You are on page 1of 21

Immunological Investigations

A Journal of Molecular and Cellular Immunology

ISSN: 0882-0139 (Print) 1532-4311 (Online) Journal homepage: http://www.tandfonline.com/loi/iimm20

Active Immunotherapy of Cancer

Thinle Chodon, Richard C. Koya & Kunle Odunsi

To cite this article: Thinle Chodon, Richard C. Koya & Kunle Odunsi (2015) Active
Immunotherapy of Cancer, Immunological Investigations, 44:8, 817-836, DOI:
10.3109/08820139.2015.1096684

To link to this article: http://dx.doi.org/10.3109/08820139.2015.1096684

Published online: 17 Nov 2015.

Submit your article to this journal

Article views: 51

View related articles

View Crossmark data

Citing articles: 1 View citing articles

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=iimm20

Download by: [Universite Laval] Date: 24 November 2015, At: 01:04


Immunological Investigations, 2015; 44(8): 817–836
! Taylor & Francis Group, LLC
ISSN: 0882-0139 print / 1532-4311 online
DOI: 10.3109/08820139.2015.1096684

Active Immunotherapy of Cancer


Thinle Chodon,1 Richard C. Koya,1 and Kunle Odunsi1,2
1
Center for Immunotherapy and
2
Department of Gynecologic Oncology, Roswell Park Cancer Institute,
Buffalo, NY, USA

Clinical progress in the field of cancer immunotherapy has been slow for many years
but within the last 5 years, breakthrough successes have brought immunotherapy to
the forefront in cancer therapy. Promising results have been observed in a variety of
cancers including solid tumors and hematological malignancies with adoptive cell
therapy using natural host tumor infiltrating lymphocytes, host cells that have been
Downloaded by [Universite Laval] at 01:04 24 November 2015

genetically engineered with antitumor T-cell receptors or chimeric antigen receptors,


immune checkpoint inhibitors like anti-CTLA-4, anti-PD-1 or PD-L1 monoclonal
antibodies and oncolytic virus-based immunotherapy. However, most treatment
modalities have shown limited efficacy with single therapy. The complex nature of
cancer with intra- and inter-tumor antigen and genomic heterogeneity coupled with the
immune suppressive microenvironment emphasizes the prospect of personalized
targeted immunotherapy to manipulate the patient’s own immune system against
cancer. For successful, robust and long-lasting cure of cancer, a multi-modal approach is
essential, combining anti-tumor cell therapy with manipulation of multiple pathways in
the tumor microenvironment to ameliorate tumor-induced immunosuppression.

Keywords Cancer vaccine, cell therapy, immunotherapy

INTRODUCTION
The development of strategies for actively stimulating immunological rejection
of tumors, previously an elusive goal, has been accelerated by recent improved
understanding of the molecular basis of immune recognition and immune
regulation of cancer cells. The origins of the field of cancer immunotherapy can
be traced back to William Coley who, in the 1890s, observed that potentially
fatal bacterial infections could induce an effective anti-tumor response in
patients with partially resected tumors (Coley, 1991; Nauts & McLaren, 1990).
Enthusiasm for this approach waxed and waned over several decades. It is now
well known that the immune system plays a pivotal role in monitoring cancer
development (Doll & Kinlen, 1970; Dunn et al., 2002; Galon et al., 2013; Slaney
et al., 2013). This concept of ‘‘cancer immunoediting’’ (Dunn et al., 2002, 2004a,
b) is supported by several lines of evidence derived from murine tumor models,
and holds that the immune system not only protects the host against
development of primary cancers, but also sculpts tumor immunogenicity
(Dunn et al., 2004a). Cancer immunoediting is a dynamic process composed of
three phases: elimination, equilibrium and escape (Dunn et al., 2004a).
Elimination represents the classical concept of cancer immunosurveillance

Correspondence: Kunle Odunsi, MD, PhD, Roswell Park Cancer Institute, Elm and
Carlton Streets, Buffalo, NY 14263, USA. Tel: 716-845-8376. Fax: 716-845-1595. E-mail:
kunle.odunsi@roswellpark.org
818 T. Chodon et al.

Table 1. Available anti-cancer immunotherapies.

Approach Licensed Reference

Tumor targeting antibodies Yes NCT01650701, NCT02095054


DC vaccination Yes NCT01338012, NCT01807065
Peptide vaccines No Refer Table 2
Immunostimulatory cytokines Yes NCT01124734, NCT00002882
Immunomodulatory antibodies Yes NCT01807065, NCT02306850
Oncolytic virotherapy Yes NCT02173171, NCT02366195
TLR agonists Yes NCT00453050
DNA and recombinant viral vaccines No Refer Table 2
Inhibitors of IDO, arginase No NCT02042430, NCT02048709
Adoptive cell therapy No Refer Table 3

(Shankaran et al., 2001; Street et al., 2004), equilibrium is the period of


Downloaded by [Universite Laval] at 01:04 24 November 2015

immune-mediated latency after incomplete tumor destruction in the elimin-


ation phase (MacKie et al., 2003), and escape refers to the final outgrowth of
tumors that have outstripped immunological restraints of the equilibrium
phase (Marincola et al., 2000). Correlative human studies also support the
concept of ‘‘cancer immunoediting’’ because of observations that tumor
infiltration by lymphocytes is a reflection of a tumor-related immune response.
Data from these studies indicate that the presence of tumor infiltrating
lymphocytes (TILs) may be associated with improved clinical outcome in
several cancers including melanoma, colorectal, breast, prostate, renal-cell,
esophageal and ovarian carcinomas (Clemente et al., 1996; Funada et al., 2003;
Guidoboni et al., 2001; Hartveit, 1998; Naito et al., 1998; Sato et al., 2005;
Schumacher et al., 2001; Zhang et al., 2003). A recent meta-analysis of 10
studies with 1815 ovarian cancer patients confirmed the observation that a
lack of intraepithelial lymphocytes (TILs) is significantly associated with a
worse survival among ovarian cancer patients (Hwang et al., 2012).
The major criteria required for the immunological destruction of tumors
include generation of sufficient numbers of effector T cells with high avidity
recognition of tumor antigens (TAs) in vivo, trafficking and infiltration into the
tumor, overcoming inhibitory networks in the tumor microenvironment and
persistence of the anti-tumor T cells. In the past decade, significant progress
has been made in the development of interventions that mediate antitumor
effects by initiating a de novo or boosting an existing immune response against
cancer cells, and some have gained regulatory approval (Table 1). These
interventions include cancer vaccines, cell-based therapy, immune checkpoint
blockade and oncolytic virus-based therapy. Here, we will briefly review these
modalities and future directions to the pathway for cure.

CANCER VACCINES
The development of approaches for analyzing humoral (Carey et al., 1976) and
cellular (Knuth et al., 1984) immune reactivity to cancer in the context of the
autologous host led to the molecular characterization of TAs recognized by
autologous CD8+ T cells (van der Bruggen et al., 1991) and/or antibodies
(Sahin et al., 1995). Some of these approaches include serological analysis of
Cancer Immunotherapy 819

recombinant cDNA expression libraries (Chen et al., 1997), differential gene


expression analysis, T-cell epitope cloning (Boon & van der Bruggen, 1996; Van
den Eynde & Boon, 1997) and bioinformatics (Alpen et al., 2002; Scanlan et al.,
2000). As a consequence of these advances, human TAs defined to date can be
classified into one or more of the following categories: (i) differentiation
antigens (that are restricted to very defined tissues), e.g. tyrosinase (Brichard
et al., 1993), Melan-A/MART-1 (Coulie et al., 1994) and gp 100 (Kawakami
et al., 1995); (ii) mutational antigens (that are altered forms of proteins), e.g.
CDK4 (Wolfel et al., 1995), b-catenin (Robbins et al., 1996), caspase-8
(Mandruzzato et al., 1997) and P53 (Scanlan et al., 1998); (iii) amplification
antigens, e.g. Her2/neu (Cheever et al., 1995) and P53 (Gnjatic et al., 1998);
(iv) splice variant antigens, e.g. NY-CO-37/PDZ-45 (Scanlan et al., 1998) and
ING1 (Jager et al., 1999); (v) glycolipid antigens; (vi) viral antigens, e.g. HPV
(Tindle, 1996), EBV (Lennette et al., 1995) and (vii) cancer-testis (CT) antigens
(that are restricted in expression to the germ line and tumors) e.g. MAGE
Downloaded by [Universite Laval] at 01:04 24 November 2015

