International Reviews of Immunology
ISSN: 0883-0185 (Print) 1563-5244 (Online) Journal homepage: https://www.tandfonline.com/loi/iiri20
Cancer immunotherapy: dawn of the death of
cancer?
Sidhant Jain & Sahil Kumar
To cite this article: Sidhant Jain & Sahil Kumar (2020): Cancer immunotherapy: dawn of the death
of cancer?, International Reviews of Immunology, DOI: 10.1080/08830185.2020.1775827
To link to this article: https://doi.org/10.1080/08830185.2020.1775827
Published online: 12 Jun 2020.
Submit your article to this journal
View related articles
View Crossmark data
Full Terms & Conditions of access and use can be found at
https://www.tandfonline.com/action/journalInformation?journalCode=iiri20
INTERNATIONAL REVIEWS OF IMMUNOLOGY
https://doi.org/10.1080/08830185.2020.1775827
REVIEW
Cancer immunotherapy: dawn of the death of cancer?
Sidhant Jaina
and Sahil Kumarb
a
Department of Zoology, University of Delhi, Delhi, India; bDepartment of Pharmacology, Maulana Azad Medical College and Lok
Nayak Hospital, New Delhi, India
ABSTRACT
ARTICLE HISTORY
Cancer is one of the proficient evaders of the immune system which claims millions of lives
every year. Developing therapeutics against cancer is extremely challenging as cancer involves
aberrations in self, most of which are not detected by the immune system. Conventional therapeutics like chemotherapy, radiotherapy are not only toxic but they significantly lower the
quality of life. Immunotherapy, which gained momentum in the 20th century, is emerging as
one of the alternatives to the conventional therapies and is relatively less harmful but more
costly. This review explores the modern advances in an array of such therapies and try to
compare them along with a limited analysis of concerns associated with them.
Received 29 December 2019
Accepted 16 May 2020
Introduction
Cancer is an extensive aberration in the cellular
behavior leading to continual unregulated proliferation of cells. Cancer generally arises from ‘gain-offunction’ mutation into proto-oncogenes converting
them to oncogenes or ‘loss-of-function’ mutation into
tumor-suppressor genes [1,2].
Normal growth promoting cellular genes are called
as proto-oncogenes and mutation in these genes can
alter their delimited function converting them into
cancer inducing cellular oncogenes. Certain viruses
like Rous Sarcoma Virus, Epstein-Barr Virus etc. contain oncogene derived from a cellular proto-oncogene
in their genome, termed as viral oncogene, which they
integrate in the cells they are infecting, inducing cancer [2; 3].
Worldwide, 8.2 million people die of cancer annually. Approximately, 14 million people suffer from it
every year, which is expected to increase to 22 million
in the next two decades [4]. Chemotherapy, hormone
therapy, radiotherapy and surgery are the conventional methods for treating cancer, however since late
20th century, immunotherapy and other targeted
therapies got approval for the treatment [5]. The
development of immunotherapy began as early as in
1890s which involved injecting bacteria or bacterial
products named as Coley’s toxins into inoperable
CONTACT: Sidhant Jain
110007, India
[email protected]
ß 2020 Taylor & Francis Group, LLC
KEYWORDS
ATCT; cancer; CAR T cell
therapy; conventional
therapy; immunotherapy;
monoclonal antibodies;
therapeutic vaccine
tumors [6; 3] but its development gained momentum
in late 20th century.
Immunotherapy involves a set of procedures which
help in elimination of a tumor by reviving, instigating
or enhancing the in-vivo anti-tumor immune response
or by neutralizing immune-inhibitory pathways like
immune checkpoints. To put simply, immunotherapy
directs the immune system to fight cancer [7; 3].
Immunotherapy encompasses an array of therapies
like Therapeutic Vaccines, Adoptive T Cell Therapy,
Monoclonal Antibodies, Inhibition of immune checkpoint etc. or a combination of these therapies
(Figure 1).
The aim of this review is multi-directional. First, it
consolidates the information available on the novel
methodologies in various fields of cancer immunotherapy, reflecting upon newer trials that are being
carried out. Second, it tries to broadly compare different immunotherapies in terms of their advantages and
disadvantages. Finally, this review, in brief, talks about
concerns related with these immunotherapies.
Earlier reviews have covered many similar aims as
this review tries to do, however, most of them
[4,8–10] concentrate on a specific aspect of cancer
immunotherapy. Other reviews (e.g. [6]) which did
cover information in various fields of immunotherapy
at one place are now somewhat old. New data from
multiple clinical trials as well as new approaches have
come up, which needs to be discussed. Instead of
Department of Zoology, University of Delhi, Gate No.:3, Chaatra Marg, North Campus, Delhi
2
S. JAIN AND S. KUMAR
Figure 1. Conventional therapies and immunotherapies for cancer treatment.
covering each therapy in detail, we try to cover the
important aspects of most of the therapies so that an
overall understanding of various immunotherapeutics
can be presented.
Cancer antigens
Majority of antigens associated with tumor cells are
self-antigens and hence are subject to tolerance inducing process which prevents auto-immunity. However,
many tumors express unique antigens where as others
inappropriately express self-antigens (e.g. overexpression of an oncogene product). Both of these types of
antigenic expression is recognized by immune system [3].
Peptide antigens can be either Tumor Associated
Antigens (TAAs) or Tumor Specific Antigens (TSAs).
TAAs are expressed on both normal and tumor cells
but they are much more in number in the later. TSAs
are specifically expressed by tumors [11]. TAAs can
have many sub categories as described in the Table 1.
Certain carbohydrates can also be TAAs. Most of
these carbohydrate antigens cannot elicit powerful
immune response as they are T cell independent.
Conjugating these carbohydrate antigens into proper
carriers can help eliciting an immune response and is
a breakthrough in carbohydrate antigen based therapeutic vaccines [13].
TSAs generally arise as a result of either genetic
mutations or viral infections (Table 2). These neoantigens are totally foreign proteins which are not
expressed on normal human cells [17]. Theoretically,
these antigens are perfect targets for developing therapeutics and have shown promising initial results.
Neo-antigen prediction became rapid with the use of
next-generation sequencing [14]. As normal cells do
not express these viral or mutated gene products,
these antigens are recognized as non-self by the
immune system.
Immunotherapeutic measures
Therapeutic vaccines
Cancer vaccination involves multiple approaches to
induce and/or amplify anti-tumor immunity by
INTERNATIONAL REVIEWS OF IMMUNOLOGY
3
Table 1. Different types of Tumor associated antigens.
Tumor Associated Antigen
Description
Examples
Differentiation Antigens
These are expressed on normal tissues and the
tumor which originate from these tissues [11].
These are expressed at a relatively small levels
in normal cells of a healthy tissue but are
amplified manifold at DNA, mRNA or/and
protein levels in tumor cells arising from that
tissue. [4]
In normal conditions, a restricted expression of
these antigens is reported in human germ
cells in testis and in trophoblast. However,
their expression is reported in a variety of
human cancers [4,11].
In spite of being expressed in normal germ line
cells and trophoblastic cells, cancer-testis
antigens can also be put under TSAs. As male
germ line cells and trophoblastics cells do not
have MHC class I molecules on their surface,
they cannot display antigens to T cells and
hence appear to be strictly tumor
specific [12].
These are normal fetal antigens which are
turned off during development but are
turned on again in some transformed
cells [3,4].
MelanA/MART-1, gp100/pmel17, CEA, gp75/TRP1, TRP-2
Her2/neu, WT1, Human mucin 1, RAGE-1,
antigens derived from PRAME gene.
Overexpressed Antigens
Cancer-Testis Antigens
Onco-fetal Antigens
NY-ESO-1/LAGE, Melanoma antigen- encoding
genes or MAGE (MAGE-A, MAGE-B, MAGE-C
family), BAGE, GAGE family of antigens
aFetoprotein (AFP)
Table 2. Different types of tumor specific antigens or neo-antigens.
Tumor Specific Antigen
Viral antigens
Description
These antigens are viral proteins and their
expression is extremely essential for the
maintenance of tumor malignancy.
Any epitope(s) derived from the open reading
frames in the viral genome becomes a
potential neo-antigen [11,14].
Mutant antigens
These antigens arise as a result of genetic
mutations hence encoding for a mutant
protein which is not expressed on the cells in
the normal conditions.
An array of genetic alterations leads to the
derivation of these mutant neo-antigens
which involve point mutations (leading to
single-nucleotide variants or causing a frameshift), gene fusions, insertions or deletions
and structural variations [12,14].
administrating tumor antigens with or without
immune modulators like Antigen Presenting Cells
(APCs) and adjuvants [4]. After going through the literature, we divided these approaches into five major
methodologies as shown in Table 3.
First methodology involves vaccination with peptide antigens. Antigenic peptides expressed by cancer
cells can be recognized by CD8þ TC cells (Cytotoxic
T lymphocytes or CTL) and CD4þ TH cells (Helper T
cells) in association with Class I MHC molecules and
Class II MHC molecules respectively [18].
Vaccines which are based upon TAAs, theoretically,
should not produce any immune response as TAAs
are self-antigens which are overexpressed in cancers.
Indeed, it was observed that vaccines against TAAs,
based on high affinity epitopes, do not produce an
Examples
Long peptides from E6 and E7 onco-proteins of
HPV-16 were used in a clinical trial for
vaccination of women who were HPV-16
positive with vulvar intraepithelial neoplasia
(grade 3).
A relief in symptoms was reported in about 3
months with adverse effects ranging up to
second grade [15]
Altered beta-catenin having an alteration in
single base pair in cDNA at 37th position and
hence coding for phenylalanine residue
instead of a serine residue [16]. Other
examples involve mutant p21/ras, mutant
EGFR VIII and mutant CDK4.
immune response as T cells to these high affinity selfepitopes must be deleted during T cell development as a
result of positive selection leading to tolerance [9]. The
solution to this problem lied in selecting low to medium
affinity peptide which can help in overcoming tolerance
[19]. Further, if antibodies are generated against such
low to medium affinity epitopes (which are overexpressed in cancer but otherwise are expressed in low
amounts by normal cells), an autoimmune response can
be expected, however, in transgenic mice models or clinical human trials, no such auto immune reactions have
been observed [19]. Two examples are here discussed to
explain the peptide antigen based vaccines.
Gp100 is a non-mutated differentiation antigen
expressed only by pigmented retinal cells and normal
melanocytes and by melanomas [20,21]. A study [22]
4
S. JAIN AND S. KUMAR
Table 3. Different methodologies for therapeutic vaccination.
Methodology
Antigenic peptides
Modified DCs
Tumor cell lysates
Viral Vectors
Naked DNA or RNA
Description
Vaccination with antigenic peptides expressed by tumor cells like gp100,
HER2/neu.
Vaccination with autologous DCs which are modified to express
tumor antigen.
Vaccination with irradiated tumor cells or tumor cell lysates along
with APCs.
Vaccination with tumor antigen DNA using viral vectors which uses
infection potential of virus.
