0% found this document useful (0 votes)
28 views13 pages

Cancer Vaccines - Toward The Next Breakthrough in Cancer Immunotherapy

Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
0% found this document useful (0 votes)
28 views13 pages

Cancer Vaccines - Toward The Next Breakthrough in Cancer Immunotherapy

Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
You are on page 1/ 13

Hindawi

Journal of Immunology Research


Volume 2020, Article ID 5825401, 13 pages
https://doi.org/10.1155/2020/5825401

Review Article
Cancer Vaccines: Toward the Next Breakthrough in
Cancer Immunotherapy

1
Yuka Igarashi and Tetsuro Sasada1,2
1
Kanagawa Cancer Center, Research Institute, Division of Cancer Immunotherapy, Japan
2
Kanagawa Cancer Center, Cancer Vaccine and Immunotherapy Center, Japan

Correspondence should be addressed to Yuka Igarashi; [email protected]

Received 22 July 2020; Revised 26 September 2020; Accepted 30 September 2020; Published 17 November 2020

Academic Editor: Fabiano Carvalho

Copyright © 2020 Yuka Igarashi and Tetsuro Sasada. This is an open access article distributed under the Creative Commons
Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work
is properly cited.

Until now, three types of well-recognized cancer treatments have been developed, i.e., surgery, chemotherapy, and radiotherapy;
these either remove or directly attack the cancer cells. These treatments can cure cancer at earlier stages but are frequently
ineffective for treating cancer in the advanced or recurrent stages. Basic and clinical research on the tumor microenvironment,
which consists of cancerous, stromal, and immune cells, demonstrates the critical role of antitumor immunity in cancer
development and progression. Cancer immunotherapies have been proposed as the fourth cancer treatment. In particular,
clinical application of immune checkpoint inhibitors, such as anti-CTLA-4 and anti-PD-1/PD-L1 antibodies, in various cancer
types represents a major breakthrough in cancer therapy. Nevertheless, accumulating data regarding immune checkpoint
inhibitors demonstrate that these are not always effective but are instead only effective in limited cancer populations. Indeed,
several issues remain to be solved to improve their clinical efficacy; these include low cancer cell antigenicity and poor
infiltration and/or accumulation of immune cells in the cancer microenvironment. Therefore, to accelerate the further
development of cancer immunotherapies, more studies are necessary. In this review, we will summarize the current status of
cancer immunotherapies, especially cancer vaccines, and discuss the potential problems and solutions for the next breakthrough
in cancer immunotherapy.

1. Introduction Additionally, the World Health Organization (WHO) rec-


ommends the administration of free mumps and varicella
When one hears the word “vaccine,” many people think of vaccines in developed countries, where vaccines are recog-
vaccines against infectious agents, such as viruses and bacte- nized as being among the most versatile and important
ria. For many years, such vaccines have protected human- preventive measures [4].
kind from catastrophic infections [1]. The mechanism The immune system is directed at maintaining homeo-
through which vaccines provide protection against an infec- stasis in living organisms by monitoring the invasion of
tion involves the artificial induction of immune responses foreign pathogens (and associated factors), as well as the
against infectious antigens by inoculating a healthy person presence of abnormal or transformed cells, for their exclu-
with attenuated/detoxified bacteria, viruses, or extracted sion. This process is called immune surveillance [5]. In daily
toxins [2]. The aim of a vaccine is to prevent or reduce the life, humans are exposed to external factors, such as bacteria,
severity of life-threatening infectious diseases (prophylactic viruses, or harmful substances. Additionally, humans are
vaccines). Acquisition of immune memory from vaccines is exposed to various factors that lead to abnormalities and
often effective over long periods of time [3]. A global system transformations in normal cells. However, it is rare that these
of routine immunization against highly prevalent infections exposures or transformations immediately lead to the devel-
has been effectively established; this has resulted in multiple opment of disease, because humans are strongly protected by
individuals developing immunity against various diseases. their immune systems. When there is an imbalance between
2 Journal of Immunology Research

extraneous stimuli and biological defense and when compo- 3. Immunological Characteristics of Cancers
nents of the immune system are unable to eliminate the path-
ogen or malfunctioning cells, conditions, such as infections Progress in the latter half of the 20th century in tumor immu-
and cancers, develop [6]. nology and molecular biology has been remarkable. Numer-
ous studies have vigorously investigated the mechanism by
which tumors evade the immune system. These efforts have
2. History of Cancer Vaccines identified a mechanism called “cancer immunoediting” as
one of the immune evasion tactics utilized by the tumors
Till now, chemotherapy, radiotherapy, and surgical exci- [16]. Cancer immunoediting appears to be the consequence
sion are the three major cancer treatment methods that of antitumor immune responses mediated by antigen recog-
directly remove or target the cancer cells. In addition, nition in the tumor environment. The interaction between
immunotherapies for the treatment of cancer by leveraging the immune system and cancer cells, which originally con-
the innate and/or adaptive immune function in humans tained specific genetic mutations, may cause a selective and
have been extensively studied. The critical role of the biased proliferation of the clones that have lost these muta-
tumor microenvironment (TME), which consists of cancer, tions, leading to tumor escape from the immune system.
stromal, and immune cells that interact with each other, is For these cancers, the immune system might not discrimi-
becoming increasingly apparent. Therefore, cancer immu- nate cancer cells that have lost specific antigens from normal
notherapies have been reconsidered and recognized as host cells, resulting in the possibility that cancer cells do not
the fourth treatment method (Figure 1) [7–9]. Preventive elicit a strong exclusionary immune response.
and therapeutic vaccines exist as representative strategies Additionally, the presentation of cancer cell antigens to
for cancer immunotherapy. The former is aimed at induc- the T cells differs from the presentation of antigens by mature
ing immune memory by administering vaccines to healthy antigen-presenting cells (APCs) in the context of the partici-
persons to prevent morbidity due to a particular cancer. pation of costimulatory molecules. Antigen presentation by
The latter is administered to patients with cancer for dis- the APCs involves the presence of a simultaneous second sig-
ease management by reinforcing or reactivating the nal from costimulatory molecules, such as CD28, for induc-
patient’s own immune system. ing T cell activation during antigen recognition. This
Clinical application of cancer vaccines has faced extreme second signal controls the subsequent T cell response [17].
hurdles despite multiyear research and development efforts When the antigen is presented to T cells without the second
by many researchers. The administration of streptococcal signal, antigen stimulation itself might be ignored; this is
organisms (Coley’s toxin) as a therapeutic vaccine in patients termed unresponsive anergy resulting in the loss of the
with sarcoma in the 1890s by Dr. William B. Coley was the antigen-specific T cell responses. Because cancer cells lack
first report of cancer immunotherapy [10]. In this strategy, such critical second signals, they may not efficiently induce
for both prophylactic and therapeutic purposes, specific T cell responses, even if strong cancer-specific antigens
immune responses were induced against certain sarcoma derived from genetic mutations are presented to T cells.
antigens. The development of this cancer vaccine was based Immunosuppressive cells (e.g., myeloid-derived suppres-
on the clinical findings that the incidence of cancer was low sor cells (MDSCs) and regulatory T (Treg) cells) are recruited
in patients with certain infectious diseases. This phenome- to the TME by chemotactic factors derived from tumor, stro-
non may reflect the fact that infection and inflammation mal, or other immune cells and convey negative signals to the
induce the exposure of antigens abnormally expressed by antitumor immune cells via the expression of inhibitory
cancer cells. It might also be a secondary effect, where the ligands (e.g., programmed death-ligand 1 (PD-L1)) and the
immunological memory acquired from past infection or secretion of immunosuppressive factors (e.g., interleukin 10
inflammation affects the cancer cells. Similarly, antibodies (IL-10), transforming growth factor-beta (TGF-b), and pros-
against abnormal cell surface-associated mucin (MUC1) pro- taglandin E2 (PGE 2)). In doing so, an environment favoring
duced during mumps infection decreases the incidence of cancer cell growth is created [18].
ovarian cancer [11]. Moreover, Bacillus Calmette-Guerin
has been used as a tuberculosis vaccine for a long time [12, 4. Current Status of Cancer Vaccines
13] and is now also widely employed as a therapeutic vaccine
against bladder cancer. The identification of the mechanisms used by the cancer cells
As some types of cancers are caused by infectious viruses, to evade the immune system has resulted in the development
prophylactic vaccines against viral infection can prevent can- of several tools including antibodies, peptides, proteins,
cer development [14, 15]. The Food and Drug Administra- nucleic acids, and immunocompetent cells (dendritic cells,
tion (FDA) has approved two types of prophylactic cancer T cells, etc.) for cancer immunotherapy. With respect to can-
vaccines for targeting the human papillomavirus (HPV) cer vaccines, these techniques fall into three major categories
and hepatitis B virus (HBV) to prevent HPV-related cancers based on format and content, i.e., cell vaccines (tumor or
and HBV-related hepatocarcinoma. However, only a few immune cells), protein/peptide vaccines, and nucleic acid
types of cancer are caused by viral infections. In addition, vaccines (DNA, RNA, or viral vector).
the global vaccination rate for these prophylactic vaccines is
not high. Thus, the number of patients in whom the antiviral 4.1. Cell Vaccines (Tumor Cell Vaccines or Dendritic Cell (DC)
vaccines have successfully prevented cancers is limited. Vaccines). An autologous tumor cell vaccine using a patient’s
Journal of Immunology Research 3