(Boon & van der Bruggen, 1996), NY-ESO-1 (Chen et al., 1997) and LAGE-1
(Lethe et al., 1998).
These TAs stimulate cellular and/or humoral responses in cancer patients,
and give rise to epitopes that are presented on tumor cells (TCs) in the context
of the major histocompatibility complex (MHC) class I or II molecules, thereby
stimulating CD8+ or CD4+ T cells, respectively. Although there are several
options in deciding which antigen to target, the fundamental requirements of
the ideal TA include: (i) Limited or no expression in normal tissues, but
aberrant expression at high frequencies in tumor; (ii) immunogenicity and
(iii) a role in tumor progression. While none of the current TAs completely
meets all of these criteria, the family of CT antigens are closest. CT antigens
are a subclass of TAs encoded by 140 genes. The criteria for placing antigens
in this category are based on several characteristic features (Old, 2001; Tureci
et al., 1998): (i) predominant expression in germ cells of the testis and
generally not in other normal tissues; (ii) expression in a proportion of
malignant tumors of different histological types; (iii) expression in malig-
nancies in a lineage nonspecific fashion; (iv) often mapping of the gene on the
X-chromosome; (v) often members of multigene families. Despite their poorly
characterized biologic function, expression of these antigens are known to be
restricted in immune privileged sites such as the testes, placenta and fetal
ovary, but not in other normal tissues. Abnormal expression of these germ-line
genes in malignant tumors may reflect the activation of a silenced ‘‘gameto-
genic program’’, which ultimately leads to tumor progression and broad
immunogenicity (Simpson et al., 2005). The immunogenicity of CT antigens
has led to the widespread development of cancer vaccines targeting these
antigens (e.g. NY-ESO-1 and MAGE) in many solid tumors.
The identification and characterization of peptide epitopes from several
TAs, along with the relative ease of production of cGMP grade peptides for
clinical use, led to a large number of human cancer vaccine studies utilizing
these peptide epitopes in several solid and hematological malignancies (see
Table 2 for examples). Additional vaccine studies have included the use of long
peptides, recombinant proteins, recombinant viral vectors and dendritic cells
(DC). Since the majority of TAs are self-proteins, and are therefore subject to
central and peripheral tolerance, an initial major question was to ask how best
Downloaded by [Universite Laval] at 01:04 24 November 2015

820

Table 2. Selected cancer vaccine studies.

Antigen Phase Disease Technology Cotherapy Sponsor Reference

NY-ESO-1 I Metastatic cancer Recombinant protein GLA-SE Immune design CT02015416


I Ovarian, fallopian tube DEC-205 fusion protein Poly-ICLC, ID O1 inhibitor Roswell Park Cancer NCT02166905
cancer Institute
I NY-ESO-1 expressing DEC-205 fusion protein/ Rapamycin Roswell Park Cancer NCT01522820
solid tumors DC Institute
T. Chodon et al.

I/II NY-ESO-1 expressing DEC-205 fusion protein Resiquimod, Poly-ICLC Celldex Therapeutics NCT00948961
tumors
I NY-ESO-1 expressing Full-length protein Montanide, Resiquimod Mount Sinai School of NCT00821652
tumors Medicine
I Ovarian, fallopian, pri- Peptide Decitabine, Doxorubicin, Roswell Park Cancer NCT01673217
mary peritoneal Montanide Institute
cancer
I NY-ESO-1/LAGE-1 Peptide CpG7909, Montanide Ludwig Institute for NCT00199836
expressing tumors Cancer Research
I Ovarian, fallopian, pri- Peptide Montanide Memorial Sloan Kettering NCT00066729
mary peritoneal Cancer Center
cancer
I Prostate cancer Peptide Baylor College of NCT00616291
Medicine
I Ovarian, fallopian, pri- Overlapping long pep- Montanide, Poly-ICLC Ludwig Institute For NCT00616941
mary peritoneal tides (OLP4) Cancer Research
cancer
I Ovarian, fallopian, pri- Vector (ALVAC(2)-NY- GM-CSF, Rapamycin Roswell Park Cancer NCT01536054
mary peritoneal ESO-1(M) TRICOM) Institute
cancer
I Ovarian, fallopian, pri- Vector (ALVAC(2)-NY- GM-CSF Ludwig Institute for NCT00803569
mary peritoneal ESO-1(M) TRICOM) Cancer Research
cancer
II Ovarian, fallopian, pri- Vector (Fowlpox-NY- Recombinant Vaccinia- Ludwig Institute for NCT00112957
mary peritoneal ESO-1) NY-ESO_1 Cancer Research
cancer
NY-ESO-1, gp100 I Metastatic melanoma Peptide Pool Montanide, anti-PD1, Moffitt Cancer Center NCT01176474
anti-CTLA4
NY-ESO-1, gp100, MART-1 I Metastatic melanoma Peptide Pool Montanide, anti-PD1 Moffitt Cancer Center NCT01176461
MART-1, gp100, Tyrosinase I Metastatic melanoma Peptide Pool Montanide, GM-CSF National cancer Institute NCT00006243
Tyrosinase I/II Metastatic melanoma Peptide Cliniques Universitaires NCT01331915
Saint-Luc-Universite
Catholique de
Louvain
Downloaded by [Universite Laval] at 01:04 24 November 2015

WT-1 I AML, MDS, NSCLC, Peptide Montanide, GM-CSF Memorial Sloan Kettering NCT00398138
Mesothelioma Cancer Center
II AML, ALL Peptide Montanide Memorial Sloan Kettering NCT01266083
Cancer Center
I AML Peptide Montanide, GM-CSF Moffitt Cancer Center NCT00665002
II Mesothelioma Peptide Montanide, GM-CSF Memorial Sloan Kettering NCT01265433
Cancer Center
I Advanced cancers Peptide Sunovion NCT01621542
I AML, CML, ALL, MDS, B Peptide Pool Montanide, GM-CSF Duke University Medical NCT00672152
cell malignancies Center
I/II AML Recombinant protein Treg depleted T lympho- Jules Bordet Institute NCT01513109
ASCI cyte infusion
MAGE-A3 I Multiple myeloma Recombinant protein AS15 Ludwig Institute For NCT01380145
ASCI Cancer Research
II Metastatic melanoma Peptide AS15, poly-ICLC Moffitt Cancer Center NCT01437605
I Bladder cancer Peptide AS15, BCG University of Lausanne NCT01498172
Hospitals
II Bladder cancer Peptide AS15 European Association of NCT01435356
Urology Research
Foundation
MAGE-12 I Metastatic cancer Peptide Montanide, IL-2 National cancer Institute NCT00020267
MUC-1 III NSCLC Peptide (Tecemotide) Cyclophos-phamide Merck KGaA NCT01015443
0 Breast cancer Peptide Poly-ICLC Case Comprehensive NCT00986609
Cancer center
II Rectal carcinoma Peptide (Tecemotide) Cyclophospha-mide, Merck KGaA NCT01507103
chemo-radiotherapy
II Colorectal carcinoma Peptide Poly-ICLC University of Pittsburgh NCT00773097
I Solid tumors Peptide (ONT-10) Oncothyreon Inc. NCT01556789
HER-2 I Breast cancer Peptide Mayo Clinic NCT01632332
I/II Breast cancer Peptide AS15, Lapatinib Duke University Medical NCT00952692
Center
III Breast cancer Peptide (NeuVax) GM-CSF Galena Biopharma NCT01479244
I Metastatic solid tumors Peptide Montanide, Nor-MDP Ohio State University NCT01376505
Comprehensive
Cancer Center
I Breast cancer Vector (Alphaviral) Duke University Medical NCT01526473
Center
Ras II NSCLC Vector (GI-4000) Memorial Sloan Kettering NCT00655161
Cancer Center
Cancer Immunotherapy
821
822 T. Chodon et al.