DNA or RNA coding for tumor antigen, under a strong viral promoter, is
directly administered, usually intra-muscularly or intra-dermally.
showed that native peptides can generate melanoma
reactive CTLs in humans. However, their efficiency
can be increased by modifying these peptides and it
was shown that the modified peptides have more
potential to produce melanoma reactive CTLs. In
another study, gp100 was injected in subcutaneous tissue with interleukin-2 (IL-2) and it was found that
the rate of response and progression free survival was
longer when these two components were given
together [23].
Human Epidermal growth factor Receptor 2
(HER2), the 185-kDa protein receptor, is overexpressed in about 25% of all primary breast carcinomas. It acts as a non-mutated over-expressed antigen
[24] and it is overexpressed in other cancers involving
ovarian, colorectal, stomach and pancreatic carcinomas as well [25]. A study [26] showed that intradermal injection of vaccines based on Her-2/neu
peptides/protein mixed with granulocyte-macrophage
colony stimulating factor (GM-CSF) in patients overexpressing these antigens developed immunity to both
Her-2/neu peptides and proteins. Phase I/II clinical
trials were conducted for another Her-2/neu based
vaccine, E75 (Nelipepimut-S), which is also injected
with GM-CSF and produced safe and clinically efficient results and is under phase III trials [27].
Second methodology for cancer vaccination
involves vaccinating patients with modified APCs like
Peripheral Blood Mononuclear Cells (PBMCs),
Dendritic cells (DCs) and activated B-lymphocytes. It
was suggested that by using DCs, improved strategies
can be designed which can boost immunity against
metastatic cancers [28]. The immunological basis of
cancer vaccines which utilizes the knowledge of DC
system has also been described [29]. DC system based
vaccines involve harvesting of DCs from cancer
patients which are modified to express tumor antigen.
Like peptide based vaccines, DC system based vaccines are being injected with adjuvants, Interleukins
etc. to produce a heightened response in the patients.
Viruses have been used to transduce peptide antigens in autologous DCs. Initial gene targeting
techniques using viruses for DNA modification in
dendritic cells have been summarized in a review [30].
These techniques are being modified to generate more
effective vaccines. In a study, phase I/II trials were
conducted for a vaccine given to patients suffering
from metastatic melanoma. This vaccine contains
autologous DCs transduced with a replication lacking
adenovirus encoding for complete melanoma antigen
MART-1. The patients showed an immunogenic positive and safe response towards this vaccine [31].
In another study, the effect of vaccination with
autologous dendritic cells was studied in ten patients
who were suffering from Acute Myeloid Leukemia
(AML) by targeting Wilm’s Tumor 1 (WT-1) antigen.
These dendritic cells were loaded with full length
WT-1 mRNA via electroporation and injected intradermally in partial/complete remission state. These
patients showed improved clinical outcomes along
with enhanced immune activation [32].
Majority of the trials which were carried out in the
past decade, involved patients at advanced stages of
cancer. It had been hypothesized that DC cell vaccination can yield better and more promising results in
patients who are at early stages of cancer with less
tumor burden and before immunological impairment
occurs as a result of chemotherapy [33]. Some reviews
summarize the clinical trials based on DC system of
vaccination which are in progress [34,35].
Third methodology for cancer vaccination is based
upon using the killed tumor cells or tumor cell lysates
supplied along with APCs and/or co-stimulatory cytokines like GM-CSF. Studies were conducted using the
aforesaid methodology and a potent anti-tumor
immunity was seen in patients in phase 1 trials suffering from metastatic melanoma when irradiated
autologous melanoma cells, which were manipulated
to secrete GM-CSF, were injected in them [36]. In
another phase I/II study, vaccination of 28 patients
suffering from stage IV metastatic melanoma, with
similar irradiated autologous tumor cells which were
retro-virally transduced with GM-CSF gene showed
heightened T-cell activation and limited degree of
INTERNATIONAL REVIEWS OF IMMUNOLOGY
toxicity against melanocyte antigens but led to induction of vitiligo in a few patients [37].
Certain studies combined the use of APCs like dendritic cells with tumor cell lysates to produce new
combinations of vaccines. Promising results came
from another study involving 17 patients, many of
whom are suffering from active metastatic melanoma,
who were immunized intra-dermally with autologous
APCs which were cultured with GM-CSF and mixed
with tumor lysate 48 hours prior to immunization
proving feasibility of such vaccines two decades back
[38]. In another study, dendritic cells obtained from
PBMC were treated with tumor lysates and immunogenic protein Keyhole Limpet Hemocyanin (KLH) invitro along with GM-CSF and Interleukin-4 and given
intra-dermally for treatment of relapsed cancer in
children with solid tumors. It was observed that the
treatment was feasible and apparently nontoxic and
was able to regress tumor in a child with metastatic
R -L, which tarfibro-sarcoma [39]. A vaccine, DCVaxV
gets Glioblastoma Multiforme (GBM) contains mature
dendritic cells along with tumor lysate or peptides is
in stage III trials [40].
Fourth methodology is based upon the use of viral
vectors. Viruses have been used to transduce antigens
in autologous APCs ex-vivo in the case of modified
DC based vaccines discussed above. Further, irradiated
autologous tumor cells can be retro-virally transduced
with genes of various factors like GM-CSF, etc. to
produce heightened immune response against cancer
antigens as described above [37]. With respect to vaccination, viral vectors carrying antigen DNA can be
designed to directly inject tumor antigens into tissue
which can be easily delivered to tissue cells owing to
infective potential of virus.
In a study, two patients suffering from hepatocellular cancer having an AFPþ tumor, received vaccination with plasmid DNA injections. Plasmid construct
had full length AFP followed by replication deficient
but AFP expressing adenovirus and it was observed
that the vaccine was largely nontoxic and well tolerated with immunogenic nature. Larger clinical trial for
this methodology is proposed [41].
Currently, Adeno-associated virus (AAV) vectors
are being studied extensively for delivery of transgenes. AAV vectors are showing increased clinical
success. The current status, applications and future
prospects for AAV vectors has been strikingly summarized in a recent review [42]. Proposed and ongoing clinical trials based on DNA vaccines utilizing
viral vectors majorly have been clubbed in another
review [43].
5
Viruses can be utilized as an effective measure
against cancer in an altogether different way.
Extensive research is being done on oncolytic viruses
to develop them as an effective therapeutic measure
against cancer. Oncolytic viruses specifically infect and
propagate via tumor cells, hence damaging them and
this process is minimally toxic to the normal cells
[44,45]. Adenoviruses are being majorly researched to
develop them as oncolytic viruses. One strategy
involves mutating the E1 genes of the Adenovirus
whose gene products are responsible for preventing
the apoptosis of the host cell. This makes the virus,
tumor selective as products of E1 genes are essential
for propagation of virus [46]. Therefore, propagation
in normal cells is aborted and host cells undergo p53
mediated apoptosis. However, in majority of tumors,
p53 is itself nonfunctional and hence E1 mutated/
deleted viruses can still spread through them which
ultimately causes their lysis [47].
These oncolytic Adeno viruses have been modified
to express immune-stimulatory cytokines. In one
study, oncolytic Adenovirus was modified to encode
TNFa and IL-2 which induced danger- and pathogenassociated signaling (DAMP/PAMP) which activated
cytosolic protein sensors like AIM2 leading to modulation of tumor microenvironment [48]. Other studies,
used oncolytic Adenovirus or Helper dependent
Adenovirus (HDAd) expressing IL-12 and these treatments inhibited cancer progression. It was also
observed that for IL-12 delivery, HDAd are safer as
compared oncolytic Adenovirus [49,50].
Similar to the expression of cytokines, oncolytic
viruses have been engineered to express PD-L1 inhibitor and GM-CSF [51]. This PD-L1 inhibitor, inhibited
PD-L1 on immune cells as well as on tumor cells
which overcame the PD-L1 mediated immunosuppression and this led to provocation of systemic tumor
neoantigen-specific T cell response. This therapy can
be extremely useful to patients resistant to PD-1/PDL1 immune checkpoint blockade (discussed ahead).
An array of clinical trials are in progress with respect
to oncolytic viruses but it would be beyond the scope
of this work to collate all of them. A recent review
can be referred [52] which summarizes many of these
clinical trials.
An interesting candidate for virus based therapies
is Zika virus. Although it causes microcephaly by killing brain cells but studies have shown its oncolytic
potential against aggressive glioblastoma stem cells
providing us with a possible brain tumor therapy [53,54].
6
S. JAIN AND S. KUMAR
Another strategy for cancer vaccination is based on
physically delivering DNA or RNA. Naked DNA vaccines for immunotherapy are made using bacterial
plasmid coding for a targeted tumor antigen under a
strong viral promoter for continuous and maximal
expression and are usually administered intra-muscularly or intra-dermally. Naked DNA vaccines are usually injected with adjuvants like polysaccharides,
liposomes, aluminum salts, nano-particles etc. which
act as immune stimulators and/or delivery system
[55,56]. Intra-muscular electroporation is also used to
deliver plasmid DNA [57]. Plasmid DNA vaccines are
nearly nontoxic and rarely integrates into host genome. The chances of insertional mutagenesis are even
less than that of spontaneous mutation [56].
In a study, plasmid DNA vaccine was made which
encoded for truncated Carcino-embryonic Antigen
(CEA) overexpressed in many cancers like colorectal,
breast, lung, ovarian cancer etc. and was fused with
T-helper epitope DNA. 10 patients suffering from
colorectal cancer were immunized intra-dermally with
this vaccine three times via jet injection. Prior to first
vaccination, cyclophosphamide was injected intravenously whereas GM-CSF was administered subcutaneously with every vaccination. Except for grade I and II
adverse effects (AEs), the vaccine showed positive
results with minimal toxicity and no signs of autoimmunity [58].
The use of mRNAs/siRNAs is also increasing in
development of therapeutics. Conventionally, RNA
vaccines are made by transcription of a template
DNA, which encodes for a particular peptide antigen
that has to be targeted, into mRNA using RNA polymerase derived from bacteriophage in-vitro. After
administration and internalization by host cells, translation directly takes place in host cells’ cytoplasm and
the translated peptide antigens are presented to APCs
and an immune response is hence generated. On the
other hand, autologous DCs can be transfected with
TAAs’ mRNA and delivered to respective patient to
generate an immune response [55,59]. In a study, DCs
were harvested from patients suffering from metastatic
prostate cancer and transfected with mRNA encoding
for Prostate Specific Antigen (PSA). PSA specific Tcell responses were recorded in these patients confirming the bioactivity of this vaccine [60]. On-going
clinical trials based on such vaccines has been summarized in a recent review [61].
In-spite of decades of research in cancer vaccination, only two cancer vaccines got US Food and
Drug Administration (FDA) approval till May, 2018.
One of them is Sipuleucel-T used for treating Prostate
cancer [62]. Sipuleucel-T uses a recombinant fusion
protein, PAP-GM-CSF (PA2024), to activate PBMCs.
