You are on page 1of 35

DOI: 10.1002/JLB.

5BT0420-585RR

REVIEW

Breakthrough concepts in immune-oncology: Cancer vaccines


at the bedside

Sohini Roy1 Tarsheen K. Sethi2 David Taylor1 Young J. Kim1


Douglas B. Johnson2

1 Department of Otolaryngology – Head & Neck

Surgery, Vanderbilt University Medical Center, Abstract


Nashville, Tennessee, USA Clinical approval of the immune checkpoint blockade (ICB) agents for multiple cancer types
2 Department of Medicine, Vanderbilt University
has reinvigorated the long-standing work on cancer vaccines. In the pre-ICB era, clinical efforts
Medical Center, Nashville, Tennessee, USA focused on the Ag, the adjuvants, the formulation, and the mode of delivery. These translational
Correspondence efforts on therapeutic vaccines range from cell-based (e.g., dendritic cells vaccine Sipuleucel-T)
Young Kim and Douglas Johnson, 777 PRB,
2220 Pierce Ave, Nashville, TN 37232, USA.
to DNA/RNA-based platforms with various formulations (liposome), vectors (Listeria monocyto-
Email: y2.kim@vumc.org; genes), or modes of delivery (intratumoral, gene gun, etc.). Despite promising preclinical results,
douglas.b.johnson@vumc.org cancer vaccine trials without ICB have historically shown little clinical activity. With the anticipa-
tion and expansion of combinatorial immunotherapeutic trials with ICB, the cancer vaccine field
has entered the personalized medicine arena with recent advances in immunogenic neoantigen-
based vaccines. In this article, we review the literature to organize the different cancer vaccines
in the clinical space, and we will discuss their advantages, limits, and recent progress to overcome
their challenges. Furthermore, we will also discuss recent preclinical advances and clinical strate-
gies to combine vaccines with checkpoint blockade to improve therapeutic outcome and present
a translational perspective on future directions.

KEYWORDS
cancer, DNA, neoantigen, nucleotide, peptide, RNA, vaccine

1 CLINICAL LANDSCAPE 440 patients enrolled between 1995 and 2004 at the National Cancer
OF CANCER VACCINES Institute presented with metastatic disease and treated with different
vaccines showed a lowly objective response rate of 2.6%.1 Early strate-
Since James Coley’s first attempt to generate therapeutic immunity gies on therapeutic cancer vaccines focused on tumor-associated Ags
against inoperable sarcoma with inoculated heat-inactivated bacteria (TAA), which include overexpressed Ags (e.g., Her2/neu), cancer testis
(“Coley’s toxins”), cancer vaccine development has lagged significantly (CT) Ags, developmentally regulated Ags (e.g., oncofetal proteins), or
behind the success of infectious disease vaccines (Fig. 1). This can mutated neoantigens that are not found in normal cells.2,3 Failure of
partly be attributed to the immunosuppressive context of cancer these trials has been attributed to generation of tolerance to TAA, low
vaccine utilization (age, high disease burden (metastasis), cancer- affinity/avidity tumor-specific T cells, and the homeostatic response
induced and cancer-therapy induced immunosuppression etc.). FDA to check hyper-responsive adaptive effectors cells.4 Other hurdles
approved prophylactic cancer vaccines are operationally infectious include suboptimal dose, mode of delivery, and improper or limited
disease vaccines that target oncogenic microbes (human papilloma adjuvant selections. However, the FDA approval of immune checkpoint
and hepatitis B viruses) prior to the transformation into neoplastic inhibitors has led to a renewed enthusiasm for therapeutic cancer
cells. These vaccines have achieved a great deal of success in prevent- vaccines. In this review, we provide an overview of the emerging field
ing these viral-associated cancers and are outside the scope of this of “Cancer Vaccine 2.0”, the challenges that remain and the factors that
review. Therapeutic cancer vaccines, in contrast, have yielded limited need to be taken into consideration for maximizing the chances of suc-
clinical benefits. For example, an earlier report on meta-analysis of cess of this mode of combinatorial immunotherapy. We will also discuss

Received: 4 November 2019 Revised: 15 April 2020 Accepted: 18 April 2020


J Leukoc Biol. 2020;1–35. www.jleukbio.org ○
c 2020 Society for Leukocyte Biology 1
2 ROY ET AL .

FIGURE 1 A timeline of vaccine development

novel vaccine platforms (oncolytic virus vaccine, induced pluripotent activity and T-VEC contains viral products and GM-CSF that can elicit
stem cell (iPSC) vaccine, personalized neoantigen vaccine) and their multiple adjuvant activity. Sipuleucel-T is a dendritic cell therapy that
combinatorial clinical trials with immune checkpoint blockade (ICB). bypasses the endogenous TAA capture. Another salient feature is the
clinical context of BCG’s success—the vaccine was used for early stage
and in situ disease, suggesting that cancer vaccine’s effectiveness
2 VACCINES IN CLINICAL DEVELOPMENT may occur primarily in the context of low tumor burden. The primary
concern with all these trials is the lack of biomarker development and
2.1 FDA approved cancer vaccines its associated mechanism of action in cancer patients.

While the clinical efficacy of therapeutic vaccines for cancer as a


monotherapy is low, the following examples of approved cancer vac-
2.2 Vaccine trials
cines illustrate the critical elements required for successful translation
and some of the hurdles/problems that need to be addressed. The ear- Many other vaccines have been tested in phase I/II/III trials, but the
liest post-Coley “tumor vaccine” to be approved was Bacille Calmette- results have been less impressive. Owing to the exhaustive list of clini-
Guerin (BCG) in 1977. This intravesically delivered, live, attenuated cal trials using different vaccine platforms, we have attempted to high-
mycobacterial extract vaccine was approved for use in prophylaxis of light the completed clinical trials from the last 5 years in Table 1. Many
early stage bladder cancer and the treatment of urothelial carcinoma of these vaccines have adopted TAA and microbial vectors that fared
in situ. The approval was based on a phase III trial that showed a 5-year well in preclinical studies, though this success did not translate into
disease free survival (DFS) of 45% with BCG.5 The FDA approval of the clinic. With the approval of ICB, the most recent iteration of tri-
Sipuleucel-T (Provenge) for the treatment of metastatic castration- als has focused on combining vaccines with ICB and other therapeutic
resistant prostate cancer (mCRPC) in 2010 was another advance in modalities. The mechanisms and current evidence in support of these
cancer immunotherapy. It is an autologous dendritic cell (DC)-based combinations have been described in detail elsewhere8,9 and also
immunotherapy containing at least 50 million autologous CD54+ cells briefly later in this review. Leveraging the targeted generation of tumor
activated with a recombinant fusion protein (PA2024) consisting of specific T-cell response from cancer vaccines and sustaining these
prostatic acid phosphatase (PAP) fused with GM-CSF (PAP-GMCSF). responses in the tumor microenvironment (TME) with ICB is an attrac-
The approval was based on a placebo-controlled phase III trial tive prospect and has led to the testing of these combinations. Mech-
with 512 patients randomized in a 2:1 ratio to receive Sipuleucel-T anistically, vaccination alone has been shown to increase the adaptive
(341 patients) or placebo (171 patients).6 More recently, talimogene immune resistance mechanism whereby IFN-𝛾 from tumor infiltrating
laherparepvec (T-VEC), an oncolytic virus-based vaccine was approved T cells can increase programmed death-ligand 1 (PD-L1) expression on
for the treatment of advanced stage melanoma in 2015. T-VEC is an tumor cells.10,11 Preclinical studies have provided evidence for a syn-
oncolytic live attenuated herpes simplex virus-1 (HSV-1) genetically ergistic effect combining vaccines and checkpoint inhibitors including
modified to replicate within the tumor and produce GM-CSF. The cytotoxic T-lymphocyte associated protein 4 (CTLA-4) inhibitors and
approval followed a phase III study that met its primary endpoint of PD-1/PD-L1 inhibitors.12-18
superior durable response rate (defined as objective response lasting Early phase studies evaluating the combination of vaccines and
> 6 months) compared with GM-CSF, 16.3% versus 2.1%, respectively immune checkpoint inhibitors have been completed or are ongoing.
(OR 8.9; P < 0.001).7 All these 3 examples underscore the importance We have separately highlighted selected completed combination trials
of adjuvants and the activation of the innate immunity for clinical effi- in the last 5 years in Table 2. A phase Ib study of Ipilimumab (CTLA-4
cacy. While it is assumed that BCG and T-VEC-mediated induction of inhibitor) and GVAX in pancreatic adenocarcinoma showed improve-
cytolytic action on the tumor cells can release endogenous TAA, both ment in OS over Ipilimumab alone.19 The combination of MART-1
do not have specific TAA formulated in the vaccine. Both BCG and T- peptide-pulsed DC and Tremelimumab showed evidence of activity in
VEC contain a rich source of Myeloid differentiation primary response a phase I trial with advanced melanoma patients.20 Patients with pre-
protein (MyD88) activating adjuvants—BCG contains TLR4 agonist treated advanced melanoma showed some responses to autologous
TA B L E 1 Selected list of completed vaccine trials in the last 5 years
ROY ET AL .

Trial
Vaccine Type Disease Trial combination NCT ID Phase Results References
1
GVAX GVAX Pancreatic cancer Cyclophosphamide NCT02243371 II NA
CRS 207
Nivolumab
GVAX Pancreatic cancer Cyclophosphamide NCT02004262 IIb Study did not meet primary 2

CRS 207 efficacy endpoint with no


chemotherapy improvement in OS in
Cy/GVAX + CRS-207
compared to chemotherapy
alone
Colon gvax mismatch repair–proficient Cyclophosphamide NCT02981524 II Gvax can modulate immune 3

(MMR-p) advanced pembrolizumab response compared to


colorectal cancer pembrolizumab alone
Dendritic cell Peritoneal Surface Celecoxib NCT02151448 I/II NA NA
Malignancies Interferon-alfa-2b
rintatolimod
NY-ESO-1(157-165) Locally Advanced Solid Aldesleukin NCT02775292 I NA NA
Peptide-pulsed Tumors Expressing Cyclophosphamide
Autologous Dendritic Cell NY-ESO-1 Fludarabine Phosphate
Vaccine Nivolumab
NY-ESO-1 Reactive TCR
Retroviral Vector
Transduced Autologous
PBL
DLK1, EPHA2, HBB, NRP1, Metastatic melanoma Dasatinib NCT01876212 II NA NA
RGS5, TEM1 peptide
Pulsed with Cancer stem cell Ovarian cancer NCT02178670 I/II NA NA
Pulsed with Cancer stem cell Lung cancer NCT02084823 I/II NA NA
Pulsed with Cancer stem cell Nasopharyngeal cancer NCT02115958 I/II NA NA
Pulsed with Cancer stem cell Pancreatic cancer NCT02074046 I/II NA NA
Pulsed with Cancer stem cell Colorectal cancer NCT02176746 I/II NA NA
Pulsed with Cancer stem cell Hepatocellular carcinoma NCT02089919 I/II NA NA
Sipuleucel-T Metastatic Tasquinimod NCT02159950 II NA NA
hormone-resistant
prostate cancer
Allogeneic DC Metastatic Renal Cell Sunitinib NCT02432846 II Complete response induced Onclive, 2019
(INTUVAX) Carcinoma by vaccine; details results
NA
INTUVAX Gastrointestinal cancer NCT02686944 I NA NA
Glioma stem cell pulsed Recurrent glioblastoma NCT02820584 I NA NA
(Continues)
3
4