to generate sufficient anti-tumor immune responses. Although this issue was


tackled by several individual investigators, one of the most remarkable efforts
was launched as the Cancer Vaccine Collaborative (CVC) Program of the
Cancer Research Institute and the Ludwig Institute for Cancer Research (Old,
2008). In this program, several parallel clinical trials focusing on the prototype
antigen NY-ESO-1 were conducted. Important issues addressed by the CVC
and others include: Which are the appropriate adjuvants? Are monovalent or
multi-antigen vaccine approaches likely to provide better results? At what
disease state is vaccination appropriate? What is the optimal frequency and
duration of vaccination? How should antigen-specific immune responses be
monitored? How should the induced immune response be sustained? A number
of current cancer vaccine strategies in clinical trials are summarized in
Table 2. This table is not exhaustive, and other vaccine approaches, either
alone or in combination with immunodulation are on-going.
The most common cancer vaccine strategy is to administer full-length
Downloaded by [Universite Laval] at 01:04 24 November 2015

recombinant protein antigens or peptides to cancer patients, most often via the
intramuscular, subcutaneous or intradermal route, together with one or more
immunostimulatory adjuvants to promote DC maturation. The rationale
behind this approach is that resident DCs or other antigen presenting cells
(APCs) acquire the ability to present the tumor-associated antigen (TAA)-
derived epitopes while maturing, hence priming a robust TAA-specific immune
response. While short peptides (8–12 amino acids) directly bind to MHC
molecules expressed on the surface of APCs, synthetic long peptides (25–30
residues) are taken up, processed and presented by APCs for eliciting an
immune response (Melief & van der Burg, 2008). Several reports indicate that
therapeutic activity of synthetic long peptides is superior to that of their short
counterparts, especially when they include epitopes recognized by both
cytotoxic and helper T cells or when conjugated to efficient adjuvants (Melief
& van der Burg 2008; Odunsi et al., 2007).
Both peptide- and DNA-based vaccines have been associated with clinical
activity in several cancer types (Galluzzi et al., 2012; Senovilla et al., 2013;
Vacchelli et al., 2012, 2014). For example the administration of a multi-peptide
vaccine after single-dose cyclophosphamide was shown to prolong overall
survival (OS) in a cohort of renal cell carcinoma patients (Walter et al., 2012).
In addition, several NY-ESO-1 vaccine clinical trials also demonstrated clinical
activity, but these studies were small and not definitive (Davis et al., 2004;
Odunsi et al., 2012). Moreover, it is now well recognized that several immune
resistance mechanisms may dampen the efficacy of vaccine-induced immune
responses (discussed further below). Consequently, no peptide- or DNA-based
anticancer vaccine is currently approved by the US FDA and EMA for use in
humans.
DCs express a number of cytokines and membrane costimulators that drive
the T-cell response and DCs ‘‘cross-present’’ antigens on MHC Class I (Savina
et al., 2006). Several forms of DC-based vaccine approaches have been
developed, most of which involve the isolation of patient-derived circulating
monocytes and their differentiation ex vivo, in the presence of agents that
promote DC maturation, such as granulocyte macrophage colony-stimulating
factor (GM-CSF). These autologous DCs are injected into cancer patients upon
exposure to a TA (protein, peptide, mRNAs, recombinant viral vectors
Cancer Immunotherapy 823

encoding TA, TC lysates). The antigen-pulsed DCs are able to prime tumor-
targeting immune responses in vivo upon administration to patients. An
additional strategy is to fuse tumor to mAbs that selectively bind to
endocytosis receptors (e.g. mannose receptor or DEC-205) on the surface of
DCs (Tsuji et al., 2011). At present, only one cellular product containing a
significant proportion of probably immature DCs (sipuleucel-T or Provenge) is
currently licensed by the US FDA and the EMA for the therapy of
asymptomatic or minimally symptomatic metastatic castration-refractory
prostate cancer (Kantoff et al., 2010). The safety and efficacy of many other
DC-based cellular vaccines are currently being investigated in clinical trials.
In a recent publication, pooled data from three matastatic melanoma trials
showed superior survival in patients receiving a therapeutic vaccine consisting
of autologous DC loaded with antigens from self-renewing, proliferating,
irradiated autologous TCs (DC-TC) compared with patients receiving the
autologous irradiated TCs alone (Dillman et al., 2015). OS was longer in 72
Downloaded by [Universite Laval] at 01:04 24 November 2015

patients treated with DC-TC compared with 71 patients treated with TC


(median OS 60 versus 22 months; 5-year OS 51 versus 32%). The OS was
better with the DC-TC in both patients with no evidence of disease and with
detectable metastasis at the time of start of this therapy. Although the success
rate of establishing an autologous cell line from each patient is 50% and the
time it takes for this varies, this is a promising approach to be used as an
adjuvant.
Advances in next-generation sequencing and epitope prediction now permit
the rapid identification of mutant tumor neoantigens. This has led to efforts in
utilizing these mutant tumor neoantigens for personalizing cancer immu-
notherapies. Indirect support for this approach comes from studies demon-
strating that (i) infusion of autologous ex vivo-expanded TILs can induce
objective clinical responses in metastatic melanoma (Dudley et al., 2013), and
(ii) the relationship between pre-therapy CD8+ T cell infiltrates and response
to checkpoint blockade in melanoma (Tumeh et al., 2014). Deep-sequencing
technologies permit easy identification of the mutations present within the
protein-encoding part of the genome (the exome) of an individual tumor
allowing for prediction of potential neoantigens. Several pre-clinical and
clinical studies have now confirmed the possibility of identifying neoantigens
on the basis of cancer exome data (Castle et al., 2012; Duan et al., 2014; Gubin
et al., 2014; Rizvi et al., 2015; Wick et al., 2014). Although there are limitations
of probing the mutational profile of a tumor in a single biopsy (Gerlinger et al.,
2012; Linnemann et al., 2015), it is evident that the vast majority of
neoantigens occur within exonic sequence and do not lead to the formation
of neoantigens that are recognized by autologous T cells (Linnemann et al.,
2015; Lu et al., 2014). Consequently, a robust pipeline for filtering the cancer
exome data is essential. Epitope presentation of neoantigens by MHC class I
molecules may be predicted using previously established algorithms that
analyze critical features such as the likelihood of proteasomal processing,
transport into the endoplasmic reticulum and affinity for the relevant MHC
class I alleles. In order to predict epitope abundance, gene and/or protein
expression levels can also be integrated into the analysis. Based on these
considerations, it becomes of interest to stimulate neoantigen-specific T cell
responses in cancer patients using two possible approaches. The first is to
824 T. Chodon et al.

synthesize long peptide vaccines that encode a set of predicted neoantigens.


The second approach is to identify and expand pre-existing neoantigen-specific
T cell populations to create either bulk neoantigen-specific T cell products or T-
cell receptor (TCR)-engineered T cells for adoptive therapy.

ADOPTIVE CELLULAR THERAPY


‘‘Adoptive cell transfer’’ (ACT) is an approach that involves: (i) the collection of
circulating or TILs T cells (Rosenberg et al., 2008), (ii) activation and
modification and/or expansion ex vivo; and (iii) their re-infusion to patients,
usually after lymphodepleting pre-conditioning chemotherapy. Initial studies
demonstrating the potential of T cell immunotherapy to eradicate solid tumors
came from the NCI in studies of adoptive transfer of in vitro-selected TILs
(Dudley et al., 2002, 2005). Unfortunately, methods of isolating and
manufacturing TILs are labor intensive and only successful in a subset of
Downloaded by [Universite Laval] at 01:04 24 November 2015

patients (Dudley et al., 2003, 2008). In order to improve the therapeutic


potential of transferred cells, peripheral blood lymphocytes (PBLs) with
unique antigen specificity (Rosenberg et al., 2008) can be genetically modified
to express: (i) a TA-specific TCR (Robbins et al., 2011), or (ii) ‘‘chimeric antigen
receptor’’ (CAR), i.e. a transmembrane protein comprising the TAA-binding
domain of an immunoglobulin linked to one or more costimulatory molecules
(Kalos et al., 2011). The TCR strategy is based on the understanding that the
binding of MHC-antigen complex by TCR is the main determinant of tumor
recognition by T cells. Genes that encode the a and b chains are cloned from
tumor-reactive T cells restricted to a particular HLA allele and then introduced
into recipient T cells to endow them with the specificity of the donor TCR.
Transduced T cells then acquire stable reactivity to the TAAs. Several studies
are on-going or completed testing CD8TCR-redirected T cells in cancer
patients. Although spectacular responses have been observed, the majority
of clinical responses are short-lived with ultimate tumor relapse. A major
explanation for this sub-optimal outcome is the relatively limited long-term
survival and effector function due to suppression or exhaustion of infused
engineered T cells.
Among various approaches focused on stimulating the immune system to
recognize and destroy tumors, ACT using TCR-engineered T cells have
resulted in objective responses in the majority of treated patients (Robbins
et al., 2011). CD19-specific CAR T cells have also demonstrated encouraging
results of inducing complete response in 70–90% of patients with relapsed or
refractory B-cell acute lymphoblastic leukemia (Davila et al., 2014; Lee et al.,
2015; Maude et al., 2014). A partial listing of completed and on-going ACT
clinical trials is provided in Table 3. No ACT protocol is currently approved by
the US FDA for use in cancer patients.
ACT using genetically engineered PBLs to express antitumor T-cell
receptors holds promise for extending the use of ACT to patients with
common epithelial cancers (Morgan et al., 2006). Sentinel-node-acquired CD4+
Th1-lymphocytes could be clonally expanded in vitro and safely administered
to all 16 colorectal cancer patients without side effects. In four out of nine stage
IV patients, complete tumor regression occurred. Median survival time was 2.6
years, as compared with 0.8 years in conventionally treated controls. A
Downloaded by [Universite Laval] at 01:04 24 November 2015

Table 3. Selected adoptive cell therapy studies.