PAP-GM-CSF is made up of Prostatic Acid
Phosphatase (PAP), an antigen expressed in prostate
cancer which is joined to the N-terminus of GM-CSF
via its C-terminus. Each dose of this vaccine has
about 50 million activated CD54þ cells [63]. Second
vaccine to be approved by FDA and first one to be
based on oncolytic virus is talimogene laherparepvec,
also known as T-VEC, or OncoVEXGM-CSF for use in
melanoma patients [64].
Adoptive T cell therapy
Adoptive T cell therapy (ATCT) involves infusion of
autologous tumor specific CTLs derived from cancer
patients for the recognition and destruction of the
cancerous cells. These T cells can be harvested and
expanded from the tumors themselves (Tumor
Infiltrating Lymphocytes or TILs) or can be harvested
from peripheral blood and tumor specificity can be
induced either via antigen specific expansion or genetic engineering [9].
The first trial with ATCT was done in a 1988
study, when TILs, extracted from melanomas were
given to 20 patients along with IL-2 after an intravenous dose of cyclophosphamide. Regression was
observed in 11 out of 20 patients which lasted for
approximately 2–13 months along with reversible toxic
effects of IL-2 [65]. Many modifications and advancements have been introduced since this trial including
treatment with T cells after depleting the immune elements which increases anti-tumor efficacy [66].
Promising results have been obtained from multiple
studies which have been done utilizing TILs for treatment, where in some of them, the other immunotherapies have failed [67,68]. A recent review
highlights the opportunities which can be harnessed
in the field of ATCT along with the challenges being
faced and possible solution to those challenges [69].
The evolution of Chimeric Antigen Receptor T cells
(CAR T cells) can be thought as most promising
development in ATCT. This therapy uses high numbers of tumor targeting T cells, produced by genetic
manipulation of autologous T cells using plasmid or
retroviral vectors which aims at introducing tumor
specific chimeric T cell receptors (TCRs) [10]. TCRs
require peptide antigen presentation derived from
proteins in association with MHC whereas CARs
exhibit MHC independent recognition of these antigens which, theoretically, can be any surface antigen
INTERNATIONAL REVIEWS OF IMMUNOLOGY
7
Figure 2. Production of CAR T cells and Generations of CARs.
i.e. a peptide, protein or phospholipid [70; reviewed
in 10].
Manufacturing CAR T cells involves a patient or a
T cell donor, from whose blood T cells are harvested
via leukapheresis. T cells are activated in presence of
magnetic beads covered with CD3 and CD28 antibodies
followed by genetic engineering (via viral transduction
to express CAR) and T cell expansion. This is followed
by bead removal and adoptive transfer or freezing for
storage (Figure 2). A conditional chemotherapy is preferred before the adoptive transfer [71,72].
The recognition of an antigen by TCRs and T cell
activation is an intricate process. It involves an antigen presenting cell/target cell to present an antigen
with MHC (class II/I) molecule to CD4þ/CD8þ T cell.
The TCR, itself, is a disulfide heterodimer made up of
two chains, a and b, with some CD3 invariant chains
(f, e, d, and c). Co-stimulation of T cell by interactions between CD 80/86 on APC/target cell and CD
28 on T cell is also required for its activation.
Adhesion Molecules’ interactions like LFA-1 to
ICAM-1 and LFA-3 to CD2 further strengthens interaction between the cells [3].
CARs are recombinant receptors made up with an
extracellular single-chain variable fragment (scFv)
obtained from an antibody linked to a trans-membrane domain, which is again joined to intracellular
T-cell signaling domain(s) [73]. A ground breaking
study led to the development of first generation CARs
where cytoplasmic domain of TCR signaling complex
is fused to variable fragment of tumor targeting antibody [74]. A first generation CAR involves fusion of
CD3f or Fc receptor c chain, which are cytoplasmic
signaling domains, to a scFv of a tumor targeting antibody via multiple hinge and trans-membrane
domains [73].
After the development of CAR T cell therapy, some
groups started targeting ovarian cancer ex-vivo and
in-vivo using monoclonal antibodies against ovarian
cancer peptides to generate CARs [75,76]. A trial
study was then conducted which involved generation
of autologous T cells against a folate receptor (FR)
antigen associated with ovarian cancer. The CARs
(first generation) were expressed by a chimeric gene
which incorporated anti-FR single chain antibody
linked to c chain of TCR/CD3 signaling complex. The
8
S. JAIN AND S. KUMAR
patients were segregated in two groups where patients
in first group received autologous T cells against FR
along with high dose IL-2 and patients in second
group received dual-specific T cells (against FR and
allogenic cells) followed by injecting with allogenic
PBMC. Though the treatment was safe, except for the
IL-2 associated toxicity, but didn’t reduce tumor burden and it was observed that such tumor reactive cells
perished soon [77].
The first generation CAR T cells undergo limited
proliferation because of lack of co-stimulatory signals
leading to anergy or activation induced cell death
[73]. To overcome this issue, second generation CARs
were developed (Figure 2) by incorporating signaling
domains of T cell co-stimulatory receptors like CD28,
OX-40 or 4-1BB because of which the signaling
strength enhanced and anti-tumor activity was manifold increased [78,79]. A further improvement to these
second generation CAR T cells, involving cytoplasmic
domains from two co-stimulatory receptors like
CD28-4-1BB or CD28-OX-40, led to production of its
third generation [73]. The fourth generation has also
been developed, termed as TRUCKs (T Cell
Redirected Universal Cytokine Killing) which, along
with the primary signal of TCR/CD3 and a second
signal from the co-stimulatory signaling domain like
CD28, have a CAR inducible transgene which usually
codes for a cytokine [80].
CAR T cell therapy has shown positive results in
anti-leukemic regime in mice models and human trials [79,81]. FDA in 2017 has approved two CAR T
cell therapies. First, Kymriah or Tisagenlecleucel, is
approved for the treatment of certain children and
young adults suffering from Acute Lymphoblastic
Leukemia. Second, Yescarta or axicabtagene ciloleucel,
is approved for the treatment of adults suffering from
large B-cell Lymphoma.
After showing its potential in hematologic malignancies, currently, CAR T cell therapy is also being
researched to analyze if it can be used to treat solid
tumors. The barriers imposed by the solid tumors
against this therapy like penetrating the stroma of
tumor, persistence and expansion within the tumor
etc. and the possible strategies to overcome such
obstacles have been abridged in a recent review [82].
Antibody based therapy
Over the last two decades, antibody based cancer therapy has established itself as one of the most effective
strategies for treating humans suffering from haematological malignancies as well as solid tumors [8].
The revolutionary process of generation of monoclonal antibodies (mABs) was documented in 1975
and involves the use of cancerous cells to produce
hybridomas [83], the very same technology is now
being used to generate mABs to target cancerous cells.
Although most mABs used in these therapies are
immunoglobulin G (IgG) but various modifications
like protein-Fc conjugates and drug conjugates are
also used [8]. mABs target cancer cells by binding to
specific antigens expressed on cell surface like CEA,
Vascular Endothelial Growth Factor (VEGF), HER2,
Cluster of Differentiation (e.g. CD20, CD33), VEGF
Receptor etc [84].
After attachment to a particular tumor antigen
there are multiple pathways by which a cancer cell
can be destroyed:
Direct tumor cell death.
Immune mediated tumor cell killing.
Vascular ablation.
Direct cell killings involve three different mechanisms: Firstly, there can be delivery of cytotoxic
drugs/radioisotopes using conjugate antibodies leading
to cell lysis [85]; secondly, apoptosis can be induced
[86–88]; lastly, signaling pathways necessary for cell
survival can be targeted and inhibited [87].
Immune mediated cell killing incorporates measures like activation of cellular phagocytosis, engagement of Antibody Dependent Cell-mediated
Cytotoxicity (ADCC) or complement mediated cell
lysis [89–92]. Further, mABs’ SCFvs can be used in
generation of CARs [75,76,93]. Finally, few mABs can
inhibit the inhibitory receptors on T lymphocytes
leading to activation of T lymphocytes [94, reviewed
in 95] and have been discussed in next section.
Angiogenesis is a necessary component for growth
and metastasis of cancer. Vascular ablation restricts
blood supply to the tumor decreasing tumor perfusion
[96]. The total number of mABs approved US FDA
for different types of cancers reached 64 in 2018 [97]
indicating its importance as a milestone therapy in
cancer therapeutics.
However, in spite of the promising results one can
achieve with these monoclonal antibodies, there exists
a major lacunae as resistance can be generated among
cancer cells to such targeted therapy due to down
regulation of pathways which are being targeted and
inhibited by mABs and up regulation of the alternative pathways. For example, Cetuximab, a mAB
against Epidermal Growth Factor Receptor (EGFR) is
an effective strategy to treat patients with colorectal
INTERNATIONAL REVIEWS OF IMMUNOLOGY
cancer but resistance to the same has been reported
due to uncharacteristic ERBB2 signaling (ERBB2 gene
amplification or hergulin production) leading to
resistance [98]. Resistance in HER-2 targeted therapy [99,100] has also been reported. To limit this
issue of resistance and increase the efficacy of targeted therapy, bi-specific antibodies (bsABs) have
been developed.
A bsAB can simultaneously bind to two antigen
like a tumor antigen and effector cell or drug [101].
bsABs can help in multiple ways:
More than one tumor cell antigen can be simultaneously engaged due to presence of different antigen
binding sites.
Non-conventional immune cells like T cells, which
are not activated by mABs due to lack of FC receptor, can be activated [102].
bsABs can be directed against receptor tyrosine
kinase (can be used in HER-2 targeted therapy),
tumor angiogenesis and even intracellular antigens [103].
Further, all the targeting methods discussed above
like delivery of cytotoxic drugs, activation of apoptosis etc. can be used in combination to target cancerous cells using bsABs [104].
Given the overabundance of combinatorial options,
bsABs can be used in multiple combinations to counter this lacunae of resistance.
Inhibition of immune checkpoint inhibitors
Immune response inhibitory interactions are common
in natural conditions like expression of CTLA-4 on T
cell after T cell activation, which like CD28 binds to
B7 (CD80/CD86) expressed on an APC but binds
with higher affinity. This interaction, instead of providing a co-stimulatory signal, provides an inhibitory
signal. CTLA-4 limits CD28-B7 interaction to reduce
hyper T cell activation which can lead to splenomegaly, lymphadenopathy, autoimmunity and even death
but it helps the unchecked growth of tumors as
CTLA-4 is constitutively expressed on cancer
cells [105,106].
Another such molecule is PD-1 which is present on
the surface of T cells. It binds to PD-L1 or PD-L2
expressed on professional APCs and this PD-1 to PDL1/PD-L2 interaction down regulate the T cell
response [107,108]. PD-L1 is overly expressed on cancer cells [108] and this combined with overexpression
of PD-1 on TILs can lead to poor surveillance by
9
immune system helping in unchecked growth of
tumor cells [109].