TA B L E 1 (Continued)

Trial
Vaccine Type Disease Trial combination NCT ID Phase Results References
Dendritic cell, Pulsed with tumor cell lysate NCT03114631 I/II NA NA
Tumor lysate or MUC-1/WT-1 peptide
DC vaccine, RNA CEA RNA Metastatic adenocarcinoma NCT00004604 I NA NA
as antigen
CEA RNA Metatstatic colon cancer NCT00003433 I/II NA NA
4
MART-1, tyrosinase, gp100, Metastatic melanoma siRNA to target NCT00672542 I In combination arm, immune
and MAGE-3 RNA immuneproteasome response remained
antigen prossessing elevated, circulating cancer
cells reduced and lytic T
cells activity was induced.
5
cytomegalovirus (CMV) Glioblastoma Multiforme Basiliximab,temozolomide, NCT00626483 I/II No significant clinical
pp65-lysosomal- radiation therapy benefit
associated membrane
protein (LAMP) mRNA and
GMCSF
6
cytomegalovirus (CMV) Glioblastoma temozolomide NCT00639639) I long-term PFS and OS in
pp65-lysosomal- vaccinated arm
associated membrane
protein (LAMP) mRNA and
GMCSF
TLR7/8-matured DCs AML in CR NCT01734304 I/II T cell responses observed. 7

electroporated with mRNA Long-term disease control and


encoding WT1, PRAME, immunological responses
and CMVpp65 are studied in the ongoing
phase II trial.
MAGE-A3, MelanA and Cutaneous melanoma KLH NCT00074230 I/II NA NA
Survivin RNA
(Continues)
ROY ET AL .
(Continued)
ROY ET AL .

TA B L E 1

Trial
Vaccine Type Disease Trial combination NCT ID Phase Results References
CMV pp65 RNA glioblastoma Autologous T cells NCT00693095 I NA John Sampson
activated in vitro with DUMC. Evaluation
CMV pp65 pulsed DC of Recovery From
Drug-Induced
Lymphopenia Using
Cytomegalovirus-
specific T-cell
Adoptive Transfer.
In:
ClinicalTrials.gov
[Internet].
Bethesda (MD):
National Library of
Medicine (US);
2000. Available
from: http://clinical
trials.gov/show/
NCT00693095_
NLM_Identifier:
NCT00693095.
Amplified autologous tumor Metastatic prostate cancer NCT00108264 I NA NA
cell RNA
8
CEA peptide or CEA RNA Metastatic Colorectal cancer KLH NCT00228189 I/II RNA pulsed DC vaccination
failed to show improved
response
9
gp100 and tyrosinase mRNA Metastatic melanoma KLH NCT00243529 I/II broad repertoire of IFN𝛾
producing TAA-specific
CD8+ and CD4+ T cell
responses in stage III
patients mainly
Amplified autologous tumor Metastatic renal cell NCT00087984 I/II NA NA
cell RNA carcinoma
10
amplified tumor RNA plus metastatic clear cell renal cell sunitinib NCT00678119 II Doubling of expected survival,
synthetic CD40L RNA carcinoma encouraging long-term and
5-year overall survival in
intermediate risk group
Autologous tumor stem cell Recurrent glioblastoma Radiation, bevacizumab NCT00890032 I Immune analysis not yet 11

amplified RNA published


Tumor RNA Advanced kidney cancer NCT00005816 I NA NA
PSA RNA Prostate cancer NCT00004211 I/II NA NA
(Continues)
5
6

TA B L E 1 (Continued)

Trial
Vaccine Type Disease Trial combination NCT ID Phase Results References
DC vaccine, DNA Autologous DC pulsed with Metastatic breast cancer NCT00197522 I NA NA
antigen Adenovirus Expressing
Her-2
Personalized Neoantigen mRNA Metastatic melanoma, NCT03480152 I/II Presence of neoepitope 12

neoantigen epithelial cancers. specific T cell responses


Neoantigen long peptides Melanoma TLR3 agonist poly-ICLC NCT01970358 I Elicited neoepitope specific T 13

Hiltonol R , anti-PD-1 cell responses, positive


after recurrence effect on recurrence
prevention; 2 patients with
metastatic disease who
relapsed benefited from
anti PD-1
14
DC loaded with neoantigen Cutaneous melanoma gp100 peptide; for first NCT00683-670, I Neoepitope specific T cell
peptide dose, influenza virus BB-IND responses found
vaccine was included as 13590
recall response
RNA based poly-neo epitope Melanoma Melanoma specific peptide Neoepitope specific 15

vaccine before polyfunctional T cell


neoantigen vaccine responses and autologous
injection; anti PD-1 if tumor cell killing found;
relapsed prevented recurrence and
regression in metastatic
lesions.
Oncolytic virus OncoVex-GMCSF Pancreatic cancer NCT00402025 I n/a n/a
T-VEC Melanoma NCT02014441 II Vaccine can be administered 16

to patients without
shedding by transmission
17,18,19,20,21
T-VEC Melanoma Compared with GMCSF NCT00769704 III T-VEC achieved higher DRR
and median OS compared
to GMCSF
22
T-VEC Melanoma NCT00289016 II Response rate 26%, abscopal
effects observed; survival
benefits recorded
T-VEC Melanoma Pembrolizumab NCT02263508 Ib Improved anti-PD-1 efficacy; 23

responders to combination
therapy had higher CD8+
and IFN-𝛾 signature
24
T-VEC Melanoma Ipilimumab NCT01740297 II ORR higher in combination
therapy with enhanced
anti-tumor activity,
abscopal effects observed
recombinant nonpathogenic Glioma NCT01491893 NA NA
polio-rhinovirus chimera
(PVSRIPO)
(Continues)
ROY ET AL .
ROY ET AL .

TA B L E 1 (Continued)

Trial
Vaccine Type Disease Trial combination NCT ID Phase Results References
Oncolytic adenovirus BCG unresponsive non NCT02365818 II Strong response and limited 25

expressing GMCSF muscle invasive Bladder progression in patients with


cancer carcinoma in situ
TBI-1401(HF10) HSV1 virus Solid tumors with NCT02428036 I Stabilized disease Annals of Oncology
superficial lesions (2017) 28
(suppl_10): x113-
x116https://doi.org/
10.1093/annonc/
mdx667
26
human-derived adenovirus. Glioblastoma and IFN-gamma NCT02197169 Ib IFN-gamma did not improve
DNX-2401 gliosarcoma survival
CAVATAK Uveal melanoma metastatic ipilimumab NCT03408587 Ib NA NA
to liver
VCN-01 Pancreatic cancer Gemcitabine NCT02045589 I NA NA
AbraxaneR

27
HF10 Metastatic melanoma ipilimumab NCT02272855 II Combination showed
favorable profile and
antitumor activity
reolysin Pancreatic cancer Gemcitabine NCT02620423 II Antitumor response in 28

Irinotecan previously treated patients


Leucovorin observed. In depth immune
5-fluorouracil analysis planned and not
pembrolizumab yet published
29
Bacterial vaccine VXM01 Recurrent glioblastoma who NCT02718443 I Induced immune
(live attenuated bacteria have failed atleast response.Increased
modified to target radiochemotherapy with CD8+/Treg ratio
VEGFR2) temozolomide Intratumoral PDL-1
decreased in 1 patient
ADU-623 Astrocytic Tumors NCT01967758 I NA NA
(live-attenuated Listeria Glioblastoma Multiforme
Monocytogenes Strain Anaplastic Astrocytoma
expressing the Brain Tumor
EGFRvIII-NY-ESO-1
vaccine)
30
JNJ-64041809 NCT02625857 I Observed immune response
(Live Attenuated did not translate to clinical
Double-deleted Listeria) response
(Continues)
7
8

TA B L E 1 (Continued)

Trial
Vaccine Type Disease Trial combination NCT ID Phase Results References
Peptide vaccine APVAC1 (off the shelf TAA glioblastoma Chemotherapy NCT02149225 I APVAC1-induced sustained 31

peptide) Poly-ICLC CD8 responses with


APVAC2 (personalized GMCSF memory phenotype
mutated peptide) APVAC2 induced CD4
responses of favorable TH1
type
GRN-1201 Resected melanoma GMCSF NCT02696356 I NA NA
((HLA)-A2-restricted
peptides)
IDO peptide Melagnant melanoma Ipilimumab NCT02077114 I NA NA
Vemurafenib
IDH1 Peptide emulsified in glioma Radiation NCT02454634 I Met its endpoint; 32,33,34

incomplete Freund’s chemotherapy immunogenicity observed


adjuvant
RV001V (RhoC peptide) Metastatic prostate NCT03199872 I/II Sustained immune response Rhovac press release
emulsified in the cancer at 3 and 6m reported.
immunoadjuvant Further analyses pending
montanide ISA-51
DNA vaccine MVA-brachyury-TRICOM Lung, breast, prostate, NCT02179515 I Induced T cell response in 35,36

vaccine ovarian vaccinated patients.


Most striking clinical outcome
observed in metastatic
EGFR-mutated lung cancer
who were on erlotinib
PROSTVAC Prostate cancer NCT02153918 II Increased CD8+/CD4+ 37

(PSA, TRICOM) peritumoral infioltrate


Immune response observed
TAA immune response in 50%
patients in peripheral blood
Increase in suppressive
markers post vaccination
observed

PROSTVAC Prostate cancer GMCSF or placebo NCT01322490 III No improvement in overall J Clin Oncol 36, 2018
(PSA, TRICOM) survival (suppl; abstr 5006)
modified vaccinia virus Recurrent Ovarian gemcitabine NCT02275039 I Combination feasible and did 38

ankara vaccine Epithelial Cancer not induce autoimmunity


expressing p53
(Continues)
ROY ET AL .
TA B L E 1 (Continued)
ROY ET AL .

