SlideShare une entreprise Scribd logo
1  sur  9
Télécharger pour lire hors ligne
REVIEW ARTICLE – TRANSLATIONAL RESEARCH AND BIOMARKERS
An Update on Immunotherapy for Solid Tumors: A Review
Toan Pham, MBBS, BMedSc, PGDipSurgAnat, FRACS1,2,3,4
, Sara Roth, BSc, MSc, PhD1
,
Joseph Kong, MBChB, MS, PhD, FRACS1,2,3,4
, Glen Guerra, MBBS, PGDipSurgAnat, FRACS1,2,3,4
,
Vignesh Narasimhan, MBChB, FRACS1,2,3,4
, Lloyd Pereira, BSc (Hons), PhD1
, Jayesh Desai, MBBS, FRACP3
,
Alexander Heriot, MB, BChir, MA, MD, MBA, FRACS, FRCS (Gen.), FRCSEd, FACS, GAICD1,2,3,4
,
and Robert Ramsay, BSc (Hons), PhD1,5
1
Divisions of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; 2
Cancer Surgery, Melbourne,
VIC, Australia; 3
Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia;
4
Department of Surgery, University of Melbourne, Melbourne, VIC, Australia; 5
Department of Pathology, University of
Melbourne, Melbourne, VIC, Australia
ABSTRACT In recent years, it has been demonstrated
that immunotherapy is an effective strategy for the man-
agement of solid tumors. The origins of immunotherapy
can be traced back to the work of William Coley, who
elicited an immune response against sarcoma by injecting
patients with a mixture of dead bacteria. Significant pro-
gress has been made since, with immune markers within
the tumor now being used as predictors of cancer prognosis
and manipulated to improve patient survival. While surgery
remains central to the management of most patients with
solid malignancies, it is important that surgeons consider
the different immunotherapy strategies that can be
employed to manage disease. Here, we highlight how the
immune system influences tumorigenesis and bring atten-
tion to how current and future immunotherapies can serve
as an adjunct to surgery.
Cancer is the leading cause of mortality worldwide,
being responsible for 8.8 million deaths in 2015.1
Despite
advances in prevention, screening, and diagnosis of cancer,
the landscape of cancer treatment has been altered by the
advent of immunotherapy, offering improved survival in
several solid cancers and establishing itself as a new
therapeutic modality.2
The concept of exploiting the host immune system to
eradicate cancer was first conceived by William Coley. In
pioneering work, a mixture of attenuated bacteria was
injected into patients in order to elicit an immune-mediated
response against sarcoma.3,4
However, significant advances
in cancer immunology have only been achieved in recent
years, with immunotherapy demonstrating clear efficacy in
clinical trials, and a synergistic effect when used in
combination.5,6
In this review, we delineate the relationship between
cancer and the immune response and discuss how this has
been used as a basis for immunotherapy. As personalized
medicine begins to enter the domain of all oncology spe-
cialties, an awareness of available therapies is important,
allowing surgeons to provide the most up-to-date care for
their patients.
THE IMMUNE SYSTEM AND PROGNOSIS:
A NOVEL STAGING SYSTEM
Our immune system continuously protects us against
environmental pathogens and malignant cells (Fig. 1).
However, malignant cells can acquire the capacity to evade
the action of the immune system and form cancer7
.
A seminal publication by Galon et al.8
demonstrated that
low infiltrating cytotoxic (CD8?
) and memory (CD45RO?
)
lymphocytes predict early relapse in colon cancer (CC). This
led to a new scoring system called Immunoscore (IS),9–11
which has been evaluated in 1336 stage I–III CC patients
and further validated in 2681 patients by an international
Toan Pham and Sara Roth have contributed equally to this study and
are co-first authors.
Ó Society of Surgical Oncology 2018
First Received: 27 February 2018
T. Pham, MBBS, BMedSc, PGDipSurgAnat, FRACS
e-mail: toan.pham@petermac.org
Ann Surg Oncol
https://doi.org/10.1245/s10434-018-6658-4
consortium. Both studies concluded that high IS is associ-
ated with longer recurrence-free survival.11,12
Moreover, IS
was able to identify high-risk patients (low IS) within
American Joint Committee on Cancer (AJCC) stage II CC,
thus making IS a powerful biomarker tool to stratify patients
to adjuvant chemotherapy. IS was translated to other tumor
types including gastric cancer13,14
and melanoma,15
in
which patients within the same tumor–node–metastasis
(TNM) stage could be stratified with enhanced accuracy to
predict long-term outcomes. Hence, IS is expected to
become a new component of the AJCC TNM system.
CANCER IMMUNOTHERAPY STRATEGIES
In cancer immunotherapy, the host immune system is
used to target and eradicate cancer cells.3,16
Tumors
express neoantigens that can be recognized as foreign by
the immune system and increase the level of T-lympho-
cytes infiltrating the tumor (TILs). This relationship can be
further described by the concept of immunoediting, which
forms the basis of modern cancer immunology research.17
In this paradigm, elimination, equilibrium, and escape are
key components. Elimination is the recognition and
destruction of tumor cells by the immune system. Subse-
quently, through immunoediting [e.g., expression of
immune checkpoints, loss of major histocompatibility
complex (MHC) expression, and/or upregulation of
T-regulatory cells], tumors create a sanctuary against the
immune system, thereby establishing an equilibrium
between tumor growth and immune destruction. In this
context, tumors escape immune control when the degree of
immunoediting allows for unchecked proliferation. This
model is underpinned by the close interaction between the
immune system and cancer cells, thus our understanding of
this relationship is an important component in the devel-
opment and considered use of immunotherapy. An outline
of the main classes of immunotherapy is shown in Fig. 2
and further described under each subheading below.
Immune Checkpoint Inhibitors: Reactivation of Tumor-
Infiltrating T-Cells
Immune checkpoints are the body’s natural brakes to
dampen the immune response and prevent autoimmunity.
Tumor cells can exploit this avenue to escape the immune
system by expressing complementary molecules that
interact with T-cells, rendering them incapable of killing.
Hence, these immune checkpoints [e.g., programmed cell
death (PD)-1, PD-L1, cytotoxic T lymphocyte-associated
antigen (CTLA)-4] form therapeutic targets.
The first immune checkpoint inhibitor (ICI) discovered
was ipilimumab (Bristol-Myers Squibb), an anti-CTLA-4
Innate Immune System
Microbes
Complement
system
Dendritic cell B cells Antibodies
T cells
Effector T cells
Blood vessel
NK cell
Infected/
transformed cell
Infected/
transformed cell
Granulocytes Monocytes/
Macrophages
Skin barrier
Hours Days
Adaptive Immune System
FIG. 1 Overview of the immune system: the immune system is
divided into the innate and adaptive arms. The innate system responds
rapidly to a conserved repertoire of pathogenic antigens, whereas the
adaptive system mounts a specific and memory response that initially
requires 4–7 days to develop
T. Pham et al.
antibody.18
It was first approved for the treatment of
unresectable metastatic melanoma, and the results were
encouraging with median overall survival of 10.1 months
versus 6.4 months in the control group.18
This equated to
improved 1- and 2-year survival rates of 46 versus 25 %
and 24 versus 14 %, respectively.18
Despite its promising
therapeutic efficacy, there were significant side effects,
including diarrhea (41.4%), colitis (15.9%), and endocrine
disorders (37.6%), and five on-trial mortalities.19
As such,
ipilimumab is now used as second-line therapy or in
combination with another ICI.
Pembrolizumab (Merck & Co.) and nivolumab (Bristol-
Myers Squibb) are the first two Food and Drug Adminis-
tration (FDA)-approved ICIs that block the interaction
between PD-1 (expressed by T-cells) and its ligands PD-L1
and PD-L2 (expressed by tumor and myeloid cells).20
These therapies have similar efficacy and long-term out-
comes to anti-CTLA-4 therapy but without significant side
effects,21
thus gaining FDA approval following landmark
clinical trials for melanoma (KEYNOTE-002),22
non-small
cell lung cancer (KEYNOTE-010),23
microsatellite unsta-
ble/mismatch repair gene deficient colorectal cancer,24
and
gastric cancer (KEYNOTE-059).25
Recently, combination anti-PD-1 and anti-CTLA-4
therapies in treatment-naı̈ve metastatic melanoma
(CHECKMATE-067)5
have demonstrated a synergistic
survival advantage over either monotherapy.5
Conse-
quently, pembrolizumab is now being tested with a variety
of other drugs, including molecular targeted therapies, such
as dabrafenib and trametinib, in BRAF-mutant advanced
melanoma (KEYNOTE-022).26
Expanding an Existing Immune Response: Adoptive
Cellular Therapy
Another emerging immunotherapy is adoptive cellular
therapy (ACT), where tumor-specific lymphocytes are
extracted from peripheral blood or resected tumors and
expanded ex vivo. These lymphocytes are then reintroduced
into the patient with or without systemic lymphodepletion
therapy. The Surgery Branch of the National Cancer Insti-
tute, led by Steven Rosenberg, has been at the forefront of
this therapy, with first successes in melanoma27,28
and cur-
rent clinical trials targeting other malignancies including
head and neck,29,30
renal cell,31
and gynecological can-
cers.32,33
Recent trials have explored the combination of
ACT with BRAF inhibitor34
and PD-1 ICI.35
ACT has even
led to an additional indication for metastasectomy which was
once considered futile, now a potential source for tumor-
specific lymphocytes used in ACT.36
a b
Dying
tumor cell
Granzyme
& perforin
Cytotoxic
T-cell
Cytotoxic
T-cell
Cytotoxic
T-cell
Cytotoxic
T-cell
Cytotoxic
T-cell
NK cell
CAR T -cell
CAR T -cell
Antigen
presenting cell
Oncolytic
virus
Radiation
Neoantigen induced
cytotoxic T-cell
Cytotoxic
T-cell
MDSC
Treg
MHC class I
presentation
Resistant
tumor cell
Tumor
surface protein
Immune
checkpoint
receptor/
ligand
interaction
Immune
checkpoint
inhibitor
Vaccine induced
cytotoxic T-cell
Recognition and Elimination
Of Malignant Cell
Radiation or Oncolytic Virus
induced Tumor Neoantigen
Adoptive CAR T-cell therapy
or Cancer Vaccination
Administration of Immune
Checkpoint Blockers
Adoptive CAR T-cell or NK cell
therapy
Depletion or Suppression of
MDSC and Tregs
Low Immunogenicity of
presented Antigen
Expression of Immune
Checkpoints
Downregulation of MHC
Class I Expression
Suppression by MDSC or
Tregs
c
NORMAL
IMMUNOSUPPRESION
THERAPY
OPTONS
FIG. 2 Tumor ‘‘escape’’ mechanisms (pink) and associated
immunotherapy counterstrategies (green): a normal recognition of
transformed cells by MHC class I presentation of tumor-associated
antigen and subsequent destruction by specific cytotoxic T-cells
(blue); b malignant cells escape immune recognition by low
immunogenicity of the presented antigen, expression of immune
checkpoints, downregulation of MHC class I, and generation of an
immunosuppressive microenvironment; c current immunotherapies
counteracting these evasion mechanisms via increasing the tumor-
specific antigens (radiation, vaccination, oncolytic viruses), immune
checkpoint blockade, CAR T-cells, and NK cell therapies, and
elimination of immunosuppressive microenvironment
Immunotherapy for Solid Tumors: A Review
Enhancing the Immunological Profile of Solid Tumors
Tumors with sparse TILs typically have low mutational
burden and consequently low neoantigen load. In order to
address this subset of challenging cancers, immune-based
strategies such as radiotherapy, oncolytic viruses, and
cancer vaccines have been developed to enhance the
immune recognition of these tumors.
Radiotherapy Radiotherapy can induce antigen expression
through tumor cell death and upregulation of MHC class I on
surviving tumor cells, both increasing their susceptibility to
cytotoxic T-cell killing.37
Single high-dose (5–10 Gy) or
hypofractionated (3 9 8 Gy) radiotherapy induces a
proinflammatory response that may result in an abscopal
effect,38
which is most pronounced in immunogenic tumors
including renal cell carcinoma (RCC), melanoma, and
hepatocellular carcinoma (HCC).39
Several clinical trials
are now investigating the combination of radiotherapy and
immune checkpoint inhibitors,40
because radiation has been
associated with PD-L1 upregulation.41
Oncolytic Viruses Oncolytic viruses (OV) are genetically
engineered or naturally occurring viruses that selectively
replicate in and kill cancer cells without collateral damage
to normal tissues. The release of antigens from the lysed
cancer cell enhances immune recognition and triggers
further immune-mediated destruction.42
The tumor
specificity of OV can be attributed to multiple factors
including the virus’s tropism for specific tissue type,
downregulation of the antiviral response of cancer cells,
and/or engineered enhancements to the virus to exploit the
different intracellular milieu of cancer cells.43
OV can also
be ‘‘armed’’ with genes encoding for immunodulators such
as granulocyte-macrophage colony-stimulating factor
(GM-CSF) or tumor-associated antigens to further
enhance their function.43
The first OV approved by the FDA was T-Vec, an
oncolytic herpes simplex virus type 1 armed with GM-
CSF, for the treatment of advanced melanoma.44
Further
oncolytic viruses such as Pexa-Vec against hepatocellular
carcinoma, G47D against glioblastoma and prostate cancer,
and CG0070 against bladder cancer show promising
results.43,45
The main disadvantage of OV is the acquired specific
immunity against the virus that effectively neutralizes any
repeat therapy. However, even partial responses leading to
downstaging may facilitate surgical resection and ICI
therapy.
Cancer Vaccines Cancer vaccines represent a different
approach of generating tumor-specific T-cells in poorly
immunogenic tumors, against either neoantigens or
differentially overexpressed self-antigens. Most cancer
vaccines to date have demonstrated limited clinical
benefit,46
which can be attributed to immune-suppressive
mechanisms within the tumor microenvironment (TME) if
administered as monotherapy. The success of ICI has led to
a renaissance of combination therapy cancer vaccine
clinical trials, with promising results.47,48
A few
encouraging examples include the FDA-approved