Clinical trials using mABs which block PD-1/PDL1 interaction, by binding to any one of them, gave
excellent responses especially for melanoma, bladder
cancer and renal cell carcinoma [110]. Pembrolizumab
(Keytruda) and Nivolumab (Opdivo) are two FDA
approved mABs for certain types of cancers which
block PD-1. Phase II/III trials are undergoing to check
the efficacy of certain mABs blocking PD-L1, which
have shown significant results in pre-clinical and
Phase-I trials [111]. Similarly, Ipilimumab (Yervoy) is
a mAB that binds to CTLA-4 and blocks its activity
[112]. In the absence of these inhibitory signals, an
array of T cells get activated and mount an immune
response against the tumor cells.
These immune checkpoint receptors are essential to
keep in check the body’s immune system to prevent it
from causing damage and harm but in the presence of
active malignancies, inhibitory signals may be dominant and needs to be curbed to generate effective
immunity against tumor cells [112]. However, as these
mABs are blocking all these inhibitory molecules, they
can pave way for a serious and even life threatening
side-effects in some people. Common side effects of
these drugs can include nausea, fatigue, itching, skin
rash, dirrahea and less often one can have serious
problems in the lungs, intestines, liver, kidneys and
endocrine glands [113].
Many other molecules are under study which are
co-inhibitory molecules which can be inhibited to
mount an effective immune response against tumor
cells. Lah-3, CD40, CD137 have already reached Phase
I or II trials whereas B7x, BTLA, and Tim-3 are under
pre-clinical study [114] clearly indicating that this
therapy is still in the very initial stages and many
more therapeutics, developed on similar lines, are
expected in near future.
Combinatorial therapies
Some cancer patients may respond to a single therapy
but for a majority of them, it is ineffective [115]. It is
a necessity to combine various cancer therapies to
achieve complete remission [116]. In this review, we
describe some of these combinatorial therapies.
Checkpoint inhibitors with conventional therapies
Linking antibodies against checkpoint inhibitors
with conventional chemotherapy may take an advantage of the fall in tumor burden caused by this
10
S. JAIN AND S. KUMAR
Table 4. Comparison of different immunotherapies.
Immunotherapy (Examples)
Vaccines (Sipuleucel-T,
OncoVEXGM-CSF)
Advantages
ATCT and CAR T cell therapy
(Kymriah, Yescarta)
mABs (Rituximab, Cetuximab)
Immune checkpoint blockade
(Pembrolizumab,
Nivolumab, Ipilimumab)
different
References/ Reviewed into
Lack of universal antigens
Relatively poor efficacy
Not Cost effective.
Expensive
Relatively more time consuming
Certain CAR T regimes are
detrimentally toxic
Neurotoxicity with CAR
T regimes
Approved CAR T cell therapies
are extremely costly
Potent anti-tumor activity
Prolonged survival
Can deal with broad range
of cancers
treatment curbing cancer. A chemotherapeutic agent,
Decarbazine was given with ipilumumab (anti-CTLA4) to patients suffering from metastatic melanoma
which was not treated previously. The survival rate
with this regime, relative to mono-treatment with
decarbazine plus placebo, was longer though the grade
3 and 4 AEs were higher [117].
In a phase III trial involving males with ‘minimum
one bone metastasis’ from castration-resistant prostate
cancer which had progressed after docetaxel treatment, 8 Gy radiotherapy, directed to bone, was provided followed by ipilimumab or placebo. Though, the
survival rates in both the groups were not much
different, certain amount of anti-tumor activity was
observed in the radiation treated group warranting
further research [118].
Combination of
point inhibitors
Disadvantages
Specificity
Engagement of host
immune system
Relatively low toxicity
Potential for durable treatment
effect owing to memory.
Specificity
High response rate
Efficacy can be increased using
lymphodepletion.
CARs exhibit MHC independent
tumor antigen identification
CARs can theoretically identify
any surface antigen
(phospholipid peptide, protein)
Specificity
Relatively low toxicity
Engagement of host
immune system.
immune
check-
The inhibition of both the checkpoint inhibitors i.e.
CTLA-4 or PD-1 leads to prevention of a heightened
T cell response but both of them use different signaling pathway to reach the outcome. CTLA-4 diminishes the initial T-cell activation whereas PD-1
obstructs effector T-cell responses in tissues [119].
Due to this, synergy is expected from a rational combination of these two antibodies. Combination of PD1 and CTLA-4 blocking (in combination with Fvax) is
more than two times as effective as anyone alone in
supporting the elimination of B16 melanomas in
mice [120].
Development of resistance.
mAB specific Hepatitis B virus
reactivation
Development of severe
infections in some cases.
Relatively few patients obtain
clinical benefit.
Serious side effects in vital
organs in a few patients.
[126–128]
[70]
[129]
[130]
[131]
[132]
[98,100,133–135]
[113]
[116]
[115]
In humans, combination therapy blocking CTLA4
and PD1 causes distinct immunologic changes as
compared to changes that occur by blocking one of
them [121]. In a phase I trial, concurrent treatment,
with nivolumab (anti PD-1) and ipilimumab (antiCTLA-4), of patients suffering from melanoma
achieved higher response rate than any of them alone
along with manageable safety profile [122]. The frequency, magnitude and onset of immune related toxicities, however, also increased in this combinatorial
approach relative to treatment with a single checkpoint blocking agent [123].
Therapeutic vaccines
point inhibitors
with
immune
check-
In a study involving mice, treatment with GVAX
(GM-CSF-expressing irradiated tumor cells) along
with CD-1 and CTLA-4 blockage induced significant
rejection of tumors and in one case 100% mice
rejected tumor [124].
In a Phase III study, patients with metastatic melanoma, were given one of the three treatments i.e.
either gp100 peptide vaccine alone, ipilimumab
alone, or both gp100 vaccine and ipilimumab to
study the overall survival. The median overall survival with gp100 peptide vaccine alone was
6.4 months and the same in combination with both
was 10 months, showing a high overall survival
[125]. Interestingly, in the same study, treatment
with ipilimumab alone gave a median overall survival of 10.1 months. Further research is required in
INTERNATIONAL REVIEWS OF IMMUNOLOGY
this combinatorial therapy to access multiple other
factors and their efficacy.
Immunotherapies at a glance
All the therapeutics described in this review have been
broadly compared in Table 4.
Concerns with immunotherapy
Cost
Cancer immunotherapies are relatively less toxic and
generate efficient responses but the primary concern
lies with their cost. Majority of these therapies are
extremely expensive [136] and hence are out of the
reach of poor individuals or the developing nations.
Taking into consideration CAR T cell therapies,
Tisagenlecleucel has a price tag of $4,75,000 whereas
Axicabtagene ciloleucel costs around $3,73,000 [132].
Owing to such high prices, CAR T cell therapy stays
out the reach of poor people. Similarly, immune
checkpoint inhibitors have shown promising results
but many of them are not economical. A study found
that use of nivolumab for metastatic head/neck cancers was not cost-effective over chemotherapy and use
of nivolumab and pembrolizumab for renal cell cancer
or bladder cancer didn’t show adequate cost-effectiveness [137]. However, the same study found few other
inhibitors economically reasonable for other types of
cancer and have noted that the patient selection criteria is an important parameter for determination of
cost-effectiveness which can alter their conclusions [137].
The first FDA approved therapeutic vaccine,
Sipuleucel-T, used for prostate cancer, is relatively
minimally toxic but costs around $93,000–$100,000
for a full therapy course given for one month
[138,139]. Abiraterone, an anti-androgen medication,
is a conventional alternative to Sipuleucel-T and it
costs almost the same though, it may cost more as the
cost depends upon the months of treatment received.
However, a study showed that with Abiraterone, the
gains in number of total life years and quality adjusted
life years are somewhat higher [140]. Hence, a conflict
between the cost, efficiency and toxicity of treatment
definitely exists.
However, with the emergence of biosimilars, the
cost is expected to come down for various immunotheraputics. Biosimilars have a comparable biological
activity to their respective reference drugs but are generally cost-effective and have the ability to increase
treatment accessibility, especially to the poor. Further,
11
they bring competition in the market making the original drug developers to bring down the cost. With
the expiry due for the patents of various immunotheraputics in coming years, biosimilars are expected to
emerge which can potentially address the cost issue.
One such assessment of the impact of biosimilars on
the cost and their safety and efficacy has been done in
a recent review [141].
Autoimmunity and infections
Vaccines, ATCT and mABs also give rise to autoimmunity against normal tissues. It can arise as a
result of therapies acting against TAAs expressed on
normal tissues, expansion of self-reactive T cell due to
inhibition of CTLA-4 or by other less understood
mechanisms [142].
mABs have been reported to increase the risk of
severe infections as well as reactivation of hepatitis B
virus (HBV). In particular, mABs for anti-epidermal
growth factor receptor, used in many malignancies,
has been reported to significantly increase the chance
of developing non-fatal but severe infections [133].
Rituximab, which is an anti-CD20 mAB, is used for
the treatment of CD20þ B-cell non-Hodgkin
Lymphoma. There exists a well-defined association
between Rituximab treatment and reactivation of
HBV and hence it is critical to precisely identify
patients at risk and therefore require prophylactic
antiviral regimes [134].
Side effects
The frequent AEs linked with Sipuleucel-T involved
pyrexia, malaise, chills, fatigue, nausea and culture
positives [143]. Severe Cytokine release syndrome
(sCRS) and neurological disorders are major toxicities
associated with CAR T cell therapy [81]. The manifestations of sCRS involve hypoxia, end organ dysfunction and hemophagocytic lymphohistiocytosis among
others whereas neurological disorders involve cognitive defects, encephalopathy, seizures and cerebral
edema [144].
Immune checkpoint blockade involves effects like
fatigue, hepatitis, vitiligo, colitis, diarrhea, hypothyroidism, myasthenia gravis [113,145]. Patients administered with PD-1/PD-L1 inhibitors reported 30–40%
of dermatological toxicities of all grades which was
50% with anti-CTLA-4 inhibitor, ipilimumab. Further,
organ specific inflammation like pneumonitis, hepatitis, autoimmune encephalitis, inflammatory arthritis,
hypophysitis, lupus nephritis and posterior reversible
12
S. JAIN AND S. KUMAR
encephalopathy syndrome, sarcoidosis, anemia are
some of the many other AEs which have been
observed with immune checkpoint inhibitors [146].
Combining mABs against immune checkpoints further
increases these toxicities [123].
Discussion
Immunotherapy is emerging as one of the leading
therapeutics to deal with certain types of cancer.
Although conventional therapies like surgery, chemotherapy and radiotherapy remain the most widely
used mechanisms and generally are the first line of
treatment in many cancers, however, immunotherapy
is slowly gaining speed and is becoming relevant for
first treatment line in certain cancers like non-small
cell lung cancer (NSCLC) [147]. Other than that,
immunotherapies are used as adjuvant therapies in
the line of cancer management in certain cases like in
advanced melanoma [148] or when first line of treatment fails. Most immunotherapeutics are highly specific, relatively less toxic and offer better survival
compared to conventional therapies. In our view,
every immunotherapy offers different niche of advantages but also has certain challenges, most common
among them being their high cost.