F
Chemotherapy
s
od Cl
et h
lm H
na O
N O HN
io y
e nt N
rap
nv HN he
co F dt
ete
O N
e
re t arg CTLA4
Th Radiotherapy O N ly PD-1
lar
C O
H3

l ec u PDL-1, etc.
P Mo
12C

Immune checkpoint inhibitor


(antibody reagent)
Surgery

Adaptive cell transfer


(NK, NKT, 𝛾𝛿T, TIL, etc)

Engineered T cell therapy


Vaccine (CAR-T, TCR-T)
(cell vaccine (tumor-cell, immune-cell),
Protein/peptide vaccine,
nucleic acid vaccine)

Fourth methods

Immunotherapy

Figure 1: Cancer treatment methods. Conventional methods for cancer treatment include surgery, chemotherapy, and radiation therapy,
which remove or directly attack the cancer cells. Recent advances in medical science have resulted in the addition of cancer
immunotherapies as a fourth treatment method, which can indirectly attack cancers by regulating the patient’s immunity.

own cancer cells is one of the vaccine strategies being evaluated. associated with the individualization of autologous tumor
In this approach, irradiated tumor cells are administered along vaccines. These vaccines have been studied in prostate [43,
with an adjuvant. As this vaccine uses tumor cells, it might be 44], breast [45], and pancreatic cancers [46]. Although
possible to induce T cells specific to any antigen expressed by homologous GVAX against metastatic castration-resistant
the used cells. However, the limitation of this strategy is that a prostate cancer (CRPC) did not achieve its phase 3 clinical
sufficient number of cells is sometimes difficult to obtain [19– trial goals, a combination strategy using allogeneic GVAX
22]. This approach has been attempted in many tumors, includ- against CRPC and an immune checkpoint inhibitor is being
ing lung cancer [22–24], colorectal cancer [20, 25–27], mela- studied [47, 48].
noma [28–30], renal cell carcinoma [31–33], and prostate A new therapeutic approach focuses on DCs that present
cancer [19, 34]. In many cases, tumor cells are genetically mod- antigens to T cells and promote immune system activation.
ified to add functions, such as cytokine production (e.g., IL-2 DC therapy has been intensively studied since the late
[35] and granulocyte-macrophage colony-stimulating factor 1990s [49], when Dr. Ralph M. Steinman, who discovered
(GM-CSF) [36–39]) and costimulation (e.g., B7-1) [32]. GVAX DCs, recognized their potential, and the possibility of using
is a cancer vaccine based on tumor cells genetically modified to DCs as a vaccine [50]. A variety of antigens, including tumor
secrete GM-CSF. It is used after cancer irradiation to stop the cells, tumor-derived proteins or peptides, and DNA/RNA/-
uncontrollable growth of cancer cells. There are two types of virus, could be potentially loaded on DCs. There are addi-
GVAX vaccine approaches, one using autologous cells tional methods, such as the fusion of DCs with tumor cells.
(patient-specific), and the other using allogeneic cells (non- Several receptor types are expressed on the surface of DCs.
patient-specific). GVAX phase 1/2 clinical trials in patients with For example, binding of an antigen to a lectin-like receptor
non-small-cell lung carcinoma have shown good results, corre- known as scavenger receptor on DCs is reported to induce
lating GM-CSF secretion and patient prognosis [40]. However, antigen-specific suppressive CD4(+) T cells. It is noteworthy
no effects have been seen in phase 3 clinical trials for prostate that not all antigen presentation by DCs contributes to
cancer [41]. Currently, several phase 2 trials of GVAX therapy immune activation [51].
for advanced pancreatic cancer have been conducted in combi- In 2010, Provenge (sipuleucel-T; Dendreon Corporation)
nation with body radiation or mesothelin-expressing Listeria was approved by the FDA as a prostate cancer vaccine and
monocytogenes vaccine or cyclophosphamide (CY), with prom- has drawn attention to the use of autologous immune cells
ising results. for immunotherapy. It is a crude leukocyte fraction recovered
An allogeneic tumor cell vaccine that includes tumor cell from the peripheral blood of an individual patient, which is
lines, such as Canvaxin [42], may overcome the limitation then cultured with a prostate carcinoma antigen (prostatic
4 Journal of Immunology Research

acid phosphatase (PAP)) in the presence of GM-CSF. DCs nostimulatory adjuvant has been developed to improve the
are the main active components of Provenge (about 11.2% responsiveness to peptide vaccines [65–68].
[52]) and display the PAP antigen to artificially stimulate
and induce antigen-specific T cells in patients. Provenge is 4.3. Nucleic Acid Vaccines (DNA, RNA, or Viral Vector).
a good example of the complexity of personalized medicine, Nucleic acid (DNA/RNA) vaccines have advantages in that
as personalized cancer vaccines can be effectively created they can simultaneously activate immunity against multiple
using this approach. Nevertheless, all of the processes epitopes [69]. Further, these vaccines are inexpensive and
involved in the production of a personalized vaccine, from can be synthesized stably. When an immunogenic viral vec-
sample collection to transporting, processing, shipping, and tor is used, the adjuvants are not as important, unlike in pep-
administration of the cells, need to be customized for each tide vaccines. However, when a viral vector is not used,
patient, leading to increased labor and cost. developing an efficient delivery method becomes an impor-
tant issue, especially as the efficiency of nucleic acid uptake
4.2. Protein/Peptide Vaccines. Protein/peptide vaccines can into cells might be low [70].
induce immunity against specific antigenic epitopes derived DNA vaccines have shown promise in several prelimi-
from the vaccinated protein/peptides that are expressed in nary studies [71, 72]. For example, VGX3100, a DNA vaccine
cancer cells (and preferably not expressed in normal tissues). for cervical cancer, is in phase 3 clinical trials
When an artificially synthesized antigen protein/peptide is (NCT03185013) [73]. RNA vaccines, unlike DNA vaccines,
administered, it is taken up by professional APCs and pre- are not incorporated into the genome, thereby preventing
sented in complex with the HLA molecules on the cell sur- carcinogenicity. Additionally, unlike DNA vaccines that need
face. When T cells recognize the antigens, cancer-specific to enter the nucleus, RNA vaccines can function in the cyto-
immune responses are induced. Antigenic epitopes derived plasm. Therefore, clearance is quick and the possibility of
from tumor-associated antigens (TAAs) capable of binding causing side effects might be low. RNA is more easily
HLA have been extensively identified. In addition, antigens degraded than DNA, but stability can be enhanced by various
derived from cancer-specific gene mutations that are not modifications, such as formulations with liposomes or stabi-
present in normal tissues have recently attracted attention lizing adjuvants [74–77]. Techniques have also been devel-
as neoantigens. To efficiently search for these neoantigens, oped to stabilize the RNA molecule itself (5 ′ cap structure,
algorithm-based computer searches are rapidly being devel- untranslated regions, and codon usage in translated regions)
oped [53–55]. [78]. Phase 1/2 studies are ongoing for melanoma and kidney
As many early protein/peptide vaccine clinical trials have cancer [79–81]. A phase I study of liposome-encapsulated
resulted in favorable results, phase 3 trials have been con- mRNA for patients with advanced melanoma is also under-
ducted to confirm the results. Unfortunately, most of these way [82]. Further development of nucleic acid delivery
trials have failed, suggesting that single-protein/peptide vac- methods will serve as a breakthrough in nucleic acid vaccines.
cines do not exert sufficient antitumor effects [56]. Even if Viral vectors are used to efficiently carry nucleic acids for
T cell responses were induced by protein-/peptide-derived vaccines. Adjuvants are not required for viral vectors, which
epitopes, the antitumor effects can rarely be achieved with can activate innate immunity and also induce immune
low response rate (less than 10%) [57, 58]. These unexpected responses to viruses. Commonly used viral vectors are derived
results may be explained by several factors, including tumor from poxvirus, vaccinia virus, adenovirus, and alphavirus and
immune escape mechanisms and immunosuppressive TMEs are attenuated or replication-defective for safety. For example,
[59]. some modified vaccinia virus Ankara (MVA) vector-based
There may be problems with the vaccine formulations; vaccines are used to target the renal cell carcinoma 5T4 and
most peptide vaccines developed thus far consist of short- MUC1 antigens [83, 84]. Recombinant adenoviruses are com-
chain peptides (SPs) restricted to MHC class I. Unlike long- monly used in cancer gene therapy because they can transduce
chain peptides (LPs), SPs are able to bind to any cells without dividing and nondividing cells and are easy to produce ([85–
processing and might induce anergy if presented to CD8 T 87], NCT00583024, NCT00197522). Herpes simplex virus
cells without the secondary costimulatory signal [60, 61]. In type 1 (HSV-1), an enveloped dsDNA virus, is used as an
these situations, immune tolerance is induced, creating an oncolytic virus. HSV-1 expressing GM-CSF (e.g., OncoV-
environment favorable for cancer progression. Furthermore, EXGM-CSF) is useful in melanoma [88, 89]. The disadvantage
MHC class I-restricted SPs cannot contribute to the activa- of viral vectors is that repeated administration might be diffi-
tion of MHC class II-restricted helper T cells, which are cult due to the induction of antiviral immune responses. For
important for efficient cytotoxic T lymphocyte (CTL) induc- this reason, a heterologous prime-boost strategy is developing.
tion. However, LPs can be processed to both MHC class I- or For example, PROSTVAC, a vaccine consisting of two poxvi-
class II-restricted antigens and presented by professional rus vectors that express tumor-associated antigen prostate-
APCs, but not by other cell types. Professional APCs that specific antigen (PSA) combined with 3 immune-enhancing
present LP-derived antigens can activate CTL or helper T costimulatory molecules collectively designated as TRICOM
cells without inducing anergy by transmitting signals via (LFA-3, ICAM-1, and B7.1), is under development by Bavar-
both T cell receptors (TCRs) and costimulatory molecules ian Nordic (Denmark) to stimulate an immune response in
[60, 62–64]. Currently, the development of LP vaccines that prostate cancer [90–92]. The results of the PROSTVAC-
contain epitopes for both CTL and helper T cells is being VF/TRICOM phase 3 trial were not as expected, but this vac-
actively pursued. In addition, a novel method with an immu- cine shows promise when combined with immune checkpoint
Journal of Immunology Research 5