Sipuleucel-T (dendritic cell vaccine) for castrate-resistant
prostate cancer,49
TG01 (mutant K-ras peptide vaccine) for
pancreatic cancer,50
and two personalized neoantigen
vaccines for melanoma.47,48
The TetMYB vaccine, a
DNA plasmid encoding a modified MYB oncoprotein
fused with tetanus antigen to break self-tolerance, is being
tested against advanced colorectal and adenoid cystic
carcinoma (NCT03287427).51
NK Cell and CAR T-Cell Therapy: Lifting the Cloak
of Invisibility
Another immune evasion technique employed by tumor
cells is downregulation of MHC class I molecules, thus
rendering the tumor invisible to cytotoxic T-cells.52,53
This
adaptation has been observed in patients treated with ICI
and cancer vaccines,48,54
and may be countered by natural
killer (NK) and chimeric antigen receptor (CAR) T-cell
therapies, as these two immune cells do not rely on MHC
class I presentation for tumor recognition.
In early-phase clinical trials, NK cell therapy has proven
to be safe and well tolerated, albeit with limited success, in
non-small cell lung,55,56
gastrointestinal,57
breast, ovar-
ian,58
and renal cell cancers, and melanoma.59
The intrinsic
lack of tumor infiltration by NK cells is often attributed to
suppressive TME. This can be mitigated by nanoparticles
that attract and expand NK cells within the tumor60
or
genetically modified NK cells.61
CAR T-cells are autologous T-cells that have undergone
ex vivo genetic modification to express a tumor-specific
hybrid receptor. Prominent successes have been achieved
in hematological malignancies, including the use of anti-
CD19 CAR T-cells in non-Hodgkin’s B cell lymphoma and
lymphoblastic leukemia, which gained FDA approval in
2017.62–64
Current target antigens include epidermal
growth factor receptor (EGFR), human epidermal growth
factor receptor 2 (HER-2), mesothelin, and carcinoembry-
onic antigen (CEA)65
on solid tumors.
Early-phase trials have shown CAR T-cells to be safe
and feasible in a variety of solid tumors, including non-
small cell lung cancer,66
neurological malignancies,67–69
breast,70
pancreatic,71,72
and metastatic colon cancer,73
and
sarcoma.74,75
Furthermore, enhanced efficacy in combina-
tion with ICI has been observed against melanoma,
Hodgkin’s lymphoma, and non-small cell lung cancers.76
T. Pham et al.
T-Regulatory and Myeloid-Derived Suppressor Cells:
Antisuppressor Therapy
T-regulatory (Treg) and myeloid-derived suppressor
cells (MDSCs) are partly responsible for the immunosup-
pressive TME,77
thus representing a potential avenue in
cancer immunotherapy.
FoxP3-expressing Treg cells induce self-tolerance and
prevent autoimmunity. Tumors can exploit Tregs to their
advantage, and current therapeutic strategies are focusing
on depleting these cells.78
Two such therapies are dacli-
zumab (IL-2 receptor blocking antibody) and denileukin
diftitox (IL-2:diptheria toxin fusion peptide), which have
demonstrated robust clinical outcomes79
in metastatic
breast cancer80,81
and chemo/immuno-naı̈ve stage IV
melanoma.82
Additionally, the checkpoint inhibitor ipili-
mumab has been reported to deplete tumor-infiltrating
Tregs via antibody-dependent cell-mediated cytotoxicity
(ADCC) in melanoma patients.83
MDSCs are immature myeloid cells that suppress
cytotoxic T-cell function and modulate the activation and
expansion of Tregs by immunosuppressive cytokines,
arginase-1, and reactive oxygen species (ROS) and nitric
oxide (NO).77
Treatments targeting MDSCs were
serendipitously discovered as a by-effect of other cancer
treatments.84
Ipilimumab and vemurafenib (BRAF inhibitor) have
been shown to reverse the immunosuppressive effect of
MDSCs in melanoma.85,86
Sunitinib (tyrosine kinase inhi-
bitor) decreases MDSC levels by reducing the expansion of
monocytic MDSCs and inducing apoptosis of granulocytic
MDSCs,87
being the first-line therapy for metastatic RCC.
All-trans retinoic acid (ATRA) and vitamin D3 promote
differentiation of MDSCs into mature nonsuppressive cells
and have shown benefit in metastatic RCC88
and head and
neck squamous cell carcinoma (SCC),89
respectively.
Sildenafil, tadalafil, and vardenafil (PDE5 inhibitors) have
been shown to inhibit arginase-1 and NO expression and
significantly reduce disease burden in myeloma patients90
and circulating MDSCs in head and neck SCC.91
Addi-
tionally, conventional chemotherapeutic drugs, namely
gemcitabine and 5-fluorouracil, have been found to
decrease MDSC and to improve antitumor immune
responses.92,93
Other Barriers Against Immunotherapy
In addition to the immune-evasive mechanism men-
tioned above, further parts of the TME can adversely affect
immune cell function.
Some tumors create a hostile TME for TILs by depleting
nutrients and oxygen and releasing acidic94
and toxic
metabolites.95
Others promote an immunosuppressive cell
infiltrate by altering the inflammatory cytokine milieu.96
Lastly, modifications of the tumor vasculature such as
downregulation of intercellular adhesion molecule 1
(ICAM-1)97
and altering the tumor stroma98,99
to retard
T-cell migration have been described.
Research into counteracting some of these mechanisms
is currently being done, however there are no clinical trials
to date.
THE RISE OF IMMUNOTHERAPY CLINICAL
TRIALS
An enquiry of the ClinicalTrials.gov database revealed
that, of the 60,903 cancer trials in the entire database, the
search string ‘‘Cancer AND Immunotherapy’’ returned
1994 (* 3%) interventional studies, and of these, 313
(* 16%) have been completed (Table 1). Recent years
have seen a rapid increase in the number of immunotherapy
trials (Fig. 3). Although earlier trials were disappointing,
more recent trials featuring combination therapies have
shown promising results. Thus, there is a need for further
evaluation of upcoming therapies as well as the potential
synergistic effects of existing therapies.
INTEGRATION WITH SURGICAL PRACTICE
Local/Regional Control
The success of treating primary solid malignancies is
highly dependent on the disease stage. Surgery has tradi-
tionally played a role in this setting, with early-stage
disease often cured with a margin-negative resection. In
contrast, in many locally advanced solid tumors, neoadju-
vant therapies such as radiotherapy and/or chemotherapy
have shown benefit in downstaging cancers to enable
resection. This is exemplified in breast cancer, where
5-year survival for stage III patients was similar between
mastectomy (96.3%) and chemoradiotherapy combined
with breast-conserving surgery (90.9%, p = 0.669).100
More recently, chemoimmunotherapy has been used suc-
cessfully to downstage prostate cancer.101
Distant/Metastatic Disease
The presence of metastatic disease has classically
implied an incurable state for patients. Surgery has tradi-
tionally had a limited role, often reserved for control of
symptoms. Chemotherapy and radiotherapy can extend
overall survival in this cohort; however, this requires bal-
anced consideration given the impact on quality of life and
treatment-related morbidity.
Harnessing what immunotherapy promises to offer, we
have the potential to turn advanced cancer into a chronic
Immunotherapy for Solid Tumors: A Review
disease, or state of equilibrium.17
This has been demon-
strated in subgroups of the CheckMate-066 and -067
melanoma trials, where clinical benefit from nivolumab is
seen beyond disease progression for advanced mela-
noma.102
Furthermore, improvement in survival has been
achieved with dual checkpoint inhibition.5
TABLE 1 Phase and modality of completed immunotherapy clinical trials against solid malignancies (compiled from ClinicalTrials.gov;
5/15/2018)
Solid cancer
type
Phase of
clinical trial
(number of
trials*)
Immunotherapy modality (number of trials*)
I II III Cancer
vaccine
Checkpoint
inhibition
Adoptive
cellular
therapy
CAR
T-cell
Oncolytic virus,
gene therapy
Anti-Treg,
anti-MDSC
Cytokine,
targeted therapy
Prostate 17 27 7 39 4 2 3 3
Melanoma 39 41 5 40 6 19 1 2 1 16
Lung 18 30 3 35 5 1 10
Head and neck 11 8 10 2 1 1 5
Neurological 26 20 3 24 1 8 4 3 9
Gynecological 12 15 1 16 3 1 8
Anal 3 2 1 1 1 2
Penile 3 1 1 1 2
Sarcoma 10 8 8 1 4 1 4
Urothelial 7 14 4 9 1 4 1 1 9
Breast 18 18 23 1 6 6
Hepatobiliary 9 4 2 6 4 2 3
Colorectal 10 13 1 17 1 2 1 3
Esophagogastric 6 4 5 1 4
Pancreas 13 11 2 20 1 1 1 3
Total 202 216 28 254 21 58 12 10 4 87
*Where a trial involves more than one therapy, it is counted in duplicate according to the number of immunotherapy modalities used
0
5
10
15
20
25
30
1995 1997 1999 2001 2003 2005 2007 2009 2011 2013 2015
Number
of
Trials
Year of Trial Commencement
The Rise of Cancer Immunotherapy Trials
FIG. 3 Trend showing the rapid
increase in number of started
cancer immunotherapy trials from
1995–2015 (compiled from
ClinicalTrials.gov; 5/15/2018)
T. Pham et al.
When examining published data, there is a changing role
for surgery. Current immunotherapeutic agents are most
effective when there is low to moderate tumor burden given
the limited immunosuppressive factors. Therefore, surgery’s
role is still foremost to achieve cure in early-stage disease,
however what was once considered futile/palliative surgery
now has the potential to offer cure when combined with
immunotherapy. This not only includes resecting tumors
after downstaging, but also removing tumors in some can-
cers, including metastatic deposits, in order to understand the
immune profile of the resistant clone, and for the expansion
of TILs. Both of these methods allow a personalized treat-
ment algorithm to be developed and modified throughout the
patient’s journey, so that the best outcomes can be achieved.
CONCLUSIONS
Immunotherapy is a promising therapeutic avenue that is
rapidly shifting the paradigm of cancer management. This
review is intended to raise awareness about the current
immunotherapeutic strategies. Immunotherapy can provide
an important adjunct to current available therapies to
improve long-term outcomes in previously resistant or
incurable cancers. With its growing use, it is important for
surgeons and surgical oncologists to embrace it, thereby
providing immunotherapy as a treatment options for their
patients.
ACKNOWLEDGMENT Not applicable.
DISCLOSURE All authors are involved in the MYPHISMO clin-
ical trial (NCT03287427).
REFERENCES
1. World Health Organization Cancer Factsheet. 2018; http://www.
who.int/mediacentre/factsheets/fs297/en/. Accessed 18 Feb
2018.
2. Weber JS, O’Day S, Urba W, et al. Phase I/II study of ipili-
mumab for patients with metastatic melanoma. J Clin Oncol.
2008;26(36):5950–56.
3. Coley WB. The treatment of inoperable sarcoma by bacterial
toxins (the mixed toxins of the Streptococcus erysipelas and the
Bacillus prodigiosus). Proc R Soc Med. 1910;3:1–48.
4. Coley WB. The treatment of malignant tumors by repeated
inoculations of erysipelas. With a report of ten original cases.
1893. Clin Orthop Relat Res. 1991(262):3–11.
5. Wolchok JD, Chiarion-Sileni V, Gonzalez R, et al. Overall
survival with combined nivolumab and ipilimumab in advanced
melanoma. N Engl J Med. 2017;377(14):1345–56.
6. Ott PA, Hodi FS, Kaufman HL, Wigginton JM, Wolchok JD.
Combination immunotherapy: a road map. J Immunother Can-
cer. 2017;5:16.
7. Dunn GP, Bruce AT, Ikeda H, Old LJ, Schreiber RD. Cancer
immunoediting: from immunosurveillance to tumor escape. Nat
Immunol. 2002;3(11):991–8.
8. Galon J, Costes A, Sanchez-Cabo F, et al. Type, density, and
location of immune cells within human colorectal tumors predict
clinical outcome. Science. 2006;313(5795):1960–4.
9. Mlecnik B, Tosolini M, Kirilovsky A, et al. Histopathologic-
based prognostic factors of colorectal cancers are associated
with the state of the local immune reaction. J Clin Oncol.
2011;29(6):610–8.
10. Galon J, Mlecnik B, Bindea G, et al. Towards the introduction of
the ‘Immunoscore’ in the classification of malignant tumours. J
Pathol. 2014;232:199–209.
11. Galon J, Mlecnik B, Marliot F, et al. Validation of the Immu-
noscore (IM) as a prognostic marker in stage I/II/III colon
cancer: Results of a worldwide consortium-based analysis of
1,336 patients. J Clin Oncol. 2016;34(15):3500–3500.
12. Pages F, Mlecnik B, Marliot F, et al. International validation of
the consensus Immunoscore for the classification of colon can-
cer: a prognostic and accuracy study. Lancet.
2018;391(10135):2128–39.
13. Fujitani K, Ando M, Sakamaki K, et al. A prospective multi-
center observational study of surgical palliation examining
postoperative quality of life in patients treated for malignant
gastric outlet obstruction caused by incurable advanced gastric
cancer. J Clin Oncol. 2017;35(4):6-6.
14. Qi X, Jiang Y, Zhang Q, et al. Prognostic and predictive value of
immunoscore signature in gastric cancer. J Clin Oncol.
2017;35(15):e15594-e15594.
15. Galon J, Fox BA, Bifulco CB, et al. Immunoscore and
immunoprofiling in cancer: an update from the melanoma and
immunotherapy bridge 2015. J Transl Med. 2016;14(1):273.
16. Pilch YH, Myers GH, Sparks FC, Golub SH. Prospects for the
immunotherapy of cancer. Part I: basic concepts of tumor
immunologyProspects for the immunotherapy of cancer. Part I:
basic concepts of tumor immunology. Curr Probl Surg.
1975;12(1):1–46.
17. Dunn GP, Old LJ, Schreiber RD. The three Es of cancer
immunoediting. Annu Rev Immunol. 2004;22:329–60.
18. Lipson EJ, Drake CG. Ipilimumab: an anti-CTLA-4 antibody for
metastatic melanoma. Clin Cancer Res. 2011;17(22):6958–62.
19. Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival
with ipilimumab in patients with metastatic melanoma. N Engl J
Med. 19 2010;363(8):711–23.
20. Ribas A. Tumor immunotherapy directed at PD-1. N Engl J
Med. 2012;366(26):2517–9.
21. Robert C, Schachter J, Long GV, et al. Pembrolizumab versus
ipilimumab in advanced melanoma. N Engl J Med.
2015;372(26):2521–32.
22. Ribas A, Puzanov I, Dummer R, et al. Pembrolizumab versus
investigator-choice chemotherapy for ipilimumab-refractory
melanoma (KEYNOTE-002): a randomised, controlled, phase 2
trial. Lancet Oncol. 2015;16(8):908–18.
23. Herbst RS, Baas P, Kim DW, et al. Pembrolizumab versus
docetaxel for previously treated, PD-L1-positive, advanced non-
small-cell lung cancer (KEYNOTE-010): a randomised con-
trolled trial. Lancet. 2016;387(10027):1540–50.
24. Le DT, Uram JN, Wang H, et al. PD-1 Blockade in tumors with