Trial
Vaccine Type Disease Trial combination NCT ID Phase Results References
Whole cell HS110 Non-Small Cell Lung Cancer Cyclophosphamide NCT02117024 II Combination in first cohort 39

vaccine (viagenpumatucel-L: Or physician’s choice of Increased PD-1, Tim-3 and


irradiated tumor cells and chemotherapy alone Ki67 was observed on
modified to overexpress CD8+ T cells isolated from
gp96-Ig) peripheral blood indicating
CD8+ response to vaccine
HS110 Lung cancer nivolumab NCT02439450 Ib/II Immune response observed in ImmunoOncology
reduced tumor cases News
60% patients in cold tumor
cohort had tumot reduction
Mixed vaccine Kidney cancer NCT03357289 I/II NA NA
Mixed vaccine Metastatic colorectal cancer NCT03357276 I/II NA NA
Mixed vaccine Metastatic sarcoma NCT03357315 I/II NA NA
VIGIL Advanced melanoma Pembrolizumab NCT02574533 I NA NA
(GMCSF and shRNA to
target furin, the
pro-protein convertase
activator for TGF-𝛽1 and
TGF-𝛽2
VIGIL high-grade papillary NCT02346747 II/III Recurrence free survival was 40

serous/clear cell/ improved in vigil arm


endometrioid ovarian,
fallopian tube, or primary
peritoneal cancer
Owing to the exhaustive list of clinical trials using different vaccine platforms, several trials could not be included in this list. Details of all such trials are readily accessible from the Clinicaltrials.gov website. For the
current review, we have included trials that were completed and met safety endpoints. We have excluded trials for HPV+ cancer types and, also, ones that were either prematurely withdrawn or terminated.
1 Le DT, Crocenzi TS, Uram JN, Lutz ER, Laheru DA, Sugar EA, Vonderheide RH, Fisher GA, Ko AH, Murphy A, McDougall K, Ferber S, Brockstedt DG, Jaffee EM. Randomized phase 2 study of the safety, efficacy,

and immune response of GVAX pancreas (with cyclophosphamide) and CRS-207 with or without nivolumab in patients with previously treated metastatic pancreatic adenocarcinoma (STELLAR). Journal of Clinical
Oncology. 2016;34(15_suppl):TPS4153-TPS. https://doi.org/10.1200/JCO.2016.34.15_suppl.TPS4153.
2
Le DT, Picozzi VJ, Ko AH, Wainberg ZA, Kindler H, Wang-Gillam A, Oberstein P, Morse MA, Zeh HJ, Weekes C, Reid T, Borazanci E, Crocenzi T, LoConte NK, Musher B, Laheru D, Murphy A, Whiting C, Nair
N, Enstrom A, Ferber S, Brockstedt DG, Jaffee EM. Results from a Phase IIb, Randomized, Multicenter Study of GVAX Pancreas and CRS-207 Compared with Chemotherapy in Adults with Previously Treated
Metastatic Pancreatic Adenocarcinoma (ECLIPSE Study). Clinical Cancer Research. 2019;25(18):5493-502. https://doi.org/10.1158/1078-0432.ccr-18-2992.
3
Yarchoan M, Ferguson AK, Durham JN, Rozich N, Rodriguez C, Huang C-Y, Browner IS, Jesus-Acosta AD, Le DT, Laheru D, Donehower RC, Jaffee EM, Zheng L, Azad NS. A phase II study of
GVAX colon vaccine with cyclophosphamide and pembrolizumab in patients with mismatch repair–proficient (MMR-p) advanced colorectal cancer. Journal of Clinical Oncology. 2019;37(4_suppl):563-.
https://doi.org/10.1200/JCO.2019.37.4_suppl.563.
4
Dannull J, Haley NR, Archer G, Nair S, Boczkowski D, Harper M, De Rosa N, Pickett N, Mosca PJ, Burchette J, Selim MA, Mitchell DA, Sampson J, Tyler DS, Pruitt SK. Melanoma immunotherapy using mature DCs
expressing the constitutive proteasome. J Clin Invest. 2013;123(7):3135-45. Epub 2013/08/13. https://doi.org/10.1172/JCI67544. PubMed PMID: 23934126; PMCID: PMC3696565.
5
Vlahovic G, Archer GE, Reap E, Desjardins A, Peters KB, Randazzo D, Healy P, Herndon JE, Friedman AH, Friedman HS, Bigner DD, Sampson JH. Phase I trial of combination of antitumor immunother-
apy targeted against cytomegalovirus (CMV) plus regulatory T-cell inhibition in patients with newly-diagnosed glioblastoma multiforme (GBM). Journal of Clinical Oncology. 2016;34(15_suppl):e13518-e.
https://doi.org/10.1200/JCO.2016.34.15_suppl.e13518.
6
Batich KA, Reap EA, Archer GE, Sanchez-Perez L, Nair SK, Schmittling RJ, Norberg P, Xie W, Herndon JE, 2nd, Healy P, McLendon RE, Friedman AH, Friedman HS, Bigner D, Vlahovic G, Mitchell DA, Sampson
JH. Long-term Survival in Glioblastoma with Cytomegalovirus pp65-Targeted Vaccination. Clin Cancer Res. 2017;23(8):1898-909. Epub 2017/04/16. https://doi.org/10.1158/1078-0432.CCR-16-2057. PubMed
PMID: 28411277; PMCID: PMC5559300.
9
10

TA B L E 1 Continued

7
Deiser K, Lichtenegger FS, Schnorfeil F, Koehnke T, Altmann T, Buecklein V, Moosmann A, Brueggemann M, Wagner B, Hiddemann W, Bigalke I, Kvalheim G, Subklewe MS. Next-Generation Dendritic Cell Vaccina-
tion in Postremission Therapy of AML: Results of a Clinical Phase I Trial. Blood. 2015;126(23):3805-. https://doi.org/10.1182/blood.V126.23.3805.3805.
8
Lesterhuis WJ, De Vries IJ, Schreibelt G, Schuurhuis DH, Aarntzen EH, De Boer A, Scharenborg NM, Van De Rakt M, Hesselink EJ, Figdor CG, Adema GJ, Punt CJ. Immunogenicity of dendritic cells pulsed with CEA
peptide or transfected with CEA mRNA for vaccination of colorectal cancer patients. Anticancer Res. 2010;30(12):5091-7. Epub 2010/12/29. PubMed PMID: 21187495.
9
Aarntzen EH, Schreibelt G, Bol K, Lesterhuis WJ, Croockewit AJ, de Wilt JH, van Rossum MM, Blokx WA, Jacobs JF, Duiveman-de Boer T, Schuurhuis DH, Mus R, Thielemans K, de Vries IJ, Figdor CG, Punt CJ, Adema
GJ. Vaccination with mRNA-electroporated dendritic cells induces robust tumor antigen-specific CD4+ and CD8+ T cells responses in stage III and IV melanoma patients. Clin Cancer Res. 2012;18(19):5460-70.
Epub 2012/08/17. https://doi.org/10.1158/1078-0432.CCR-11-3368. PubMed PMID: 22896657.
10 Amin A, Dudek AZ, Logan TF, Lance RS, Holzbeierlein JM, Knox JJ, Master VA, Pal SK, Miller WH, Jr., Karsh LI, Tcherepanova IY, DeBenedette MA, Williams WL, Plessinger DC, Nicolette CA, Figlin RA. Survival

with AGS-003, an autologous dendritic cell-based immunotherapy, in combination with sunitinib in unfavorable risk patients with advanced renal cell carcinoma (RCC): Phase 2 study results. J Immunother Cancer.
2015;3:14. Epub 2015/04/23. https://doi.org/10.1186/s40425-015-0055-3. PubMed PMID: 25901286; PMCID: PMC4404644.
11 Dunn-Pirio A, Peters K, DesJardins A, Randazzo D, Friedman H, Healy P, Herndon J, Reap E, Archer G, Li Q-J, Sampson J, Vlahovic G. Tumor stem cell RNA-loaded dendritic cell vaccine for recurrent glioblastoma:

a phase 1 trial (S41.004). Neurology. 2017;88(16 Supplement):S41.004.


12 Cafri G, Gartner JJ, Hopson K, Meehan RS, Zaks TZ, Robbins P, Rosenberg SA. Immunogenicity and tolerability of personalized mRNA vaccine mRNA-4650 encoding defined neoantigens expressed by the

autologous cancer. Journal of Clinical Oncology. 2019;37(15_suppl):2643-. https://doi.org/10.1200/JCO.2019.37.15_suppl.2643.


13 Ott PA, Hu Z, Keskin DB, Shukla SA, Sun J, Bozym DJ, Zhang W, Luoma A, Giobbie-Hurder A, Peter L, Chen C, Olive O, Carter TA, Li S, Lieb DJ, Eisenhaure T, Gjini E, Stevens J, Lane WJ, Javeri I, Nellaiappan

K, Salazar AM, Daley H, Seaman M, Buchbinder EI, Yoon CH, Harden M, Lennon N, Gabriel S, Rodig SJ, Barouch DH, Aster JC, Getz G, Wucherpfennig K, Neuberg D, Ritz J, Lander ES, Fritsch EF, Hacohen N,
Wu CJ. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature. 2017;547(7662):217-21. Epub 2017/07/06. https://doi.org/10.1038/nature22991. PubMed PMID: 28678778; PMCID:
PMC5577644.
14
Carreno BM, Magrini V, Becker-Hapak M, Kaabinejadian S, Hundal J, Petti AA, Ly A, Lie WR, Hildebrand WH, Mardis ER, Linette GP. Cancer immunotherapy. A dendritic cell vaccine increases the breadth and
diversity of melanoma neoantigen-specific T cells. Science. 2015;348(6236):803-8. Epub 2015/04/04. https://doi.org/10.1126/science.aaa3828. PubMed PMID: 25837513; PMCID: PMC4549796.
15
Sahin U, Derhovanessian E, Miller M, Kloke BP, Simon P, Lower M, Bukur V, Tadmor AD, Luxemburger U, Schrors B, Omokoko T, Vormehr M, Albrecht C, Paruzynski A, Kuhn AN, Buck J, Heesch S, Schreeb KH,
Muller F, Ortseifer I, Vogler I, Godehardt E, Attig S, Rae R, Breitkreuz A, Tolliver C, Suchan M, Martic G, Hohberger A, Sorn P, Diekmann J, Ciesla J, Waksmann O, Bruck AK, Witt M, Zillgen M, Rothermel A, Kasemann
B, Langer D, Bolte S, Diken M, Kreiter S, Nemecek R, Gebhardt C, Grabbe S, Holler C, Utikal J, Huber C, Loquai C, Tureci O. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against
cancer. Nature. 2017;547(7662):222-6. Epub 2017/07/06. https://doi.org/10.1038/nature23003. PubMed PMID: 28678784.
16
Andtbacka RHI, Amatruda T, Nemunaitis J, Zager JS, Walker J, Chesney JA, Liu K, Hsu CP, Pickett CA, Mehnert JM. Biodistribution, shedding, and transmissibility of the oncolytic virus talimogene laherparepvec
in patients with melanoma. EBioMedicine. 2019;47:89-97. Epub 2019/08/15. https://doi.org/10.1016/j.ebiom.2019.07.066. PubMed PMID: 31409575.
17
Andtbacka RH, Kaufman HL, Collichio F, Amatruda T, Senzer N, Chesney J, Delman KA, Spitler LE, Puzanov I, Agarwala SS, Milhem M, Cranmer L, Curti B, Lewis K, Ross M, Guthrie T, Linette GP, Daniels GA,
Harrington K, Middleton MR, Miller WH, Jr., Zager JS, Ye Y, Yao B, Li A, Doleman S, VanderWalde A, Gansert J, Coffin RS. Talimogene Laherparepvec Improves Durable Response Rate in Patients With Advanced
Melanoma. J Clin Oncol. 2015;33(25):2780-8. Epub 2015/05/28. https://doi.org/10.1200/JCO.2014.58.3377. PubMed PMID: 26014293.
18
Harrington KJ, Andtbacka RH, Collichio F, Downey G, Chen L, Szabo Z, Kaufman HL. Efficacy and safety of talimogene laherparepvec versus granulocyte-macrophage colony-stimulating factor in patients with
stage IIIB/C and IVM1a melanoma: subanalysis of the Phase III OPTiM trial. Onco Targets Ther. 2016;9:7081-93. Epub 2016/11/30. https://doi.org/10.2147/OTT.S115245. PubMed PMID: 27895500; PMCID:
PMC5119624.
19 Kaufman HL, Andtbacka RHI, Collichio FA, Amatruda T, Senzer NN, Chesney J, Delman KA, Spitler LE, Puzanov I, Ye Y, Li A, Gansert JL, Coffin R, Ross MI. Primary overall survival (OS) from OPTiM, a randomized

phase III trial of talimogene laherparepvec (T-VEC) versus subcutaneous (SC) granulocyte-macrophage colony-stimulating factor (GM-CSF) for the treatment (tx) of unresected stage IIIB/C and IV melanoma.
Journal of Clinical Oncology. 2014;32(15_suppl):9008a-a. https://doi.org/10.1200/jco.2014.32.15_suppl.9008a.
20 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. 2010;6(6):941-9. Epub 2010/06/10.

https://doi.org/10.2217/fon.10.66. PubMed PMID: 20528232.