In the development process of therapeutic vaccination, new antigens are being tested regularly for a
potential vaccination candidate. Multiple ways have
been developed for therapeutic vaccination involving
infusion of antigens, irradiated tumor cells, mutated
cells which express specific antigen, or even the antigen DNA with different types of vector systems.
Relative to other immunotherapies, vaccines are less
toxic but also are less efficient. The development of
oncolytic viruses can be considered as a major breakthrough in therapeutic vaccination for their very ability to selectively infect and propagate via tumor cells
and causing minimal harm to the normal cells.
However, lack of universal tumor antigens hinder this
development. In spite of these obstacles, vaccines are
being developed targeting both TAAs and TSAs.
ATCT and specifically CAR T cell therapy are
evolving at a rapid rate. Since its inception in 1988,
ATCT has demonstrated positive results all along. The
rapid evolution of CAR T cell therapy can only be
judged from a simple fact that there now exists four
generations of CAR T cell. CAR T cell therapy is
highly efficient, specific and has a high response rate.
However, research and development in CAR T cell
therapy regime seems to benefit only a few as of now
owing to its extremely high cost of administration.
Further, toxicity related to CAR T cell therapy also
possesses great challenges. sCRS, measured in terms of
cytokine maximum fold change, fever, hypoxia, neurological disorders etc., is related with CAR T cell treatment. This sCRS requires immediate medical
intervention and steroid treatment in the conventional
strategy [81]. Davila and his group studied efficacy of
using tocilizumab, a monoclonal antibody blocking
IL-6 receptor as a measure to control sCRS against
steroids which lead to five-fold decline in CAR T cells
and got positive results [81]. More of such novel alternatives are required while dealing with such
severe toxicities.
Monoclonal antibodies are one of the most rapidly
rising regime in the field of immunotherapy. Owing
to the fact that it is extremely specific and relatively
less toxic, it can soon become most widely accepted
treatment regime in near future. The issue of development of resistance toward these mABs can be managed by using bi-specific antibodies which can target
more than one cancer antigen or signaling pathway at
the same time. Along with targeting cancer antigen,
same bsABs can also be used for cytotoxic drug delivery or apoptosis activation and multiple such combinations can be made. However, more research is
required to address of the risk factors which lead to
development of severe infections and accurate screening to prevent HBV reactivation.
Immune check point blocking is one of the most
efficient techniques in terms of its potency and ability
to deal with a range of cancers. However, this therapy
faces a serious drawback of life threatening side effects
which, at present, is hindering its development as a
major immunotherapeutic.
Combinatorial therapies can culminate the advantages of two therapies and can generate a heightened
response against cancer. However, recent trials which
involved combination of ipilumumab with therapeutic
vaccine or radiotherapy, didn’t generate promising
results. Further, the combination of two immune
check point blockers might have given positive results
due to the expected synergy but the issue of increased
toxicity has to be answered. Hence these combinatorial therapies call for extensive research to develop a
combinatorial therapy which can cater to the needs, is
cost effective and least toxic.
An important aspect with immunotherapies is
choosing the appropriate therapy for a particular type
of cancer. Biomarkers can provide insights about genetic makeup and immune system interactions of a
particular type of cancer in individual patient. They
are also used to monitor the response of a patient
INTERNATIONAL REVIEWS OF IMMUNOLOGY
toward a particular treatment regime. Serum proteins,
tumor micro-environment factors, circulating tumor
cells, immune cells are some of the important biomarkers among many others [149]. A recent clinical
trial involving patients suffering from gastrointestinal
stromal tumor showed that circulating tumor DNA
(ctDNA) indicated disease activity in these patients
presenting its potential as prospective biomarker for
future [150]. Similarly, a study predicted plasmatic
biomarkers in patients suffering from NSCLC who
were being treated with nivolumab [151]. These biomarkers are a rapidly developing focus in the field of
immunotherapy which holds the key to more efficient
and safe treatments.
A special mention is required here for those chemotherapeutics which boost the natural immunity
against cancer forming the very basis of immunotherapy. Myeloid Derived Suppressor Cells (MDSCs) can
mount up in tumors during its growth and can help
in cancer immune tolerance by inhibiting function of
CD8þ T cells. Experiments in mice and cell lines
showed that Gemcitabine and 5-Flurouracil are selectively cytotoxic to MDSCs allowing effector T cells to
function more efficiently [152]. Regulatory T cells
(Tregs), suppress high avidity tumor-specific T cells.
Cyclophosphamide can be used to supress Tregs for
effective anti-tumor immune response [153]. Hence,
depletion of the immunosuppressive activities of Tregs
and MDSCs should lead to an effective induction of
anti-tumor T cell response [115].
The future prospects of immunotherapy seem very
bright. Given their specificity and efficiency, these can
be widely used in future to deal with a range of cancers. However, along with the concerns like cost and
toxicity, other parameters like heterogeneity in tumors
as well as in cancer patients have to be taken in consideration. As cancer is unstable genetically, next generation sequencing and other bioinformatics tools
are of utmost importance in the identification of neoantigens with high immunogenic potential as well as
for the development of safer and efficient therapies
in future. Similarly, oncolytic viruses hold a great
potential in differential killing of cancer cells.
Combinatorial treatments have shown a high level of
synergy, especially by using two immune check point
inhibitors. If future research can take care of the
added toxicities in these combinatorial therapies, a
major outbreak in the cancer treatment can be
reached. Personalized combinatorial therapies can be a
major attraction in this field which can address the
issue of heterogeneity to a great extent.
13
In spite of discussing all the positive outcomes
from multiple areas of immunotherapeutics, it is still
difficult to say if these therapeutics can actually bring
the dawn of the death of cancer.
Disclosure statement
No potential
the author(s).
conflict
of
interest
was
reported
by
ORCID
Sidhant Jain
Sahil Kumar
http://orcid.org/0000-0003-2596-9566
http://orcid.org/0000-0003-0834-9330
References
1.
2.
3.
4.
5.
6.
7.
8.
9.
10.
11.
12.
13.
14.
Cooper GM, Hausman RE. The Cell: A Molecular
Approach. 4th ed. USA: Sinauer Associates; 2007.
Lodish H, Berk A, Zipursky SL, Matsudira P,
Baltimore D, Darnell JE. Molecular Cell Biology. 5th
ed. W. H. Freeman. USA. 2003: 973.
Owen JA, Punt J, Stranford SA. Kuby Immunology.
7th ed. Susan Winslow. New York, USA. 2013: 692.
Butterfield LH. Cancer vaccines. BMJ. 2015;350:h988
doi:10.1136/bmj.h988.
Sudhakar A. History of cancer, ancient and modern
treatment methods. J Cancer Sci Ther. 2009;1(2):1–4.
doi:10.4172/1948-5956.100000e2.
Kirkwood JM, Butterfield LH, Tarhini AA, Zarour
H, Kalinski P, Ferrone S. Immunotherapy of cancer
in 2012. CA Cancer J Clin. 2012;62(5):309–335. doi:
10.3322/caac.20132.
Couzin-Frankel J. Breakthrough of the year 2013.
Cancer immunotherapy. Science. 2013;342(6165):
1432–1433. doi:10.1126/science.342.6165.1432.
Scott AM, Wolchok JD, Old LJ. Antibody therapy of
cancer. Nat Rev Cancer. 2012;12(4):278–287. doi:10.
1038/nrc3236.
Perica K, Varela JC, Oelke M, Schneck J. Adoptive T
cell
immunotherapy
for
cancer.
Rambam
Maimonides Med J. 2015;6(1):e0004 doi:10.5041/
RMMJ.10179.
Almåsbak H, Aarvak T, Vemuri MC. CAR T cell
therapy: a game changer in cancer treatment. J
Immunol Res. 2016;2016:5474602 doi:10.1155/2016/
5474602.
Yang J, Zhang Q, Li K, Yin H, Zheng JN. Composite
peptide-based vaccines for cancer immunotherapy
(Review). Int J Mol Med. 2015;35(1):17–23. doi:10.
3892/ijmm.2014.2000.
Vigneron N. Human tumor antigens and cancer
immunotherapy. Biomed Res Int. 2015;2015:948501
doi:10.1155/2015/948501.
Liu CC, Ye XS. Carbohydrate-based cancer vaccines:
target cancer with sugar bullets. Glycoconj J. 2012;
29(5–6):259–271. doi:10.1007/s10719-012-9399-9.
Jiang T, Shi T, Zhang H, et al. Tumor neoantigens:
from basic research to clinical applications. J
14
15.
16.
17.
18.
19.
20.
21.
22.
23.
24.
25.
26.
27.
S. JAIN AND S. KUMAR
Hematol Oncol. 2019;12(1):93 doi:10.1186/s13045019-0787-5.
Kenter GG, Welters MJ, Valentijn AR, et al.
Vaccination against HPV-16 oncoproteins for vulvar
intraepithelial neoplasia. N Engl J Med. 2009;361(19):
1838–1847. doi:10.1056/NEJMoa0810097.
Robbins PF, El-Gamil M, Li YF, et al. A mutated
beta-catenin gene encodes a melanoma-specific antigen recognized by tumor infiltrating lymphocytes. J
Exp Med. 1996;183(3):1185–1192. doi:10.1084/jem.
183.3.1185.
Bocchia M, Wentworth PA, Southwood S, et al.
Specific binding of leukemia oncogene fusion protein
peptides to HLA class I molecules. Blood. 1995;
85(10):2680–2684.
doi:10.1182/blood.V85.10.2680.
bloodjournal85102680.
Slingluff CL. The present and future of peptide vaccines for cancer: single or multiple, long or short,
alone or in combination? Cancer J. 2011;17(5):
343–350. doi:10.1097/PPO.0b013e318233e5b2.
Lazoura E, Apostolopoulos V. Insights into peptidebased vaccine design for cancer immunotherapy.
Curr Med Chem. 2005;12(13):1481–1494. doi:10.
2174/0929867054039017.
Kawakami Y, Eliyahu S, Delgado CH, et al.
Identification of a human melanoma antigen recognized by tumor-infiltrating lymphocytes associated
with in vivo tumor rejection. Proc Natl Acad Sci Usa.
1994;91(14):6458–6462. doi:10.1073/pnas.91.14.6458.
Rosenberg SA, Yang JC, Schwartzentruber DJ, et al.
Immunologic and therapeutic evaluation of a synthetic peptide vaccine for the treatment of patients
with metastatic melanoma. Nat Med. 1998;4(3):
321–327. doi:10.1038/nm0398-321.
Salgaller ML, Marincola FM, Cormier JN, Rosenberg
SA. Immunization against epitopes in the human
melanoma antigen gp100 following patient immunization with synthetic peptides. Cancer Res. 1996;
56(20):4749–4757.
Schwartzentruber DJ, Lawson DH, Richards JM, et al.
gp100 peptide vaccine and interleukin-2 in patients
with advanced melanoma. N Engl J Med. 2011;
364(22):2119–2127. doi:10.1056/NEJMoa1012863.