mismatch-repair deficiency. N Engl J Med.
2015;372(26):2509–20.
25. Fuchs CS, Doi T, Jang RW-J, et al. KEYNOTE-059 cohort 1:
Efficacy and safety of pembrolizumab (pembro) monotherapy in
patients with previously treated advanced gastric cancer. J Clin
Oncol. 2017;35(15):4003-4003.
26. Ribas A, Hodi FS, Lawrence D, et al. 1216OKEYNOTE-022
update: phase 1 study of first-line pembrolizumab (pembro) plus
dabrafenib (D) and trametinib (T) for BRAF-mutant advanced
melanoma. Vol 282017.
Immunotherapy for Solid Tumors: A Review
27. Dudley ME, Wunderlich J, Nishimura MI, et al. Adoptive
transfer of cloned melanoma-reactive T lymphocytes for the
treatment of patients with metastatic melanoma. J Immunother.
2001;24(4):363–73.
28. Dudley ME, Wunderlich JR, Yang JC, et al. A phase I study of
nonmyeloablative chemotherapy and adoptive transfer of
autologous tumor antigen-specific T lymphocytes in patients
with metastatic melanoma. J Immunother. 2002;25(3):243–51.
29. Zhou P, Tian S, Li J, et al. Paradoxes in thyroid carcinoma
treatment: analysis of the SEER database 2010–2013. Onco-
target. 2017;8(1):345–53.
30. Ostrom QT, Gittleman H, Kruchko C, et al. Completeness of
required site-specific factors for brain and CNS tumors in the
Surveillance, Epidemiology and End Results (SEER) 18 data-
base (2004–2012, varying). J Neurooncol. 2016;130(1):31–42.
31. Liao Z, Rodrigues MC, Poynter JN, Amatruda JF, Rodriguez-
Galindo C, Frazier AL. Risk of second malignant neoplasms in
women and girls with germ cell tumors. Ann Oncol.
2017;28(2):329–32.
32. Yao N, Alcala HE, Anderson R, Balkrishnan R. Cancer dis-
parities in rural Appalachia: incidence, early detection, and
survivorship. J Rural Health. 2017;33(4):375–81.
33. Shah BK, Kandel P, Khanal A. Second primary malignancies in
hepatocellular cancer - A US population-based study. Anti-
cancer Res. 2016;36(7):3511–4.
34. Deniger DC, Kwong ML, Pasetto A, et al. A pilot trial of the
combination of vemurafenib with adoptive cell therapy in
patients with metastatic melanoma. Clin Cancer Res.
2017;23(2):351–62.
35. Bista A, Sharma S, Shah BK. Disparities in receipt of radio-
therapy and survival by age, sex, and ethnicity among patient
with stage I follicular lymphoma. Front Oncol. 2016;6:101.
36. Crompton JG, Klemen N, Kammula US. Metastasectomy for
tumor-infiltrating lymphocytes: an emerging operative indica-
tion in surgical oncology. Ann Surg Oncol. 2018;25(2):565–72.
37. Reits EA, Hodge JW, Herberts CA, et al. Radiation modulates
the peptide repertoire, enhances MHC class I expression, and
induces successful antitumor immunotherapy. J Exp Med.
2006;203(5):1259–71.
38. Haikerwal SJ, Hagekyriakou J, MacManus M, Martin OA,
Haynes NM. Building immunity to cancer with radiation ther-
apy. Cancer Lett. 2015;368(2):198–208.
39. Reynders K, Illidge T, Siva S, Chang JY, De Ruysscher D. The
abscopal effect of local radiotherapy: using immunotherapy to
make a rare event clinically relevant. Cancer Treat Rev.
2015;41(6):503–10.
40. Ko EC, Formenti SC. Radiotherapy and checkpoint inhibitors: a
winning new combination? Ther Adv Med Oncol.
2018;10:1758835918768240.
41. Dovedi SJ, Illidge TM. The antitumor immune response gen-
erated by fractionated radiation therapy may be limited by tumor
cell adaptive resistance and can be circumvented by PD-L1
blockade. Oncoimmunology. 2015;4(7):e1016709.
42. Lawler SE, Speranza MC, Cho CF, Chiocca EA. Oncolytic
viruses in cancer treatment: a review. JAMA Oncol.
2017;3(6):841–9.
43. Fukuhara H, Ino Y, Todo T. Oncolytic virus therapy: A new era
of cancer treatment at dawn. Cancer Sci. 2016;107(10):1373–9.
44. Kaufman HL, Bines SD. OPTIM trial: a Phase III trial of an
oncolytic herpes virus encoding GM-CSF for unresectable stage
III or IV melanoma. Future Oncol. Jun 2010;6(6):941–9.
45. Breitbach CJ, Moon A, Burke J, Hwang TH, Kirn DH. A phase
2, open-label, randomized study of Pexa-Vec (JX-594) admin-
istered by intratumoral injection in patients with
unresectable primary hepatocellular carcinoma. Methods Mol
Biol. 2015;1317:343–57.
46. Dalgleish AG, Whelan MA. Cancer vaccines as a therapeutic
modality: The long trek. Cancer Immunol Immunother.
2006;55(8):1025–32.
47. Ott PA, Hu Z, Keskin DB, et al. An immunogenic personal
neoantigen vaccine for patients with melanoma. Nature.
2017;547(7662):217–21.
48. Sahin U, Derhovanessian E, Miller M, et al. Personalized RNA
mutanome vaccines mobilize poly-specific therapeutic immunity
against cancer. Nature. 2017;547(7662):222–6.
49. Kantoff PW, Higano CS, Shore ND, et al. Sipuleucel-T
immunotherapy for castration-resistant prostate cancer. N Engl J
Med. 2010;363(5):411–22.
50. Weden S, Klemp M, Gladhaug IP, et al. Long-term follow-up of
patients with resected pancreatic cancer following vaccination
against mutant K-ras. Int J Cancer. 2011;128(5):1120–8.
51. MYPHISMO: MYB and PD-1 Immunotherapies against multi-
ple oncologies trial. https://ClinicalTrials.gov/show/
NCT03287427.
52. Garrido F, Perea F, Bernal M, Sanchez-Palencia A, Aptsiauri N,
Ruiz-Cabello F. The escape of cancer from T cell-mediated
immune surveillance: HLA class I loss and tumor tissue archi-
tecture. Vaccines (Basel). 2017;5(1):7.
53. Garrido F, Ruiz-Cabello F, Aptsiauri N. Rejection versus
escape: the tumor MHC dilemma. Cancer Immunol Immunother.
2017;66(2):259–71.
54. Zaretsky JM, Garcia-Diaz A, Shin DS, et al. Mutations associ-
ated with acquired resistance to PD-1 blockade in melanoma. N
Engl J Med. 2016;375(9):819–29.
55. Tonn T, Schwabe D, Klingemann HG, et al. Treatment of
patients with advanced cancer with the natural killer cell line
NK-92. Cytotherapy. 2013;15(12):1563–70.
56. Yang YJ, Park JC, Kim HK, Kang JH, Park SY. A trial of
autologous ex vivo-expanded NK cell-enriched lymphocytes
with docetaxel in patients with advanced non-small cell lung
cancer as second- or third-line treatment: phase IIa study. An-
ticancer Res. 2013;33(5):2115–22.
57. Sakamoto N, Ishikawa T, Kokura S, et al. Phase I clinical trial of
autologous NK cell therapy using novel expansion method in
patients with advanced digestive cancer. J Transl Med.
2015;13:277.
58. Geller MA, Cooley S, Judson PL, et al. A phase II study of
allogeneic natural killer cell therapy to treat patients with
recurrent ovarian and breast cancer. Cytotherapy.
2011;13(1):98–107.
59. Arai S, Meagher R, Swearingen M, et al. Infusion of the allo-
geneic cell line NK-92 in patients with advanced renal cell
cancer or melanoma: a phase I trial. Cytotherapy.
2008;10(6):625–32.
60. Ammam M. Immunotherapy based on natural killer cells may
soon begin clinical studies. In: M. Nace (Ed) Immuno-Oncology
News. Dallas, TX: BioNews Services, LLC; 2017.
61. Rezvani K, Rouce R, Liu E, Shpall E. Engineering natural killer
cells for cancer immunotherapy. Mol Ther. 2017;25(8):1769–81.
62. Porter DL, Kalos M, Zheng Z, Levine B, June C. Chimeric
antigen receptor therapy for B-cell malignancies. J Cancer.
2011;2:331–2.
63. Porter DL, Hwang WT, Frey NV, et al. Chimeric antigen
receptor T cells persist and induce sustained remissions in
relapsed refractory chronic lymphocytic leukemia. Sci Transl
Med. 2015;7(303):303ra139.
64. FDA US. FDA approval brings first gene therapy to the United
States. CAR T-cell therapy approved to treat certain children
and young adults with B-cell acute lymphoblastic leukemia:
FDA, USA; 2017.
65. Yu S, Li A, Liu Q, et al. Chimeric antigen receptor T cells: a
novel therapy for solid tumors. J Hematol Oncol. 2017;10(1):78.
T. Pham et al.
66. Feng K, Guo Y, Dai H, et al. Chimeric antigen receptor-modi-
fied T cells for the immunotherapy of patients with EGFR-
expressing advanced relapsed/refractory non-small cell lung
cancer. Sci China Life Sci. 2016;59(5):468–79.
67. A phase I trial of T cells expressing an anti-GD2 chimeric
antigen receptor in children and young adults with GD2?
solid
tumors. https://ClinicalTrials.gov/show/NCT02107963.
68. CMV-specific cytotoxic T lymphocytes expressing CAR tar-
geting HER2 in patients with gbm. https://ClinicalTrials.gov/sh
ow/NCT01109095.
69. Pilot study of autologous anti-EGFRvIII CAR T cells in recur-
rent glioblastoma multiforme. https://ClinicalTrials.gov/show/
NCT02844062.
70. Kawalec P, Paszulewicz A, Holko P, Pilc A. Sipuleucel-T
immunotherapy for castration-resistant prostate cancer. A sys-
tematic review and meta-analysis. Arch Med Sci.
2012;8(5):767–75.
71. A study of mesothelin redirected autologous T cells for
advanced pancreatic carcinoma. https://ClinicalTrials.gov/show/
NCT02706782.
72. Pilot study of autologous T-cells in patients with metastatic
pancreatic cancer. https://ClinicalTrials.gov/show/
NCT02465983.
73. Katz SC, Burga RA, McCormack E, et al. Phase I hepatic
immunotherapy for metastases study of intra-arterial chimeric
antigen receptor-modified T-cell therapy for CEA?
liver
metastases. Clin Cancer Res. 2015;21(14):3149–59.
74. Ahmed N, Brawley VS, Hegde M, et al. Human epidermal
growth factor receptor 2 (HER2)-specific chimeric antigen
receptor-modified T cells for the immunotherapy of HER2-
positive sarcoma. J Clin Oncol. 2015;33(15):1688–96.
75. Her2 chimeric antigen receptor expressing T cells in advanced
sarcoma. https://ClinicalTrials.gov/show/NCT00902044.
76. Hegde UP, Mukherji B. Current status of chimeric antigen
receptor engineered T cell-based and immune checkpoint
blockade-based cancer immunotherapies. Cancer Immunol
Immunother. 2017;66(9):1113–21.
77. Lindau D, Gielen P, Kroesen M, Wesseling P, Adema GJ. The
immunosuppressive tumour network: myeloid-derived suppres-
sor cells, regulatory T cells and natural killer T cells.
Immunology. 2013;138(2):105–15.
78. Tanaka A, Sakaguchi S. Regulatory T cells in cancer
immunotherapy. Cell Res. 2017;27(1):109–18.
79. Liu C, Workman CJ, Vignali DA. Targeting regulatory T cells in
tumors. FEBS J. 2016;283(14):2731–48.
80. Rech AJ, Vonderheide RH. Clinical use of anti-CD25 antibody
daclizumab to enhance immune responses to tumor antigen
vaccination by targeting regulatory T cells. Ann N Y Acad Sci.
2009;1174:99–106.
81. Rech AJ, Mick R, Martin S, et al. CD25 blockade depletes and
selectively reprograms regulatory T cells in concert with
immunotherapy in cancer patients. Sci Transl Med.
2012;4(134):134–62.
82. Telang S, Rasku MA, Clem AL, et al. Phase II trial of the
regulatory T cell-depleting agent, denileukin diftitox, in patients
with unresectable stage IV melanoma. BMC Cancer.
2011;11:515.
83. Romano E, Kusio-Kobialka M, Foukas PG, et al. Ipilimumab-
dependent cell-mediated cytotoxicity of regulatory T cells
ex vivo by nonclassical monocytes in melanoma patients. Proc
Natl Acad Sci U S A. 2015;112(19):6140–5.
84. Tobin RP, Davis D, Jordan KR, McCarter MD. The clinical
evidence for targeting human myeloid-derived suppressor cells
in cancer patients. J Leukoc Biol. 2017;102(2):381–91.
85. Schilling B, Sucker A, Griewank K, et al. Vemurafenib reverses
immunosuppression by myeloid derived suppressor cells. Int J
Cancer. 2013;133(7):1653–63.
86. de Coana YP, Wolodarski M, Poschke I, et al. Ipilimumab
treatment decreases monocytic MDSCs and increases CD8
effector memory T cells in long-term survivors with advanced
melanoma. Oncotarget. 2017;8(13):21539–53.
87. Ko JS, Rayman P, Ireland J, et al. Direct and differential sup-
pression of myeloid-derived suppressor cell subsets by sunitinib
is compartmentally constrained. Cancer Res.
2010;70(9):3526–36.
88. Mirza N, Fishman M, Fricke I, et al. All-trans-retinoic acid
improves differentiation of myeloid cells and immune response
in cancer patients. Cancer Res. 2006;66(18):9299–307.
89. Lathers DM, Clark JI, Achille NJ, Young MR. Phase 1B study to
improve immune responses in head and neck cancer patients
using escalating doses of 25-hydroxyvitamin D3. Cancer
Immunol Immunother. 2004;53(5):422–30.
90. Noonan KA, Ghosh N, Rudraraju L, Bui M, Borrello I. Tar-
geting immune suppression with PDE5 inhibition in end-stage
multiple myeloma. Cancer Immunol Res. 2014;2(8):725–31.
91. Weed DT, Vella JL, Reis IM, et al. Tadalafil reduces myeloid-
derived suppressor cells and regulatory T cells and promotes
tumor immunity in patients with head and neck squamous cell
carcinoma. Clin Cancer Res. 2015;21(1):39–48.
92. Annels NE, Shaw VE, Gabitass RF, et al. The effects of gem-
citabine and capecitabine combination chemotherapy and of
low-dose adjuvant GM-CSF on the levels of myeloid-derived
suppressor cells in patients with advanced pancreatic cancer.
Cancer Immunol Immunother. 2014;63(2):175–83.
93. Kanterman J, Sade-Feldman M, Biton M, et al. Adverse
immunoregulatory effects of 5FU and CPT11 chemotherapy on
myeloid-derived suppressor cells and colorectal cancer out-
comes. Cancer Res. 2014;74(21):6022–35.
94. Fischer K, Hoffmann P, Voelkl S, et al. Inhibitory effect of
tumor cell-derived lactic acid on human T cells. Blood.
2007;109(9):3812–9.
95. Chang CH, Qiu J, O’Sullivan D, et al. Metabolic competition in
the tumor microenvironment is a driver of cancer progression.
Cell. 2015;162(6):1229–41.
96. Allavena P, Germano G, Marchesi F, Mantovani A. Chemokines
in cancer related inflammation. Exp Cell Res.
2011;317(5):664–73.
97. Griffioen AW, Damen CA, Martinotti S, Blijham GH, Groe-
newegen G. Endothelial intercellular adhesion molecule-1
expression is suppressed in human malignancies: the role of
angiogenic factors. Cancer Res. 1996;56(5):1111–7.
98. Lider O, Mekori YA, Miller T, et al. Inhibition of T lymphocyte
heparanase by heparin prevents T cell migration and T cell-
mediated immunity. Eur J Immunol. 1990;20(3):493–9.
99. Kalluri R, Zeisberg M. Fibroblasts in cancer. Nat Rev Cancer.
2006;6(5):392–401.
100. Shin HC, Han W, Moon HG, et al. Breast-conserving surgery
after tumor downstaging by neoadjuvant chemotherapy is
oncologically safe for stage III breast cancer patients. Ann Surg
Oncol. 2013;20(8):2582–9.
101. Vuky J, Corman JM, Porter C, Olgac S, Auerbach E, Dahl K.
Phase II trial of neoadjuvant docetaxel and CG1940/CG8711
followed by radical prostatectomy in patients with high-risk
clinically localized prostate cancer. Oncologist.
2013;18(6):687–8.
102. Long GV, Weber JS, Larkin J, et al. Nivolumab for patients with
advanced melanoma treated beyond progression: analysis of 2
phase 3 clinical trials. JAMA Oncol. 2017;3(11):1511–19.
Immunotherapy for Solid Tumors: A Review