Target Phase Disease Technology Cotherapy Sponsor Reference

NY-ESO-1 II Metastatic melanoma TCR engineered CD62L+ Aldesleukin, condition- National Cancer Institute NCT02062359
T cells ing chemotherapy
I Sarcoma TCR engineered T cells Radiotherapy Fred Hutchinson Cancer NCT02319824
Research Center
I/II Multiple myeloma TCR engineered T cells Adapt immune NCT01352286
I/II Metastatic melanoma TCR engineered T cells Conditioning Adapt immune NCT01350401
chemotherapy
I Synovial sarcoma TCR engineered T cells Conditioning chemo- Adapt immune NCT01343043
therapy, Denileukin,
Diffitox
I Metastatic cancer (mel- TCR engineered T cells Aldesleukin, condition- National Cancer Institute NCT00670748
anoma, renal cell ing chemotherapy
cancer
I/II Ovarian cancer TCR engineered T cells Conditioning Adapt immune NCT01567891
chemotherapy
I Liposarcoma, synovial TCR engineered T cells Conditioning Fred Hutchinson Cancer NCT01477021
sarcoma chemotherapy Research Center
II Advanced malignancies TCR engineered T cells Aldesleukin, condition- Jonsson Comprehensive NCT01697527
ing chemotherapy, Cancer Center
DC vaccine
I Solid tumors TCR engineered T cells Conditioning Mie University NCT02366546
chemotherapy
I Solid tumors TCR engineered T cells Aldesleukin, condition- Jonsson Comprehensive NCT02070406
ing chemotherapy, Cancer Center
DC vaccine,
Ipilimumab
MAGE-A3 I/II Metastatic melanoma TCR engineered T cells Aldesleukin, condition- National Cancer Institute NCT02153905
ing chemotherapy
MAGE-A3/12 I/II Metastatic cancer TCR engineered T cells Aldesleukin, condition- National Cancer Institute NCT01273181
ing chemotherapy
MAGE-A4 I Solid tumors (melanoma TCR engineered T cells Aldesleukin, peptide Tianjin Medical University NCT01694472
& lung cancer) vaccine Cancer Institute &
Hospital
(Continued )
Cancer Immunotherapy
825
Downloaded by [Universite Laval] at 01:04 24 November 2015

826

Table 3. Continued

Target Phase Disease Technology Cotherapy Sponsor Reference

MART-1 II Metastatic melanoma TCR engineered T cells Aldesleukin, condition- National Cancer Institute NCT00509288
ing chemotherapy
T. Chodon et al.

MART-1 II Metastatic melanoma TCR engineered T cells Aldesleukin, condition- Jonsson Comprehensive NCT00910650
ing chemotherapy, Cancer Center
DC vaccine
MART-1 II Metastatic melanoma TCR engineered T cells Aldesleukin, condition- National Cancer Institute NCT00706992
ing chemotherapy,
peptide & ALVAC
vaccine
WT-1 I Ovarian, fallopian, pri- TCR engineered T cells Conditioning chemo- Memorial Sloan Kettering NCT00562640
mary peritoneal therapy, Filgrastim Cancer Center
cancer
I/II NSCLC, mesothelioma TCR engineered CD8+ Aldesleukin, condition- Fred Hutchinson Cancer NCT02408016
Tcm/Tn T cells ing chemotherapy Research center
HER-2 I/II Breast cancer TCR engineered T cells Conditioning chemo- University of Washington NCT00791037
therapy, GM-CSF
CD20 I B cell malignancies CAR T cells Aldesleukin, condition- Fred Hutchinson Cancer NCT00621452
ing chemotherapy Research center
CD19 I ALL CAR T cells Conditioning Memorial Sloan Kettering NCT01044069
chemotherapy Cancer Center
I B cell leukemia & CAR T cells Abramson Cancer NCT01029366
lymphoma Center, University of
Pennsylvania
I B cell lymphoma CAR T cells Conditioning National Cancer Institute NCT00924326
chemotherapy
I/II B cell CMV/EBV bi-specific Fred Hutchinson Cancer NCT01475058
CAR T cells (Tcm) Research center
malignancies
I B cell malignancies CAR T cells Conditioning chemo- National cancer Institute NCT01087294
therapy, Pentostatin
I ALL, CLL, NHL CMV/EBV/Adeno trivirus- Baylor College of NCT00840853
specific CAR T cells Medicine
PSMA I Prostate cancer CAR T cells Conditioning Roger Williams Medical NCT00664196
chemotherapy Center
Downloaded by [Universite Laval] at 01:04 24 November 2015

GD2 I Neuroblastoma EBV specific CAR T cells Baylor College of NCT00085930


Medicine
I Neuroblastoma Trivirus-specific CAR T Conditioning Children’s Mercy NCT01460901
cells chemotherapy Hospital Kansas City
Mesothelin & CD19 I Pancreatic Cancer CAR T cells Conditioning University of NCT02465983
chemotherapy Pennsylvania
VEGFR2 I/II Metastatic cancer (mel- CD8+ CAR T cells Conditioning National Cancer Institute NCT01218867
anoma, renal cell chemotherapy
cancer
EGFRvIII I/II Malignant Glioma, CAR T cells Aldesleukin, condition- National Cancer Institute NCT01454596
Glioblastoma ing chemotherapy
BCMA I Multiple Myeloma CAR T cells Conditioning National cancer Institute NCT02215967
chemotherapy
CEA I Breast Cancer CAR T cells Aldesleukin Roger Williams Medical NCT00673829
Center

ALL: Acute Lymphoblastic leukemia, CLL: Chronic Lymphocytic Leukemia, DC: Dendritic Cells, NHL: Non Hodgkin’s Lymphoma, NSCLC: Non-Small Cell Lung
Carcinoma.
Cancer Immunotherapy
827
828 T. Chodon et al.

dose-dependent effect with regards to reduced tumor burden and long-term


survival was observed (Karlsson et al., 2010). Interestingly, a recent experi-
mental mouse model supports the efficacy of CD4 T-helper lymphocytes in
adoptive immunotherapy. Mice with established tumors were treated with
adoptive transfer using in vitro-activated T lymphocytes harvested from
tumor-draining lymph nodes. The results demonstrated that expanded sorted
CD4+ T-helper lymphocytes from the tumor-draining lymph node have
therapeutic efficacy on their own, and a synergistic effect was found when
CD4+ T-helper lymphocytes were used in combination with expanded cytotoxic
CD8+ T-lymphocytes (Wang et al., 2007). In addition, adoptive transfer of a TA-
specific CD4+ T-cell clone to one patient with metastatic malignant melanoma
was reported to remain disease free 2 years later (Hunder et al., 2008).
From the transgenic cell manufacture point of view, large numbers of
tumor-specific T cells for ACT can be manufactured by retroviral or lentiviral
genetic engineering of autologous PBLs and expanding them over several
Downloaded by [Universite Laval] at 01:04 24 November 2015