Criscitiello C. Tumor-associated antigens in breast
cancer. Breast Care (Basel)). 2012;7(4):262–266. doi:
10.1159/000342164.
Peres LP, da Luz FA, Pultz BA, et al. Peptide vaccines in breast cancer: the immunological basis for
clinical response. Biotechnol Adv. 2015;33(8):
1868–1877. doi:10.1016/j.biotechadv.2015.10.013.
Disis ML, Gooley TA, Rinn K, et al. Generation of
T-cell immunity to the HER-2/neu protein after
active immunization with HER-2/neu peptide-based
vaccines. J Clin Oncol. 2002;20(11):2624–2632. doi:
10.1200/JCO.2002.06.171.
Mittendorf EA, Clifton GT, Holmes JP, et al. Final
report of the phase I/II clinical trial of the E75 (nelipepimut-S) vaccine with booster inoculations to prevent disease recurrence in high-risk breast cancer
patients. Ann Oncol. 2014;25(9):1735–1742. doi:10.
1093/annonc/mdu211.
28.
29.
30.
31.
32.
33.
34.
35.
36.
37.
38.
39.
40.
41.
Palucka K, Ueno H, Fay J, Banchereau J. Dendritic
cells and immunity against cancer. J Intern Med.
2011;269(1):64–73.
doi:10.1111/j.1365-2796.2010.
02317.x.
Palucka K, Banchereau J. Dendritic-cell-based therapeutic cancer vaccines. Immunity. 2013;39(1):38–48.
doi:10.1016/j.immuni.2013.07.004.
Kirk CJ, Mule JJ. Gene-modified dendritic cells for
use in tumor vaccines. Hum Gene Ther. 2000;11(6):
797–806. doi:10.1089/10430340050015419.
Butterfield LH, Comin-Anduix B, Vujanovic L, et al.
Adenovirus MART-1-engineered autologous dendritic cell vaccine for metastatic melanoma. J
Immunother. 2008;31(3):294–309. doi:10.1097/CJI.
0b013e31816a8910.
Van Tendeloo VF, Van de Velde A, Van Driessche
A, et al. Induction of complete and molecular remissions in acute myeloid leukemia by Wilms’ tumor 1
antigen-targeted dendritic cell vaccination. Proc Natl
Acad Sci Usa. 2010;107(31):13824–13829. doi:10.
1073/pnas.1008051107.
Galati D, Zanotta S. Hematologic neoplasms: dendritic cells vaccines in motion. Clin Immunol. 2017;
183:181–190. doi:10.1016/j.clim.2017.08.016.
Galluzzi L, Senovilla L, Vacchelli E, et al. Trial
watch: dendritic cell-based interventions for cancer
therapy. Oncoimmunology. 2012;1(7):1111–1134. doi:
10.4161/onci.21494.
Vacchelli E, Vitale I, Eggermont A, et al. Trial watch:
dendritic cell-based interventions for cancer therapy.
Oncoimmunology. 2013;2(10):e25771 doi:10.4161/
onci.25771.
Soiffer R, Lynch T, Mihm M, et al. Vaccination with
irradiated autologous melanoma cells engineered to
secrete human granulocyte-macrophage colony-stimulating factor generates potent antitumor immunity
in patients with metastatic melanoma. Proc Natl
Acad Sci Usa. 1998;95(22):13141–13146. doi:10.1073/
pnas.95.22.13141.
Luiten RM, Kueter EW, Mooi W, et al.
Immunogenicity, including vitiligo, and feasibility of
vaccination with autologous GM-CSF-transduced
tumor cells in metastatic melanoma patients. J Clin
Oncol. 2005;23(35):8978–8991. doi:10.1200/JCO.2005.
01.6816.
Chakraborty NG, Sporn JR, Tortora AF, et al.
Immunization with a tumor-cell-lysate-loaded
autologous-antigen-presenting-cell-based vaccine in
melanoma. Cancer Immunol Immunother. 1998;
47(1):58–64. doi:10.1007/s002620050504.
Geiger J, Hutchinson R, Hohenkirk L, McKenna E,
Chang A, Mule J. Treatment of solid tumours in
children with tumour-lysate-pulsed dendritic cells.
Lancet.
2000;356(9236):1163–1165.
doi:10.1016/
S0140-6736(00)02762-8.
R -L-developed by
Polyzoidis S, Ashkan K. DCVaxV
Northwest Biotherapeutics. Hum Vaccin Immunother.
2014;10(11):3139–3145. doi:10.4161/hv.29276.
Butterfield LH, Economou JS, Gamblin TC, Geller
DA. Alpha fetoprotein DNA prime and adenovirus
boost immunization of two hepatocellular cancer
INTERNATIONAL REVIEWS OF IMMUNOLOGY
42.
43.
44.
45.
46.
47.
48.
49.
50.
51.
52.
53.
54.
55.
56.
patients. J Transl Med. 2014;12:86 doi:10.1186/14795876-12-86.
Santiago-Ortiz JL, Schaffer DV. Adeno-associated
virus (AAV) vectors in cancer gene therapy. J
Control Release. 2016;240:287–301. doi:10.1016/j.
jconrel.2016.01.001.
Pol J, Bloy N, Obrist F, et al. Trial watch: DNA vaccines for cancer therapy. Oncoimmunology. 2014;
3(1):e28185 doi:10.4161/onci.28185.
Wilcox ME, Yang W, Senger D, et al. Reovirus as an
oncolytic agent against experimental human malignant gliomas. Journal of the National Cancer
Institute. 2001;93(12):903–912. doi:10.1093/jnci/93.12.
903.
Russell SJ, Peng KW, Bell JC. Oncolytic virotherapy.
Nat Biotechnol. 2012;30(7):658–670. doi:10.1038/nbt.
2287.
Ko D, Hawkins L, Yu DC. Development of transcriptionally regulated oncolytic adenoviruses.
Oncogene. 2005;24(52):7763–7774. doi:10.1038/sj.onc.
1209048.
Russell SJ, Peng KW. Viruses as anticancer drugs.
Trends Pharmacol Sci. 2007;28(7):326–333. doi:10.
1016/j.tips.2007.05.005.
Heini€o C, Havunen R, Santos J, et al. TNFa and IL2
encoding oncolytic adenovirus activates pathogen
and danger-associated immunological signaling.
Cells. 2020;9(4):798. doi:10.3390/cells9040798.
Poutou J, Bunuales M, Gonzalez-Aparicio M, et al.
Safety and antitumor effect of oncolytic and helperdependent adenoviruses expressing interleukin-12
variants in a hamster pancreatic cancer model. Gene
Ther. 2015;22(9):696–706. doi:10.1038/gt.2015.45.
Nguyen HM, Guz-Montgomery K, Saha D.
Oncolytic Virus encoding a master pro-inflammatory
cytokine interleukin 12 in cancer immunotherapy.
Cells. 2020;9(2):400. doi:10.3390/cells9020400.
Wang G, Kang X, Chen KS, et al. An engineered oncolytic virus expressing PD-L1 inhibitors activates tumor
neoantigen-specific T cell responses. Nat Commun.
2020;11(1):1395 doi:10.1038/s41467-020-15229-5.
Eissa IR, Bustos-Villalobos I, Ichinose T, et al. The
current status and future prospects of oncolytic
viruses in clinical trials against melanoma, glioma,
pancreatic, and breast cancers. Cancers. 2018;10(10):
356. doi:10.3390/cancers10100356.
Zhu Z, Gorman MJ, McKenzie LD, et al. Zika virus
has oncolytic activity against glioblastoma stem cells.
J Exp Med. 2017;214(10):2843–2857. doi:10.1084/jem.
20171093.
Zhu Z, Mesci P, Bernatchez JA, et al. Zika Virus
Targets Glioblastoma Stem Cells through a SOX2Integrin avb5 Axis. Cell Stem Cell. 2020;26(2):
187–204.e10. doi:10.1016/j.stem.2019.11.016.
McNamara MA, Nair SK, Holl EK. RNA-based vaccines in cancer immunotherapy. J Immunol Res.
2015;2015:794528 doi:10.1155/2015/794528.
Tiptiri-Kourpeti A, Spyridopoulou K, Pappa A,
Chlichlia K. DNA vaccines to attack cancer: strategies for improving immunogenicity and efficacy.
Pharmacol Ther. 2016;165:32–49. doi:10.1016/j.
pharmthera.2016.05.004.
57.
58.
59.
60.
61.
62.
63.
64.
65.
66.
67.
68.
69.
15
Ahmad S, Casey G, Sweeney P, Tangney M,
O’Sullivan GC. Optimised electroporation mediated
DNA vaccination for treatment of prostate cancer.
Genet Vaccines Ther. 2010;8(1):1. doi:10.1186/14790556-8-1.
Staff C, Mozaffari F, Haller BK, Wahren B, Liljefors
M. A Phase I safety study of plasmid DNA immunization targeting carcinoembryonic antigen in colorectal cancer patients. Vaccine. 2011;29(39):
6817–6822. doi:10.1016/j.vaccine.2010.12.063.
Abraham RS, Mitchell DA. Gene-modified dendritic
cell vaccines for cancer. Cytotherapy. 2016;18(11):
1446–1455. doi:10.1016/j.jcyt.2016.09.009.
Heiser A, Coleman D, Dannull J, et al. Autologous
dendritic cells transfected with prostate-specific antigen RNA stimulate CTL responses against metastatic
prostate tumors. J Clin Invest. 2002;109(3):409–417.
doi:10.1172/JCI0214364.
Bloy N, Buque A, Aranda F, et al. Trial watch: naked
and vectored DNA-based anticancer vaccines.
Oncoimmunology. 2015;4(5):e1026531 doi:10.1080/
2162402X.2015.1026531.
Kantoff PW, Higano CS, Shore ND, et al. SipuleucelT immunotherapy for castration-resistant prostate
cancer. N Engl J Med. 2010;363(5):411–422. doi:10.
1056/NEJMoa1001294.
Paller CJ, Antonarakis ES. Sipuleucel-T for the treatment of metastatic prostate cancer: promise and
challenges. Hum Vaccin Immunother. 2012;8(4):
509–519. doi:10.4161/hv.18860.
Pol J, Kroemer G, Galluzzi L. First oncolytic virus
approved
for
melanoma
immunotherapy.
Oncoimmunology. 2016;5(1):e1115641 doi:10.1080/
2162402X.2015.1115641.
Rosenberg SA, Packard BS, Aebersold PM, Solomon
D, et al. Use of tumor-infiltrating lymphocytes and
interleukin-2 in the immunotherapy of patients with
metastatic melanoma. A preliminary report. N Engl J
Med.
1988;319(25):1676–1680.
doi:10.1056/
NEJM198812223192527.
Gattinoni L, Finkelstein SE, Klebanoff CA, et al.
Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8þ T cells. J Exp Med.
2005;202(7):907–912. doi:10.1084/jem.20050732.
Besser MJ, Shapira-Frommer R, Itzhaki O, et al.