21 Kaufman HL, Kim DW, DeRaffele G, Mitcham J, Coffin RS, Kim-Schulze S. Local and distant immunity induced by intralesional vaccination with an oncolytic herpes virus encoding GM-CSF in patients with stage

IIIc and IV melanoma. Ann Surg Oncol. 2010;17(3):718-30. Epub 2009/11/17. https://doi.org/10.1245/s10434-009-0809-6. PubMed PMID: 19915919.
22 Senzer NN, Kaufman HL, Amatruda T, Nemunaitis M, Reid T, Daniels G, Gonzalez R, Glaspy J, Whitman E, Harrington K, Goldsweig H, Marshall T, Love C, Coffin R, Nemunaitis JJ. Phase II clinical trial of a

granulocyte-macrophage colony-stimulating factor-encoding, second-generation oncolytic herpesvirus in patients with unresectable metastatic melanoma. J Clin Oncol. 2009;27(34):5763-71. Epub 2009/11/04.
https://doi.org/10.1200/JCO.2009.24.3675. PubMed PMID: 19884534.
23
Ribas A, Dummer R, Puzanov I, VanderWalde A, Andtbacka RHI, Michielin O, Olszanski AJ, Malvehy J, Cebon J, Fernandez E, Kirkwood JM, Gajewski TF, Chen L, Gorski KS, Anderson AA, Diede SJ,
Lassman ME, Gansert J, Hodi FS, Long GV. Oncolytic Virotherapy Promotes Intratumoral T Cell Infiltration and Improves Anti-PD-1 Immunotherapy. Cell. 2017;170(6):1109-19 e10. Epub 2017/09/09.
https://doi.org/10.1016/j.cell.2017.08.027. PubMed PMID: 28886381.
24
Chesney J, Puzanov I, Collichio F, Singh P, Milhem MM, Glaspy J, Hamid O, Ross M, Friedlander P, Garbe C, Logan TF, Hauschild A, Lebbe C, Chen L, Kim JJ, Gansert J, Andtbacka RHI, Kaufman HL. Randomized,
Open-Label Phase II Study Evaluating the Efficacy and Safety of Talimogene Laherparepvec in Combination With Ipilimumab Versus Ipilimumab Alone in Patients With Advanced, Unresectable Melanoma. J Clin
Oncol. 2018;36(17):1658-67. Epub 2017/10/06. https://doi.org/10.1200/JCO.2017.73.7379. PubMed PMID: 28981385; PMCID: PMC6075852.
ROY ET AL .
TA B L E 1 Continued
ROY ET AL .

25
Packiam VT, Lamm DL, Barocas DA, Trainer A, Fand B, Davis RL, 3rd, Clark W, Kroeger M, Dumbadze I, Chamie K, Kader AK, Curran D, Gutheil J, Kuan A, Yeung AW, Steinberg GD. An open label, single-arm, phase
II multicenter study of the safety and efficacy of CG0070 oncolytic vector regimen in patients with BCG-unresponsive non-muscle-invasive bladder cancer: Interim results. Urol Oncol. 2018;36(10):440-7. Epub
2017/08/02. https://doi.org/10.1016/j.urolonc.2017.07.005. PubMed PMID: 28755959.
26 Lang FF, Tran ND, Puduvalli VK, Elder JB, Fink KL, Conrad CA, Yung WKA, Penas-Prado M, Gomez-Manzano C, Peterkin J, Fueyo J. Phase 1b open-label randomized study of the oncolytic adenovirus DNX-2401

administered with or without interferon gamma for recurrent glioblastoma. Journal of Clinical Oncology. 2017;35(15_suppl):2002-. https://doi.org/10.1200/JCO.2017.35.15_suppl.2002.
27 Andtbacka RHI, Ross MI, Agarwala SS, Taylor MH, Vetto JT, Neves RI, Daud A, Khong HT, Ungerleider RS, Tanaka M, Grossmann KF. Final results of a phase II multicenter trial of HF10, a replication-

competent HSV-1 oncolytic virus, and ipilimumab combination treatment in patients with stage IIIB-IV unresectable or metastatic melanoma. Journal of Clinical Oncology. 2017;35(15_suppl):9510-.
https://doi.org/10.1200/JCO.2017.35.15_suppl.9510.
28 Mahalingam D, Fountzilas C, Moseley JL, Noronha N, Cheetham K, Dzugalo A, Nuovo G, Gutierrez A, Arora SP. A study of REOLYSIN in combination with pembrolizumab and chemotherapy in patients (pts) with

relapsed metastatic adenocarcinoma of the pancreas (MAP). Journal of Clinical Oncology. 2017;35(15_suppl):e15753-e. https://doi.org/10.1200/JCO.2017.35.15_suppl.e15753.
29 Wick W, Wick A, Sahm F, Riehl D, Deimling Av, Bendszus M, Kickingereder P, Beckhove P, Schmitz-Winnenthal FH, Jungk C, Wieckowski S, Podola L, Herold-Mende C, Unterberg A, Platten M. VXM01 phase I

study in patients with progressive glioblastoma: Final results. Journal of Clinical Oncology. 2018;36(15_suppl):2017-. https://doi.org/10.1200/JCO.2018.36.15_suppl.2017.
30 Drake CG, Pachynski RK, Subudhi SK, McNeel DG, Antonarakis ES, Bauer TM, Patricia D, Wade M, Zudaire E, Mason G, Knoblauch RE, Stone NL, Infante JR, Gottardis M, Fong L. Safety and preliminary

immunogenicity of JNJ-64041809, a live attenuated, double-deleted Listeria monocytogenes-based immunotherapy, in metastatic castration-resistant prostate cancer (mCRPC). Journal of Clinical Oncology.
2019;37(15_suppl):e16509-e. https://doi.org/10.1200/JCO.2019.37.15_suppl.e16509.
31
Wick W, Dietrich P-Y, Kuttruff S, Hilf N, Frenzel K, Admon A, Burg SHvd, Deimling Av, Gouttefangeas C, Kroep JR, Martinez-Ricarte F, Okada H, Ottensmeier CHH, Ponsati B, Poulsen HS, Stevanovic S, Tabatabai
G, Rammensee H-G, Sahin U, Singh H, Consortium G. GAPVAC-101: First-in-human trial of a highly personalized peptide vaccination approach for patients with newly diagnosed glioblastoma. Journal of Clinical
Oncology. 2018;36(15_suppl):2000-. https://doi.org/10.1200/JCO.2018.36.15_suppl.2000.
32
Platten M, Schilling D, Bunse T, Sahm F, Hueckelhoven A, Schenkel I, Stevanovic S, Schmitt A, Laumann M, Steinbach JP, Misch M, Tabatabai G, Weyerbrock A, Schnell O, Krex D, Hense J, Bendszus M, Deim-
ling Av, Schmitt M, Wick W. A mutation-specific peptide vaccine targeting IDH1R132H in patients with newly diagnosed malignant astrocytomas: A first-in-man multicenter phase I clinical trial of the German
Neurooncology Working Group (NOA-16). Journal of Clinical Oncology. 2016;34(15_suppl):TPS2082-TPS. https://doi.org/10.1200/JCO.2016.34.15_suppl.TPS2082.
33
Platten M, Schilling D, Bunse L, Wick A, Bunse T, Riehl D, Karapanagiotou-Schenkel I, Harting I, Sahm F, Schmitt A, Steinbach JP, Weyerbrock A, Hense J, Misch M, Krex D, Stevanovic S, Tabatabai G, Deim-
ling Av, Schmitt M, Wick W. A mutation-specific peptide vaccine targeting IDH1R132H in patients with newly diagnosed malignant astrocytomas: A first-in-man multicenter phase I clinical trial of the German
Neurooncology Working Group (NOA-16). Journal of Clinical Oncology. 2018;36(15_suppl):2001-. https://doi.org/10.1200/JCO.2018.36.15_suppl.2001.
34
Platten M, Schilling D, Bunse L, Wick A, Bunse T, Riehl D, Green E, Sanghvi K, Karapanagiotou-Schenkel I, Harting I, Sahm F, Steinbach J, Weyerbrock A, Hense J, Misch M, Krex D, Stevanovic S,
Tabatabai G, von Deimling A, Schmitt M, Wick W. ATIM-33. NOA-16: A FIRST-IN-MAN MULTICENTER PHASE I CLINICAL TRIAL OF THE GERMAN NEUROONCOLOGY WORKING GROUP EVAL-
UATING A MUTATION-SPECIFIC PEPTIDE VACCINE TARGETING IDH1R132H IN PATIENTS WITH NEWLY DIAGNOSED MALIGNANT ASTROCYTOMAS. Neuro-Oncology. 2018;20(suppl_6):vi8-vi9.
https://doi.org/10.1093/neuonc/noy148.028.
35 Heery CR, Donahue R, Lepone L, Grenga I, Richards J, Metenou S, Fernando RI, Dirmeier U, Singh H, Madan R, Gulley JL, Schlom J. Phase I, dose-escalation, clinical trial of MVA-Brachyury-TRICOM vaccine

demonstrating safety and brachyury-specific T cell responses. Journal for Immunotherapy of Cancer. 2015;3(Suppl 2):P132-P. https://doi.org/10.1186/2051-1426-3-S2-P132. PubMed PMID: PMC4645490.
36 eery CR, Palena C, McMahon S, Donahue RN, Lepone LM, Grenga I, Dirmeier U, Cordes L, Marte J, Dahut W, Singh H, Madan RA, Fernando RI, Hamilton DH, Schlom J, Gulley JL. Phase I Study of a Poxviral TRICOM-

Based Vaccine Directed Against the Transcription Factor Brachyury. Clin Cancer Res. 2017;23(22):6833-45. Epub 2017/09/01. https://doi.org/10.1158/1078-0432.CCR-17-1087. PubMed PMID: 28855356;
PMCID: PMC5690815.
37 Gulley JL, Sater HA, Marte J, Donahue RN, Rodriguez BW, Heery CR, Stern M, Thompson D, Cordes LM, Chun G, Karzai F, Bilusic M, Schlom J, Dahut WL, Madan RA, Pinto PA. Effect of rilimogene gal-

vacirepvec/rilimogene glafolivec on intra/peritumoral immune infiltrate in patients with localized prostate cancer undergoing radical prostatectomy. Journal of Clinical Oncology. 2018;36(15_suppl):5083-.
https://doi.org/10.1200/JCO.2018.36.15_suppl.5083.
38 Cristea MC, Hardwick N, Frankel PH, Morgan R, Leong LA, Diamond DJ. A phase I study of a p53MVA vaccine in combination with gemcitabine (GEM) in recurrent ovarian cancer (OC). Journal of Clinical Oncology.