inhibitors (NCT02933255, NCT02506114). In addition to approach. For TIL therapy, T cells that recognize cancer-
viruses, bacteria and yeasts are also attracting attention as specific antigens are collected from tumor tissues in patients
new vaccine carriers [93, 94]. with cancer, artificially reactivated by using T cell-
stimulating agents, such as a high IL-2 concentration, and
4.4. Combination Therapy with Cancer Vaccines. Until are then returned to the patients. This approach is potentially
recently, monotherapies using cancer vaccines often had min- simple because genetic modifications are not required, but
imal clinical effects except for certain specific cancer types. the clinical effects might be dependent on the amount and
The relatively low efficacy of monotherapies was attributed quality of infiltrating lymphocytes collected from tumor tis-
to the multifaceted immune evasion mechanisms of cancer, sues. Dudley et al. [103] have reported much information
which are difficult to control by either cancer vaccine alone. on TIL therapy in patients with cancer. Many researchers
As described earlier, the immunosuppressive TME [95] may and doctors are fascinated by their reports on the potential
override any antitumor effects elicited by the cancer vaccine. of TIL therapy in melanoma [104–106]. In the latest method,
In accordance with the development of various immuno- IL-2 and TILs are simultaneously administered to patients to
therapy types, more attention has focused on combination enhance clinical effects [107–109]. In addition, the same
therapies. Several different approaches, including conven- group has conducted similar TIL studies for advanced cervi-
tional chemotherapy/radiation therapy or the latest antibody cal cancer with potential success [110].
therapies [96, 97], have been attempted in combination
either simultaneously or in sequence with immunotherapies. 5.2. TCR/CAR-T Cell Therapy. The availability and perfor-
mance of TIL therapy might be dependent on whether suffi-
4.5. Issues in the Clinical Development of Cancer Vaccines. cient numbers of high-quality antigen-responsive T cells can
During the development of many conventional cancer vac- be secured from tumor tissues in individual patients. To cir-
cines for clinical use, the test designs may have been flawed. cumvent such limitations, peripheral blood mononuclear
For example, clinical effects of cancer vaccines were often cell- (PBMC-) derived lymphocytes, which artificially express
evaluated in patients with a terminal diagnosis whose a desired TCR or chimeric antigen receptor (CAR), have
immune conditions were already substantially compromised been devised and clinically applied as a novel T cell therapy.
by exposure to several other treatments, such as surgery and TCR-T therapy is a therapeutic method using T cells trans-
chemotherapy/radiotherapy, or by progression of the disease. duced with antigen-specific TCRs [111]. CAR-T therapy is
In addition, it is essential to develop accompanying tech- a method of administering T cells with a CAR gene, which
nologies such as adjuvants, manufacturing, and delivery is composed of a fragment derived from a cancer antigen-
methods to employ vaccines and to obtain expected results recognizing antibody gene, gene fragments from intracellular
in clinical settings [98, 99]. The introduction of such state- TCR domains, and other T cell costimulatory molecules.
of-the-art technology may create additional hurdles due to CAR-T therapy is extremely effective in blood cancers
current drug regulations, which are controlled by regulatory [112]. In 2017, the FDA approved CD19 CAR-T therapy
authorities in individual countries, including the FDA, Euro- for B cell acute lymphoblastic leukemia (ALL) which has
pean Medicines Agency (EMA), and Ministry of Health, become refractory to first-line treatment or has recurred
Welfare, and Labour in Japan [100, 101]. These hurdles are more than once [113, 114]. Although CAR-T therapy is
especially important in the development of drugs related to highly effective, the issues surrounding the cost of care with
cancer immunotherapy, where it can be difficult to set up a CAR-T therapy (more than $500,000 for one administration)
primary endpoint to evaluate the effectiveness of a therapy. have yet to be resolved. Notably, CAR-T is effective when a
Thus far, some cytotoxic drugs under investigation have been tumor antigen is monolithic or tumor tissue heterogeneity
known to generally prolong disease-free survival (DFS), but is low (e.g., a genetically uniform tumor, which is often the
not overall survival (OS), but drugs that extend OS without case with blood tumors). However, the efficacy of CAR-T
improving DFS are not very common. Nevertheless, regard- therapy to solid tumors remains limited, because these
ing immunotherapy approaches, a situation often arises tumors generally show more heterogeneity [115].
where OS is extended even if tumor reduction is not observed Another problem with TCR/CAR-T cell therapy in solid
[102]. In addition, as the immune status of individual tumors is the suppressive TME, which inhibits effective infil-
patients may be affected by various factors such as age and tration and/or accumulation of administered T cells inside
past treatment history, it is difficult to adequately predict the tumors. Therefore, a method for effectively driving infil-
the antitumor effects in nonclinical studies. tration of the genetically modified cells into the TME remains
out of reach. Moreover, since target antigens are not uni-
5. The Current Status of Other formly expressed in solid tumors and are different depending
Cancer Immunotherapies on the cancer type, stage, and patient, current TCR/CAR-T
cell therapy is not widely used in patients with cancer. Fur-
In addition to cancer vaccines, other types of cancer immu- thermore, commonly available target antigens similar to
notherapies are in development. These have been described CD19 in blood cancers remain to be identified in solid can-
below. cers [116].

5.1. Tumor-Infiltrating Lymphocyte (TIL) Therapy. When 5.3. Immune Checkpoint Inhibitors. In the 1990s, immune
tumor tissues are available, TIL therapy is a promising checkpoint molecules, including cytotoxic T-lymphocyte-
6 Journal of Immunology Research

Table 1: A list of currently approved cancer immunotherapies.

FDA/EMA MHLW (Japan)


Melanoma Melanoma
non-small cell lung cancer non-small cell lung cancer
renal cell carcinoma renal cell carcinoma
Hodgkin's lymphoma Hodgkin's lymphoma
Nivolumab (Anti-PD-1 Ab) Head neck cancer Head neck cancer
MSI-H/dMMR colorectal cancer gastric cancer
hepatocellular carcinoma diffuse malignant pleural mesothelioma
small cell lung cancer Esophageal cancer
MSI-high colorectal cancer
Melanoma Melanoma
non-small cell lung cancer non-small cell lung cancer
Head neck cancer Urothelial cancer
Hodgkin's lymphoma MSI-high solid tumor
Urothelial cancer renal cell carcinoma ∗ (combination)
MSI-high colorectal cancer Head neck cancer ∗∗ (mono/combination)
Pembrolizumab (Anti-PD-1 Ab) MSI-high cancer
gastric cancer
cervical cancer
hepatocellular carcinoma
Merkel cell carcinoma
renal cell carcinoma
endometrial cancer
Merkel cell carcinoma Merkel cell carcinoma
Avelmab (Anti-PD-L1 Ab) renal cell carcinoma renal cell carcinoma ∗ (combination)
Urothelial cancer
Urothelial cancer non-small cell lung cancer
non-small cell lung cancer extensive-disease small cell lung cancer.
Atezolizumab (Anti-PD-L1 Ab)
breast cencer triple negative breast cancer
small cell lung cancer
Urothelial cancer non-small cell lung cancer (stage 3)
Durvalumab (Anti-PD-L1 Ab)
non-small cell lung cancer
Melanoma Melanoma ∗∗∗ (mono/combination)
Ipilimumab (Anti-CTLA4 Ab) renal cell carcinoma renal cell carcinoma ∗∗∗∗ (combination)
MSI-H/dMMR colorectal cancer
B-ALL (<25 yars-old) B-ALL (<25 yars-old)
Kymriah (CAR-T)
DLBCL (Hodgkin's lymphoma)
Yescarta (CAR-T) DLBCL (Hodgkin's lymphoma) not approved
Sipuleucel-T (Provenge) (DC-vaccine) Prostate cancer not approved
∗ ∗∗ ∗∗∗
Combination with axitinib, monotherapy or combination with chemotherapy, monotherapy or combination with nivolumab, ∗∗∗∗ combination with
nivolumab. MSI-H/dMMR: microsatellite instability-high/deficient mismatch repair; B-ALL: B cell acute lymphoblastic leukemia; DLBCL: diffuse large B
cell lymphoma; Ab: antibody; PD-1: programmed death-1; PD-L1: programmed death ligand-1; CAR: chimeric antigen receptor; DC: dendritic cell; FDA:
Food and Drug Administration; EMA: European Medicines Agency; MHLW: Ministry of Health, Labour and Welfare.