weeks but emphasis should be on preparing young T cells with minimum


duration of ex vivo manipulation which allows quick therapy for cancer
patients (Chodon et al., 2014). Preparation of double cell therapy with TCR-
engineered T cells and adjuvant DC vaccine is feasible in 6–7 days.
Administration of freshly manufactured TCR transgenic T cells resulted in a
higher persistence of antigen-specific T cells in the blood as compared with
cryopreserved (Chodon et al., 2014). However, the convenience of cryopre-
served product is valuable for a centralized manufacture facility model and
transgenic T cell persistence could be promoted by combining with checkpoint
inhibitors and TGFb blockade.
In preparation for the ACT, patients usually receive lymphodepleting
conditioning chemotherapy and low or high dose IL2 is administered after
the transfer for T cell expansion. As ACT becomes more common, it becomes
very important that all centers are familiar with potential adverse events
that may occur and its management. Cytokine release syndrome, typically
occurring between post-infusion days 2 and 10, manifested as fever,
hypotension and respiratory insufficiency, can be managed with
Tocilizumab, an IL6-receptor antagonist and general supportive treatment
in an ICU setting. High-dose steroids are administered to life-threatening
cases. In patients who have had undergone multiple cycles of chemotherapy
in the past may end up with persistent pancytopenia and therefore advisable
to decrease the dose of conditioning chemotherapy and cryopreserve stem cell
reserve for backup.
Previous ACT trials have focused on the use of CD8TCR but not CD4TCR.
Because CD4+ T cells maintain CD8+ T-cell responses (Matloubian et al., 1994;
Schoenberger et al., 1998) and rescue exhausted T cells (Aubert et al., 2011),
long-lasting anti-tumor responses are expected by the synergy of CD8TCR-
and CD4TCR-engineered T cells. Recently, two types of TA (NY-ESO-1)-specific
CD4+ T cells, tumor recognizing and nontumor recognizing CD4+ T cells have
been published, that are considered to play distinct roles at the local tumor site
(Matsuzaki et al., 2014). Whereas both CD4+ T cell types recognize exogenous
NY-ESO-1 protein that is processed and presented by APCs, only tumor
recognizing CD4 directly recognize cancer cells in MHC class II-restricted and
antigen-specific manner (Matsuzaki et al., 2014; Tsuji et al., 2012).
Cancer Immunotherapy 829

Recent reports indicate that T cells that are expanded ex vivo to maintain
more stem like T cell populations known as T stem cell memory cells are
capable of a more sustained response by replenishing effectors (Gattinoni
et al., 2011). A clear benefit of transferring less mature, more stem-like cells is
likely due to increased persistence and replenishing capability of these cells
in vivo. Conceptually, the regenerative nature of hHSCs may provide a long-
lasting, potentially life-long supply of effector T cells engineered against TAAs
by TCR genes. Incorporation of suicide genes in the gene-modification could
add an extra level of safety. Genetic labeling with bioluminescence imaging
and positron emitting tomography reporter genes will allow real-time visual-
ization of transgenic T cells trafficking into the tumor and its correlation with
antitumor responses (Koya et al., 2010).

IMMUNE MODULATION
Downloaded by [Universite Laval] at 01:04 24 November 2015

Immune modulation is designed to reinstate an existing anticancer immune


response or elicit novel responses as a result of antigen spreading. This has
been achieved through four general strategies: (i) the inhibition of immuno-
suppressive receptors expressed by activated T lymphocytes, such as cytotoxic
T lymphocyte-associated protein 4 (CTLA4) and programmed cell death 1 (PD-
1), (ii) the inhibition of the principal ligands of these receptors, such as the PD-
1 ligand CD274 (PD-L1 or B7-H1), (iii) the activation of costimulatory
receptors expressed on the surface of immune effector cells such as tumor
necrosis factor receptor superfamily, member 4 (TNFRSF4 or OX40),
TNFRSF9 (CD137 or 4-1BB) and TNFRSF18 (GITR) and (iv) the neutraliza-
tion of immunosuppressive factors released in the tumor microenvironment,
such as transforming growth factor b1.
Inhibition of immunosuppressive receptors expressed by activated T
lymphocytes is commonly referred to as ‘‘checkpoint blockade’’. This has
been shown to induce robust and durable responses in patients with a variety
of solid tumors. Antibody blockade of PD1 and PDL1 has demonstrated
enhanced antitumor immunity in mouse models (Blank et al., 2004; Dong
et al., 2002; Iwai et al., 2002), with less immune toxicity as compared with
CTLA blockade. Initial clinical results have been extremely promising
(Brahmer et al., 2010) with durable responses in multiple cancers including
colon, renal, lung carcinoma and melanoma with significant increases in
lymphocyte infiltration into tumor lesions. A number of checkpoint-blocking
mAbs were recently approved by the FDA and other international regulatory
agencies for use in humans, namely: the anti-CTLA4 mAb ipilimumab
(Yervoy), which was licensed by the US FDA for use in individuals with
unresectable or metastatic melanoma; the anti-PD-1 mAb pembrolizumab
(Keytruda), which received accelerated approval by the US FDA for the
treatment of advanced or unresectable melanoma patients who fail to respond
to other therapies; and nivolumab (Opvido), another PD-1-targeting mAb
licensed by the FDA and Japanese Ministry of Health and Welfare for use in
humans. Blockade of additional inhibitory receptors are in various phases of
clinical development and include LAG3, B7-H3, B7-H4 and TIM3. Emerging
evidence suggest that the clinical efficacy of immunomodulatory mAbs
(especially checkpoint blockers) may be profoundly influenced by the
830 T. Chodon et al.

mutational burden and ‘‘neoantigens’’ specific to the neoplasm (Snyder et al.,


2014). The higher neoantigen load leads to recruitment of a diverse repertoire
of neoantigen specific T cells, and checkpoint blockade restores a favorable
balance of Teff/Treg ratio, leading to more effective tumor control.

ONCOLYTIC VIRUS-BASED THERAPY


Oncolytic viruses are nonpathogenic viral strains that specifically infect cancer
cells, triggering their demise. The anti-neoplastic potential of oncolytic viruses
can be innate via a cytopathic effect, or these viruses can mediate an oncolytic
activity because of gene products that are potentially lethal for the host cell,
irrespective of their capacity to massively replicate and cause a cytopathic
effect. Increasing preclinical and clinical evidence indicate that the therapeutic
activity of oncolytic viruses is also related to their ability to elicit tumor-
targeting immune responses as they (i) reprogram the inflammatory tumor
Downloaded by [Universite Laval] at 01:04 24 November 2015

microenvironment to be more immunogenic and (ii) promote the release of TAs


in this immunostimulatory environment, leading to efficient cross presenta-
tion. These viruses can be genetically engineered to endow them with
additional attributes such as antagonist of chemokine–chemokine ligand
interaction (Gil et al., 2014), or sequences coding for enzymes that convert an
innocuous pro-drug into a cytotoxic agent.
The results of immunotherapy with talimogene laherparepvec (T-VEC), a
modified form of herpes simplex virus type-1, that provides direct tumor lysis,
and simultaneously produce GM-CSF were recently reported (Andtbacka
et al., 2015). Researchers randomized 436 patients with aggressive, inoperable
malignant melanoma to receive either an injection of the viral therapy T-VEC,
or a control immunotherapy in a phase III trial. 16.3 percent of the group given
T-VEC showed a durable treatment response of more than 6 months. Some
patients had a response extending past 3 years.
Combination therapy with adoptive T-cell therapy and oncolytic viral
delivery may have beneficial synergistic effect. In mouse models, it has been
shown that antigen-nonspecific T cells loaded with oncolytic vesicular stoma-
titis virus efficiently delivered the virus to metastatic lymph nodes leading to
tumor clearance associated with antitumor immune priming (Qiao et al.,
2008a). The loading of antigen-specific T cells with vesicular stomatitis virus
enhanced the delivery of the virus to lung tumors (Qiao et al., 2008b) and the
associated pro-inflammatory tumor microenvironment enhanced antigen-
specific T-cell proliferation and survival within the tumor.