2016;34(15_suppl):e17040-e. https://doi.org/10.1200/JCO.2016.34.15_suppl.e17040.
39 Cohen RB, Morgensztern D, Walsh WV, Bazhenova L, Melnyk AM, Beck JT, Nemunaitis JJ, Schreiber TH, Price ML. First interim exploratory analysis of immune response in patients with advanced

non-small cell lung cancer receiving viagenpumatucel-l (HS-110) in combination with low-dose cyclophosphamide in an ongoing phase II trial. Journal of Clinical Oncology. 2015;33(15_suppl):3077-.
https://doi.org/10.1200/jco.2015.33.15_suppl.3077.
40
Oh J, Barve M, Matthews CM, Koon EC, Heffernan TP, Fine B, Grosen E, Bergman MK, Fleming EL, DeMars LR, West L, Spitz DL, Goodman H, Hancock KC, Wallraven G, Kumar P, Bognar E, Manning L, Pap-
pen BO, Adams N, Senzer N, Nemunaitis J. Phase II study of Vigil(R) DNA engineered immunotherapy as maintenance in advanced stage ovarian cancer. Gynecol Oncol. 2016;143(3):504-10. Epub 2016/09/30.
https://doi.org/10.1016/j.ygyno.2016.09.018. PubMed PMID: 27678295.
11
12 ROY ET AL .

monocyte-derived mRNA electroporated dendritic cells (trimixdc- (trade name Yervoy) against CTLA-4 and 6 different agents against
mel) plus Ipilimumab in a phase II trial.21 PROSTVAC (a viral based PD-1 (Nivolumab and Pembrolizumab) and PD-L1 (Atezolizumab,
vaccine directed against the prostate specific Ag, PSA) was found to Avelumab, Durvalumab, and Cemiplimab), respectively. CTLA-4 binds
be safe in combination with ICB in a phase I study22 and its efficacy CD28 on DC and limits T cell functionality whereas PD-1 binding to
is currently being studied in mCRPC patients in several phase II trials PD-L1 disrupts the co-stimulatory signaling downstream of CD28.
(NCT02506114, NCT02933255). A phase II study evaluated the Clinical administration of monoclonal antibodies against CTLA-4 and
combination of an HPV E6/E7 peptide vaccine, ISA101 and Nivolumab PD-1/PD-L1 axis has shown transformative benefits in a minority of
and showed promising OS and overall response rates (ORR) compared patients with >15 cancers.36-48 These drugs are thought to remove
with retrospective data with Nivolumab in advanced HPV-16 positive the inhibitory brakes induced by the tumor cells within the TME to
cancer (22 out of 24 reported patients had oropharyngeal cancer).23 A prevent cell killing potential of activated T cells. Modest benefits
few additional phase I studies have reported safety of anti PD-1 combi- from these combinations were also noted in other cancers including
nations including p53MVA combined with Pembrolizumab (advanced non-small cell lung carcinoma,49-52 small cell lung cancer,53 prostate,54
solid cancers) and peptide vaccine with Nivolumab (melanoma).24-26 esophagogastric cancer,55 and Hodgkin’s lymphoma.56 Trials studying
Several ongoing studies are testing similar combinations and have other checkpoint blockade molecules in addition to anti PD-1/PD-L1
recently been well described by Curran et al.27 and anti-CTLA-4 are currently ongoing. For example, Monalizumab, a
blocking anti-human NKG2A mAb has already been tested in combina-
tion with cetuximab57 and metastatic microsatellite-stable colorectal
2.3 Personalized cancer vaccines—the next
cancer (MSS-CRC).58 LAG-3 (NCT04140500) and TIM-359 as well
generation of vaccines
as co-stimulatory molecules like OX4060 are also currently being
Recent years have seen major advances in next generation sequenc- evaluated in clinical trials. It is still unclear whether combination ICB
ing (NGS) providing the opportunity for successful crosstalk between can alter the existing TCR repertoire to foster the expansion of new
our ever-expanding knowledge base of oncologic mutations and tumor tumor Ag specific T cell clones.61,62
immunology. Targeting tumor-specific Ags (TSA or neoantigens) based Another mechanism of resistance is the global immunosuppressive
on unique tumor somatic mutations (neoantigens) has been one strat- TME. Unfortunately, despite the success stories observing durable
egy toward vaccines “personalized” to the tumor.28-30 This approach responses in a subset of patients, even in the immunologically “hot”
has the theoretical advantage of inherently escaping central toler- tumors characterized by high level of inflammation and high T cell
ance to generate high affinity T cell engagement. Recognition of the infiltration with elevated expression of PD-1/PD-L1 that benefit
tumor mutanome led to the next challenge of identifying which muta- from ICB, the percentage of responders is far from optimal due to
tions to target in the individual tumor. Leveraging bioinformatics tools, acquired resistance or co-expression of other inhibitory molecules.
one proposed framework used an RNA-based approach against mul- Non-immunogenic “cold” cancers like pancreatic, ovarian, breast,
tiple neo-epitopes and this method showed promise in a pilot cohort and glioblastoma that have poor immune infiltration are mostly
of melanoma patients.31 Combination trials of neoantigen based vac- non-responsive to ICB.63 Proposed mechanism of action for this resis-
cines with immune modulators including checkpoint inhibitors are in tance is the enrichment of immunosuppressive cells (myeloid derived
progress (NCT02287528, NCT03199040, NCT03380871). We have suppressor cells (MDSC) and regulatory T cells (Tregs) as well as the
provided a list of selected ongoing trials in personalized vaccine plat- dynamic temporo-spatial architecture of the TME that allows the
form in Table 3. tumor cells to outgrow the immune response generated against the
tumor. Cancer classification system relying on tumor characteristics,
2.3.1 Immune checkpoint blockade and immunoscore such as size of the primary tumor (T), spread to regional lymph nodes
Despite significant improvements in the therapeutic cancer vaccine (N), and distant metastases (M) (TNM staging) and other markers
platforms, they have still failed to provide meaningful clinical benefit (e.g., tumor cell of origin, tumor gene signatures, etc.) fails to provide
as monotherapy. This can be attributed to the TME intrinsic resis- complete prognostic information about the immune fitness of the
tance mechanisms discussed later in this review. Several studies have tumor and therefore may not correctly predict or stratify patients
highlighted that even in vaccinated patients, T and NK cells are less who will respond to a certain therapy. Currently, patient stratification
functional in the tumor.32-35 This can partly arise from increased based on Immunoscore and immune contexture that indicate the
expression of multiple inhibitory checkpoint molecules such as CTLA- type, density, and spatial distribution of the two types of immune
4, IDO (Indoleamine 2,3-dioxygenase), KIR (Killer Ig-like receptors), cells (cytolytic T-cells versus immunosuppressive Tregs and myeloid
LAG-3 (Lymphocyte activation protein 3), TIM-3 (T cell immunoglob- cells) in the pre-existing tumor are investigated to potentially help
ulin and mucin domain containing protein 3), SIGLEC-7 (Sialic acid predict which patients are more likely to benefit from ICB and direct
binding immunoglobulin like lectin-7), PD-1 (programmed cell death therapeutic intervention.64 This immune based classification system
1), PD-L1 (/programmed death-ligand 1), and NKG2A resulting in identifies four types of tumors: (i) hot (high immune cells infiltration
T cell exhaustion, NK cell impairment, and/or recruitment of Tregs with high expression of PD-1/PD-L1), (ii) excluded (T cells at margin,
in the tumor. Currently, there are two FDA approved pathways for indicating the system could potentially mount an anti-tumor response
targeting multiple inhibitors of checkpoint blockade (ICB): Ipilimumab but fails to do so due to the presence of tumor-imposed physical
ROY ET AL . 13

TA B L E 2 Selected ongoing trials evaluating vaccine combinations

Vaccine name and type Trial combination NCT number Disease Trialphase
Intravesical BCG Durvalumab +EBRT NCT03317158 Bladder 1/2
(Bacterial cancer vaccine)
DPX-Survivac Pembrolizumab + NCT03029403 Ovarian, Peritoneal, 2
(peptide vaccine) Cyclophosphamide or fallopian tube
cancer
CDX-1401 Atezolizumab + NCT03206047 Ovarian cancer 2
(protein vaccine) Guadecitabine
EGFRvIII, EphA2, Bevacizumab NCT02754362 Glioblastoma 2
Her2/neu peptide
(peptide vaccine)
ISA101b Utomilumab NCT03258008 HPV16+ 2
(HPV16 E6/E7 peptide vaccine) (4–1BB agonist) oropharyn
xcancer
Intuva Sunitinib NCT02432846 RCC 2
x(allogenic cell-based therapy)
GVAX (GM-CSF-secreting tumor cells) Nivolumab+ NCT02451982 Pancreatic cancer 1/2
Cyclophosphamide
GVAX Nivolumab + ipilimumab+ NCT03190265 Pancreatic cancer 2
CRS-207 +
Cyclophosphamide
GVAX Neoadjuvant NCT03161379 Pancreatic cancer 2
cyclophosphamide +
SBRT + Nivolumab
GVAX Adjuvant NCT02648282 Pancreatic cancer 2
Cyclophosphamide +
SBRT + Pembrolizumab
GVAX Epaccadostat + NCT03006302 Pancreatic cancer 2
Pembrolizumab +
CRS-207 ± GVAX
Dendritic cell-based p53 vaccine Nivolumab + NCT03406715 Small cell lung cancer 2
ipilimumab
PROSTVAC (poxviral vector expressing Nivolumab NCT02933255 Prostate cancer 1/2
PSA)
PROSTVAC Ipilimumab NCT02506114 Prostate cancer 2
MVA-HPV16E6/E7-IL2 vaccine TG4001 Avelumab NCT03260023 HPV16+ 1/2
(vaccinia virus vector) oropharyn
xcancer
MEDI0457 Durvalumab NCT03162224 p16 or 1/2
(DNA vaccine) HPV16/18+
HNSCC
HPV E6/E7 GX-188E pembrolizumab NCT03444376 HPV16/18 1/2
(DNA vaccine) cervical cancer

pTVG-HP Pembrolizumab NCT02499835 Prostate cancer 1/2


(DNA Vaccine)
EBRT, external beam radiation therapy; SBRT, stereotactic body radiation therapy; RCC, renal cell cancer; HNSCC, head and neck squamous cell carcinoma.

barrier like aberrant vasculature and dense stroma), (iii) immunosup- radiation, or chemotherapy. This would reverse this immunosup-
pressed (few T cell infiltrate inside tumor, suggesting the absence pressive TME by inducing immunogenic cell death (ICD) to expand
of physical barrier but presence of immunosuppressive factors), and the Ag repertoire through epitope spreading and increase the pro-
(iv) cold (no T cell inside or at margin, low mutation burden, absence inflammatory TME. Radiation and specific chemotherapy induced cell
of PD-1, PD-L1, and poor expression of Ag presentation machinery death can release DNA or damage- associated molecular patterns
like MHCI).64-78 (DAMPs) from dying cells that can trigger the STING mediated Type
Understandably, the “cold” tumors with low immunoscore present I IFNs pathway and increase intratumoral infiltration of DC and
most challenges to effective immunotherapy. In order to overcome the cytotoxic T cells. Several preclinical studies across multiple tumor
absence of pre-existing immune responses in these tumors, patients types have highlighted that multifactorial approach of combining ICB
would likely require a priming strategy, for example, vaccine therapy, with vaccination therapies and/or radiation and chemotherapy can
14 ROY ET AL .