Contenu connexe

Tendances

July 2015 Cancer immunotherapy update
July 2015 Cancer immunotherapy updateJuly 2015 Cancer immunotherapy update
July 2015 Cancer immunotherapy update
Yujia Sun
 
ImMucin-2014 12 carmon et al bjh13245 (2)
ImMucin-2014 12 carmon et al bjh13245 (2)ImMucin-2014 12 carmon et al bjh13245 (2)
ImMucin-2014 12 carmon et al bjh13245 (2)
Leor Mann
 
WP_ClinDev-Immunotherapy_d03-final edits
WP_ClinDev-Immunotherapy_d03-final editsWP_ClinDev-Immunotherapy_d03-final edits
WP_ClinDev-Immunotherapy_d03-final edits
Chris Learn, Ph.D, PMP
 
Immunotherapy Web Presentation
Immunotherapy   Web PresentationImmunotherapy   Web Presentation
Immunotherapy Web Presentation
anthonymaida
 
Cancer Immunotherapies (Focus on Melanoma & Lung Cancers)
Cancer Immunotherapies (Focus on Melanoma & Lung Cancers)Cancer Immunotherapies (Focus on Melanoma & Lung Cancers)
Cancer Immunotherapies (Focus on Melanoma & Lung Cancers)
Zeena Nackerdien
 
Cancer immunotherapy nivedita shah msc.biotech- 13937
Cancer immunotherapy   nivedita shah  msc.biotech- 13937Cancer immunotherapy   nivedita shah  msc.biotech- 13937
Cancer immunotherapy nivedita shah msc.biotech- 13937
eureka1
 

Tendances (20)

July 2015 Cancer immunotherapy update
July 2015 Cancer immunotherapy updateJuly 2015 Cancer immunotherapy update
July 2015 Cancer immunotherapy update
 
Immune checkpoints sot2016
Immune checkpoints sot2016Immune checkpoints sot2016
Immune checkpoints sot2016
 
Cancer testis antigens and NY-BR-1 expression in primary breast cancer: prog...
Cancer testis antigens and NY-BR-1 expression in  primary breast cancer: prog...Cancer testis antigens and NY-BR-1 expression in  primary breast cancer: prog...
Cancer testis antigens and NY-BR-1 expression in primary breast cancer: prog...
 
Immunotherapy for cancer
Immunotherapy for cancerImmunotherapy for cancer
Immunotherapy for cancer
 
Immunotherapy Update in Oncology
Immunotherapy Update in OncologyImmunotherapy Update in Oncology
Immunotherapy Update in Oncology
 
ImMucin-2014 12 carmon et al bjh13245 (2)
ImMucin-2014 12 carmon et al bjh13245 (2)ImMucin-2014 12 carmon et al bjh13245 (2)
ImMucin-2014 12 carmon et al bjh13245 (2)
 
Overexpression of primary microRNA 221/222 in acute myeloid leukemia
Overexpression of primary microRNA 221/222 in acute myeloid leukemiaOverexpression of primary microRNA 221/222 in acute myeloid leukemia
Overexpression of primary microRNA 221/222 in acute myeloid leukemia
 
Principles of cancer immunotherapy
Principles of cancer immunotherapyPrinciples of cancer immunotherapy
Principles of cancer immunotherapy
 
Multicentric and multifocal versus unifocal breast cancer: differences in the...
Multicentric and multifocal versus unifocal breast cancer: differences in the...Multicentric and multifocal versus unifocal breast cancer: differences in the...
Multicentric and multifocal versus unifocal breast cancer: differences in the...
 
Cancer immunotherapy
Cancer immunotherapy Cancer immunotherapy
Cancer immunotherapy
 
WP_ClinDev-Immunotherapy_d03-final edits
WP_ClinDev-Immunotherapy_d03-final editsWP_ClinDev-Immunotherapy_d03-final edits
WP_ClinDev-Immunotherapy_d03-final edits
 
Cancer immunotherapy different modes of action - astra zeneca - jordan
Cancer immunotherapy   different modes of action - astra zeneca - jordanCancer immunotherapy   different modes of action - astra zeneca - jordan
Cancer immunotherapy different modes of action - astra zeneca - jordan
 
Immunotherapy Web Presentation
Immunotherapy   Web PresentationImmunotherapy   Web Presentation
Immunotherapy Web Presentation
 
Overexpression of YAP 1 contributes to progressive features and poor prognosi...
Overexpression of YAP 1 contributes to progressive features and poor prognosi...Overexpression of YAP 1 contributes to progressive features and poor prognosi...
Overexpression of YAP 1 contributes to progressive features and poor prognosi...
 
Integrative Cancer - New theories and Advances in Treatment From Hippocrates ...
Integrative Cancer - New theories and Advances in Treatment From Hippocrates ...Integrative Cancer - New theories and Advances in Treatment From Hippocrates ...
Integrative Cancer - New theories and Advances in Treatment From Hippocrates ...
 
Gastroenterology Medicine & Research-Crimson Publishers: Can we Optimize Immu...
Gastroenterology Medicine & Research-Crimson Publishers: Can we Optimize Immu...Gastroenterology Medicine & Research-Crimson Publishers: Can we Optimize Immu...
Gastroenterology Medicine & Research-Crimson Publishers: Can we Optimize Immu...
 
Cancer Immunotherapies (Focus on Melanoma & Lung Cancers)
Cancer Immunotherapies (Focus on Melanoma & Lung Cancers)Cancer Immunotherapies (Focus on Melanoma & Lung Cancers)
Cancer Immunotherapies (Focus on Melanoma & Lung Cancers)
 
Cancer immunotherapy nivedita shah msc.biotech- 13937
Cancer immunotherapy   nivedita shah  msc.biotech- 13937Cancer immunotherapy   nivedita shah  msc.biotech- 13937
Cancer immunotherapy nivedita shah msc.biotech- 13937
 
Immunotherapy
ImmunotherapyImmunotherapy
Immunotherapy
 
Asbestos-related diseases - mechanisms and causation at Helsinki Asbestos 2014
Asbestos-related diseases - mechanisms and causation at Helsinki Asbestos 2014Asbestos-related diseases - mechanisms and causation at Helsinki Asbestos 2014
Asbestos-related diseases - mechanisms and causation at Helsinki Asbestos 2014
 

Similaire à Pham2018

Novel_technologies_and _emerging_biomarkers_for_personalized_cancer_immunothe...
Novel_technologies_and _emerging_biomarkers_for_personalized_cancer_immunothe...Novel_technologies_and _emerging_biomarkers_for_personalized_cancer_immunothe...
Novel_technologies_and _emerging_biomarkers_for_personalized_cancer_immunothe...
TOKBLS
 
Ultrasound Technology as a Novel Treatment Strategy in Pancreatic Cancer_Crim...
Ultrasound Technology as a Novel Treatment Strategy in Pancreatic Cancer_Crim...Ultrasound Technology as a Novel Treatment Strategy in Pancreatic Cancer_Crim...
Ultrasound Technology as a Novel Treatment Strategy in Pancreatic Cancer_Crim...
CrimsonpublishersCancer
 
Engineered T Cell Therapy for Gynecologic Malignancies Challenges and Opportu...
Engineered T Cell Therapy for
Gynecologic Malignancies
Challenges and Opportu...Engineered T Cell Therapy for
Gynecologic Malignancies
Challenges and Opportu...
Engineered T Cell Therapy for Gynecologic Malignancies Challenges and Opportu...
RudrikaChandra1
 

Similaire à Pham2018 (20)

Immunotherapy the Present and Future of Cancer Treatment
Immunotherapy the Present and Future of Cancer TreatmentImmunotherapy the Present and Future of Cancer Treatment
Immunotherapy the Present and Future of Cancer Treatment
 
Immuno oncology wp
Immuno oncology wpImmuno oncology wp
Immuno oncology wp
 
Novel_technologies_and _emerging_biomarkers_for_personalized_cancer_immunothe...
Novel_technologies_and _emerging_biomarkers_for_personalized_cancer_immunothe...Novel_technologies_and _emerging_biomarkers_for_personalized_cancer_immunothe...
Novel_technologies_and _emerging_biomarkers_for_personalized_cancer_immunothe...
 
Introduction to cancer vaccines
Introduction to cancer vaccinesIntroduction to cancer vaccines
Introduction to cancer vaccines
 
Nanomedicine based cancer immunotherapy
Nanomedicine based cancer immunotherapyNanomedicine based cancer immunotherapy
Nanomedicine based cancer immunotherapy
 
Crimson Publishers-Immune Checkpoint Inhibitors in Triple Negative Breast Can...
Crimson Publishers-Immune Checkpoint Inhibitors in Triple Negative Breast Can...Crimson Publishers-Immune Checkpoint Inhibitors in Triple Negative Breast Can...
Crimson Publishers-Immune Checkpoint Inhibitors in Triple Negative Breast Can...
 
Limitations of Immunotherapeutic Approaches for Cancer Treatment_Crimson Publ...
Limitations of Immunotherapeutic Approaches for Cancer Treatment_Crimson Publ...Limitations of Immunotherapeutic Approaches for Cancer Treatment_Crimson Publ...
Limitations of Immunotherapeutic Approaches for Cancer Treatment_Crimson Publ...
 
Immunotherapy for Hepatocellular Carcinoma_Crimson Publishers
Immunotherapy for Hepatocellular Carcinoma_Crimson PublishersImmunotherapy for Hepatocellular Carcinoma_Crimson Publishers
Immunotherapy for Hepatocellular Carcinoma_Crimson Publishers
 
Immunotherapy and gene therapy
Immunotherapy and gene therapyImmunotherapy and gene therapy
Immunotherapy and gene therapy
 
Session 490003
Session 490003Session 490003
Session 490003
 
Zauderer, M.G., et al. Clinical Cancer Research, 2017.
Zauderer, M.G., et al. Clinical Cancer Research, 2017.Zauderer, M.G., et al. Clinical Cancer Research, 2017.
Zauderer, M.G., et al. Clinical Cancer Research, 2017.
 