associated protein 4 (CTLA-4) [117] and programmed death and prevent autoimmune responses. The activity of T cells
1 (PD-1) [118], were discovered. Both molecules suppress T is suppressed by ligands binding to CTLA-4 and PD-1. For
cell activation, which is important for exerting antitumor example, the costimulatory molecule CD80/CD86 expressed
effects. The expression of these molecules increases in pro- on APCs enhances T cell activation by simultaneously bind-
portion to T cell activation, and they function as a defense ing to CD28 on T cells during antigen presentation. In con-
system for organisms to inhibit excessive T cell activation trast, CTLA-4 is induced on activated T cells and inhibits
Journal of Immunology Research 7

Hot tumor
Type A

Cold tumor

Cancer
cell Dendritic
cell

B cell

T cell

Macrophage
(inhibitory)

Tertiary lymphoid
structures (TLS)
Type B

Figure 2: Classification of tumors by immune cell infiltration. Tumor types can be classified by the level of immune cell infiltration into
tumors. “Cold tumor,” characterized by the poor infiltration of immune cells, is reported to be one of the reasons why immune
checkpoint inhibitors are ineffective. Contrastingly, a “hot tumor” is characterized by the abundant infiltration of immunocompetent cells,
showing good responses to immune checkpoint inhibitors. Recently, aggregated infiltration of immune cells, known as the tertiary
lymphoid structure (hot tumor, type B), has gained increasing attention compared to separated infiltration of immune cells (hot tumor,
type A).

the costimulatory signal mediated by CD80/CD86 through approved immune checkpoint inhibitors and their indica-
competition with CD28. Additionally, antigen-stimulated T tions. These immune checkpoint inhibitors often show only
cells express PD-1, which suppresses excessive T cell activa- limited and/or transient efficacy, reflecting the complexities
tion by binding to its ligand, PD-L1 or programmed death- of antitumor immunity [129, 130]. For example, immune
ligand 2 (PD-L2) [119]. Additional constituents of the cancer checkpoint inhibitors are less effective in microsatellite stable
microenvironment, such as DCs [120], macrophages [121], tumors [131]. Such tumors often express only minor genetic
and fibroblasts [122], can also express PD-L1 and/or PD- abnormalities and thus possess little antigenic capacity.
L2, forming an immunosuppressive environment where can- Therefore, these tumors fail to induce cancer antigen-
cer is more prone to progress. specific T cells, resulting in unresponsiveness to immune
In addition to evading the immune system by deleting checkpoint inhibitors. In addition, the immunosuppressive
cell surface molecules required for antigen recognition, can- TME might also affect the clinical effects of immune check-
cer cells can often directly use (e.g., hijack) the immunosup- point inhibitors.
pression system, such as immune checkpoints. Some cancer
cells strongly express ligands for immune checkpoint mole-
cules, such as PD-L1 and PD-L2 [123, 124]. A remarkable 6. Challenges for the Future Development of
antitumor effect can thus be observed in some patients by Cancer Immunotherapies: Requirement of
administering treatments that block inhibitory molecules in Lymphocyte Infiltration/Accumulation
T cells [125, 126]. Once antitumor activity is induced and within Tumors
immune memory is established by immune checkpoint
inhibitors in patients with cancer, their clinical effects should Analysis of the interaction among tumor infiltrating immune
be long-lasting. cells (e.g., DCs, MDSCs, CD4/8T cells, and Tregs), stromal
In 2011, both the FDA and EMA approved anti-CTLA-4 cells, and tumor cells is essential to understand the relation-
antibody treatment (ipilimumab) for patients with mela- ship between the TME and the clinical effects of cancer
noma [127, 128]. Since then, several immune checkpoint immunotherapies. Especially, many clinical trials using can-
inhibitors, anti-PD-1, and anti-PD-L1 antibodies have been cer immunotherapies indicate that TIL abundance in the
approved every year. Table 1 shows several currently tumor is an important prognostic factor [132]. For example,
8 Journal of Immunology Research

the magnitude of lymphocyte infiltration to tumors signifi- develop novel approaches, including cancer vaccines. We
cantly contributes to immune checkpoint inhibitor effective- hope that this review will facilitate the further development
ness [133, 134]. The effectiveness of immune checkpoint of cancer immunotherapies.
inhibitors is high against inflammatory-type tumors (hot
tumors) [135, 136], where immunocompetent cells are suffi- Conflicts of Interest
ciently present. In contrast, in immune desert-type tumors
(cold tumors) with less intratumoral lymphocyte infiltration The authors declare that there is no conflict of interest
[137], immune checkpoint inhibitors are less effective. regarding the publication of this paper.
The difference between these tumor types might depend
on whether the immune responses to tumors are maintained Acknowledgments
or not, suggesting that the recruitment/accumulation of
tumor-specific T cells is the limiting factor for therapeutic This research was supported by JSPS KAKENHI Grant
effects. Treatment could thus be optimized by the develop- Number 19K09110 and AMED under Grant Number
ment of techniques to increase lymphocyte infiltration into JP20ae0101076.
tumors either before or during immunotherapy treatment
(Figure 2). References
For the development of more effective CAR-T cell thera-
pies, modification with cytokine/chemokine-related genes [1] WHO, Assessment reports of the Global Vaccine Action Plan,
was recently reported to efficiently drive infiltration of World Health Organization, Geneva, 2018, https://apps.who
.int/iris/bitstream/handle/10665/276967/WHO-IVB-18.11-
administered CAR-T cells into tumors [138]. Nevertheless,
eng.pdf?ua=1.
few studies are available on the development of methods for
[2] “Immunology and Vaccine-Preventable Diseases,” in Princi-
changing cold tumors to hot tumors. Thus, more studies ples of VaccinationPink Bookhttps://www.cdc.gov/vaccines/
are needed to develop a method to efficiently recruit/accu- Pubs/pinkbook/downloads/prinvac.pdf.
mulate lymphocytes within tumors in combination with [3] F. Sallusto, A. Lanzavecchia, K. Araki, and R. Ahmed, “From
immunotherapeutic approaches, including cancer vaccines vaccines to memory and back,” Immunity, vol. 33, no. 4,
to stimulate antigen-specific immunocompetent cells, pp. 451–463, 2010.
antigen-specific cytotoxic T cell therapies using ex vivo [4] WHO, Vaccines and immunizationWorld Health Organiza-
genetic modification, or blockade of inhibitory signals from tionhttps://www.who.int/health-topics/vaccines-and-
the tumor. immunization#tab=tab_1.
[5] R. R. Rich, “The human immune response,” in Clinical
6.1. Induction of Tertiary Lymphoid Structures (TLS). As Immunology, pp. 3–17, 2019.
shown in “hot tumor (type B)” in Figure 2, the accumulation [6] R. S. Goldszmid, A. Dzutsev, and G. Trinchieri, “Host
of various immune cell types, especially the formation of immune response to infection and cancer: unexpected com-
lymphoid follicles where immunocompetent cells can monalities,” Cell Host & Microbe, vol. 15, no. 3, pp. 295–
exchange information in the tumor, may be important 305, 2014.
[139, 140]. Indeed, lymphoid follicles in tumors, known as [7] A. D. Waldman, J. M. Fritz, and M. J. Lenardo, “A guide to
TLS, are associated with better prognosis in several cancers cancer immunotherapy: from T cell basic science to clinical
and have recently attracted attention. Several TLS-inducing practice,” Nature Reviews Immunology, pp. 1–18, 2020.
factors have been reported, including CCL19, CCL21, [8] K. Esfahani, L. Roudaia, N. Buhlaiga, S. del Rincon,
CXCL12, CXCL13, LIGHT, and lymphotoxin [141–144]. N. Papneja, and W. H. Miller Jr, “A review of cancer immu-
While some basic studies for inducing lymphoid follicles notherapy: from the past, to the present, to the future,” Cur-
have been conducted in mice, there are no promising rent Oncology, vol. 27, Supplement 2, pp. S87–S97, 2020.
methods that are clinically applicable for human therapy. [9] P. S. Hegde and D. S. Chen, “Top 10 challenges in cancer
Since efforts so far have been generally focused on a single immunotherapy,” Immunity, vol. 52, no. 1, pp. 17–35, 2020.
factor, they might fail to reproduce an induction of TLS [10] W. B. Coley, “II. Contribution to the knowledge of sarcoma,”
due to the complicated mechanisms within the TME. A novel Annals of Surgery, vol. 14, no. 3, pp. 199–220, 1891.
method to efficiently induce TLS in humans, combined with [11] D. W. Cramer, A. F. Vitonis, S. P. Pinheiro et al., “Mumps
and ovarian cancer: modern interpretation of an historic
other immunotherapies, such as cancer vaccines and
association,” Cancer Causes & Control, vol. 21, no. 8,
immune checkpoint inhibitors, may be a promising approach
pp. 1193–1201, 2010.
for tumor control.
[12] D. E. Morales, D. Eidinger, and A. W. Bruce, “Intracavitary
Bacillus Calmette-Guerin in the treatment of superficial blad-
7. Conclusions der tumors,” Journal of Urology, vol. 116, no. 2, pp. 180–182,
1976.
The clinical application of immune checkpoint inhibitors has [13] M. Kamat, J. Bellmunt, M. D. Galsky et al., “Society for
greatly advanced cancer treatment. However, their effects are immunotherapy of cancer consensus statement on immuno-
limited because tumor cells use various mechanisms to evade therapy for the treatment of bladder carcinoma,” Journal for
antitumor effects. To overcome these mechanisms and to ImmunoTherapy of Cancer, vol. 5, no. 1, p. 68, 2017.
improve the versatility of current cancer immunotherapies, [14] A. R. Garbuglia, D. Lapa, C. Sias, M. R. Capobianchi, and
it is necessary to understand the TME in more detail and P. del Porto, “The use of both therapeutic and prophylactic
Journal of Immunology Research 9