Adoptive transfer of tumor-infiltrating lymphocytes
in patients with metastatic melanoma: intent-to-treat
analysis and efficacy after failure to prior immunotherapies. Clin Cancer Res. 2013;19(17):4792–4800.
doi:10.1158/1078-0432.CCR-13-0380.
Andersen R, Donia M, Ellebaek E, et al. Long-lasting
complete responses in patients with metastatic melanoma after adoptive cell therapy with tumor-infiltrating lymphocytes and an attenuated IL2 regimen.
Clin Cancer Res. 2016;22(15):3734–3745. doi:10.1158/
1078-0432.CCR-15-1879.
Baruch EN, Berg AL, Besser MJ, Schachter J, Markel
G. Adoptive T cell therapy: an overview of obstacles
and opportunities. Cancer. 2017;123(S11):2154–2162.
doi:10.1002/cncr.30491.
16
70.
71.
72.
73.
74.
75.
76.
77.
78.
79.
80.
81.
82.
83.
S. JAIN AND S. KUMAR
Yun CO, Nolan KF, Beecham EJ, Reisfeld RA,
Junghans RP. Targeting of T lymphocytes to melanoma cells through chimeric anti-GD3 immunoglobulin T-cell receptors. Neoplasia. 2000;2(5):449–459.
doi:10.1038/sj.neo.7900108.
Hollyman D, Stefanski J, Przybylowski M, et al.
Manufacturing validation of biologically functional T
cells targeted to CD19 antigen for autologous adoptive cell therapy. J Immunother. 2009;32(2):169–180.
doi:10.1097/CJI.0b013e318194a6e8.
Maus MV, June CH. Making better chimeric antigen
receptors for adoptive T-cell therapy. Clin Cancer
Res. 2016;22(8):1875–1884. doi:10.1158/1078-0432.
CCR-15-1433.
Park JH, Brentjens RJ. Are all chimeric antigen
receptors created equal? J Clin Oncol. 2015;33(6):
651–653. doi:10.1200/JCO.2014.57.5472.
Gross G, Waks T, Eshhar Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc
Natl Acad Sci Usa. 1989;86(24):10024–10028. doi:10.
1073/pnas.86.24.10024.
Hwu P, Shafer GE, Treisman J, et al. Lysis of ovarian
cancer cells by human lymphocytes redirected with a
chimeric gene composed of an antibody variable
region and the Fc receptor gamma chain. J Exp Med.
1993;178(1):361–366. doi:10.1084/jem.178.1.361.
Kershaw MH, Westwood JA, Hwu P. Dual-specific T
cells combine proliferation and antitumor activity.
Nat Biotechnol. 2002;20(12):1221–1227. doi:10.1038/
nbt756.
Kershaw MH, Westwood JA, Parker LL, et al. A
phase I study on adoptive immunotherapy using
gene-modified T cells for ovarian cancer. Clin
Cancer Res. 2006;12(20 Pt 1):6106–6115. doi:10.1158/
1078-0432.CCR-06-1183.
Kowolik CM, Topp MS, Gonzalez S, et al. CD28 costimulation provided through a CD19-specific chimeric antigen receptor enhances in vivo persistence
and antitumor efficacy of adoptively transferred T
cells. Cancer Res. 2006;66(22):10995–11004. doi:10.
1158/0008-5472.CAN-06-0160.
Milone MC, Fish JD, Carpenito C, et al. Chimeric
receptors containing CD137 signal transduction
domains mediate enhanced survival of T cells and
increased antileukemic efficacy in vivo. Mol Ther.
2009;17(8):1453–1464. doi:10.1038/mt.2009.83.
Chmielewski M, Abken H. TRUCKs: the fourth generation of CARs. Expert Opin Biol Ther. 2015;15(8):
1145–1154. doi:10.1517/14712598.2015.1046430.
Davila ML, Riviere I, Wang X, et al. Efficacy and toxicity management of 19-28z CAR T cell therapy in B
cell acute lymphoblastic leukemia. Sci Transl Med.
2014;6(224):224ra25 doi:10.1126/scitranslmed.3008226.
Kakarla S, Gottschalk S. CAR T cells for solid
tumors: armed and ready to go? Cancer J. 2014;
20(2):151–155. doi:10.1097/PPO.0000000000000032.
K€
ohler G, Milstein C. Continuous cultures of
fused cells secreting antibody of predefined specificity. Nature. 1975;256(5517):495–497. doi:10.
1038/256495a0.
84.
85.
86.
87.
88.
89.
90.
91.
92.
93.
94.
95.
96.
97.
Coulson A, Levy A, Gossell-Williams M. Monoclonal
antibodies in cancer therapy: mechanisms, successes
and limitations. West Indian Med J. 2014;63(6):
650–654. doi:10.7727/wimj.2013.241.
Vaklavas C, Forero-Torres A. Safety and efficacy of
brentuximab vedotin in patients with Hodgkin
lymphoma or systemic anaplastic large cell lymphoma. Ther Adv Hematol. 2012;3(4):209–225. doi:10.
1177/2040620712443076.
Kaminski MS, Estes J, Zasadny KR, et al.
Radioimmunotherapy with iodine (131)I tositumomab for relapsed or refractory B-cell non-Hodgkin
lymphoma: updated results and long-term follow-up
of the University of Michigan experience. Blood.
2000;96(4):1259–1266.
Nguyen TH, Havari E, McLaren R, et al.
Alemtuzumab induction of intracellular signaling
and apoptosis in malignant B lymphocytes. Leuk
Lymphoma.
2012;53(4):699–709.
doi:10.3109/
10428194.2011.623253.
Lynce F, Swain SM. Pertuzumab for the treatment of
breast cancer. Cancer Invest. 2014;32(8):430–438. doi:
10.3109/07357907.2014.922570.
Adams GP, Weiner LM. Monoclonal antibody therapy of cancer. Nat Biotechnol. 2005;23(9):1147–1157.
doi:10.1038/nbt1137.
Weiner GJ. Rituximab: mechanism of action. Semin
Hematol. 2010;47(2):115–123. doi:10.1053/j.seminhematol.2010.01.011.
Seimetz D. Novel monoclonal antibodies for cancer
treatment: the trifunctional antibody catumaxomab
(removab). J Cancer. 2011;2:309–316. doi:10.7150/jca.
2.309.
H€
orl S, Banki Z, Huber G, et al. Complement factor
H-derived short consensus repeat 18–20 enhanced
complement-dependent cytotoxicity of ofatumumab
on
chronic
lymphocytic
leukemia
cells.
Haematologica. 2013;98(12):1939–1947. doi:10.3324/
haematol.2013.089615.
Miotti S, Canevari S, Menard S, et al.
Characterization of human ovarian carcinoma-associated antigens defined by novel monoclonal antibodies with tumor-restricted specificity. Int J Cancer.
1987;39(3):297–303. doi:10.1002/ijc.2910390306.
Yang JC, Hughes M, Kammula U, et al. Ipilimumab
(anti-CTLA4 antibody) causes regression of metastatic renal cell cancer associated with enteritis and
hypophysitis. J Immunother. 2007;30(8):825–830. doi:
10.1097/CJI.0b013e318156e47e.
Callahan MK, Postow MA, Wolchok JD.
Immunomodulatory therapy for melanoma: ipilimumab and beyond. Clin Dermatol. 2013;31(2):191–199.
doi:10.1016/j.clindermatol.2012.08.006.
Willett CG, Boucher Y, di Tomaso E, et al. Direct
evidence that the VEGF-specific antibody bevacizumab has antivascular effects in human rectal cancer.
Nat Med. 2004;10(2):145–147. doi:10.1038/nm988.
Tsumoto K, Isozaki Y, Yagami H, Tomita M. Future
perspectives of therapeutic monoclonal antibodies.
Immunotherapy. 2019;11(2):119–127. doi:10.2217/
imt-2018-0130.
INTERNATIONAL REVIEWS OF IMMUNOLOGY
98.
99.
100.
101.
102.
103.
104.
105.
106.
107.
108.
109.
110.
111.
112.
Yonesaka K, Zejnullahu K, Okamoto I, et al.
Activation of ERBB2 signaling causes resistance to
the EGFR-directed therapeutic antibody cetuximab.
Sci Transl Med. 2011;3(99):99ra86 doi:10.1126/scitranslmed.3002442.
Nahta R, Yu D, Hung MC, Hortobagyi GN, Esteva
FJ. Mechanisms of disease: understanding resistance
to HER2-targeted therapy in human breast cancer.
Nat Clin Pract Oncol. 2006;3(5):269–280. doi:10.
1038/ncponc0509.
Rexer BN, Arteaga CL. Intrinsic and acquired resistance to HER2-targeted therapies in HER2 geneamplified breast cancer: mechanisms and clinical
implications. Crit Rev Oncog. 2012;17(1):1–16. doi:
10.1615/critrevoncog.v17.i1.20.
Coloma MJ, Morrison SL. Design and production of
novel tetravalent bispecific antibodies. Nat Biotechnol.
1997;15(2):159–163. doi:10.1038/nbt0297-159.
Fournier P, Schirrmacher V. Bispecific antibodies
and trispecific immunocytokines for targeting the
immune system against cancer: preparing for the
future. BioDrugs. 2013;27(1):35–53. doi:10.1007/
s40259-012-0008-z.
Weidle UH, Tiefenthaler G, Weiss EH, Georges G,
Brinkmann U. The intriguing options of multispecific antibody formats for treatment of cancer.
Cancer Genomics Proteomics. 2013;10(1):1–18.
Jachimowicz RD, Borchmann S, Rothe A. Multi-specific antibodies for cancer immunotherapy. BioDrugs.
2014;28(4):331–343. doi:10.1007/s40259-014-0091-4.
Hodi FS, Mihm MC, Soiffer RJ, et al. Biologic activity of cytotoxic T lymphocyte-associated antigen 4
antibody blockade in previously vaccinated metastatic melanoma and ovarian carcinoma patients.
Proc Natl Acad Sci USA. 2003;100(8):4712–4717. doi:
10.1073/pnas.0830997100.
Contardi E, Palmisano GL, Tazzari PL, et al. CTLA4 is constitutively expressed on tumor cells and can
trigger apoptosis upon ligand interaction. Int J
Cancer. 2005;117(4):538–550. doi:10.1002/ijc.21155.
Tseng SY, Otsuji M, Gorski K, et al. B7-DC, a new
dendritic cell molecule with potent costimulatory
properties for T cells. J Exp Med. 2001;193(7):
839–846. doi:10.1084/jem.193.7.839.
Lote H, Cafferkey C, Chau I. PD-1 and PD-L1
blockade in gastrointestinal malignancies. Cancer
Treat Rev. 2015;41(10):893–903. doi:10.1016/j.ctrv.
2015.09.004.
Ohaegbulam KC, Assal A, Lazar-Molnar E, Yao Y,
Zang X. Human cancer immunotherapy with antibodies to the PD-1 and PD-L1 pathway. Trends Mol Med.
2015;21(1):24–33. doi:10.1016/j.molmed.2014.10.009.
