, Cordeiro MF, Anderson IK, Khaw PT. Evaluation of anti-TGF-b2 antibody as a new postoperative anti-scarring agent in glaucoma surgery. Invest Ophthalmol Vis Sci. 2003 Aug; 44(8):3394-3401. 120. Nemunaitis J, Dillman RO, Schwarzenberger PO, Senzer N, Cunningham C, Cutler J et al. Phase II study of belagenpumatucel-L, a transforming growth factor beta-2 antisense gene- modified allogeneic tumor cell vaccine in non-small-cell lung cancer. J Clin Oncol. 2006 Oct 10; 24(29): 4721–4730. 121. Fakhrai H, Mantil JC, Liu L, Nicholson GL, Murphy-Satter CS, Ruppert J, Shawler DL. Phase I clinical trial of a TGF-beta antisense-modified tumor cell vaccine in patients with advanced glioma. Cancer Gene Ther. 2006 Dec ; 13(12): 1052–1060. 122. Gadir N, Jackson DN, Lee E, Foster DA. Defective TGF-beta signaling sensitizes human cancer cells to rapamycin. Oncogene. 2008 Feb 14; 27(8): 1055-62. 123. Candido J, Hagemann T. Cancer-related inflammation. J Clin Immunol. 2013 Jan;33(1):79- 84.