vaccines in the therapy of papillomavirus disease,” Frontiers [28] A. Baars, J. M. G. H. van Riel, M. A. Cuesta, E. H. Jaspars,
in Immunology, vol. 11. H. M. Pinedo, and A. J. M. van den Eertwegh, “Metastasect-
[15] M. H. Nguyen, G. Wong, E. Gane, J. H. Kao, and omy and active specific immunotherapy for a large single
G. Dusheiko, “Hepatitis B virus: advances in prevention, melanoma metastasis,” Hepato-Gastroenterology, vol. 49,
diagnosis, and therapy,” Clinical Microbiology Reviews, no. 45, pp. 691–693, 2002.
vol. 33, no. 2, 2020. [29] D. Berd, H. C. Maguire, P. McCue, and M. J. Mastrangelo,
[16] G. P. Dunn, A. T. Bruce, H. Ikeda, L. J. Old, and R. D. “Treatment of metastatic melanoma with an autologous
Schreiber, “Cancer immunoediting: from immunosurveil- tumor-cell vaccine: clinical and immunologic results in 64
lance to tumor escape,” Nature Immunology, vol. 3, patients,” Journal of Clinical Oncology, vol. 8, no. 11,
no. 11, pp. 991–998, 2002. pp. 1858–1867, 1990.
[17] L. Chen and D. B. Flies, “Molecular mechanisms of T cell co- [30] R. Méndez, F. Ruiz-Cabello, T. Rodríguez et al., “Identifica-
stimulation and co-inhibition,” Nature Reviews Immunology, tion of different tumor escape mechanisms in several metas-
vol. 13, no. 4, pp. 227–242, 2013. tases from a melanoma patient undergoing
immunotherapy,” Cancer Immunology Immunotherapy,
[18] D. Lindau, P. Gielen, M. Kroesen, P. Wesseling, and G. J.
vol. 56, no. 1, pp. 88–94, 2007.
Adema, “The immunosuppressive tumour network:
myeloid-derived suppressor cells, regulatory T cells and nat- [31] S. J. Antonia, J. Seigne, J. Diaz et al., “Phase I trial of a B7-1
ural killer T cells,” Immunology, vol. 138, no. 2, pp. 105– (CD80) gene modified autologous tumor cell vaccine in com-
115, 2013. bination with systemic interleukin-2 in patients with meta-
static renal cell carcinoma,” Journal of Urology, vol. 167,
[19] M. Berger, F. T. Kreutz, J. L. Horst, A. C. Baldi, and W. J. Koff, no. 5, pp. 1995–2000, 2002.
“Phase I study with an autologous tumor cell vaccine for
locally advanced or metastatic prostate cancer,” Journal of [32] M. Fishman, T. B. Hunter, H. Soliman et al., “Phase II trial of
Pharmacy & Pharmaceutical Sciences, vol. 10, no. 2, B7-1 (CD-86) transduced, cultured autologous tumor cell
pp. 144–152, 2007. vaccine plus subcutaneous interleukin-2 for treatment of
stage IV renal cell carcinoma,” Journal of Immunotherapy,
[20] J. E. Harris, L. Ryan, H. C. Hoover et al., “Adjuvant active spe- vol. 31, no. 1, pp. 72–80, 2008.
cific immunotherapy for stage II and III colon cancer with an
[33] Y. Kinoshita, T. Kono, R. Yasumoto et al., “Antitumor effect
autologous tumor cell vaccine: Eastern Cooperative Oncol-
on murine renal cell carcinoma by autologous tumor vaccines
ogy Group Study E5283,” Journal of Clinical Oncology,
genetically modified with granulocyte-macrophage colony-
vol. 18, no. 1, pp. 148–157, 2000.
stimulating factor and interleukin-6 cells,” Journal of Immu-
[21] C. Maver and M. McKneally, “Preparation of autologous notherapy, vol. 24, no. 3, pp. 205–211, 2001.
tumor cell vaccine from human lung cancer,” Cancer
[34] K. Tani, M. Azuma, Y. Nakazaki et al., “Phase I study of
Research, vol. 39, no. 8, 1979.
autologous tumor vaccines transduced with the GM-CSF
[22] R. S. Schulof, D. Mai, M. A. Nelson et al., “Active specific gene in four patients with stage IV renal cell cancer in Japan:
immunotherapy with an autologous tumor cell vaccine in clinical and immunological findings,” Molecular Therapy,
patients with resected non-small cell lung cancer,” Molecular vol. 10, no. 4, pp. 799–816, 2004.
Biotherapy, vol. 1, no. 1, pp. 30–36, 1988. [35] H. Asada, T. Kishida, H. Hirai et al., “Significant antitumor
[23] J. Nemunaitis and J. Nemunaitis, “Granulocyte-macrophage effects obtained by autologous tumor cell vaccine engineered
colony-stimulating factor gene-transfected autologous tumor to secrete interleukin (IL)-12 and IL-18 by means of the
cell vaccine: focus on non-small-cell lung cancer,” Clinical EBV/lipoplex,” Molecular Therapy, vol. 5, no. 5, pp. 609–
Lung Cancer, vol. 5, no. 3, pp. 148–157, 2003. 616, 2002.
[24] D. Rüttinger, N. K. van den Engel, H. Winter et al., “Adjuvant [36] G. Dranoff, E. Jaffee, A. Lazenby et al., “Vaccination with irra-
therapeutic vaccination in patients with non-small cell lung diated tumor cells engineered to secrete murine granulocyte-
cancer made lymphopenic and reconstituted with autologous macrophage colony-stimulating factor stimulates potent,
PBMC: first clinical experience and evidence of an immune specific, and long-lasting anti-tumor immunity,” Proceedings
response,” Journal of Translational Medicine, vol. 5, no. 1, of the National Academy of Sciences of the United States of
p. 43, 2007. America, vol. 90, no. 8, pp. 3539–3543, 1993.
[25] V. A. de Weger, A. W. Turksma, Q. J. M. Voorham et al., [37] N. Mach, S. Gillessen, S. B. Wilson, C. Sheehan, M. Mihm,
“Clinical effects of adjuvant active specific immunotherapy and G. Dranoff, “Differences in dendritic cells stimulated
differ between patients with microsatellite-stable and in vivo by tumors engineered to secrete granulocyte-
microsatellite-instable colon cancer,” Clinical Cancer macrophage colony-stimulating factor or Flt3-ligand,” Can-
Research, vol. 18, no. 3, pp. 882–889, 2012. cer Research, vol. 60, no. 12, pp. 3239–3246, 2000.
[26] M. G. Hanna, H. C. Hoover, J. B. Vermorken, J. E. Harris, [38] R. Salgia, T. Lynch, A. Skarin et al., “Vaccination with irradi-
and H. M. Pinedo, “Adjuvant active specific immunother- ated autologous tumor cells engineered to secrete
apy of stage II and stage III colon cancer with an autolo- granulocyte-macrophage colony-stimulating factor aug-
gous tumor cell vaccine: first randomized phase III trials ments antitumor immunity in some patients with metastatic
show promise,” Vaccine, vol. 19, no. 17–19, pp. 2576– non-small-cell lung carcinoma,” Journal of Clinical Oncology,
2582, 2001. vol. 21, no. 4, pp. 624–630, 2003.
[27] D. Ockert, V. Schirrmacher, N. Beck et al., “Newcastle disease [39] E. M. Jaffee, R. H. Hruban, B. Biedrzycki et al., “Novel alloge-
virus-infected intact autologous tumor cell vaccine for adju- neic granulocyte-macrophage colony-stimulating factor-
vant active specific immunotherapy of resected colorectal secreting tumor vaccine for pancreatic cancer: a phase I trial
carcinoma,” Clinical Cancer Research, vol. 2, no. 1, pp. 21– of safety and immune activation,” Journal of Clinical Oncol-
28, 1996. ogy, vol. 19, no. 1, pp. 145–156, 2001.
10 Journal of Immunology Research