Current Targets of Immune Suppression in Tumor Microenvironment_Crimson Publi...
Current Targets of Immune Suppression in Tumor Microenvironment_Crimson Publi...Current Targets of Immune Suppression in Tumor Microenvironment_Crimson Publi...
Current Targets of Immune Suppression in Tumor Microenvironment_Crimson Publi...
 
Ultrasound Technology as a Novel Treatment Strategy in Pancreatic Cancer_Crim...
Ultrasound Technology as a Novel Treatment Strategy in Pancreatic Cancer_Crim...Ultrasound Technology as a Novel Treatment Strategy in Pancreatic Cancer_Crim...
Ultrasound Technology as a Novel Treatment Strategy in Pancreatic Cancer_Crim...
 
Engineered T Cell Therapy for Gynecologic Malignancies Challenges and Opportu...
Engineered T Cell Therapy for
Gynecologic Malignancies
Challenges and Opportu...Engineered T Cell Therapy for
Gynecologic Malignancies
Challenges and Opportu...
Engineered T Cell Therapy for Gynecologic Malignancies Challenges and Opportu...
 
Tumour Immunology
Tumour ImmunologyTumour Immunology
Tumour Immunology
 
Tumour immunology
Tumour immunologyTumour immunology
Tumour immunology
 
In Vitro Evaluation of Biofield Treatment on Cancer Biomarkers Involved in En...
In Vitro Evaluation of Biofield Treatment on Cancer Biomarkers Involved in En...In Vitro Evaluation of Biofield Treatment on Cancer Biomarkers Involved in En...
In Vitro Evaluation of Biofield Treatment on Cancer Biomarkers Involved in En...
 
Can the Addition of Immunotherapy to Multimodal Management of Stage I-III NSC...
Can the Addition of Immunotherapy to Multimodal Management of Stage I-III NSC...Can the Addition of Immunotherapy to Multimodal Management of Stage I-III NSC...
Can the Addition of Immunotherapy to Multimodal Management of Stage I-III NSC...
 
Cancer therapy
Cancer therapyCancer therapy
Cancer therapy
 
The Future Development of ADC For Cancer.pdf
The Future Development of ADC For Cancer.pdfThe Future Development of ADC For Cancer.pdf
The Future Development of ADC For Cancer.pdf
 

Dernier

Russian Call Girls In Pune 👉 Just CALL ME: 9352988975 ✅❤️💯low cost unlimited ...
Russian Call Girls In Pune 👉 Just CALL ME: 9352988975 ✅❤️💯low cost unlimited ...Russian Call Girls In Pune 👉 Just CALL ME: 9352988975 ✅❤️💯low cost unlimited ...
Russian Call Girls In Pune 👉 Just CALL ME: 9352988975 ✅❤️💯low cost unlimited ...
chanderprakash5506
 
Cara Menggugurkan Kandungan Dengan Cepat Selesai Dalam 24 Jam Secara Alami Bu...
Cara Menggugurkan Kandungan Dengan Cepat Selesai Dalam 24 Jam Secara Alami Bu...Cara Menggugurkan Kandungan Dengan Cepat Selesai Dalam 24 Jam Secara Alami Bu...
Cara Menggugurkan Kandungan Dengan Cepat Selesai Dalam 24 Jam Secara Alami Bu...
Cara Menggugurkan Kandungan 087776558899
 
👉 Chennai Sexy Aunty’s WhatsApp Number 👉📞 7427069034 👉📞 Just📲 Call Ruhi Colle...
👉 Chennai Sexy Aunty’s WhatsApp Number 👉📞 7427069034 👉📞 Just📲 Call Ruhi Colle...👉 Chennai Sexy Aunty’s WhatsApp Number 👉📞 7427069034 👉📞 Just📲 Call Ruhi Colle...
👉 Chennai Sexy Aunty’s WhatsApp Number 👉📞 7427069034 👉📞 Just📲 Call Ruhi Colle...
rajnisinghkjn
 
Difference Between Skeletal Smooth and Cardiac Muscles
Difference Between Skeletal Smooth and Cardiac MusclesDifference Between Skeletal Smooth and Cardiac Muscles
Difference Between Skeletal Smooth and Cardiac Muscles
MedicoseAcademics
 
❤️ Chandigarh Call Girls☎️98151-579OO☎️ Call Girl service in Chandigarh ☎️ Ch...
❤️ Chandigarh Call Girls☎️98151-579OO☎️ Call Girl service in Chandigarh ☎️ Ch...❤️ Chandigarh Call Girls☎️98151-579OO☎️ Call Girl service in Chandigarh ☎️ Ch...
❤️ Chandigarh Call Girls☎️98151-579OO☎️ Call Girl service in Chandigarh ☎️ Ch...
Rashmi Entertainment
 

Dernier (20)

7 steps How to prevent Thalassemia : Dr Sharda Jain & Vandana Gupta
7 steps How to prevent Thalassemia : Dr Sharda Jain & Vandana Gupta7 steps How to prevent Thalassemia : Dr Sharda Jain & Vandana Gupta
7 steps How to prevent Thalassemia : Dr Sharda Jain & Vandana Gupta
 
Russian Call Girls In Pune 👉 Just CALL ME: 9352988975 ✅❤️💯low cost unlimited ...
Russian Call Girls In Pune 👉 Just CALL ME: 9352988975 ✅❤️💯low cost unlimited ...Russian Call Girls In Pune 👉 Just CALL ME: 9352988975 ✅❤️💯low cost unlimited ...
Russian Call Girls In Pune 👉 Just CALL ME: 9352988975 ✅❤️💯low cost unlimited ...
 
💰Call Girl In Bangalore☎️63788-78445💰 Call Girl service in Bangalore☎️Bangalo...
💰Call Girl In Bangalore☎️63788-78445💰 Call Girl service in Bangalore☎️Bangalo...💰Call Girl In Bangalore☎️63788-78445💰 Call Girl service in Bangalore☎️Bangalo...
💰Call Girl In Bangalore☎️63788-78445💰 Call Girl service in Bangalore☎️Bangalo...
 
ANATOMY AND PHYSIOLOGY OF RESPIRATORY SYSTEM.pptx
ANATOMY AND PHYSIOLOGY OF RESPIRATORY SYSTEM.pptxANATOMY AND PHYSIOLOGY OF RESPIRATORY SYSTEM.pptx
ANATOMY AND PHYSIOLOGY OF RESPIRATORY SYSTEM.pptx
 
Race Course Road } Book Call Girls in Bangalore | Whatsapp No 6378878445 VIP ...
Race Course Road } Book Call Girls in Bangalore | Whatsapp No 6378878445 VIP ...Race Course Road } Book Call Girls in Bangalore | Whatsapp No 6378878445 VIP ...
Race Course Road } Book Call Girls in Bangalore | Whatsapp No 6378878445 VIP ...
 
Call Girls Kathua Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Kathua Just Call 8250077686 Top Class Call Girl Service AvailableCall Girls Kathua Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Kathua Just Call 8250077686 Top Class Call Girl Service Available
 
Cara Menggugurkan Kandungan Dengan Cepat Selesai Dalam 24 Jam Secara Alami Bu...
Cara Menggugurkan Kandungan Dengan Cepat Selesai Dalam 24 Jam Secara Alami Bu...Cara Menggugurkan Kandungan Dengan Cepat Selesai Dalam 24 Jam Secara Alami Bu...
Cara Menggugurkan Kandungan Dengan Cepat Selesai Dalam 24 Jam Secara Alami Bu...
 
Call Girls Wayanad Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Wayanad Just Call 8250077686 Top Class Call Girl Service AvailableCall Girls Wayanad Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Wayanad Just Call 8250077686 Top Class Call Girl Service Available
 
Call 8250092165 Patna Call Girls ₹4.5k Cash Payment With Room Delivery
Call 8250092165 Patna Call Girls ₹4.5k Cash Payment With Room DeliveryCall 8250092165 Patna Call Girls ₹4.5k Cash Payment With Room Delivery
Call 8250092165 Patna Call Girls ₹4.5k Cash Payment With Room Delivery
 
👉 Chennai Sexy Aunty’s WhatsApp Number 👉📞 7427069034 👉📞 Just📲 Call Ruhi Colle...
👉 Chennai Sexy Aunty’s WhatsApp Number 👉📞 7427069034 👉📞 Just📲 Call Ruhi Colle...👉 Chennai Sexy Aunty’s WhatsApp Number 👉📞 7427069034 👉📞 Just📲 Call Ruhi Colle...
👉 Chennai Sexy Aunty’s WhatsApp Number 👉📞 7427069034 👉📞 Just📲 Call Ruhi Colle...
 
Call Girls in Lucknow Just Call 👉👉8630512678 Top Class Call Girl Service Avai...
Call Girls in Lucknow Just Call 👉👉8630512678 Top Class Call Girl Service Avai...Call Girls in Lucknow Just Call 👉👉8630512678 Top Class Call Girl Service Avai...
Call Girls in Lucknow Just Call 👉👉8630512678 Top Class Call Girl Service Avai...
 
Difference Between Skeletal Smooth and Cardiac Muscles
Difference Between Skeletal Smooth and Cardiac MusclesDifference Between Skeletal Smooth and Cardiac Muscles
Difference Between Skeletal Smooth and Cardiac Muscles
 
Call girls Service Phullen / 9332606886 Genuine Call girls with real Photos a...
Call girls Service Phullen / 9332606886 Genuine Call girls with real Photos a...Call girls Service Phullen / 9332606886 Genuine Call girls with real Photos a...
Call girls Service Phullen / 9332606886 Genuine Call girls with real Photos a...
 
❤️ Chandigarh Call Girls☎️98151-579OO☎️ Call Girl service in Chandigarh ☎️ Ch...
❤️ Chandigarh Call Girls☎️98151-579OO☎️ Call Girl service in Chandigarh ☎️ Ch...❤️ Chandigarh Call Girls☎️98151-579OO☎️ Call Girl service in Chandigarh ☎️ Ch...
❤️ Chandigarh Call Girls☎️98151-579OO☎️ Call Girl service in Chandigarh ☎️ Ch...
 
Circulatory Shock, types and stages, compensatory mechanisms
Circulatory Shock, types and stages, compensatory mechanismsCirculatory Shock, types and stages, compensatory mechanisms
Circulatory Shock, types and stages, compensatory mechanisms
 
Call Girls Bangalore - 450+ Call Girl Cash Payment 💯Call Us 🔝 6378878445 🔝 💃 ...
Call Girls Bangalore - 450+ Call Girl Cash Payment 💯Call Us 🔝 6378878445 🔝 💃 ...Call Girls Bangalore - 450+ Call Girl Cash Payment 💯Call Us 🔝 6378878445 🔝 💃 ...
Call Girls Bangalore - 450+ Call Girl Cash Payment 💯Call Us 🔝 6378878445 🔝 💃 ...
 
💞 Safe And Secure Call Girls Coimbatore🧿 6378878445 🧿 High Class Coimbatore C...
💞 Safe And Secure Call Girls Coimbatore🧿 6378878445 🧿 High Class Coimbatore C...💞 Safe And Secure Call Girls Coimbatore🧿 6378878445 🧿 High Class Coimbatore C...
💞 Safe And Secure Call Girls Coimbatore🧿 6378878445 🧿 High Class Coimbatore C...
 
Call Girls Mussoorie Just Call 8854095900 Top Class Call Girl Service Available
Call Girls Mussoorie Just Call 8854095900 Top Class Call Girl Service AvailableCall Girls Mussoorie Just Call 8854095900 Top Class Call Girl Service Available
Call Girls Mussoorie Just Call 8854095900 Top Class Call Girl Service Available
 
Bhawanipatna Call Girls 📞9332606886 Call Girls in Bhawanipatna Escorts servic...
Bhawanipatna Call Girls 📞9332606886 Call Girls in Bhawanipatna Escorts servic...Bhawanipatna Call Girls 📞9332606886 Call Girls in Bhawanipatna Escorts servic...
Bhawanipatna Call Girls 📞9332606886 Call Girls in Bhawanipatna Escorts servic...
 