CONCLUSIONS AND FUTURE DIRECTIONS


Cancer immunotherapy is evolving quickly and understanding the dynamics of
the response to this therapy to find ways to overcome immune suppression and
counter-regulation will lead to development of effective personalized targeted
approaches to treat cancer which could one day become standard of care for
cancer patients. Immunotherapy is expected to mediate tumor destruction and
drive local inflammation in the tumor microenvironment, but also trigger
coordinated induction of multiple counter-regulatory and suppressive path-
ways like IDO, TGFb, PD-L1 and Tregs. Concomitant blockade of these
Cancer Immunotherapy 831

suppressive pathways-mediated immune suppression at the time of vaccin-


ation or T cell transfer will allow inflammation-induced transformation of the
tumor milieu from a tolerogenic to an immunogenic signature.
Based on the promising results of blockade of the PD-1/PD-L1 pathway, it is
important to consider opportunities for combination therapies. These include
dual checkpoint blockade e.g. the combination of CTLA-4 and PD-1 blockade.
Ipilimumab removes a physiological brake on T cells during activation,
whereas anti-PD-1 removes a brake on activation during T-cell effector
function. This combination may also overcome resistance to CTLA-4 blockade
mediated by tumor PD-L1 expression or resistance to PD-1 blockade mediated
by T-cell downregulation through the coexpression of CTLA-4. In a recent
clinical trial in patients with metastatic melanoma, nivolumab alone or
combined with ipilimumab resulted in significantly longer progression-free
survival than ipilimumab alone (Larkin et al., 2015a). Another potential
checkpoint combination therapy is blockade of PD-1 and LAG-3, an approach
Downloaded by [Universite Laval] at 01:04 24 November 2015

that has demonstrated excellent results in pre-clinical models of ovarian


cancer and melanoma (Huang et al., 2015; Kelleher et al., 2015; Matsuzaki
et al., 2010; Mikucki et al., 2015, Woo et al., 2012; Zsiros et al., 2015).
Additional potential combinations include targeted agents (e.g. BRAF and
EGFR-targeted agents) (Larkin et al., 2015b), chemotherapies with potential
to cause immunogenic cell death, vaccine combinations and VEGFR tyrosine
kinase inhibitor combinations.

DECLARATION OF INTEREST
The authors report no conflicts of interest. This work was supported in part by
grants from the National Institutes of Health R01CA158318-01A1 and Roswell
Park Cancer Institute-University of Pittsburgh Cancer Institute Ovarian
Cancer Specialized Program of Research Excellence National Institutes of
Health P50CA159981-01A1; the Roswell Park Alliance Foundation. The
funders had no role in study design, data collection and analysis, decision to
publish, or preparation of the manuscript.

REFERENCES
Alpen B, Gure AO, Scanlan MJ, et al. (2002). A new member of the NY-ESO-1 gene
family is ubiquitously expressed in somatic tissues and evolutionarily conserved.
Gene, 297, 141–9.
Andtbacka RH, Kaufman HL, Collichio F, et al. (2015). Talimogene laherparepvec
improves durable response rate in patients with advanced melanoma. J Clin Oncol,
33, 2780–8. doi:10.1200/JCO.2014.58.3377.
Aubert RD, Kamphorst AO, Sarkar S, et al. (2011). Antigen-specific CD4 T-cell help
rescues exhausted CD8 T cells during chronic viral infection. Proc Natl Acad Sci U S
A, 108, 21182–7.
Blank C, Brown I, Peterson AC, et al. (2004). PD-L1/B7H-1 inhibits the effector phase of
tumor rejection by T cell receptor (TCR) transgenic CD8+ T cells. Cancer Res, 64,
1140–5.
Boon T, van der Bruggen P. (1996). Human tumor antigens recognized by T
lymphocytes. J Exp Med, 183, 725–9.
Brahmer JR, Drake CG, Wollner I, et al. (2010). Phase I study of single-agent anti-
programmed death-1 (MDX-1106) in refractory solid tumors: Safety, clinical activity,
pharmacodynamics, and immunologic correlates. J Clin Oncol, 28, 3167–75.
832 T. Chodon et al.

Brichard V, Van Pel A, Wolfel T, et al. (1993). The tyrosinase gene codes for an antigen
recognized by autologous cytolytic T lymphocytes on HLA-A2 melanomas. J Exp Med,
178, 489–95.
Carey TE, Takahashi T, Resnick LA, et al. (1976). Cell surface antigens of human
malignant melanoma: Mixed hemadsorption assays for humoral immunity to
cultured autologous melanoma cells. Proc Natl Acad Sci U S A, 73, 3278–82.
Castle JC, Kreiter S, Diekmann J, et al. (2012). Exploiting the mutanome for tumor
vaccination. Cancer Res, 72, 1081–91.
Cheever MA, Disis ML, Bernhard H, et al. (1995). Immunity to oncogenic proteins.
Immunol Rev, 145, 33–59.
Chen YT, Scanlan MJ, Sahin U, et al. (1997). A testicular antigen aberrantly expressed
in human cancers detected by autologous antibody screening. Proc Natl Acad Sci U S
A, 94, 1914–18.
Chodon T, Comin-Anduix B, Chmielowski B, et al. (2014). Adoptive transfer of MART-1
T-cell receptor transgenic lymphocytes and dendritic cell vaccination in patients with
metastatic melanoma. Clin Cancer Res, 20, 2457–65.
Clemente CG, Mihm Jr MC, Bufalino R, et al. (1996). Prognostic value of tumor
Downloaded by [Universite Laval] at 01:04 24 November 2015

infiltrating lymphocytes in the vertical growth phase of primary cutaneous melan-


oma. Cancer, 77, 1303–10.
Coley WB. (1991). The treatment of malignant tumors by repeated inoculations of
erysipelas. With a report of ten original cases. 1893. Clin Orthop, 262, 3–11.
Coulie PG, Brichard V, Van Pel A, et al. (1994). A new gene coding for a differentiation
antigen recognized by autologous cytolytic T lymphocytes on HLA-A2 melanomas.
J Exp Med, 180, 35–42.
Davila ML, Riviere I, Wang X, et al. (2014). Efficacy and toxicity management of 19-28z
CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med, 6,
224ra25.
Davis ID, Chen W, Jackson H, et al. (2004). Recombinant NY-ESO-1 protein with
ISCOMATRIX adjuvant induces broad integrated antibody and CD4+ and CD8+
T cell responses in humans. Proc Natl Acad Sci U S A, 101, 10697–702.
Dillman RO, McClay EF, Barth NM, et al. (2015). Dendritic versus tumor cell
presentation of autologous tumor antigens for active specific immunotherapy in
metastatic melanoma: Impact on long-term survival by extent of disease at the time of
treatment. Cancer Biother Radiopharm, 30, 187–94.
Doll R, Kinlen L. (1970). Immunosurveillance and cancer: Epidemiological evidence.
Br Med J, 4, 420–2.
Dong H, Strome SE, Salomao DR, et al. (2002). Tumor-associated B7-H1 promotes T-cell
apoptosis: A potential mechanism of immune evasion. Nat Med, 8, 793–800.
Duan F, Duitama J, Al Seesi S, et al. (2014). Genomic and bioinformatic profiling of
mutational neoepitopes reveals new rules to predict anticancer immunogenicity.
J Exp Med, 211, 2231–48.
Dudley ME, Gross CA, Somerville RP, et al. (2013). Randomized selection design trial
evaluating CD8+-enriched versus unselected tumor-infiltrating lymphocytes for
adoptive cell therapy for patients with melanoma. J Clin Oncol, 31, 2152–9.
Dudley ME, Wunderlich JR, Robbins PF, et al. (2002). Cancer regression and
autoimmunity in patients after clonal repopulation with antitumor lymphocytes.
Science, 298, 850–4.
Dudley ME, Wunderlich JR, Shelton TE, et al. (2003). Generation of tumor-infiltrating
lymphocyte cultures for use in adoptive transfer therapy for melanoma patients.
J Immunother, 26, 332–42.
Dudley ME, Wunderlich JR, Yang JC, et al. (2005). Adoptive cell transfer ther-
apy following non-myeloablative but lymphodepleting chemotherapy for the
treatment of patients with refractory metastatic melanoma. J Clin Oncol, 23,
2346–57.
Dudley ME, Yang JC, Sherry R, et al. (2008). Adoptive cell therapy for patients with
metastatic melanoma: Evaluation of intensive myeloablative chemoradiation pre-
parative regimens. J Clin Oncol, 26, 5233–9.
Cancer Immunotherapy 833
Dunn GP, Bruce AT, Ikeda H, et al. (2002). Cancer immunoediting: From immuno-
surveillance to tumor escape. Nat Immunol, 3, 991–8.
Dunn GP, Old LJ, Schreiber RD. (2004a). The immunobiology of cancer immunosur-
veillance and immunoediting. Immunity, 21, 137–48.
Dunn GP, Old LJ, Schreiber RD. (2004b). The three Es of cancer immunoediting. Annu
Rev Immunol, 22, 329–60.
Funada Y, Noguchi T, Kikuchi R, et al. (2003). Prognostic significance of CD8+ T cell
and macrophage peritumoral infiltration in colorectal cancer. Oncol Rep, 10, 309–13.
Galluzzi L, Senovilla L, Vacchelli E, et al. (2012). Trial watch: Dendritic cell-based
interventions for cancer therapy. Oncoimmunology, 1, 1111–34.
Galon J, Angell HK, Bedognetti D, Marincola, FM. (2013). The continuum of cancer
immunosurveillance: Prognostic, predictive, and mechanistic signatures. Immunity,
39, 11–26.
Gattinoni L, Lugli E, Ji Y, et al. (2011). A human memory T cell subset with stem cell-
like properties. Nat Med, 17, 1290–7.
Gerlinger M, Rowan AJ, Horswell S, et al. (2012). Intratumor heterogeneity
and branched evolution revealed by multiregion sequencing. N Engl J Med, 366,
Downloaded by [Universite Laval] at 01:04 24 November 2015