TA B L E 3 Ongoing trials evaluating personalized vaccines

Vaccine Name and Type Trial Combination NCT Number Disease TrialPhase
Personalized Temozolomide+ RT+ NCT02287428 Glioblastoma 1
Neoantigen Peptide pembrolizumab
vaccine (NeoVax)
NeoVax Ipilimumab NCT02950766 Clear cell RCC 1
Personalized peptide-based vaccine CD40 agonist antibody NCT03597282 Melanoma 1
(NEO-PV-01) (APX005M) or ipilimumab
with nivolumab
NEO-PV-01 Pembrolizumab NCT03380871 NSCLC 1
NEO-PV-01 Nivolumab NCT02897765 Melanoma, lung cancer or 1
bladder cancer
NEO-PV-01 TLR-3 agonist poly-ICLC NCT01970358 Melanoma 1
Personalized Peptide Vaccine Pembrolizumab NCT02600949 Pancreatic carcinoma or 1
colorectal cancer
Personalized peptide-based Bevacizumab NCT01814813 Glioblastoma 2
Vaccine
(HSPPC-96)
Neoantigen DNA vaccine With or without durvalumab NCT03199040 TNBC 1
Personalized RNA mutanome vaccine Atezolizumab NCT03289962 Melanoma, NSCLC, TNBC, 1
(RO7198457) HNSCC, colorectal cancer,
and RCC
Personalized RNA mutanome vaccine Pembrolizumab NCT03815058 Melanoma 2
(RO7198457)
RT, radiation therapy; RCC, renal cell cancer; NSCLC, non-small cell lung cancer; TNBC, triple-negative breast cancer, HNSCC, head and neck squamous cell
carcinoma; HSPPC-96, heat shock protein peptide complex-96.

reinforce the therapeutic margin of one another and are currently However, one must appreciate that the above discussed type of
being evaluated in clinical trials.13,16,17,79–85 Another attractive thera- tumor stratification into “hot” or “cold” phenotypes is clearly an over-
peutic approach would be to combine vaccines with adoptive chimeric simplification of the complex events occurring in the TME. It is possible
Ag receptor (CAR) T cell therapy. Although its success is mainly limited that even a tumor that appears “hot” may actually have an extremely
to hematological malignancies, a recent work by Ma et al. showed that immunosuppressed microenvironment due to co-existence of multi-
combination of vaccine and CAR-T cells eradicated 60% of solid tumors ple parallel oncogenic mechanisms and may thus not respond to the
in mice through robust activation of the latter in lymph nodes.86 approaches tailored for “hot” tumors. Also, tumors evolve their resis-
Another recent study reported robust CAR T cell activation in murine tance mechanisms over time, adding an extra level of complexity.
tumors by combination with a nanoparticulate RNA vaccine,87 which Hence, exploitation of a combined evaluation of the spatiotemporal
opens the intriguing prospective of exploiting a combined approach of interplay of immune cell types (immune contexture and immunoscore)
therapeutic cancer vaccines and CAR T therapy against solid tumors in and other parameters (morphology, chromosomal instability, tumor
the future. These strategies along with supporting preclinical data have cell of origin, oncogenic driver genes signature, etc.) can help improve
been exhaustively discussed elsewhere and is beyond the scope of prediction of clinical outcomes and stratify patients for successful ther-
this review.63,88,89 apeutic intervention.
Based on the immunome, the altered “excluded” or “immunosup-
pressed” tumors that present T cells at the borders or poor infiltration
could benefit from targeted therapies to increase chemokines 3 CANCER VACCINE PLATFORMS
or cytokines or soluble factor inhibitors to either enhance T cell
mobilization and trafficking inside the tumor or deplete MDSC With this 30,000-feet aerial view of the clinical landscape of cancer
and/or Tregs. Another important factor that contributes to poor vaccines from its infancy to latest generation of neoantigen-based
T cell infiltration is the presence of a “physical barrier” by either platforms, we will survey some of the major classes of vaccines,
aberrant vasculature or dense stroma, as in pancreatic cancer and particularly those that have formidably crossed over the chiasmic
could be targeted by either anti-angiogenic therapies or by agents divide into the clinical phase. Vaccine platforms are vast, and there
disrupting the dense matrix barrier. A multitude of these agents are many excellent reviews that highlight their basic components with
(anti-angiogenic, stromal targeting agents) are either in preclinical their advantages or limitations27,91,92 (Fig. 2). In the following sections,
stage or are being evaluated as monotherapy or as part of a multi- we will highlight the recent developments, challenges, and future
pronged treatment approach. These have been discussed in more prospect for some of the major classes of vaccines. The companion
detail elsewhere.88,90 review will focus on novel developments on formulation.
ROY ET AL . 15

F I G U R E 2 (A) Next generation cancer vaccine platforms. The essential components of an effective therapeutic cancer vaccine are illustrated
here: (i) source of tumor Ag: this can be either tumor associated or neoantigens. Ags can be derived from either whole cell lysate, peptides, over-
expressed protein, tissue differentiation, cancer testis, oncofetal proteins, and/or DNA or RNA. (ii) Formulation: vaccines can be cell based (whole
cell or loaded into DC), protein or peptide based, nucleic acid based, or microbial organism based. (iii) Adjuvants and/or cytokines used: different
types of adjuvants and/or cytokines like TLR ligands, STING agonists, Type I IFNs, IL-12, GM-CSF are used to improve vaccine efficacy and adap-
tive immunity activation. (iv) Delivery vehicles: vaccines are bioengineered and formulated into emulsions, liposomes, virosomes, or nanodiscs and
other biological scaffolds to improve bioavailability and targeting to the desired site. (B) Mechanism of T cell priming and cancer cell killing. Depend-
ing on the vaccine platform and Ag source used, the APCs are primed and/or activated. This is followed by specific binding of a TCR on T cells in the
tumor draining lymph nodes to its cognate peptide-major histocompatibility (MHC) complex displayed on an APC, like, M𝜙 or dendritic cell. This,
in combination with co-stimulatory signals and inflammatory cytokine release from activated APC can successfully transform resting naïve T cells
(CD4+ , CD8+ ) to activated effector cancer cell killing T cells and result in tumor regression. Co-stimulation may be improved by using adjuvants
that activate the pattern recognition receptors on APC and upregulate the co-stimulatory ligands, as well as by agonist antibodies that can engage
with the co-stimulatory molecules on the T cells and trigger an activation signaling cascade. Within the tumor, T cells up-regulate the expression
of checkpoint inhibitory molecules including CTLA-4 and PD-1 and this can dampen the vaccine efficacy. Combining vaccine therapy with immune
checkpoint blockade has therefore emerged as a potential treatment modality in wide variety of cancers, unfortunately with limited efficacy so far
16 ROY ET AL .

3.1 Cellular vaccines TGF-𝛽, IL-10, VEGF, Arg-1, IDO interfere with DC maturation and
arrest them in a dysfunctional state. Additional factors like expression
The cellular vaccine comes in two variants: whole tumor cell based and
of high level of coinhibitory molecules like TIM3, PDL-1, PD-1 on DCs,
autologous dendritic cell based, pulsed with either tumor cell derived
metabolic stress as well as Ag masking and faulty Ag presentation
lysates, proteins, peptides, patient specific neoantigens, viruses, or
facilitate the switch from inflammatory to tolerogenic phenotype.
nucleic acids.
Hence, over the past decade, much effort was invested to understand
the complex interplay and biology of DCs with other immune cells in
3.2 Whole cell vaccines the tumor microenvironment and modulate them for activation of T
cells against the tumor.
Whole tumor cell vaccines are essentially prepared from whole tumor
Since DC can activate Ag specific T cells, the goal of DC vac-
cells or tumor cell lysates generated by irradiation or repeated freeze
cine is to repair the immune suppression by administering “trained”
thaw cycles, derived from either autologous patient tumor tissues or
DCs and activate the adaptive immune responses in the patient. Typ-
heterologous tumor cell lines. These are often genetically modified to
ically, monocytes are isolated from patients via leukapheresis and
produce different cytokines/chemokines (GM-CSF) that can augment
then differentiated using GM-CSF and IL-4 in vitro to yield imma-
adjuvant function (recruit DCs and facilitate Ag uptake, cross priming
ture DC. These are then loaded with either (i) whole tumor prepa-
and presentation to CD8+ T cells). Irradiated tumor cells can release
ration, (ii) DC-tumor cell fusion, (iii) virus, (iv) defined, shared tumor
DAMP molecules such as HMGB1, pentraxin-3 (PTX3), heat shock pro-
associated Ags (peptides, nucleic acid), or (v) unique neoantigens from
teins (HSP 70/90), and uric acid that can stimulate DC maturation and
the tumor cells. The Ag-loaded DCs are then further matured using
T cell responses against the tumor.93 One advantage of using whole
a cytokine mixture that essentially makes them competent for pro-
cell vaccine is that the cells would serve as an unbiased source of a
cesses like lymph node homing, Ag cross presentation and activation
broad spectrum of tumor Ags, thereby eliminating the requirement of
of T cells as well as production of pro-inflammatory cytokines.128-130
identifying an immunogenic Ag or potential concerns of Ag loss.93-95
These ex vivo matured DCs are then infused back to the patient.120,131
As mentioned earlier, tumor cell vaccines are often genetically mod-
To date, DC vaccines have been applied to a variety of malignan-
ified to express different cytokines or chemokines or co-stimulatory
cies (over 240 completed clinical trials) including melanoma, prostate,
molecules to stimulate more robust immune response to the vac-
renal cell carcinoma, glioblastoma, and pancreatic cancer.117,123,129
cine. Of different cytokines tested, a superior response was observed
In the earliest trials, the first-generation DC vaccines from blood
with irradiated melanoma cells transduced with GM-CSF. This vac-
monocytes that were only loaded with synthetic TAA peptides or
cine (GVAX) showed promising results in early preclinical studies96-103
tumor cell lysates was found to be safe and immunogenic and were
that laid the groundwork for numerous clinical trials targeting a vari-
tested in a variety of cancers,132-142 but only 3.3% patients experi-
ety of cancers including prostate, melanoma, NSCLC, and pancreatic
enced tumor regression.123,128,129,135,143,144 In many studies, it was
cancer.104,105 However, in most of the clinical trials, it failed to live up to
reported that Treg-mediated immunosuppression hindered the effi-
its expectations.106-110 Despite these disappointing results, clinicians
cacy of DC vaccines.143 Subsequently, the DCs were matured using
continue to test the efficacy of GVAX in combination with different
a defined cocktail of cytokines and these second-generation vaccines
treatment strategies, especially ICB molecules.
performed better with an overall objective response rate (ORR) in the
There are other whole cell tumor vaccines: Oncovax (colon can-
range of 8–15%. There is also an interesting strategy to use DC and
cer; Vaccinogen),111-113 and Reniale (renal cancer; Liponova)114,115 to
tumor cell hybrid, and these improved immune response in patients
name a few. Of these, Oncovax, prepared by irradiating patient tumor
(NCT01096602).145,146
cells and administered with BCG bacteria as adjuvant, significantly
Despite some modest clinical efficacy for DC vaccines, the early tri-
reduced the chances of recurrence post-surgery in several early phase
als raised some questions and challenges. One concern is that the spe-
trials in colon cancer patients.111-113 Based on these promising results,
cific details of DC vaccine manufacturing vary considerably and cover
a randomized, multicenter Phase IIIb clinical trial (NCT02448173)
various aspects like the source and starting cell population, culture
is ongoing to test if Oncovax improves disease free survival post-
condition, maturation cocktail used, choice of Ag and Ag loading tech-
surgery. Unfortunately, the main disadvantage of an autologous whole
nique, as well as time and route of administration. The DC subtypes are
cell vaccine approach is its inherent logistic hurdle—it is time con-
transcriptionally and functionally distinct in their developmental ori-
suming, expensive, and variable.96,100,116 Alternatively, allogeneic cells
gin, and localization.147,148 A recent single cell RNA sequencing study
derived from different tumors could be used with the likelihood that
identified 6 different classes of circulating DC in peripheral blood.149
the patient’s tumor will share Ags with that of the vaccine.
Some researchers argue in favor of using peripheral blood derived DC
to better retain their physiological functionality following transfusion
3.3 DC vaccines to patients. Indeed, studies report transcriptional and phenotypic
Dendritic cells (DC) are the most potent APCs, and depending on differences in monocyte derived (moDC) and conventional DCs (cDC)
the context, DC also provide either co-stimulatory or co-inhibitory that translate to poor migratory capacity of moDC to lymph nodes
signals to T cells for activation or suppression.117-127 In the tumor and T cell priming.150,151 Heterogeneity of human DC population is
microenvironment, immunosuppressive molecules and cytokines like further highlighted by studies showing potent Ag presentation by
ROY ET AL . 17