ANATOMY AND PHYSIOLOGY OF REPRODUCTIVE SYSTEM.pptx
ANATOMY AND PHYSIOLOGY OF REPRODUCTIVE SYSTEM.pptxANATOMY AND PHYSIOLOGY OF REPRODUCTIVE SYSTEM.pptx
ANATOMY AND PHYSIOLOGY OF REPRODUCTIVE SYSTEM.pptx
 

Pham2018

  • 1. REVIEW ARTICLE – TRANSLATIONAL RESEARCH AND BIOMARKERS An Update on Immunotherapy for Solid Tumors: A Review Toan Pham, MBBS, BMedSc, PGDipSurgAnat, FRACS1,2,3,4 , Sara Roth, BSc, MSc, PhD1 , Joseph Kong, MBChB, MS, PhD, FRACS1,2,3,4 , Glen Guerra, MBBS, PGDipSurgAnat, FRACS1,2,3,4 , Vignesh Narasimhan, MBChB, FRACS1,2,3,4 , Lloyd Pereira, BSc (Hons), PhD1 , Jayesh Desai, MBBS, FRACP3 , Alexander Heriot, MB, BChir, MA, MD, MBA, FRACS, FRCS (Gen.), FRCSEd, FACS, GAICD1,2,3,4 , and Robert Ramsay, BSc (Hons), PhD1,5 1 Divisions of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; 2 Cancer Surgery, Melbourne, VIC, Australia; 3 Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia; 4 Department of Surgery, University of Melbourne, Melbourne, VIC, Australia; 5 Department of Pathology, University of Melbourne, Melbourne, VIC, Australia ABSTRACT In recent years, it has been demonstrated that immunotherapy is an effective strategy for the man- agement of solid tumors. The origins of immunotherapy can be traced back to the work of William Coley, who elicited an immune response against sarcoma by injecting patients with a mixture of dead bacteria. Significant pro- gress has been made since, with immune markers within the tumor now being used as predictors of cancer prognosis and manipulated to improve patient survival. While surgery remains central to the management of most patients with solid malignancies, it is important that surgeons consider the different immunotherapy strategies that can be employed to manage disease. Here, we highlight how the immune system influences tumorigenesis and bring atten- tion to how current and future immunotherapies can serve as an adjunct to surgery. Cancer is the leading cause of mortality worldwide, being responsible for 8.8 million deaths in 2015.1 Despite advances in prevention, screening, and diagnosis of cancer, the landscape of cancer treatment has been altered by the advent of immunotherapy, offering improved survival in several solid cancers and establishing itself as a new therapeutic modality.2 The concept of exploiting the host immune system to eradicate cancer was first conceived by William Coley. In pioneering work, a mixture of attenuated bacteria was injected into patients in order to elicit an immune-mediated response against sarcoma.3,4 However, significant advances in cancer immunology have only been achieved in recent years, with immunotherapy demonstrating clear efficacy in clinical trials, and a synergistic effect when used in combination.5,6 In this review, we delineate the relationship between cancer and the immune response and discuss how this has been used as a basis for immunotherapy. As personalized medicine begins to enter the domain of all oncology spe- cialties, an awareness of available therapies is important, allowing surgeons to provide the most up-to-date care for their patients. THE IMMUNE SYSTEM AND PROGNOSIS: A NOVEL STAGING SYSTEM Our immune system continuously protects us against environmental pathogens and malignant cells (Fig. 1). However, malignant cells can acquire the capacity to evade the action of the immune system and form cancer7 . A seminal publication by Galon et al.8 demonstrated that low infiltrating cytotoxic (CD8? ) and memory (CD45RO? ) lymphocytes predict early relapse in colon cancer (CC). This led to a new scoring system called Immunoscore (IS),9–11 which has been evaluated in 1336 stage I–III CC patients and further validated in 2681 patients by an international Toan Pham and Sara Roth have contributed equally to this study and are co-first authors. Ó Society of Surgical Oncology 2018 First Received: 27 February 2018 T. Pham, MBBS, BMedSc, PGDipSurgAnat, FRACS e-mail: toan.pham@petermac.org Ann Surg Oncol https://doi.org/10.1245/s10434-018-6658-4
  • 2. consortium. Both studies concluded that high IS is associ- ated with longer recurrence-free survival.11,12 Moreover, IS was able to identify high-risk patients (low IS) within American Joint Committee on Cancer (AJCC) stage II CC, thus making IS a powerful biomarker tool to stratify patients to adjuvant chemotherapy. IS was translated to other tumor types including gastric cancer13,14 and melanoma,15 in which patients within the same tumor–node–metastasis (TNM) stage could be stratified with enhanced accuracy to predict long-term outcomes. Hence, IS is expected to become a new component of the AJCC TNM system. CANCER IMMUNOTHERAPY STRATEGIES In cancer immunotherapy, the host immune system is used to target and eradicate cancer cells.3,16 Tumors express neoantigens that can be recognized as foreign by the immune system and increase the level of T-lympho- cytes infiltrating the tumor (TILs). This relationship can be further described by the concept of immunoediting, which forms the basis of modern cancer immunology research.17 In this paradigm, elimination, equilibrium, and escape are key components. Elimination is the recognition and destruction of tumor cells by the immune system. Subse- quently, through immunoediting [e.g., expression of immune checkpoints, loss of major histocompatibility complex (MHC) expression, and/or upregulation of T-regulatory cells], tumors create a sanctuary against the immune system, thereby establishing an equilibrium between tumor growth and immune destruction. In this context, tumors escape immune control when the degree of immunoediting allows for unchecked proliferation. This model is underpinned by the close interaction between the immune system and cancer cells, thus our understanding of this relationship is an important component in the devel- opment and considered use of immunotherapy. An outline of the main classes of immunotherapy is shown in Fig. 2 and further described under each subheading below. Immune Checkpoint Inhibitors: Reactivation of Tumor- Infiltrating T-Cells Immune checkpoints are the body’s natural brakes to dampen the immune response and prevent autoimmunity. Tumor cells can exploit this avenue to escape the immune system by expressing complementary molecules that interact with T-cells, rendering them incapable of killing. Hence, these immune checkpoints [e.g., programmed cell death (PD)-1, PD-L1, cytotoxic T lymphocyte-associated antigen (CTLA)-4] form therapeutic targets. The first immune checkpoint inhibitor (ICI) discovered was ipilimumab (Bristol-Myers Squibb), an anti-CTLA-4 Innate Immune System Microbes Complement system Dendritic cell B cells Antibodies T cells Effector T cells Blood vessel NK cell Infected/ transformed cell Infected/ transformed cell Granulocytes Monocytes/ Macrophages Skin barrier Hours Days Adaptive Immune System FIG. 1 Overview of the immune system: the immune system is divided into the innate and adaptive arms. The innate system responds rapidly to a conserved repertoire of pathogenic antigens, whereas the adaptive system mounts a specific and memory response that initially requires 4–7 days to develop T. Pham et al.
  • 3. antibody.18 It was first approved for the treatment of unresectable metastatic melanoma, and the results were encouraging with median overall survival of 10.1 months versus 6.4 months in the control group.18 This equated to improved 1- and 2-year survival rates of 46 versus 25 % and 24 versus 14 %, respectively.18 Despite its promising therapeutic efficacy, there were significant side effects, including diarrhea (41.4%), colitis (15.9%), and endocrine disorders (37.6%), and five on-trial mortalities.19 As such, ipilimumab is now used as second-line therapy or in combination with another ICI. Pembrolizumab (Merck & Co.) and nivolumab (Bristol- Myers Squibb) are the first two Food and Drug Adminis- tration (FDA)-approved ICIs that block the interaction between PD-1 (expressed by T-cells) and its ligands PD-L1 and PD-L2 (expressed by tumor and myeloid cells).20 These therapies have similar efficacy and long-term out- comes to anti-CTLA-4 therapy but without significant side effects,21 thus gaining FDA approval following landmark clinical trials for melanoma (KEYNOTE-002),22 non-small cell lung cancer (KEYNOTE-010),23 microsatellite unsta- ble/mismatch repair gene deficient colorectal cancer,24 and gastric cancer (KEYNOTE-059).25 Recently, combination anti-PD-1 and anti-CTLA-4 therapies in treatment-naı̈ve metastatic melanoma (CHECKMATE-067)5 have demonstrated a synergistic survival advantage over either monotherapy.5 Conse- quently, pembrolizumab is now being tested with a variety of other drugs, including molecular targeted therapies, such as dabrafenib and trametinib, in BRAF-mutant advanced melanoma (KEYNOTE-022).26 Expanding an Existing Immune Response: Adoptive Cellular Therapy Another emerging immunotherapy is adoptive cellular therapy (ACT), where tumor-specific lymphocytes are extracted from peripheral blood or resected tumors and expanded ex vivo. These lymphocytes are then reintroduced into the patient with or without systemic lymphodepletion therapy. The Surgery Branch of the National Cancer Insti- tute, led by Steven Rosenberg, has been at the forefront of this therapy, with first successes in melanoma27,28 and cur- rent clinical trials targeting other malignancies including head and neck,29,30 renal cell,31 and gynecological can- cers.32,33 Recent trials have explored the combination of ACT with BRAF inhibitor34 and PD-1 ICI.35 ACT has even led to an additional indication for metastasectomy which was once considered futile, now a potential source for tumor- specific lymphocytes used in ACT.36 a b Dying tumor cell Granzyme & perforin Cytotoxic T-cell Cytotoxic T-cell Cytotoxic T-cell Cytotoxic T-cell Cytotoxic T-cell NK cell CAR T -cell CAR T -cell Antigen presenting cell Oncolytic virus Radiation Neoantigen induced cytotoxic T-cell Cytotoxic T-cell MDSC Treg MHC class I presentation Resistant tumor cell Tumor surface protein Immune checkpoint receptor/ ligand interaction Immune checkpoint inhibitor Vaccine induced cytotoxic T-cell Recognition and Elimination Of Malignant Cell Radiation or Oncolytic Virus induced Tumor Neoantigen Adoptive CAR T-cell therapy or Cancer Vaccination Administration of Immune Checkpoint Blockers Adoptive CAR T-cell or NK cell therapy Depletion or Suppression of MDSC and Tregs Low Immunogenicity of presented Antigen Expression of Immune Checkpoints Downregulation of MHC Class I Expression Suppression by MDSC or Tregs c NORMAL IMMUNOSUPPRESION THERAPY OPTONS FIG. 2 Tumor ‘‘escape’’ mechanisms (pink) and associated immunotherapy counterstrategies (green): a normal recognition of transformed cells by MHC class I presentation of tumor-associated antigen and subsequent destruction by specific cytotoxic T-cells (blue); b malignant cells escape immune recognition by low immunogenicity of the presented antigen, expression of immune checkpoints, downregulation of MHC class I, and generation of an immunosuppressive microenvironment; c current immunotherapies counteracting these evasion mechanisms via increasing the tumor- specific antigens (radiation, vaccination, oncolytic viruses), immune checkpoint blockade, CAR T-cells, and NK cell therapies, and elimination of immunosuppressive microenvironment Immunotherapy for Solid Tumors: A Review
  • 4. Enhancing the Immunological Profile of Solid Tumors Tumors with sparse TILs typically have low mutational burden and consequently low neoantigen load. In order to address this subset of challenging cancers, immune-based strategies such as radiotherapy, oncolytic viruses, and cancer vaccines have been developed to enhance the immune recognition of these tumors. Radiotherapy Radiotherapy can induce antigen expression through tumor cell death and upregulation of MHC class I on surviving tumor cells, both increasing their susceptibility to cytotoxic T-cell killing.37 Single high-dose (5–10 Gy) or hypofractionated (3 9 8 Gy) radiotherapy induces a proinflammatory response that may result in an abscopal effect,38 which is most pronounced in immunogenic tumors including renal cell carcinoma (RCC), melanoma, and hepatocellular carcinoma (HCC).39 Several clinical trials are now investigating the combination of radiotherapy and immune checkpoint inhibitors,40 because radiation has been associated with PD-L1 upregulation.41 Oncolytic Viruses Oncolytic viruses (OV) are genetically engineered or naturally occurring viruses that selectively replicate in and kill cancer cells without collateral damage to normal tissues. The release of antigens from the lysed cancer cell enhances immune recognition and triggers further immune-mediated destruction.42 The tumor specificity of OV can be attributed to multiple factors including the virus’s tropism for specific tissue type, downregulation of the antiviral response of cancer cells, and/or engineered enhancements to the virus to exploit the different intracellular milieu of cancer cells.43 OV can also be ‘‘armed’’ with genes encoding for immunodulators such as granulocyte-macrophage colony-stimulating factor (GM-CSF) or tumor-associated antigens to further enhance their function.43 The first OV approved by the FDA was T-Vec, an oncolytic herpes simplex virus type 1 armed with GM- CSF, for the treatment of advanced melanoma.44 Further oncolytic viruses such as Pexa-Vec against hepatocellular carcinoma, G47D against glioblastoma and prostate cancer, and CG0070 against bladder cancer show promising results.43,45 The main disadvantage of OV is the acquired specific immunity against the virus that effectively neutralizes any repeat therapy. However, even partial responses leading to downstaging may facilitate surgical resection and ICI therapy. Cancer Vaccines Cancer vaccines represent a different approach of generating tumor-specific T-cells in poorly immunogenic tumors, against either neoantigens or differentially overexpressed self-antigens. Most cancer vaccines to date have demonstrated limited clinical benefit,46 which can be attributed to immune-suppressive