883–92.
Gil M, Komorowski MP, Seshadri M, et al. (2014). CXCL12/CXCR4 blockade by
oncolytic virotherapy inhibits ovarian cancer growth by decreasing immunosuppres-
sion and targeting cancer-initiating cells. J Immunol, 193, 5327–37.
Gnjatic S, Cai Z, Viguier M, et al. (1998). Accumulation of the p53 protein allows
recognition by human CTL of a wild-type p53 epitope presented by breast carcinomas
and melanomas. J Immunol, 160, 328–33.
Gubin MM, Zhang X, Schuster H, et al. (2014). Checkpoint blockade cancer immuno-
therapy targets tumour-specific mutant antigens. Nature, 515, 577–81.
Guidoboni M, Gafa R, Viel A, et al. (2001). Microsatellite instability and high content of
activated cytotoxic lymphocytes identify colon cancer patients with a favorable
prognosis. Am J Pathol, 159, 297–304.
Hartveit F. (1998). Breast cancer: Poor short-term prognosis in cases with moderate
lymphocyte infiltration at the tumour edge: A preliminary report. Oncol Rep, 5,
423–6.
Huang RY, Eppolito C, Lele S, et al. (2015). LAG3 and PD1 co-inhibitory molecules
collaborate to limit CD8+ T cell signaling and dampen antitumor immunity in a
murine ovarian cancer model’’. Oncotarget (Epub ahead of print, July 23, 2015).
Hunder NN, Wallen H, Cao J, et al. (2008). Treatment of metastatic melanoma with
autologous CD4+ T cells against NY-ESO-1. N Engl J Med, 358, 2698–703.
Hwang WT, Adams SF, Tahirovic E, et al. (2012). Prognostic significance of tumor-
infiltrating T cells in ovarian cancer: A meta-analysis. Gynecol Oncol, 124, 192–8.
Iwai Y, Ishida M, Tanaka Y, et al. (2002). Involvement of PD-L1 on tumor cells in the
escape from host immune system and tumor immunotherapy by PD-L1 blockade.
Proc Natl Acad Sci U S A, 99, 12293–7.
Jager E, Jager D, Knuth A. (1999). CTL-defined cancer vaccines: Perspectives for active
immunotherapeutic interventions in minimal residual disease. Cancer Metastasis
Rev, 18, 143–50.
Kalos M, Levine BL, Porter DL, et al. (2011). T cells with chimeric antigen receptors
have potent antitumor effects and can establish memory in patients with advanced
leukemia. Sci Transl Med, 3, 95ra73.
Kantoff PW, Higano CS, Shore ND, et al. (2010). Sipuleucel-T immunotherapy for
castration-resistant prostate cancer. N Engl J Med, 363, 411–22.
Karlsson M, Marits P, Dahl K, et al. (2010). Pilot study of sentinel-node-based adoptive
immunotherapy in advanced colorectal cancer. Ann Surg Oncol, 17, 1747–57.
Kawakami Y, Eliyahu S, Jennings C, et al. (1995). Recognition of multiple epitopes in
the human melanoma antigen gp100 by tumor-infiltrating T lymphocytes associated
with in vivo tumor regression. J Immunol, 154, 3961–8.
Kelleher RJ, Balu-Iyer S, Loyall JL, et al. (2015). Extracellular vesicles present in
human ovarian tumor microenvironments induce a phosphatidylserine dependent
834 T. Chodon et al.

arrest in the T cell signaling cascade. Cancer Immunol Res (Epub ahead of print, June
25, 2015). doi:10.1158/2326-6066.CIR-15-0086.
Knuth A, Danowski B, Oettgen HF, Old LJ. (1984). T-cell-mediated cytotoxicity against
autologous malignant melanoma: Analysis with interleukin 2-dependent T-cell
cultures. Proc Natl Acad Sci U S A, 81, 3511–15.
Koya RC, Mok S, Comin-Anduix B, et al. (2010). Kinetic phases of distribution and
tumor targeting by T cell receptor engineered lymphocytes inducing robust antitumor
responses. Proc Natl Acad Sci U S A, 107, 14286–91.
Larkin J, Chiarion-Sileni V, Gonzalez R, et al. (2015a). Combined nivolumab
and ipilimumab or monotherapy in untreated melanoma. N Engl J Med,
373, 23–34.
Larkin J, Lao CD, Urba WJ, et al. (2015b). Efficacy and safety of nivolumab in patients
with BRAF V600 mutant and BRAF wild-type advanced melanoma: A pooled analysis
of 4 clinical trials. JAMA Oncol, 1, 433–40.
Lee DW, Kochenderfer JN, Stetler-Stevenson M, et al. (2015). T cells expressing CD19
chimeric antigen receptors for acute lymphoblastic leukaemia in children and young
adults: a phase 1 dose-escalation trial. Lancet, 385, 517–28.
Downloaded by [Universite Laval] at 01:04 24 November 2015

Lennette ET, Winberg G, Yadav M, et al. (1995). Antibodies to LMP2A/2B in EBV-


carrying malignancies. Eur J Cancer, 31A, 1875–8.
Lethe B, Lucas S, Michaux L, et al. (1998). LAGE-1, a new gene with tumor specificity.
Int J Cancer, 76, 903–8.
Linnemann C, van Buuren MM, Bies L, et al. (2015). High-throughput epitope
discovery reveals frequent recognition of neo-antigens by CD4+ T cells in human
melanoma. Nat Med, 21, 81–5.
Lu YC, Yao X, Crystal JS, et al. (2014). Efficient identification of mutated cancer
antigens recognized by T cells associated with durable tumor regressions. Clin Cancer
Res, 20, 3401–10.
MacKie RM, Reid R, Junor B. (2003). Fatal melanoma transferred in a donated kidney
16 years after melanoma surgery. N Engl J Med, 348, 567–8.
Mandruzzato S, Brasseur F, Andry G, et al. (1997). A CASP-8 mutation recognized by
cytolytic T lymphocytes on a human head and neck carcinoma. J Exp Med, 186,
785–93.
Marincola FM, Jaffee EM, Hicklin DJ, Ferrone S. (2000). Escape of human solid tumors
from T-cell recognition: Molecular mechanisms and functional significance. Adv
Immunol, 74, 181–273.
Matloubian M, Concepcion RJ, Ahmed R. (1994). CD4+ T cells are required to
sustain CD8+ cytotoxic T-cell responses during chronic viral infection. J Virol, 68,
8056–63.
Matsuzaki J, Gnjatic S, Mhawech-Fauceglia P, et al. (2010). Tumor-infiltrating NY-
ESO-1-specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human
ovarian cancer. Proc Natl Acad Sci U S A, 107, 7875–80.
Matsuzaki J, Tsuji T, Luescher I, et al. (2014). Nonclassical antigen-processing
pathways are required for MHC class II-restricted direct tumor recognition by
NY-ESO-1-specific CD4(+) T cells. Cancer Immunol Res, 2, 341–50.
Maude SL, Frey N, Shaw PA, et al. (2014). Chimeric antigen receptor T cells for
sustained remissions in leukemia. N Engl J Med, 371, 1507–17.
Melief CJ, van der Burg SH. (2008). Immunotherapy of established (pre)malignant
disease by synthetic long peptide vaccines. Nat Rev Cancer, 8, 351–60.
Mikucki ME, Fisher DT, Matsuzaki J, et al. (2015). Non-redundant requirement for
CXCR3 signalling during tumoricidal T-cell trafficking across tumour vascular
checkpoints. Nat Commun, 6, 7458.
Morgan RA, Dudley ME, Wunderlich JR, et al. (2006). Cancer regression in patients
after transfer of genetically engineered lymphocytes. Science, 314, 126–9.
Naito Y, Saito K, Shiiba K, et al. (1998). CD8+ T cells infiltrated within cancer cell
nests as a prognostic factor in human colorectal cancer. Cancer Res, 58, 3491–4.
Nauts HC, McLaren JR. (1990). Coley toxins—the first century. Adv Exp Med Biol, 267,
483–500.
Cancer Immunotherapy 835
Odunsi K, Matsuzaki J, Karbach J, et al. (2012). Efficacy of vaccination with
recombinant vaccinia and fowlpox vectors expressing NY-ESO-1 antigen in ovarian
cancer and melanoma patients. Proc Natl Acad Sci U S A, 109, 5797–802.
Odunsi K, Qian F, Matsuzaki J, et al. (2007). Vaccination with an NY-ESO-1 peptide of
HLA class I/II specificities induces integrated humoral and T cell responses in
ovarian cancer. Proc Natl Acad Sci U S A, 104, 12837–42.
Old LJ. (2001). Cancer/testis (CT) antigens—a new link between gametogenesis and
cancer. Cancer Immun, 1, 1.
Old LJ. (2008). Cancer vaccines: An overview. Cancer Immun, 8 (Suppl. 1), 1.
Qiao J, Kottke T, Willmon C, et al. (2008a). Purging metastases in lymphoid organs
using a combination of antigen-nonspecific adoptive T cell therapy, oncolytic
virotherapy and immunotherapy. Nat Med, 14, 37–44.
Qiao J, Wang H, Kottke T, et al. (2008b). Loading of oncolytic vesicular stomatitis virus
onto antigen-specific T cells enhances the efficacy of adoptive T-cell therapy of tumors.
Gene Ther, 15, 604–16.
Rizvi NA, Hellmann MD, Snyder A, et al. (2015). Cancer immunology. Mutational
landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer.
Downloaded by [Universite Laval] at 01:04 24 November 2015