moDC152,153 whereas some studies indicated potent Ag presentation significantly affect the DC vaccine efficacy are the choice and method
by BDCA1/CD1c+ moDCs and BDCA3/CD141+ moDCs.123,154 The of Ag loading. While a broad range of Ags have been explored includ-
use of cDC harvested from circulation is limited owing to their poor ing defined peptides, whole tumor cell lysate, nucleic acids, irradiated
availability in the peripheral blood of cancer patients and the potential tumor cells, neoantigenic peptides, and viruses, how to generate the
need for repeated dosing in a therapeutic setting as well as their best immunogenic Ag and its loading technique to ensure maximum
compromised capacity to present Ags and secrete pro-inflammatory clinical response remains an open question. Oxidizing whole tumor
cytokines.155 Still in its infancy, cDC vaccines are being tested as pilot lysate with hypochlorous acid or infecting the tumor cell with New-
trials in the European study, “Professional cross-priming for ovarian Castle Disease virus significantly improved DC vaccination outcome
and prostate cancer” (PROCROP). A recent trial, however, used autol- in the preclinical setting.174-176 Combining chemotherapeutic drugs or
ogous naturally circulating DC loaded with HLA-A2.1 restricted tumor oncolytic viruses that induce immunogenic cell death of tumor cells is
Ag for stage IV melanoma patients (NCT01690377). Four out of 14 another way to improve DC vaccine outcome. Lastly, the cost of pro-
patients showed long-term progression-free survival that correlated duction and the complexity associated with its manufacture has hin-
with the development of cytolytic, multifunctional effector T cells.156 dered its wide-spread clinical application.
Scientists have also attempted to use plasmacytoid DC as vaccines.157 Despite these challenges, there are several novel and state of the art
Another important variable that dictates DC vaccine efficacy is the DC-based vaccines that are currently being tested in different phases
combination of cytokines used to mature the DC. Scientists have differ- of clinical trials. For example, a placebo-controlled phase III trial is test-
entiated DC from CD34+ stem cells or cord blood by adding molecules ing an autologous DC vaccine loaded with tumor lysate (DCVax-L) in
like FLT3L, Stem Cell Factor (SCF), GM-CSF, Thrombopoietin, IL-7, glioblastoma patients in combination with radiation and chemotherapy
Notch ligand Delta Like-1 (DL1), and IL-4, at the same time attempt- (NCT00045968). The initial reports show DC vaccination improved
ing to recapitulate physiological DC phenotype ex vivo by culturing the overall survival to 23 months compared to 15–17 months in case of
cells for different time periods (1 or 3 week).151,158–162 Unfortunately, patients who received standard of care therapy in earlier studies.177
there is no consensus for optimal DC maturation protocols and the Other phase III studies include investigating the efficacy of autologous
maturation cocktail used. Earlier, the gold standard was to use a cock- RNA loaded DC vaccine as an adjuvant to standard of care in uveal
tail containing TNF-𝛼, IL-1𝛽, and IL-6 in combination with PGE2.163 melanoma (NCT01983748) and that of BDCA3+ naturally circulating
However, PGE2 has been shown to induce Tregs, increase the expres- DC in stage 3 melanoma patients (NCT02993315). The patients will be
sion of IDO and reduce the secretion of IL-12.164,165 Scientists and clin- assessed for 2-year recurrence free survival. Other trials are testing
icians have explored alternative maturation cocktails. These include the efficacy of DC vaccines in combination with radiation, chemother-
triggering of co-stimulatory pathways (e.g., CD40-CD40L), activation apy, targeted therapies, depletion of myeloid cells, and Tregs. In
of TLR by using agonists like poly IC (TLR3), resiquimod (TLR7/8) addition, the emergence of neoantigens has initiated the development
pathway, and 3-O-deacylated monophosphoryl lipid A (MPLA) either and use of personalized neoantigen DC vaccines.178 A recent meta-
as single agents or in combination.166-168 For instance, DC trans- analysis reported that the objective response rate to DC vaccines in
fected on the TriMix platform with mRNA encoding constitutively melanoma and glioblastoma was 8.5% and 15.6%, respectively.129
active TLR4, CD40L, and CD70 has been clinically tested in melanoma Hence, strategies to incorporate neoantigens into DC vaccines are
patients.21,169 However, combination of different agonists in varying already ongoing179 (NCT03300843). Lastly, in the light of current
doses and varying maturation time may potentially alter the expres- understanding, like many other vaccine platforms, DC vaccines have
sion of inflammatory genes and chemokines that will translate to the been combined with ICB,20 including in combination with different PD-
efficacy of the DC vaccine in vivo.170 Recently, a novel method of trans- 1 or CTLA-4 inhibitors in several cancers including glioblastoma (and
fecting DC with a mRNA cocktail encoding co-stimulatory molecules other brain tumors), colorectal cancer, and melanoma. DC vaccines can
(CD40L, CD70, and a constitutively active TLR4) has shown promising also be combined with other ICB like Abs targeting Tim-3, Lag-3, and
result in melanoma patients (NCT00074230, NCT01066390).21,171 NKG2A that are currently in different stages of clinical development.
Another possible reason for DC vaccines to have shown limited effi-
cacy is that many of the clinical trials were done with patients having
late stage disease where dominant immunosuppressive mechanisms
3.4 Peptide vaccines in cancer
can dampen responses to DC vaccines and T cell activation. This is Peptide-based vaccines that initially targeted tumor enriched
reflected in earlier studies that noted T cell activation in higher fraction Ags,180,181 can be classified into two distinct categories:
of patients receiving adjuvant DC vaccines compared to those with tumor-associated Ags (TAA) and neoantigens (tumor specific Ags,
metastatic disease. This indicates that DC vaccines may perform better TSA). TAAs are enriched on cancerous cells but also expressed
in adjuvant settings.145,146,172 Another potential for improvement is on some normal cells and are further subcategorized to (a) aber-
DC mobilization. Intradermal injection allows ≤5% of DCs to migrate to rantly overexpressed Ags (e.g., Her2/neu182 and survivin183 ), (b)
lymph nodes whereas intravenous route distributes DC to non-specific lineage-restricted Ags (e.g., gp100184 , prostate-specific Ag,185 and
locations like spleen, lungs, liver, and bone marrow. Attempts have tyrosinase186 ), (c) cancer-testis (CT) Ags (e.g., MAGE family187 and
been made to manipulate DC by viral vectors to overexpress molecules NY-ESO-1188 ), (d) oncofetal Ags (e.g., Carcinoembryonic Ag189-191 ),
to improve their trafficking to the tumor in vivo.173 Other factors that and (e) oncogenic viral Ags (HPV E6/E7). Neoantigens, on the other
18 ROY ET AL .