mechanisms within the tumor microenvironment (TME) if administered as monotherapy. The success of ICI has led to a renaissance of combination therapy cancer vaccine clinical trials, with promising results.47,48 A few encouraging examples include the FDA-approved Sipuleucel-T (dendritic cell vaccine) for castrate-resistant prostate cancer,49 TG01 (mutant K-ras peptide vaccine) for pancreatic cancer,50 and two personalized neoantigen vaccines for melanoma.47,48 The TetMYB vaccine, a DNA plasmid encoding a modified MYB oncoprotein fused with tetanus antigen to break self-tolerance, is being tested against advanced colorectal and adenoid cystic carcinoma (NCT03287427).51 NK Cell and CAR T-Cell Therapy: Lifting the Cloak of Invisibility Another immune evasion technique employed by tumor cells is downregulation of MHC class I molecules, thus rendering the tumor invisible to cytotoxic T-cells.52,53 This adaptation has been observed in patients treated with ICI and cancer vaccines,48,54 and may be countered by natural killer (NK) and chimeric antigen receptor (CAR) T-cell therapies, as these two immune cells do not rely on MHC class I presentation for tumor recognition. In early-phase clinical trials, NK cell therapy has proven to be safe and well tolerated, albeit with limited success, in non-small cell lung,55,56 gastrointestinal,57 breast, ovar- ian,58 and renal cell cancers, and melanoma.59 The intrinsic lack of tumor infiltration by NK cells is often attributed to suppressive TME. This can be mitigated by nanoparticles that attract and expand NK cells within the tumor60 or genetically modified NK cells.61 CAR T-cells are autologous T-cells that have undergone ex vivo genetic modification to express a tumor-specific hybrid receptor. Prominent successes have been achieved in hematological malignancies, including the use of anti- CD19 CAR T-cells in non-Hodgkin’s B cell lymphoma and lymphoblastic leukemia, which gained FDA approval in 2017.62–64 Current target antigens include epidermal growth factor receptor (EGFR), human epidermal growth factor receptor 2 (HER-2), mesothelin, and carcinoembry- onic antigen (CEA)65 on solid tumors. Early-phase trials have shown CAR T-cells to be safe and feasible in a variety of solid tumors, including non- small cell lung cancer,66 neurological malignancies,67–69 breast,70 pancreatic,71,72 and metastatic colon cancer,73 and sarcoma.74,75 Furthermore, enhanced efficacy in combina- tion with ICI has been observed against melanoma, Hodgkin’s lymphoma, and non-small cell lung cancers.76 T. Pham et al.
  • 5. T-Regulatory and Myeloid-Derived Suppressor Cells: Antisuppressor Therapy T-regulatory (Treg) and myeloid-derived suppressor cells (MDSCs) are partly responsible for the immunosup- pressive TME,77 thus representing a potential avenue in cancer immunotherapy. FoxP3-expressing Treg cells induce self-tolerance and prevent autoimmunity. Tumors can exploit Tregs to their advantage, and current therapeutic strategies are focusing on depleting these cells.78 Two such therapies are dacli- zumab (IL-2 receptor blocking antibody) and denileukin diftitox (IL-2:diptheria toxin fusion peptide), which have demonstrated robust clinical outcomes79 in metastatic breast cancer80,81 and chemo/immuno-naı̈ve stage IV melanoma.82 Additionally, the checkpoint inhibitor ipili- mumab has been reported to deplete tumor-infiltrating Tregs via antibody-dependent cell-mediated cytotoxicity (ADCC) in melanoma patients.83 MDSCs are immature myeloid cells that suppress cytotoxic T-cell function and modulate the activation and expansion of Tregs by immunosuppressive cytokines, arginase-1, and reactive oxygen species (ROS) and nitric oxide (NO).77 Treatments targeting MDSCs were serendipitously discovered as a by-effect of other cancer treatments.84 Ipilimumab and vemurafenib (BRAF inhibitor) have been shown to reverse the immunosuppressive effect of MDSCs in melanoma.85,86 Sunitinib (tyrosine kinase inhi- bitor) decreases MDSC levels by reducing the expansion of monocytic MDSCs and inducing apoptosis of granulocytic MDSCs,87 being the first-line therapy for metastatic RCC. All-trans retinoic acid (ATRA) and vitamin D3 promote differentiation of MDSCs into mature nonsuppressive cells and have shown benefit in metastatic RCC88 and head and neck squamous cell carcinoma (SCC),89 respectively. Sildenafil, tadalafil, and vardenafil (PDE5 inhibitors) have been shown to inhibit arginase-1 and NO expression and significantly reduce disease burden in myeloma patients90 and circulating MDSCs in head and neck SCC.91 Addi- tionally, conventional chemotherapeutic drugs, namely gemcitabine and 5-fluorouracil, have been found to decrease MDSC and to improve antitumor immune responses.92,93 Other Barriers Against Immunotherapy In addition to the immune-evasive mechanism men- tioned above, further parts of the TME can adversely affect immune cell function. Some tumors create a hostile TME for TILs by depleting nutrients and oxygen and releasing acidic94 and toxic metabolites.95 Others promote an immunosuppressive cell infiltrate by altering the inflammatory cytokine milieu.96 Lastly, modifications of the tumor vasculature such as downregulation of intercellular adhesion molecule 1 (ICAM-1)97 and altering the tumor stroma98,99 to retard T-cell migration have been described. Research into counteracting some of these mechanisms is currently being done, however there are no clinical trials to date. THE RISE OF IMMUNOTHERAPY CLINICAL TRIALS An enquiry of the ClinicalTrials.gov database revealed that, of the 60,903 cancer trials in the entire database, the search string ‘‘Cancer AND Immunotherapy’’ returned 1994 (* 3%) interventional studies, and of these, 313 (* 16%) have been completed (Table 1). Recent years have seen a rapid increase in the number of immunotherapy trials (Fig. 3). Although earlier trials were disappointing, more recent trials featuring combination therapies have shown promising results. Thus, there is a need for further evaluation of upcoming therapies as well as the potential synergistic effects of existing therapies. INTEGRATION WITH SURGICAL PRACTICE Local/Regional Control The success of treating primary solid malignancies is highly dependent on the disease stage. Surgery has tradi- tionally played a role in this setting, with early-stage disease often cured with a margin-negative resection. In contrast, in many locally advanced solid tumors, neoadju- vant therapies such as radiotherapy and/or chemotherapy have shown benefit in downstaging cancers to enable resection. This is exemplified in breast cancer, where 5-year survival for stage III patients was similar between mastectomy (96.3%) and chemoradiotherapy combined with breast-conserving surgery (90.9%, p = 0.669).100 More recently, chemoimmunotherapy has been used suc- cessfully to downstage prostate cancer.101 Distant/Metastatic Disease The presence of metastatic disease has classically implied an incurable state for patients. Surgery has tradi- tionally had a limited role, often reserved for control of symptoms. Chemotherapy and radiotherapy can extend overall survival in this cohort; however, this requires bal- anced consideration given the impact on quality of life and treatment-related morbidity. Harnessing what immunotherapy promises to offer, we have the potential to turn advanced cancer into a chronic Immunotherapy for Solid Tumors: A Review
  • 6. disease, or state of equilibrium.17 This has been demon- strated in subgroups of the CheckMate-066 and -067 melanoma trials, where clinical benefit from nivolumab is seen beyond disease progression for advanced mela- noma.102 Furthermore, improvement in survival has been achieved with dual checkpoint inhibition.5 TABLE 1 Phase and modality of completed immunotherapy clinical trials against solid malignancies (compiled from ClinicalTrials.gov; 5/15/2018) Solid cancer type Phase of clinical trial (number of trials*) Immunotherapy modality (number of trials*) I II III Cancer vaccine Checkpoint inhibition Adoptive cellular therapy CAR T-cell Oncolytic virus, gene therapy Anti-Treg, anti-MDSC Cytokine, targeted therapy Prostate 17 27 7 39 4 2 3 3 Melanoma 39 41 5 40 6 19 1 2 1 16 Lung 18 30 3 35 5 1 10 Head and neck 11 8 10 2 1 1 5 Neurological 26 20 3 24 1 8 4 3 9 Gynecological 12 15 1 16 3 1 8 Anal 3 2 1 1 1 2 Penile 3 1 1 1 2 Sarcoma 10 8 8 1 4 1 4 Urothelial 7 14 4 9 1 4 1 1 9 Breast 18 18 23 1 6 6 Hepatobiliary 9 4 2 6 4 2 3 Colorectal 10 13 1 17 1 2 1 3 Esophagogastric 6 4 5 1 4 Pancreas 13 11 2 20 1 1 1 3 Total 202 216 28 254 21 58 12 10 4 87 *Where a trial involves more than one therapy, it is counted in duplicate according to the number of immunotherapy modalities used 0 5 10 15 20 25 30 1995 1997 1999 2001 2003 2005 2007 2009 2011 2013 2015 Number of Trials Year of Trial Commencement The Rise of Cancer Immunotherapy Trials FIG. 3 Trend showing the rapid increase in number of started cancer immunotherapy trials from 1995–2015 (compiled from ClinicalTrials.gov; 5/15/2018) T. Pham et al.
  • 7. When examining published data, there is a changing role for surgery. Current immunotherapeutic agents are most effective when there is low to moderate tumor burden given the limited immunosuppressive factors. Therefore, surgery’s role is still foremost to achieve cure in early-stage disease, however what was once considered futile/palliative surgery now has the potential to offer cure when combined with immunotherapy. This not only includes resecting tumors after downstaging, but also removing tumors in some can- cers, including metastatic deposits, in order to understand the immune profile of the resistant clone, and for the expansion of TILs. Both of these methods allow a personalized treat- ment algorithm to be developed and modified throughout the patient’s journey, so that the best outcomes can be achieved. CONCLUSIONS Immunotherapy is a promising therapeutic avenue that is rapidly shifting the paradigm of cancer management. This review is intended to raise awareness about the current immunotherapeutic strategies. Immunotherapy can provide an important adjunct to current available therapies to improve long-term outcomes in previously resistant or incurable cancers. With its growing use, it is important for surgeons and surgical oncologists to embrace it, thereby providing immunotherapy as a treatment options for their patients. ACKNOWLEDGMENT Not applicable. DISCLOSURE All authors are involved in the MYPHISMO clin- ical trial (NCT03287427). REFERENCES 1. World Health Organization Cancer Factsheet. 2018; http://www. who.int/mediacentre/factsheets/fs297/en/. Accessed 18 Feb 2018. 2. Weber JS, O’Day S, Urba W, et al. Phase I/II study of ipili- mumab for patients with metastatic melanoma. J Clin Oncol. 2008;26(36):5950–56. 3. Coley WB. The treatment of inoperable sarcoma by bacterial toxins (the mixed toxins of the Streptococcus erysipelas and the Bacillus prodigiosus). Proc R Soc Med. 1910;3:1–48. 4. Coley WB. The treatment of malignant tumors by repeated inoculations of erysipelas. With a report of ten original cases. 1893. Clin Orthop Relat Res. 1991(262):3–11. 5. Wolchok JD, Chiarion-Sileni V, Gonzalez R, et al. Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med. 2017;377(14):1345–56. 6. Ott PA, Hodi FS, Kaufman HL, Wigginton JM, Wolchok JD. Combination immunotherapy: a road map. J Immunother Can- cer. 2017;5:16. 7. Dunn GP, Bruce AT, Ikeda H, Old LJ, Schreiber RD. Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol. 2002;3(11):991–8. 8. Galon J, Costes A, Sanchez-Cabo F, et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science. 2006;313(5795):1960–4. 9. Mlecnik B, Tosolini M, Kirilovsky A, et al. Histopathologic- based prognostic factors of colorectal cancers are associated with the state of the local immune reaction. J Clin Oncol. 2011;29(6):610–8. 10. Galon J, Mlecnik B, Bindea G, et al. Towards the introduction of the ‘Immunoscore’ in the classification of malignant tumours. J Pathol. 2014;232:199–209. 11. Galon J, Mlecnik B, Marliot F, et al. Validation of the Immu- noscore (IM) as a prognostic marker in stage I/II/III colon cancer: Results of a worldwide consortium-based analysis of 1,336 patients. J Clin Oncol. 2016;34(15):3500–3500. 12. Pages F, Mlecnik B, Marliot F, et al. International validation of the consensus Immunoscore for the classification of colon can- cer: a prognostic and accuracy study. Lancet. 2018;391(10135):2128–39. 13. Fujitani K, Ando M, Sakamaki K, et al. A prospective multi- center observational study of surgical palliation examining postoperative quality of life in patients treated for malignant gastric outlet obstruction caused by incurable advanced gastric cancer. J Clin Oncol. 2017;35(4):6-6. 14. Qi X, Jiang Y, Zhang Q, et al. Prognostic and predictive value of immunoscore signature in gastric cancer. J Clin Oncol. 2017;35(15):e15594-e15594. 15. Galon J, Fox BA, Bifulco CB, et al. Immunoscore and immunoprofiling in cancer: an update from the melanoma and immunotherapy bridge 2015. J Transl Med. 2016;14(1):273. 16. Pilch YH, Myers GH, Sparks FC, Golub SH. Prospects for the immunotherapy of cancer. Part I: basic concepts of tumor immunologyProspects for the immunotherapy of cancer. Part I: basic concepts of tumor immunology. Curr Probl Surg. 1975;12(1):1–46. 17. Dunn GP, Old LJ, Schreiber RD. The three Es of cancer immunoediting. Annu Rev Immunol. 2004;22:329–60. 18. Lipson EJ, Drake CG. Ipilimumab: an anti-CTLA-4 antibody for metastatic melanoma. Clin Cancer Res. 2011;17(22):6958–62. 19. Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 19 2010;363(8):711–23. 20. Ribas A. Tumor immunotherapy directed at PD-1. N Engl J Med. 2012;366(26):2517–9. 21. Robert C, Schachter J, Long GV, et al. Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med. 2015;372(26):2521–32. 22. Ribas A, Puzanov I, Dummer R, et al. Pembrolizumab versus investigator-choice chemotherapy for ipilimumab-refractory melanoma (KEYNOTE-002): a randomised, controlled, phase 2 trial. Lancet Oncol. 2015;16(8):908–18. 23. Herbst RS, Baas P, Kim DW, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non- small-cell lung cancer (KEYNOTE-010): a randomised con- trolled trial. Lancet. 2016;387(10027):1540–50. 24. Le DT, Uram JN, Wang H, et al. PD-1 Blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015;372(26):2509–20. 25. Fuchs CS, Doi T, Jang RW-J, et al. KEYNOTE-059 cohort 1: Efficacy and safety of pembrolizumab (pembro) monotherapy in patients with previously treated advanced gastric cancer. J Clin Oncol. 2017;35(15):4003-4003. 26. Ribas A, Hodi FS, Lawrence D, et al. 1216OKEYNOTE-022 update: phase 1 study of first-line pembrolizumab (pembro) plus dabrafenib (D) and trametinib (T) for BRAF-mutant advanced melanoma. Vol 282017. Immunotherapy for Solid Tumors: A Review