Science, 348, 124–8.


Robbins PF, El-Gamil M, Li YF, et al. (1996). A mutated beta-catenin gene encodes a
melanoma-specific antigen recognized by tumor infiltrating lymphocytes. J Exp Med,
183, 1185–92.
Robbins PF, Morgan RA, Feldman SA, et al. (2011). Tumor regression in patients with
metastatic synovial cell sarcoma and melanoma using genetically engineered
lymphocytes reactive with NY-ESO-1. J Clin Oncol, 29, 917–24.
Rosenberg SA, Restifo NP, Yang JC, et al. (2008). Adoptive cell transfer: A clinical path
to effective cancer immunotherapy. Nat Rev Cancer, 8, 299–308.
Sahin U, Tureci O, Schmitt H, et al. (1995). Human neoplasms elicit multiple specific
immune responses in the autologous host. Proc Natl Acad Sci U S A, 92, 11810–13.
Sato E, Olson SH, Ahn J, et al. (2005). Intraepithelial CD8+ tumor-infiltrating
lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable
prognosis in ovarian cancer. Proc Natl Acad Sci U S A, 102, 18538–43.
Savina A, Jancic C, Hugues S, et al. (2006). NOX2 controls phagosomal pH to regulate
antigen processing during crosspresentation by dendritic cells. Cell, 126, 205–18.
Scanlan MJ, Altorki NK, Gure AO, et al. (2000). Expression of cancer-testis antigens in
lung cancer: Definition of bromodomain testis-specific gene (BRDT) as a new CT gene,
CT9. Cancer Lett, 150, 155–64.
Scanlan MJ, Chen YT, Williamson B, et al. (1998). Characterization of human colon
cancer antigens recognized by autologous antibodies. Int J Cancer, 76, 652–8.
Schoenberger SP, Toes RE, van der Voort EI, et al. (1998). T-cell help for cytotoxic
T lymphocytes is mediated by CD40-CD40L interactions. Nature, 393, 480–3.
Schumacher K, Haensch W, Roefzaad C, Schlag PM. (2001). Prognostic significance of
activated CD8(+) T cell infiltrations within esophageal carcinomas. Cancer Res, 61,
3932–6.
Senovilla L, Vacchelli E, Garcia P, et al. (2013). Trial watch: DNA vaccines for cancer
therapy. Oncoimmunology, 2, e23803.
Shankaran V, Ikeda H, Bruce AT, et al. (2001). IFNgamma and lymphocytes prevent
primary tumour development and shape tumour immunogenicity. Nature, 410,
1107–11.
Simpson AJ, Caballero OL, Jungbluth A, et al. (2005). Cancer/testis antigens,
gametogenesis and cancer. Nat Rev Cancer, 5, 615–25.
Slaney CY, Rautela J, Parker BS. (2013). The emerging role of immunosurveillance in
dictating metastatic spread in breast cancer. Cancer Res, 73, 5852–7.
Snyder A, Makarov V, Merghoub T, et al. (2014). Genetic basis for clinical response to
CTLA-4 blockade in melanoma. N Engl J Med, 371, 2189–99.
Street SE, Hayakawa Y, Zhan Y, et al. (2004). Innate immune surveillance of
spontaneous B cell lymphomas by natural killer cells and gammadelta T cells.
J Exp Med, 199, 879–84.
836 T. Chodon et al.

Tindle RW. (1996). Human papillomavirus vaccines for cervical cancer. Curr Opin
Immunol, 8, 643–50.
Tsuji T, Matsuzaki J, Caballero OL, et al. (2012). Heat shock protein 90-mediated
peptide-selective presentation of cytosolic tumor antigen for direct recognition of
tumors by CD4(+) T cells. J Immunol, 188, 3851–8.
Tsuji T, Matsuzaki J, Kelly MP, et al. (2011). Antibody-targeted NY-ESO-1 to mannose
receptor or DEC-205 in vitro elicits dual human CD8+ and CD4+ T cell responses
with broad antigen specificity. J Immunol, 186, 1218–27.
Tumeh PC, Harview CL, Yearley YH, et al. (2014). PD-1 blockade induces responses by
inhibiting adaptive immune resistance. Nature, 515, 568–71.
Tureci O, Sahin U, Zwick C, et al. (1998). Identification of a meiosis-specific protein as a
member of the class of cancer/testis antigens. Proc Natl Acad Sci U S A, 95, 5211–16.
Vacchelli E, Aranda F, Eggermont A, et al. (2014). Trial watch: Tumor-targeting
monoclonal antibodies in cancer therapy. OncoImmunology, 3, e27048.
Vacchelli E, Martins I, Eggermont A, et al. (2012). Trial watch: Peptide vaccines in
cancer therapy. Oncoimmunology, 1, 1557–76.
Van den Eynde BJ, Boon T. (1997). Tumor antigens recognized by T lymphocytes. Int J
Downloaded by [Universite Laval] at 01:04 24 November 2015

Clin Lab Res, 27, 81–6.


van der Bruggen P, Traversari C, Chomez P, et al. (1991). A gene encoding an antigen
recognized by cytolytic T lymphocytes on a human melanoma. Science, 254, 1643–7.
Walter S, Weinschenk T, Stenzl A, et al. (2012). Multipeptide immune response to
cancer vaccine IMA901 after single-dose cyclophosphamide associates with longer
patient survival. Nat Med, 18, 1254–61.
Wang LX, Shu S, Disis ML, Plautz GE. (2007). Adoptive transfer of tumor-primed,
in vitro-activated, CD4+ T effector cells (TEs) combined with CD8+ TEs provides
intratumoral TE proliferation and synergistic antitumor response. Blood, 109,
4865–76.
Wick DA, Webb JR, Nielsen JS, et al. (2014). Surveillance of the tumor mutanome by
T cells during progression from primary to recurrent ovarian cancer. Clin Cancer Res,
20, 1125–34.
Wolfel T, Hauer M, Schneider J, et al. (1995). A p16INK4a-insensitive CDK4 mutant
targeted by cytolytic T lymphocytes in a human melanoma. Science, 269, 1281–4.
Woo SR, Turnis ME, Goldberg MV, et al. (2012). Immune inhibitory molecules LAG-3
and PD-1 synergistically regulate T-cell function to promote tumoral immune escape.
Cancer Res, 72, 917–27.
Zhang L, Conejo-Garcia JR, Katsaros D, et al. (2003). Intratumoral T cells, recurrence,
and survival in epithelial ovarian cancer. N Engl J Med, 348, 203–13.
Zsiros E, Tsuji T, Odunsi K. (2015). Adoptive T-cell therapy is a promising salvage
approach for advanced or recurrent metastatic cervical cancer. J Clin Oncol, 33,
1521–2.

You might also like