[40] J. Nemunaitis, T. Jahan, H. Ross et al., “Phase 1/2 trial of [54] L. C. Tsoi, B. Wolf, and Y. A. Chen, “The promise of genomic
autologous tumor mixed with an allogeneic GVAX® vaccine studies on human diseases: from basic science to clinical
in advanced-stage non-small-cell lung cancer,” Cancer Gene application,” International Journal of Genomics, vol. 2017,
Therapy, vol. 13, no. 6, pp. 555–562, 2006. Article ID 7983236, 2 pages, 2017.
[41] P. M. Arlen, M. Mohebtash, R. A. Madan, and J. L. Gulley, [55] V. Roudko, B. Greenbaum, and N. Bhardwaj, “Computa-
“Promising novel immunotherapies and combinations for tional prediction and validation of tumor-associated neoanti-
prostate cancer,” Future Oncology, vol. 5, no. 2, pp. 187– gens,” Frontiers in Immunology, vol. 11, p. 27, 2020.
196, 2009. [56] L. Buonaguro, A. Petrizzo, M. L. Tornesello, and F. M. Buo-
[42] D. L. Morton, L. J. Foshag, D. S. B. Hoon et al., “Prolongation naguro, “Translating tumor antigens into cancer vaccines,”
of Survival in Metastatic Melanoma after Active Specific Clinical & Vaccine Immunology, vol. 18, no. 1, pp. 23–34,
Immunotherapy With a New Polyvalent Melanoma Vac- 2011.
cine,” Annals of Surgery, vol. 216, no. 4, pp. 463–482, 1992. [57] I. Melero, G. Gaudernack, W. Gerritsen et al., “Therapeutic
[43] J. W. Simons, “Phase I/II trial of an allogeneic cellular immu- vaccines for cancer: an overview of clinical trials,” Nature
notherapy in Hormone-Naive prostate cancer,” Clinical Can- Reviews Clinical Oncology, vol. 11, no. 9, pp. 509–524, 2014.
cer Research, vol. 12, no. 11, pp. 3394–3401, 2006. [58] C. J. M. Melief and S. H. van der Burg, “Immunotherapy of
[44] E. J. Small, N. Sacks, J. Nemunaitis et al., “Granulocyte mac- established (pre)malignant disease by synthetic long peptide
rophage colony-stimulating Factor-Secreting allogeneic cel- vaccines,” Nature Reviews Cancer, vol. 8, no. 5, pp. 351–
lular immunotherapy for hormone-refractory prostate 360, 2008.
cancer,” Clinical Cancer Research, vol. 13, no. 13, pp. 3883– [59] F. M. Marincola, E. M. Jaffee, D. J. Hicklin, and S. Ferrone,
3891, 2007. “Escape of human solid tumors from T-cell recognition:
[45] L. A. Emens, J. M. Asquith, J. M. Leatherman et al., “Timed molecular mechanisms and functional significance,”
sequential treatment with cyclophosphamide, doxorubicin, Advances in Immunology, vol. 74, pp. 181–273, 1999.
and an allogeneic granulocyte-macrophage colony- [60] M. S. Bijker, S. J. F. van den Eeden, K. L. Franken, C. J. M.
stimulating factor-secreting breast tumor vaccine: a chemo- Melief, S. H. van der Burg, and R. Offringa, “Superior induc-
therapy dose-ranging factorial study of safety and immune tion of anti-tumor CTL immunity by extended peptide vac-
activation,” Journal of Clinical Oncology, vol. 27, no. 35, cines involves prolonged, DC-focused antigen
pp. 5911–5918, 2009. presentation,” European Journal of Immunology, vol. 38,
[46] E. Lutz, C. J. Yeo, K. D. Lillemoe et al., “A lethally irradiated no. 4, pp. 1033–1042, 2008.
allogeneic granulocyte-macrophage colony stimulating [61] Y. Hailemichael, Z. Dai, N. Jaffarzad et al., “Persistent antigen
factor-secreting tumor vaccine for pancreatic adenocarci- at vaccination sites induces tumor-specific CD8+ T cell
noma. A phase II trial of safety, efficacy, and immune activa- sequestration, dysfunction and deletion,” Nature Medicine,
tion,” Annals of Surgery, vol. 253, no. 2, pp. 328–335, 2011. vol. 19, no. 4, pp. 465–472, 2013.
[47] A. J. M. van den Eertwegh, J. Versluis, H. P. van den Berg [62] M. S. Bijker, S. J. F. van den Eeden, K. L. Franken, C. J. M.
et al., “Combined immunotherapy with granulocyte- Melief, R. Offringa, and S. H. van der Burg, “CD8+ CTL
macrophage colony-stimulating factor-transduced allogeneic Priming by Exact Peptide Epitopes in Incomplete Freund’s
prostate cancer cells and ipilimumab in patients with meta- Adjuvant Induces a Vanishing CTL Response, whereas Long
static castration-resistant prostate cancer: a phase 1 dose- Peptides Induce Sustained CTL Reactivity,” The Journal of
escalation trial,” Lancet Oncology, vol. 13, no. 5, pp. 509– Immunology, vol. 179, no. 8, pp. 5033–5040, 2007.
517, 2012. [63] E. M. Janssen, N. M. Droin, E. E. Lemmens et al., “CD4+ T-
[48] X.-Y. Wang, D. Zuo, D. Sarkar, and P. B. Fisher, “Blockade of cell help controls CD8+ T-cell memory via TRAIL-
cytotoxic T-lymphocyte antigen-4 as a new therapeutic mediated activation-induced cell death,” Nature, vol. 434,
approach for advanced melanoma,” Expert Opinion on Phar- no. 7029, pp. 88–93, 2005.
macotherapy, vol. 12, no. 17, pp. 2695–2706, 2011. [64] R. A. Rosalia, E. D. Quakkelaar, A. Redeker et al., “Dendritic
[49] F. O. Nestle, S. Alijagic, M. Gilliet et al., “Vaccination of mel- cells process synthetic long peptides better than whole pro-
anoma patients with peptide- or tumorlysate-pulsed den- tein, improving antigen presentation and T-cell activation,”
dritic cells,” Nature Medicine, vol. 4, no. 3, pp. 328–332, 1998. European Journal of Immunology, vol. 43, no. 10, pp. 2554–
[50] R. M. Steinman and J. Banchereau, “Taking dendritic cells 2565, 2013.
into medicine,” Nature, vol. 449, no. 7161, pp. 419–426, [65] C. G. Drake, E. J. Lipson, and J. R. Brahmer, “Breathing new
2007. life into immunotherapy: review of melanoma, lung and kid-
[51] D. Li, G. Romain, A.-L. Flamar et al., “Targeting self- and for- ney cancer,” Nature Reviews Clinical Oncology, vol. 11, no. 1,
eign antigens to dendritic cells via DC-ASGPR generates IL- pp. 24–37, 2014.
10-producing suppressive CD4+ T cells,” Journal of Experi- [66] L. Liu, Q. Chen, C. Ruan et al., “Platinum-based nanovectors
mental Medicine, vol. 209, no. 1, pp. 109–121, 2012. engineered with Immuno-modulating adjuvant for inhibiting
[52] E. J. Small, P. Fratesi, D. M. Reese et al., “Immunotherapy of tumor growth and promoting immunity,” Theranostics,
hormone-refractory prostate cancer with antigen-loaded vol. 8, no. 11, pp. 2974–2987, 2018.
dendritic cells,” Journal of Clinical Oncology, vol. 18, no. 23, [67] B. J. Hos, E. Tondini, S. I. van Kasteren, and F. Ossendorp,
pp. 3894–3903, 2000. “Approaches to improve chemically defined synthetic pep-
[53] N. Meena, P. Mathur, K. M. Medicherla, and P. Suravajhala, tide vaccines,” Frontiers in Immunology, vol. 9, pp. 884–891,
“A bioinformatics pipeline for whole exome sequencing: 2018.
overview of the processing and steps from raw data to down- [68] E. M. Varypataki, N. Benne, J. Bouwstra, W. Jiskoot, and
stream analysis,” bioRxiv. F. Ossendorp, “Efficient eradication of established tumors in
Journal of Immunology Research 11