  • 8. 27. Dudley ME, Wunderlich J, Nishimura MI, et al. Adoptive transfer of cloned melanoma-reactive T lymphocytes for the treatment of patients with metastatic melanoma. J Immunother. 2001;24(4):363–73. 28. Dudley ME, Wunderlich JR, Yang JC, et al. A phase I study of nonmyeloablative chemotherapy and adoptive transfer of autologous tumor antigen-specific T lymphocytes in patients with metastatic melanoma. J Immunother. 2002;25(3):243–51. 29. Zhou P, Tian S, Li J, et al. Paradoxes in thyroid carcinoma treatment: analysis of the SEER database 2010–2013. Onco- target. 2017;8(1):345–53. 30. Ostrom QT, Gittleman H, Kruchko C, et al. Completeness of required site-specific factors for brain and CNS tumors in the Surveillance, Epidemiology and End Results (SEER) 18 data- base (2004–2012, varying). J Neurooncol. 2016;130(1):31–42. 31. Liao Z, Rodrigues MC, Poynter JN, Amatruda JF, Rodriguez- Galindo C, Frazier AL. Risk of second malignant neoplasms in women and girls with germ cell tumors. Ann Oncol. 2017;28(2):329–32. 32. Yao N, Alcala HE, Anderson R, Balkrishnan R. Cancer dis- parities in rural Appalachia: incidence, early detection, and survivorship. J Rural Health. 2017;33(4):375–81. 33. Shah BK, Kandel P, Khanal A. Second primary malignancies in hepatocellular cancer - A US population-based study. Anti- cancer Res. 2016;36(7):3511–4. 34. Deniger DC, Kwong ML, Pasetto A, et al. A pilot trial of the combination of vemurafenib with adoptive cell therapy in patients with metastatic melanoma. Clin Cancer Res. 2017;23(2):351–62. 35. Bista A, Sharma S, Shah BK. Disparities in receipt of radio- therapy and survival by age, sex, and ethnicity among patient with stage I follicular lymphoma. Front Oncol. 2016;6:101. 36. Crompton JG, Klemen N, Kammula US. Metastasectomy for tumor-infiltrating lymphocytes: an emerging operative indica- tion in surgical oncology. Ann Surg Oncol. 2018;25(2):565–72. 37. Reits EA, Hodge JW, Herberts CA, et al. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J Exp Med. 2006;203(5):1259–71. 38. Haikerwal SJ, Hagekyriakou J, MacManus M, Martin OA, Haynes NM. Building immunity to cancer with radiation ther- apy. Cancer Lett. 2015;368(2):198–208. 39. Reynders K, Illidge T, Siva S, Chang JY, De Ruysscher D. The abscopal effect of local radiotherapy: using immunotherapy to make a rare event clinically relevant. Cancer Treat Rev. 2015;41(6):503–10. 40. Ko EC, Formenti SC. Radiotherapy and checkpoint inhibitors: a winning new combination? Ther Adv Med Oncol. 2018;10:1758835918768240. 41. Dovedi SJ, Illidge TM. The antitumor immune response gen- erated by fractionated radiation therapy may be limited by tumor cell adaptive resistance and can be circumvented by PD-L1 blockade. Oncoimmunology. 2015;4(7):e1016709. 42. Lawler SE, Speranza MC, Cho CF, Chiocca EA. Oncolytic viruses in cancer treatment: a review. JAMA Oncol. 2017;3(6):841–9. 43. Fukuhara H, Ino Y, Todo T. Oncolytic virus therapy: A new era of cancer treatment at dawn. Cancer Sci. 2016;107(10):1373–9. 44. Kaufman HL, Bines SD. OPTIM trial: a Phase III trial of an oncolytic herpes virus encoding GM-CSF for unresectable stage III or IV melanoma. Future Oncol. Jun 2010;6(6):941–9. 45. Breitbach CJ, Moon A, Burke J, Hwang TH, Kirn DH. A phase 2, open-label, randomized study of Pexa-Vec (JX-594) admin- istered by intratumoral injection in patients with unresectable primary hepatocellular carcinoma. Methods Mol Biol. 2015;1317:343–57. 46. Dalgleish AG, Whelan MA. Cancer vaccines as a therapeutic modality: The long trek. Cancer Immunol Immunother. 2006;55(8):1025–32. 47. Ott PA, Hu Z, Keskin DB, et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature. 2017;547(7662):217–21. 48. Sahin U, Derhovanessian E, Miller M, et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature. 2017;547(7662):222–6. 49. Kantoff PW, Higano CS, Shore ND, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363(5):411–22. 50. Weden S, Klemp M, Gladhaug IP, et al. Long-term follow-up of patients with resected pancreatic cancer following vaccination against mutant K-ras. Int J Cancer. 2011;128(5):1120–8. 51. MYPHISMO: MYB and PD-1 Immunotherapies against multi- ple oncologies trial. https://ClinicalTrials.gov/show/ NCT03287427. 52. Garrido F, Perea F, Bernal M, Sanchez-Palencia A, Aptsiauri N, Ruiz-Cabello F. The escape of cancer from T cell-mediated immune surveillance: HLA class I loss and tumor tissue archi- tecture. Vaccines (Basel). 2017;5(1):7. 53. Garrido F, Ruiz-Cabello F, Aptsiauri N. Rejection versus escape: the tumor MHC dilemma. Cancer Immunol Immunother. 2017;66(2):259–71. 54. Zaretsky JM, Garcia-Diaz A, Shin DS, et al. Mutations associ- ated with acquired resistance to PD-1 blockade in melanoma. N Engl J Med. 2016;375(9):819–29. 55. Tonn T, Schwabe D, Klingemann HG, et al. Treatment of patients with advanced cancer with the natural killer cell line NK-92. Cytotherapy. 2013;15(12):1563–70. 56. Yang YJ, Park JC, Kim HK, Kang JH, Park SY. A trial of autologous ex vivo-expanded NK cell-enriched lymphocytes with docetaxel in patients with advanced non-small cell lung cancer as second- or third-line treatment: phase IIa study. An- ticancer Res. 2013;33(5):2115–22. 57. Sakamoto N, Ishikawa T, Kokura S, et al. Phase I clinical trial of autologous NK cell therapy using novel expansion method in patients with advanced digestive cancer. J Transl Med. 2015;13:277. 58. Geller MA, Cooley S, Judson PL, et al. A phase II study of allogeneic natural killer cell therapy to treat patients with recurrent ovarian and breast cancer. Cytotherapy. 2011;13(1):98–107. 59. Arai S, Meagher R, Swearingen M, et al. Infusion of the allo- geneic cell line NK-92 in patients with advanced renal cell cancer or melanoma: a phase I trial. Cytotherapy. 2008;10(6):625–32. 60. Ammam M. Immunotherapy based on natural killer cells may soon begin clinical studies. In: M. Nace (Ed) Immuno-Oncology News. Dallas, TX: BioNews Services, LLC; 2017. 61. Rezvani K, Rouce R, Liu E, Shpall E. Engineering natural killer cells for cancer immunotherapy. Mol Ther. 2017;25(8):1769–81. 62. Porter DL, Kalos M, Zheng Z, Levine B, June C. Chimeric antigen receptor therapy for B-cell malignancies. J Cancer. 2011;2:331–2. 63. Porter DL, Hwang WT, Frey NV, et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci Transl Med. 2015;7(303):303ra139. 64. FDA US. FDA approval brings first gene therapy to the United States. CAR T-cell therapy approved to treat certain children and young adults with B-cell acute lymphoblastic leukemia: FDA, USA; 2017. 65. Yu S, Li A, Liu Q, et al. Chimeric antigen receptor T cells: a novel therapy for solid tumors. J Hematol Oncol. 2017;10(1):78. T. Pham et al.
  • 9. 66. Feng K, Guo Y, Dai H, et al. Chimeric antigen receptor-modi- fied T cells for the immunotherapy of patients with EGFR- expressing advanced relapsed/refractory non-small cell lung cancer. Sci China Life Sci. 2016;59(5):468–79. 67. A phase I trial of T cells expressing an anti-GD2 chimeric antigen receptor in children and young adults with GD2? solid tumors. https://ClinicalTrials.gov/show/NCT02107963. 68. CMV-specific cytotoxic T lymphocytes expressing CAR tar- geting HER2 in patients with gbm. https://ClinicalTrials.gov/sh ow/NCT01109095. 69. Pilot study of autologous anti-EGFRvIII CAR T cells in recur- rent glioblastoma multiforme. https://ClinicalTrials.gov/show/ NCT02844062. 70. Kawalec P, Paszulewicz A, Holko P, Pilc A. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. A sys- tematic review and meta-analysis. Arch Med Sci. 2012;8(5):767–75. 71. A study of mesothelin redirected autologous T cells for advanced pancreatic carcinoma. https://ClinicalTrials.gov/show/ NCT02706782. 72. Pilot study of autologous T-cells in patients with metastatic pancreatic cancer. https://ClinicalTrials.gov/show/ NCT02465983. 73. Katz SC, Burga RA, McCormack E, et al. Phase I hepatic immunotherapy for metastases study of intra-arterial chimeric antigen receptor-modified T-cell therapy for CEA? liver metastases. Clin Cancer Res. 2015;21(14):3149–59. 74. Ahmed N, Brawley VS, Hegde M, et al. Human epidermal growth factor receptor 2 (HER2)-specific chimeric antigen receptor-modified T cells for the immunotherapy of HER2- positive sarcoma. J Clin Oncol. 2015;33(15):1688–96. 75. Her2 chimeric antigen receptor expressing T cells in advanced sarcoma. https://ClinicalTrials.gov/show/NCT00902044. 76. Hegde UP, Mukherji B. Current status of chimeric antigen receptor engineered T cell-based and immune checkpoint blockade-based cancer immunotherapies. Cancer Immunol Immunother. 2017;66(9):1113–21. 77. Lindau D, Gielen P, Kroesen M, Wesseling P, Adema GJ. The immunosuppressive tumour network: myeloid-derived suppres- sor cells, regulatory T cells and natural killer T cells. Immunology. 2013;138(2):105–15. 78. Tanaka A, Sakaguchi S. Regulatory T cells in cancer immunotherapy. Cell Res. 2017;27(1):109–18. 79. Liu C, Workman CJ, Vignali DA. Targeting regulatory T cells in tumors. FEBS J. 2016;283(14):2731–48. 80. Rech AJ, Vonderheide RH. Clinical use of anti-CD25 antibody daclizumab to enhance immune responses to tumor antigen vaccination by targeting regulatory T cells. Ann N Y Acad Sci. 2009;1174:99–106. 81. Rech AJ, Mick R, Martin S, et al. CD25 blockade depletes and selectively reprograms regulatory T cells in concert with immunotherapy in cancer patients. Sci Transl Med. 2012;4(134):134–62. 82. Telang S, Rasku MA, Clem AL, et al. Phase II trial of the regulatory T cell-depleting agent, denileukin diftitox, in patients with unresectable stage IV melanoma. BMC Cancer. 2011;11:515. 83. Romano E, Kusio-Kobialka M, Foukas PG, et al. Ipilimumab- dependent cell-mediated cytotoxicity of regulatory T cells ex vivo by nonclassical monocytes in melanoma patients. Proc Natl Acad Sci U S A. 2015;112(19):6140–5. 84. Tobin RP, Davis D, Jordan KR, McCarter MD. The clinical evidence for targeting human myeloid-derived suppressor cells in cancer patients. J Leukoc Biol. 2017;102(2):381–91. 85. Schilling B, Sucker A, Griewank K, et al. Vemurafenib reverses immunosuppression by myeloid derived suppressor cells. Int J Cancer. 2013;133(7):1653–63. 86. de Coana YP, Wolodarski M, Poschke I, et al. Ipilimumab treatment decreases monocytic MDSCs and increases CD8 effector memory T cells in long-term survivors with advanced melanoma. Oncotarget. 2017;8(13):21539–53. 87. Ko JS, Rayman P, Ireland J, et al. Direct and differential sup- pression of myeloid-derived suppressor cell subsets by sunitinib is compartmentally constrained. Cancer Res. 2010;70(9):3526–36. 88. Mirza N, Fishman M, Fricke I, et al. All-trans-retinoic acid improves differentiation of myeloid cells and immune response in cancer patients. Cancer Res. 2006;66(18):9299–307. 89. Lathers DM, Clark JI, Achille NJ, Young MR. Phase 1B study to improve immune responses in head and neck cancer patients using escalating doses of 25-hydroxyvitamin D3. Cancer Immunol Immunother. 2004;53(5):422–30. 90. Noonan KA, Ghosh N, Rudraraju L, Bui M, Borrello I. Tar- geting immune suppression with PDE5 inhibition in end-stage multiple myeloma. Cancer Immunol Res. 2014;2(8):725–31. 91. Weed DT, Vella JL, Reis IM, et al. Tadalafil reduces myeloid- derived suppressor cells and regulatory T cells and promotes tumor immunity in patients with head and neck squamous cell carcinoma. Clin Cancer Res. 2015;21(1):39–48. 92. Annels NE, Shaw VE, Gabitass RF, et al. The effects of gem- citabine and capecitabine combination chemotherapy and of low-dose adjuvant GM-CSF on the levels of myeloid-derived suppressor cells in patients with advanced pancreatic cancer. Cancer Immunol Immunother. 2014;63(2):175–83. 93. Kanterman J, Sade-Feldman M, Biton M, et al. Adverse immunoregulatory effects of 5FU and CPT11 chemotherapy on myeloid-derived suppressor cells and colorectal cancer out- comes. Cancer Res. 2014;74(21):6022–35. 94. Fischer K, Hoffmann P, Voelkl S, et al. Inhibitory effect of tumor cell-derived lactic acid on human T cells. Blood. 2007;109(9):3812–9. 95. Chang CH, Qiu J, O’Sullivan D, et al. Metabolic competition in the tumor microenvironment is a driver of cancer progression. Cell. 2015;162(6):1229–41. 96. Allavena P, Germano G, Marchesi F, Mantovani A. Chemokines in cancer related inflammation. Exp Cell Res. 2011;317(5):664–73. 97. Griffioen AW, Damen CA, Martinotti S, Blijham GH, Groe- newegen G. Endothelial intercellular adhesion molecule-1 expression is suppressed in human malignancies: the role of angiogenic factors. Cancer Res. 1996;56(5):1111–7. 98. Lider O, Mekori YA, Miller T, et al. Inhibition of T lymphocyte heparanase by heparin prevents T cell migration and T cell- mediated immunity. Eur J Immunol. 1990;20(3):493–9. 99. Kalluri R, Zeisberg M. Fibroblasts in cancer. Nat Rev Cancer. 2006;6(5):392–401. 100. Shin HC, Han W, Moon HG, et al. Breast-conserving surgery after tumor downstaging by neoadjuvant chemotherapy is oncologically safe for stage III breast cancer patients. Ann Surg Oncol. 2013;20(8):2582–9. 101. Vuky J, Corman JM, Porter C, Olgac S, Auerbach E, Dahl K. Phase II trial of neoadjuvant docetaxel and CG1940/CG8711 followed by radical prostatectomy in patients with high-risk clinically localized prostate cancer. Oncologist. 2013;18(6):687–8. 102. Long GV, Weber JS, Larkin J, et al. Nivolumab for patients with advanced melanoma treated beyond progression: analysis of 2 phase 3 clinical trials. JAMA Oncol. 2017;3(11):1511–19. Immunotherapy for Solid Tumors: A Review