Lipson EJ, Forde PM, Hammers HJ, Emens LA,
Taube JM, Topalian SL. Antagonists of PD-1 and
PD-L1 in cancer treatment. Semin Oncol. 2015;42(4):
587–600. doi:10.1053/j.seminoncol.2015.05.013.
Swaika A, Hammond WA, Joseph RW. Current state
of anti-PD-L1 and anti-PD-1 agents in cancer therapy. Mol Immunol. 2015;67(2 Pt A):4–17. doi:10.
1016/j.molimm.2015.02.009.
Callahan MK, Wolchok JD. At the bedside: CTLA-4and PD-1-blocking antibodies in cancer immunotherapy.
113.
114.
115.
116.
117.
118.
119.
120.
121.
122.
123.
124.
125.
126.
127.
17
J Leukoc Biol. 2013;94(1):41–53. doi:10.1189/jlb.
1212631.
Postow MA. Managing immune checkpoint-blocking
antibody side effects. Am Soc Clin Oncol Educ Book.
2015;35:76–83. doi:10.14694/EdBook_AM.2015.35.76.
Assal A, Kaner J, Pendurti G, Zang X. Emerging targets in cancer immunotherapy: beyond CTLA-4 and
PD-1. Immunotherapy. 2015;7(11):1169–1186. doi:10.
2217/imt.15.78.
Farkona S, Diamandis EP, Blasutig IM. Cancer
immunotherapy: the beginning of the end of cancer?
BMC Med. 2016;14:73 doi:10.1186/s12916-016-0623-5.
Mahoney KM, Rennert PD, Freeman GJ.
Combination cancer immunotherapy and new
immunomodulatory targets. Nat Rev Drug Discov.
2015;14(8):561–584. doi:10.1038/nrd4591.
Robert C, Thomas L, Bondarenko I, et al.
Ipilimumab plus dacarbazine for previously
untreated metastatic melanoma. N Engl J Med. 2011;
364(26):2517–2526. doi:10.1056/NEJMoa1104621.
Kwon ED, Drake CG, Scher HI, et al. Ipilimumab
versus placebo after radiotherapy in patients with
metastatic castration-resistant prostate cancer that
had progressed after docetaxel chemotherapy
(CA184-043): a multicentre, randomised, doubleblind, phase 3 trial. Lancet Oncol. 2014;15(7):
700–712. doi:10.1016/S1470-2045(14)70189-5.
Topalian SL, Weiner GJ, Pardoll DM. Cancer
immunotherapy comes of age. J Clin Oncol. 2011;
29(36):4828–4836. doi:10.1200/JCO.2011.38.0899.
Curran MA, Montalvo W, Yagita H, Allison JP. PD1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid
cells within B16 melanoma tumors. Proc Natl Acad
Sci Usa. 2010;107(9):4275–4280. doi:10.1073/pnas.
0915174107.
Das R, Verma R, Sznol M, et al. Combination therapy with anti-CTLA-4 and anti-PD-1 leads to distinct immunologic changes in vivo. J Immunol. 2015;
194(3):950–959. doi:10.4049/jimmunol.1401686.
Wolchok JD, Kluger H, Callahan MK, et al.
Nivolumab plus ipilimumab in advanced melanoma.
N Engl J Med. 2013;369(2):122–133. doi:10.1056/
NEJMoa1302369.
Chen DS, Mellman I. Oncology meets immunology:
the cancer-immunity cycle. Immunity. 2013;39(1):
1–10. doi:10.1016/j.immuni.2013.07.012.
Duraiswamy J, Kaluza KM, Freeman GJ, Coukos G.
Dual blockade of PD-1 and CTLA-4 combined with
tumor vaccine effectively restores T-cell rejection
function in tumors. Cancer Res. 2013;73(12):
3591–3603. doi:10.1158/0008-5472.CAN-12-4100.
Hodi FS, O’Day SJ, McDermott DF, et al. Improved
survival with ipilimumab in patients with metastatic
melanoma. N Engl J Med. 2010;363(8):711–723. doi:
10.1056/NEJMoa1003466.
Emens LA. Cancer vaccines: on the threshold of success. Expert Opin Emerg Drugs. 2008;13(2):295–308.
doi:10.1517/14728214.13.2.295.
Graff JN, Chamberlain ED. Sipuleucel-T in the treatment of prostate cancer: an evidence-based review of
18
128.
129.
130.
131.
132.
133.
134.
135.
136.
137.
138.
139.
140.
141.
S. JAIN AND S. KUMAR
its place in therapy. Core Evid. 2015;10:1–10. doi:10.
2147/CE.S54712.
Yaddanapudi K, Mitchell RA, Eaton JW. Cancer vaccines: looking to the future. Oncoimmunology. 2013;
2(3):e23403 doi:10.4161/onci.23403.
Yee C. Adoptive T cell therapy: addressing challenges in cancer immunotherapy. J Transl Med.
2005;3(1):17doi:10.1186/1479-5876-3-17.
Morgan RA, Yang JC, Kitano M, Dudley ME,
Laurencot CM, Rosenberg SA. Case report of a serious adverse event following the administration of T
cells transduced with a chimeric antigen receptor
recognizing ERBB2. Mol Ther. 2010;18(4):843–851.
doi:10.1038/mt.2010.24.
Choi D, Kim TG, Sung YC. The past, present, and
future of adoptive T cell therapy. Immune Netw.
2012;12(4):139–147. doi:10.4110/in.2012.12.4.139.
Worcester S. CAR T-cell therapy: Moving from cost
to value 2017. 2017. https://www.mdedge.com/hematologynews/article/152563/all-acute-lymphoblastic-leukemia/car-t-cell-therapy-moving-cost-value. Published
November 21, 2017. Accessed November 8, 2018.
Qi WX, Fu S, Zhang Q, Guo XM. Incidence and risk
of severe infections associated with anti-epidermal
growth factor receptor monoclonal antibodies in
cancer patients: a systematic review and meta-analysis. BMC Med. 2014;12(1):203. doi:10.1186/s12916014-0203-5.
Riedell P, Carson KR. A drug safety evaluation of
rituximab and risk of hepatitis B. Expert Opin Drug
Saf. 2014;13(7):977–987. doi:10.1517/14740338.2014.
918948.
Simpson A, Caballero O. Monoclonal antibodiesfor
the therapy of cancer. BMC Proceedings. 2014;
8(Suppl 4):O6. https://doi.org/10.1186/1753-6561-8S4-O6
Geynisman DM, Chien CR, Smieliauskas F, Shen C,
Shih YC. Economic evaluation of therapeutic cancer
vaccines and immunotherapy: a systematic review.
Hum Vaccin Immunother. 2014;10(11):3415–3424.
doi:10.4161/hv.29407.
Verma V, Sprave T, Haque W, et al. A systematic
review of the cost and cost-effectiveness studies of
immune checkpoint inhibitors. J Immunotherapy
Cancer. 2018;6:128.
Anassi E, Ndefo UA. Sipuleucel-T (provenge) injection: the first immunotherapy agent (vaccine) for
hormone-refractory prostate cancer. PT. 2011;36(4):
197–202.
Caram M, Ross R, Lin P, Mukherjee B. Factors associated with use of Sipuleucel-T to treat patients with
advanced prostate cancer. JAMA Netw Open. 2019;
2(4):e192589 doi:10.1001/jamanetworkopen.2019.2589.
Gong CL, Hay JW. Cost-effectiveness analysis of
abiraterone and sipuleucel-T in asymptomatic metastatic castration-resistant prostate cancer. J Natl
Compr Canc Netw. 2014;12(10):1417–-1425. doi:10.
6004/jnccn.2014.0139.
Chopra R, Lopes G. Improving Access to Cancer
Treatments: The Role of Biosimilars. J Glob Oncol.
2017;3(5):596–610. doi:10.1200/JGO.2016.008607.
142.
143.
144.
145.
146.
147.
148.
149.
150.
151.
152.
153.
Amos SM, Duong CP, Westwood JA, et al.
Autoimmunity associated with immunotherapy of
cancer. Blood. 2011;118(3):499–509. doi:10.1182/
blood-2011-01-325266.
Dores GM, Bryant-Genevier M, Perez-Vilar S.
Adverse events associated with the use of SipuleucelT reported to the US food and drug administration’s
adverse event reporting system, 2010-2017. JAMA
Netw Open. 2019;2(8):e199249 doi:10.1001/jamanetworkopen.2019.9249.
Brudno JN, Kochenderfer JN. Recent advances in
CAR T-cell toxicity: mechanisms, manifestations and
management. Blood Rev. 2019;34:45–45. doi:10.1016/
j.blre.2018.11.002.
Alatrash G, Jakher H, Stafford PD, Mittendorf EA.
Cancer immunotherapies, their safety and toxicity.
Expert Opin Drug Saf. 2013;12(5):631–645. doi:10.
1517/14740338.2013.795944.
Puzanov I, Diab A, Abdallah K, et al. Managing toxicities associated with immune checkpoint inhibitors:
consensus recommendations from the Society for
Immunotherapy of Cancer (SITC) Toxicity
Management Working Group. J Immunother Cancer.
2017;5(1):95.
Martinez P, Peters S, Stammers T, Soria JC.
Immunotherapy for the first-line treatment of
patients with metastatic non-small cell lung cancer.
Clin Cancer Res. 2019;25(9):2691–2698. doi:10.1158/
1078-0432.CCR-18-3904.
Kalani AD, Jack A, Montenegro G, Degliuomini J,
Wallack MK. Immunotherapy as an adjuvant therapy
in the management of advanced, surgically resected,
melanoma. G Ital Dermatol Venereol. 2008;143(1):
59–70.
Spencer KR, Wang J, Silk AW, Ganesan S, Kaufman
HL, Mehnert JM. Biomarkers for Immunotherapy:
Current Developments and Challenges. Am Soc Clin
Oncol Educ Book. 2016;35:e493–-503. doi:10.1200/
EDBK_160766.
Jilg S, Rassner M, Maier J, et al. Circulating cKIT
and PDGFRA DNA indicates disease activity in
Gastrointestinal Stromal Tumor (GIST). Int J
Cancer.
2019;145(8):2292–2303.
doi:10.1002/ijc.
32282.
Costantini A, Julie C, Dumenil C, et al. Predictive
role of plasmatic biomarkers in advanced non-small
cell
lung
cancer
treated
by
nivolumab.
Oncoimmunology. 2018. 2018;7(8):e1452581 doi:10.
1080/2162402X.2018.1452581.
Vincent J, Mignot G, Chalmin F, et al. 5Fluorouracil selectively kills tumor-associated myeloid-derived suppressor cells resulting in enhanced T
cell-dependent antitumor immunity. Cancer Res.
2010;70(8):3052–3061. doi:10.1158/0008-5472.CAN09-3690.
Le DT, Jaffee EM. Regulatory T-cell modulation
using cyclophosphamide in vaccine approaches: a
current perspective. Cancer Res. 2012;72(14):
3439–3444. doi:10.1158/0008-5472.CAN-11-3912.