mice with cationic liposome-based synthetic long-peptide randomized, double-blind, placebo-controlled phase III
vaccines,” Cancer Immunology Research, vol. 5, no. 3, study,” Clinical Cancer Research, vol. 16, no. 22, pp. 5539–
pp. 222–233, 2017. 5547, 2010.
[69] L. Aurisicchio and G. Ciliberto, “Genetic cancer vaccines: [84] S. Oudard, O. Rixe, B. Beuselinck et al., “A phase II study of
current status and perspectives,” Expert Opinion on Biological the cancer vaccine TG4010 alone and in combination with
Therapy, vol. 12, no. 8, pp. 1043–1058, 2012. cytokines in patients with metastatic renal clear-cell carci-
[70] S. H. T. Jorritsma, E. J. Gowans, B. Grubor-Bauk, and D. K. noma: clinical and immunological findings,” Cancer Immu-
Wijesundara, “Delivery methods to increase cellular uptake nology, Immunotherapy, vol. 60, no. 2, pp. 261–271, 2011.
and immunogenicity of DNA vaccines,” Vaccine, vol. 34, [85] S. K. Das, S. Sarkar, R. Dash et al., “Cancer terminator viruses
no. 46, pp. 5488–5494, 2016. and approaches for enhancing therapeutic outcomes,”
[71] B. Ferraro, M. P. Morrow, N. A. Hutnick, T. H. Shin, C. E. Advances in Cancer Research, vol. 115, pp. 1–38, 2012.
Lucke, and D. B. Weiner, “Clinical applications of DNA vac- [86] T.-C. Liu, T.-H. Hwang, J. C. Bell, and D. H. Kirn, “Transla-
cines: current progress,” Clinical Infectious Diseases, vol. 53, tion of targeted oncolytic virotherapeutics from the lab into
no. 3, pp. 296–302, 2011. the clinic, and back again: a high-value iterative loop,” Molec-
[72] N. Y. Sardesai and D. B. Weiner, “Electroporation delivery of ular Therapy, vol. 16, no. 6, pp. 1006–1008, 2008.
DNA vaccines: prospects for success,” Current Opinion in [87] J. Raty, J. Pikkarainen, T. Wirth, and S. Yla-Herttuala, “Gene
Immunology, vol. 23, no. 3, pp. 421–429, 2011. therapy: the first approved gene-based medicines, molecular
[73] C. L. Trimble, M. P. Morrow, K. A. Kraynyak et al., “Safety, mechanisms and clinical indications,” Current Molecular
efficacy, and immunogenicity of VGX-3100, a therapeutic syn- Pharmacology, vol. 1, no. 1, pp. 13–23, 2008.
thetic DNA vaccine targeting human papillomavirus 16 and 18 [88] N. N. Senzer, H. L. Kaufman, T. Amatruda et al., “Phase II
E6 and E7 proteins for cervical intraepithelial neoplasia 2/3: a clinical trial of a granulocyte-macrophage colony-
randomised, double-blind, placebo-controlled phase 2b trial,” stimulating factor-encoding, second-generation oncolytic
Lancet, vol. 386, no. 10008, pp. 2078–2088, 2015. herpesvirus in patients with unresectable metastatic mela-
[74] S. Espuelas, A. Roth, C. Thumann, B. Frisch, and F. Schuber, noma,” Journal of Clinical Oncology, vol. 27, no. 34,
“Effect of synthetic lipopeptides formulated in liposomes on pp. 5763–5771, 2009.
the maturation of human dendritic cells,” Molecular Immu- [89] H. L. Kaufman and S. D. Bines, “OPTIM trial: a phase III trial
nology, vol. 42, no. 6, pp. 721–729, 2005. of an oncolytic herpes virus encoding GM-CSF for unresect-
[75] M. Fotin-Mleczek, K. Zanzinger, R. Heidenreich et al., able stage III or IV melanoma,” Future Oncology, vol. 6, no. 6,
“Highly potent mRNA based cancer vaccines represent an pp. 941–949, 2010.
attractive platform for combination therapies supporting an [90] J. W. Hodge, M. Chakraborty, C. Kudo-Saito, C. T. Garnett,
improved therapeutic effect,” Journal of Gene Medicine, and J. Schlom, “Multiple costimulatory modalities enhance
vol. 14, no. 6, pp. 428–439, 2012. CTL avidity,” Journal of Immunology, vol. 174, no. 10,
[76] P. Qiu, P. Ziegelhoffer, J. Sun, and N. S. Yang, “Gene gun pp. 5994–6004, 2005.
delivery of mRNA in situ results in efficient transgene expres- [91] P. W. Kantoff, T. J. Schuetz, B. A. Blumenstein et al., “Overall
sion and genetic immunization,” Gene Therapy, vol. 3, no. 3, survival analysis of a phase II randomized controlled trial of a
pp. 262–268, 1996. poxviral-based PSA-targeted immunotherapy in metastatic
[77] B. Scheel, R. Teufel, J. Probst et al., “Toll-like receptor- castration-resistant prostate cancer,” Journal of Clinical
dependent activation of several human blood cell types by Oncology, vol. 28, no. 7, pp. 1099–1105, 2010.
protamine-condensed mRNA,” European Journal of Immu- [92] J. L. Gulley, P. M. Arlen, R. A. Madan et al., “Immunologic
nology, vol. 35, no. 5, pp. 1557–1566, 2005. and prognostic factors associated with overall survival
[78] S. Pascolo, “Vaccination with messenger RNA (mRNA),” employing a poxviral-based PSA vaccine in metastatic
Handbook of Experimental Pharmacology, vol. 183, no. 183, castrate-resistant prostate cancer,” Cancer Immunology,
pp. 221–235, 2008. Immunotherapy, vol. 59, no. 5, pp. 663–674, 2010.
[79] B. Weide, J.-P. Carralot, A. Reese et al., “Results of the first [93] C. Remondo, V. Cereda, S. Mostböck et al., “Human den-
phase I/II clinical vaccination trial with direct injection of dritic cell maturation and activation by a heat-killed recombi-
mRNA,” Journal of Immunotherapy, vol. 31, no. 2, pp. 180– nant yeast (Saccharomyces cerevisiae) vector encoding
188, 2008. carcinoembryonic antigen,” Vaccine, vol. 27, no. 7, pp. 987–
[80] B. Weide, S. Pascolo, B. Scheel et al., “Direct injection of 994, 2009.
protamine-protected mRNA: results of a phase 1/2 vaccina- [94] E. K. Wansley, M. Chakraborty, K. W. Hance et al., “Vaccina-
tion trial in metastatic melanoma patients,” Journal of Immu- tion with a recombinant Saccharomyces cerevisiae expressing
notherapy, vol. 32, no. 5, pp. 498–507, 2009. a tumor antigen breaks immune tolerance and elicits thera-
[81] H. Oshiumi, M. Matsumoto, K. Funami, T. Akazawa, and peutic antitumor responses,” Clinical Cancer Research,
T. Seya, “TICAM-1, an adaptor molecule that participates vol. 14, no. 13, pp. 4316–4325, 2008.
in toll-like receptor 3-mediated interferon-beta induction,” [95] W. Zou, “Immunosuppressive networks in the tumour envi-
Nature Immunology, vol. 4, no. 2, pp. 161–167, 2003. ronment and their therapeutic relevance,” Nature Reviews
[82] L. M. Kranz, M. Diken, H. Haas et al., “Systemic RNA deliv- Cancer, vol. 5, no. 4, pp. 263–274, 2005.
ery to dendritic cells exploits antiviral defence for cancer [96] Y. Yan, A. B. Kumar, H. Finnes et al., “Combining immune
immunotherapy,” Nature, vol. 534, no. 7607, pp. 396–401, checkpoint inhibitors with conventional cancer therapy,”
2016. Frontiers in Immunology, vol. 9, 2018.
[83] R. J. Amato, R. E. Hawkins, H. L. Kaufman et al., “Vaccina- [97] W. L. Hwang, L. R. G. Pike, T. J. Royce, B. A. Mahal, and J. S.
tion of metastatic renal cancer patients with MVA-5T4: a Loeffler, “Safety of combining radiotherapy with immune-
12 Journal of Immunology Research

checkpoint inhibition,” Nature Reviews Clinical Oncology, [111] Z. Eshhar, T. Waks, G. Gross, and D. G. Schindler, “Specific
vol. 15, no. 8, pp. 477–494, 2018. activation and targeting of cytotoxic lymphocytes through
[98] A. Bolhassani, S. Safaiyan, and S. Rafati, “Improvement of chimeric single chains consisting of antibody-binding
different vaccine delivery systems for cancer therapy,” Molec- domains and the gamma or zeta subunits of the immuno-
ular Cancer, vol. 10, no. 1, p. 3, 2011. globulin and T-cell receptors,” Proceedings of the National
Academy of Sciences of the United States of America, vol. 90,
[99] R. S. Riley, C. H. June, R. Langer, and M. J. Mitchell, “Delivery
no. 2, pp. 720–724, 1993.
technologies for cancer immunotherapy,” Nature Reviews
Drug Discovery, vol. 18, no. 3, pp. 175–196, 2019. [112] D. L. Porter, B. L. Levine, M. Kalos, A. Bagg, and C. H. June,
“Chimeric antigen receptor-modified T cells in chronic lym-
[100] European Medicines Agency, Guideline on the evaluation of
phoid leukemia,” New England Journal of Medicine, vol. 365,
anticancer medicinal products in man: EMA/CHM-
no. 8, pp. 725–733, 2011.
P/205/95/Rev.4, 2012, http://www.ema.europa.eu/docs/en_
GB/document_library/Scientific_guideline/2013/01/ [113] U.S. Food and Drug Administration FDA approves CAR-T cell
WC500137128.pdf. therapy to treat adults with certain types of large B-cell lym-
phoma, 2017, https://www.fda.gov/newsevents/newsroom/
[101] FDA, Guidance for industry: clinical considerations for thera-
pressannouncements/ucm581216.htm.
peutic cancer vaccines, 2011, http://www.fda.gov/downloads/
B i o l o g i c s B l o o d V a c c i n e s / [114] V. A. Chow, M. Shadman, and A. K. Gopal, “Translating anti-
GuidanceComplianceRegulatoryInformation/Guidances/ CD19 CAR T-cell therapy into clinical practice for relapse-
Vaccines/UCM278673.pdf. d/refractory diffuse large B-cell lymphoma,” Blood, vol. 132,
no. 8, pp. 777–781, 2018.
[102] R. A. Madan, J. L. Gulley, T. Fojo, and W. L. Dahut, “Thera-
peutic cancer vaccines in prostate cancer: the paradox of [115] M. Martinez and E. K. Moon, “CAR T cells for solid tumors:
improved survival without changes in time to progression,” new strategies for finding, infiltrating, and surviving in the
The Oncologist, vol. 15, no. 9, pp. 969–975, 2010. tumor microenvironment,” Frontiers in Immunology,
vol. 10, p. 128, 2019.
[103] M. E. Dudley, J. R. Wunderlich, J. C. Yang et al., “Adoptive
cell transfer therapy following non-myeloablative but lym- [116] A. Schmidts and M. V. Maus, “Making CAR T cells a solid
phodepleting chemotherapy for the treatment of patients option for solid tumors,” Frontiers in Immunology, vol. 9, 2018.
with refractory metastatic melanoma,” Journal of Clinical [117] D. R. Leach, M. F. Lrimmel, and J. P. Allison, “Enhancement
Oncology, vol. 23, no. 10, pp. 2346–2357, 2005. of antitumor immunity by CTLA-4 blockade,” Science,
[104] S. Stevanović, L. M. Draper, M. M. Langhan et al., “Complete vol. 271, no. 5256, pp. 1734–1736, 1996.
regression of metastatic cervical cancer after treatment with [118] Y. Ishida, Y. Agata, K. Shibahara, and T. Honjo, “Induced
human papillomavirus-targeted tumor-infiltrating T cells,” expression of PD-1, a novel member of the immunoglobulin
Journal of Clinical Oncology, vol. 33, no. 14, pp. 1543–1550, gene superfamily, upon programmed cell death,” The EMBO
2015. Journal, vol. 11, no. 11, pp. 3887–3895, 1992.
[105] M. W. Rohaan, J. H. van den Berg, P. Kvistborg, and J. B. A. [119] S. C. Wei, C. R. Duffy, and J. P. Allison, “Fundamental mech-
G. Haanen, “Adoptive transfer of tumor-infiltrating lympho- anisms of immune checkpoint blockade therapy,” Cancer
cytes in melanoma: a viable treatment option,” Journal for Discovery, vol. 8, no. 9, pp. 1069–1086, 2018.
ImmunoTherapy of Cancer, vol. 6, no. 1, p. 102, 2018. [120] T. J. Curiel, S. Wei, H. Dong et al., “Blockade of B7-H1
[106] G. U. Mehta, P. Malekzadeh, T. Shelton et al., “Outcomes of improves myeloid dendritic cell-mediated antitumor immu-
adoptive cell transfer with tumor-infiltrating lymphocytes nity,” Nature Medicine, vol. 9, no. 5, pp. 562–567, 2003.
for metastatic melanoma patients with and without brain [121] K. Wu, I. Kryczek, L. Chen, W. Zou, and T. H. Welling,
metastases,” Journal of Immunotherapy, vol. 41, no. 5, “Kupffer cell suppression of CD8+ T cells in human hepato-
pp. 241–247, 2018. cellular carcinoma is mediated by B7-H1/programmed
[107] M. A. Forget, C. Haymaker, K. R. Hess et al., “Prospective death-1 interactions,” Cancer Research, vol. 69, no. 20,
analysis of adoptive TIL therapy in patients with metastatic pp. 8067–8075, 2009.
melanoma: response, impact of anti-CTLA4, and biomarkers [122] M. R. Nazareth, L. Broderick, M. R. Simpson-Abelson, R. J.
to predict clinical outcome,” Clinical Cancer Research, vol. 24, Kelleher Jr., S. J. Yokota, and R. B. Bankert, “Characterization
no. 18, pp. 4416–4428, 2018. of human lung tumor-associated fibroblasts and their ability
[108] M. J. Besser, R. Shapira-Frommer, A. J. Treves et al., “Clinical to modulate the activation of tumor-associated T cells,” Jour-
responses in a phase II study using adoptive transfer of short- nal of Immunology, vol. 178, no. 9, pp. 5552–5562, 2007.
term cultured tumor infiltration lymphocytes in metastatic [123] M. E. Keir, M. J. Butte, G. J. Freeman, and A. H. Sharpe, “PD-
melanoma patients,” Clinical Cancer Research, vol. 16, 1 and its ligands in tolerance and immunity,” Annual Review
no. 9, pp. 2646–2655, 2010. of Immunology, vol. 26, no. 1, pp. 677–704, 2008.
[109] M. E. Dudley, J. C. Yang, R. Sherry et al., “Adoptive cell ther- [124] W. Zou and L. Chen, “Inhibitory B7-family molecules in the
apy for patients with metastatic melanoma: evaluation of tumour microenvironment,” Nature Reviews. Immunology,
intensive myeloablative chemoradiation preparative regi- vol. 8, no. 6, pp. 467–477, 2008.
mens,” Journal of Clinical Oncology, vol. 26, no. 32, [125] J. R. Brahmer, C. G. Drake, I. Wollner et al., “Phase I study of
pp. 5233–5239, 2008. single-agent anti-programmed death-1 (MDX-1106) in
[110] L. G. Radvanyi, C. Bernatchez, M. Zhang et al., “Specific lym- refractory solid tumors: safety, clinical activity, pharmacody-
phocyte subsets predict response to adoptive cell therapy namics, and immunologic correlates,” Journal of Clinical
using expanded autologous tumor-infiltrating lymphocytes Oncology, vol. 28, no. 19, pp. 3167–3175, 2010.
in metastatic melanoma patients,” Clinical Cancer Research, [126] W. Zou, J. D. Wolchok, and L. Chen, “PD-L1 (B7-H1) and
vol. 18, no. 24, pp. 6758–6770, 2012. PD-1 pathway blockade for cancer therapy: mechanisms,
Journal of Immunology Research 13

response biomarkers, and combinations,” Science Transla- [142] C. Sautès-Fridman, M. Lawand, N. A. Giraldo et al., “Tertiary
tional Medicine, vol. 8, no. 328, p. 328rv4, 2016. lymphoid structures in cancers: prognostic value, regulation,
[127] F. S. Hodi, S. J. O'Day, D. F. McDermott et al., “Improved sur- and manipulation for therapeutic intervention,” Frontiers in
vival with ipilimumab in patients with metastatic mela- Immunology, vol. 7, 2016.
noma,” New England Journal of Medicine, vol. 363, no. 8, [143] H. Tang, M. Zhu, J. Qiao, and Y.-X. Fu, “Lymphotoxin signal-
pp. 711–723, 2010. ling in tertiary lymphoid structures and immunotherapy,”
[128] E. J. Lipson and C. G. Drake, “Ipilimumab: an anti-CTLA-4 Cellular & Molecular Immunology, vol. 14, no. 10, pp. 809–
antibody for metastatic melanoma,” Clinical Cancer 818, 2017.
Research, vol. 17, no. 22, pp. 6958–6962, 2011. [144] F. Jing and E. Y. Choi, “Potential of cells and cytokines/che-
[129] M. D. Vesely and R. D. Schreiber, “Cancer immunoediting: mokines to regulate tertiary lymphoid structures in human
antigens, mechanisms, and implications to cancer immuno- diseases,” Immune Network, vol. 16, no. 5, pp. 271–280, 2016.
therapy,” Annals of the New York Academy of Sciences,
vol. 1284, no. 1, pp. 1–5, 2013.
[130] C. M. Koebel, W. Vermi, J. B. Swann et al., “Adaptive immu-
nity maintains occult cancer in an equilibrium state,” Nature,
vol. 450, no. 7171, pp. 903–907, 2007.
[131] D. T. Le, J. N. Uram, H. Wang et al., “PD-1 blockade in
tumors with mismatch-repair deficiency,” New England Jour-
nal of Medicine, vol. 372, no. 26, pp. 2509–2520, 2015.
[132] P. S. Hegde, V. Karanikas, and S. Evers, “The where, the
when, and the how of immune monitoring for cancer immu-
notherapies in the era of checkpoint inhibition,” Clinical
Cancer Research, vol. 22, no. 8, pp. 1865–1874, 2016.
[133] A. Ribas, R. Dummer, I. Puzanov et al., “Oncolytic virother-
apy promotes intratumoral T cell infiltration and improves
anti-PD-1 immunotherapy,” Cell, vol. 170, no. 6, pp. 1109–
1119.e10, 2017.
[134] S. Mariathasan, S. J. Turley, D. Nickles et al., “TGFβ attenu-
ates tumour response to PD-L1 blockade by contributing to
exclusion of T cells,” Nature, vol. 554, no. 7693, pp. 544–
548, 2018.
[135] J. E. Rosenberg, J. Hoffman-Censits, T. Powles et al., “Atezo-
lizumab in patients with locally advanced and metastatic
urothelial carcinoma who have progressed following treat-
ment with platinum-based chemotherapy: a single-arm, mul-
ticentre, phase 2 trial,” Lancet, vol. 387, no. 10031, pp. 1909–
1920, 2016.
[136] E. B. Garon, N. A. Rizvi, R. Hui et al., “Pembrolizumab for the
treatment of non-small-cell lung cancer,” New England Jour-
nal of Medicine, vol. 372, no. 21, pp. 2018–2028, 2015.
[137] R. S. Herbst, J. C. Soria, M. Kowanetz et al., “Predictive corre-
lates of response to the anti-PD-L1 antibody MPDL3280A in
cancer patients,” Nature, vol. 515, no. 7528, pp. 563–567,
2014.
[138] K. Adachi, Y. Kano, T. Nagai, N. Okuyama, Y. Sakoda, and
K. Tamada, “IL-7 and CCL19 expression in CAR-T cells
improves immune cell infiltration and CAR-T cell survival
in the tumor,” Nature Biotechnology, vol. 36, no. 4, pp. 346–
351, 2018.
[139] N. Hiraoka, Y. Ino, and R. Yamazaki-Itoh, “Tertiary lym-
phoid organs in cancer tissues,” Frontiers in Immunology,
vol. 7, 2016.
[140] M.-C. Dieu-Nosjean, J. Goc, N. A. Giraldo, C. Sautès-Frid-
man, and W. H. Fridman, “Tertiary lymphoid structures in
cancer and beyond,” Trends in Immunology, vol. 35, no. 11,
pp. 571–580, 2014.
[141] E. J. Colbeck, A. Ager, A. Gallimore, and G. W. Jones, “Ter-
tiary lymphoid structures in cancer: drivers of antitumor
immunity, immunosuppression, or bystander sentinels in
disease?,” Frontiers in Immunology, vol. 8, 2017.

You might also like