hand, are developed from somatic mutations (e.g., insertions, deletions, elsewhere.217,220 While there are examples of naked RNA vaccine
translocations)192 in the tumor triggered by DNA-damage events, that codes for carcinoembryonic Ag (CEA) exhibiting strong immune
resulting in specific expression in the tumor. responses when challenged with CEA overexpressing cancer cells,221
Early clinical trials utilized TAA-based vaccines alone, such as stability of RNA has been an issue. Some of this work has been adopted
the MAGEA3 vaccine,193 to treat established cancers and gener- by the bioengineers as a delivery problem and various forms of alter-
ally resulted in good safety profiles but lacked objective clinical native vaccine delivery method (“gene gun,” protamine condensation,
responses.194 Various forms of adjuvants (Montanide, TLR agonists, encapsulation) have been used.222-225 An example of this is CV9103
incomplete Freund’s adjuvant, etc.) are an essential component comprising of mRNA encoding four prostate cancer associated Ags
of these vaccines,195-197 and some early studies showed modest that was condensed with protamine for castration resistant prostate
signs of clinical benefits for recurrence and disease-free survival cancer. The vaccine was well tolerated and showed Ag specific T cell
compared to non-vaccinated patients.198 However, even with responses in 36 patients with metastatic prostate cancer, and the
the added anticancer benefits from immunostimulants or deliv- vaccine showed a median overall survival of 30 months compared to
ery vehicles, no peptide-based vaccine has been FDA approved predicted 16.5 months (EudraCT number: 2011-006314-14).181,226
to treat cancer. One biological reason may be the low immuno- A similar approach for lung cancer patients is ongoing.227-229 To
genicity of the TAAs, which is why it may be more beneficial to improve RNA half-life and delivery in vivo, naked RNA IVT is often
target neoantigens for peptide vaccines. An illustrative exam- encapsulated in nanoparticles and cationic lipid complexes for deliv-
ple of this is the Kras-based neoantigen vaccine that showed no ery for patients230 (NCT03313778). Details about similar studies
clinical benefit.199-201 can be found on the ClinicalTrials.gov website (NCT03289962,
To address immunosuppression from advanced tumor burden, NCT03815058).
combination strategies have been adopted to increase immunogenic The RNA and DC platform can be combined, whereby DCs can be
cancer cell death, for example, by using approved chemotherapeutic pulsed with either total tumor RNA, TAA RNA, or neoantigen RNA.
agents like gemcitabine, cyclophosphamide, fluorouracil (5-FU), or There are currently over 24 ongoing clinical trials investigating RNA
folfirinox among others. These treatments were also reported to sup- loaded DC vaccines. The advantage (or the disadvantage) with using
press the tumor promoting functions of Tregs and MDSCs.202-211 For whole tumor RNA for DC priming is it presents the entire constellation
instance, gemcitabine was shown to specifically reduce the number of of Ags without the requirement of identification of immunogenic
Gr1+ MDSC in spleens of tumor bearing mice. This decrease in splenic epitopes. Most of these early studies have shown little toxicity and
MDSCs was accompanied by an increase in the antitumor activity of potential to elicit antitumor immune response.231-235 In a more
CD8+ T cells and NK cells.202 Similarly, 5-FU significantly contracted recent Phase II trial with Rocapuldencel-T or AGS-003, moDC was
the number of MDSC in spleen and tumor bed and concomitantly co-electroporated with whole tumor RNA plus RNA encoding a cos-
increased IFN-𝛾 production by CD8+ T cells and promoted T cell timulatory gene CD40L and administered to treatment naïve patients
dependent antitumor activity.206 The effect of chemotherapy on Tregs with clear cell renal cell carcinoma and evaluated in combination with
is also appreciated. Chemotherapy can selectively reduce the number sunitinib. The combination was well tolerated and yielded supportive
of Tregs in the tumor bed, tumor draining lymph nodes as well as immunologic responses and significant increase in OS.236 Alterna-
peripheral blood.204,212,213 Cyclophosphamide may also reduce the tively, DCs can also be loaded with single or multiple TAA encoding
number Tregs by inducing apoptosis and inhibiting proliferation.204 IVT mRNAs and infused back into the patients. With this method, the
Based on these encouraging results, multiple trials have employed this Ag must be immunogenic enough to activate the T cells, and these TAA
combinatorial strategy but have failed to show any significant clinical pulsed DCs have been tested in multiple clinical trials.237-241 DCs have
effect. For example, a phase II clinical study for patients with resected also been stimulated with mRNA encoding melanoma Ags (MAGEA3,
pancreatic cancer determined that gemcitabine plus OCV-C02, a melanoma-associated Ag C2 (MAGEC2), tyrosinase and melanocyte
3-peptide cocktail, had no significant effect on DFS compared to lineage-specific Ag GP100, and other mRNAs encoding immune
gemcitabine alone.214 Another strategy explored is the combination of co-stimulatory genes like CD40 ligand (CD40L), a constitutively active
peptide vaccines and ICB because of ICB’s ability to intensify immuno- mutant form of TLR4 and CD70 (NCT01066390).21,169,242,243 Trimix
genic responses. This combination strategy is still in its infancy, and vaccine is currently being tested in a placebo controlled clinical trial in
these clinical trials are still ongoing. breast cancer patients (NCT03788083).
DNA based vaccine is the other flavor of nucleic acid vaccines, and
much like RNA, their clinical development is focused around delivery
3.5 Nucleic acid vaccines and combination with ICB. Biological basis of DNA based vaccines are
Nucleic acids have been well recognized as potent adjuvants,215,216 reviewed elsewhere.216,244 The commonly used techniques to deliver
and these also come in multiple flavors.217,218 Both RNA and DNA the vaccine are electroporation, sonoporation, gene gun, or DNA
have been used as adjuvants, but these also offer the ability to code tattooing. For gene gun mediated intradermal injection, Langerhans
for TAA. For RNA vaccine, RNA is transcribed in vitro (IVT) by a DNA cells, the APCs residing in skin directly engulf the plasmids and express
template encoding the Ag and bacteriophage RNA polymerase.219 the Ag, which is then presented by MHCI to prime naïve T cells in
Pertinent biological factors for these platforms have been discussed the lymphoid tissues. For intramuscular route, myocytes take up the
ROY ET AL . 19

plasmids. However, they are not potent inducers of immune activation. Currently, there are numerous OV that are being tested in clinical
Instead, they recruit DCs that phagocytose the infected cells and then trials for the treatment of different types of cancer.260 To date,
mount T cell response and memory. These techniques have been com- FDA has approved the HSV-1 based oncolytic virus, talimogene
prehensively reviewed elsewhere.245 Unfortunately, DNA vaccines laherparepvec, or T-VEC (ImlygicTM ) for the treatment of metastatic
as a monotherapy have often showed limited success in the clinic.244 melanoma that are surgically nonresectable. TVEC is composed of
Several clinical trials are testing the potential of DNA vaccines in genetically modified HSV-1 made to overexpress GM-CSF.20,261–263
combination with ICB in a variety of tumors (Clinicaltrials.gov). There are 28 trials that are active and testing TVEC in a variety of
cancers including pancreatic cancer, sarcoma, breast cancer, and
melanoma among others, either alone or in combination with radia-
tion, chemotherapy, or immune therapy. Another example of oncolytic
3.6 Microbial vector vaccines—oncolytic
virus therapy that has shown promising results in early phase trials is
and bacterial a genetically modified poliovirus: rhinovirus chimera, PVSRIPO. The
Oncolytic viruses (OV) have recently emerged as one of the most virus elicited effective antitumor immune responses in patients with
promising treatment options in cancer therapy. In brief, oncolytic recurrent glioblastoma.264,265 A phase II trial is currently under way
viruses infect and replicate within tumor cells causing cell death and testing the efficacy of PVSRIPO with or without the chemotherapy
release of Ags and viral debris.245,246 Phenotypic “hallmarks of can- drug Iomustine (Gleostine
R
) in glioblastoma patients. Based on the
cer” including the high metabolic activity within tumor cells and driver promising results, PVSRIPO was granted “breakthrough” status by
mutations have been shown to increase the replication of viruses inside the FDA. Other examples of OV are the human Orthoreovirus that
the cancer cells. The major advantage of oncolytic viruses is that they can cross the blood–brain barrier to activate cytotoxic T cell response
can specifically target and lyse neoplastic tissue while sparing healthy against brain tumors,266 and Maraba virus isolated from a variety of
cells, thus having limited systemic toxicity theoretically. Also, in con- sandfly in Brazil to treat triple negative breast cancer (TNBC).267
trast to drug pharmacokinetics that decreases with time, the viral dose With the expansion of OV in clinical trials, there is now an urgent
in tumor increases due to in situ replication. Viral receptors can be need for biomarker discovery, optimal dosing, and combinatorial
overexpressed by tumor cells, and recombinant viruses can be modi- pathways268,269 to identify patients that may benefit more from the
fied to target tumor-specific receptors and to overexpress cancer spe- virus therapy either as stand-alone drug or in combination with other
cific genes (e.g., apoptosis inducing or immune stimulating), to amplify immunomodulating agents. With the wealth of well characterized
the antitumor effects. OV can be either DNA or RNA virus and either microbial agents, several other viral vectors are currently being inves-
single stranded or double stranded. They establish lytic cycle in can- tigated in preclinical phases for translation.
cer cells by either replicating preferentially in cancer cells by using The other microbial vector is the bacterium, and other reviews
activated oncogenic pathways, or have been genetically modified to are available that discuss this platform in detail.270-275 Commonly
replicate selectively in malignant cells. Details of oncolytic viruses have used strains include Clostridium, Salmonella, and Listeria. A mod-
been exhaustively reviewed in depth elsewhere.246-251 ified Salmonella vaccine was tested for dose, safety, tolerability,
In brief, immune stimulatory properties of OV stem from their abil- and immune response in patients with refractory solid tumors
ity to induce immunogenic cell death like programmed necrosis or (NCT00006254, NCT00004216). Salmonella modified to express IL-
necroptosis252-256 to activate immune cells against the infected malig- 2 has been orally administered to patients with refractory hepatic
nant cells as well as prime de novo T cell response against tumor associ- metastases from solid tumors in a phase I study (NCT01099631).
ated as well as neoantigens that were not exposed earlier. OV-induced One ongoing phase I trial will test the safety and dosage of orally
lysis of cancer cells releases endogenous nuclear and cellular contents administered live Salmonella vaccine modified to produce survivin
including pathogen-associated molecular patterns (PAMPs), DAMPs, in patients with multiple myeloma and induce survivin Ag-specific
viral proteins, and nucleic acids, tumor associated Ags, immunogenic T cell response (NCT03762291). In a phase I clinical trial with 1
neoepitopes,248,257,258 and activate antitumor immune response cas- patient presenting retroperitoneal leiomyosarcoma, Clostridium vac-
cades. PAMPs, DAMPs, tumor Ags thus released can be phagocytosed cine showed extensive tumor necrosis and improved quality of life
by M𝜙s or DCs and fuel the release of inflammatory and immune acti- (NCT01924689).273 Clostridium vaccine is currently being tested in
vating cytokines. These then can activate immune cells against the combination with pembrolizumab in solid tumors (NCT03435952).
infected malignant cells as well as prime de novo T cell response against Lastly, Listeria strains modified to express the tumor Ag mesothelin
tumor associated as well as neoantigens that were not exposed ear- (CRS207) have also been tested in mesothelioma, lung, pancreatic, or
lier. Type I IFNs and DAMPs can also directly activate NK cells that will ovarian cancers in early phase trials. Both the vaccines were well toler-
now effectively kill cancer cells that have down-regulated their expres- ated in patients and induced tumor specific T and NK cell responses.276
sion of MHC I, a common occurrence in most cancers. OV can also CRS207 was next investigated in a phase II trial in combination with
be genetically modified to express anti angiogenic molecules to target GVAX and cyclophosphamide (NCT02004262) in previously treated
tumor angiogenesis.259 OVs have also been reported to produce absco- pancreatic metastatic patients. A phase I clinical trial is ongoing to
pal effect—the regression or delayed progression of metastatic sites test the safety, dose, and immune response of a live Listeria vac-
distant from the site of inoculation.259 cine (ADU623) expressing EGFRvIII and NY-ESO-1 to treat patients
20 ROY ET AL .

with astrocytic tumors (NCT01967758). Ongoing trials are focusing on “foreignness” may have been the reason for failure of many vaccine
combining Listeria vaccines with other conventional treatment options studies in the past. As the cancer evolves, it accumulates mutations
or immunotherapies (Clinicaltrials.gov). For example, Listeria vaccine is that alter the amino acid sequences in proteins. The most common
currently being tested in combination with anti-PD-1 (NCT03847519) types of mutations are single nucleotide variation (SNV), insertion,
or as an adjuvant after chemotherapy (NCT01675765) in non-small deletion, and fusion or duplications. These mutations can either result
cell lung carcinoma and malignant mesothelioma, respectively. Worth in single amino acid mutation or change the open reading frame
mentioning is another study by Aduro Biotech Inc. that is aimed to yielding mutated peptides/proteins called neoepitopes. These can vary
test the efficacy of a personalized live attenuated Listeria vaccine in within different regions of the same tumor and are truly tumor specific
metastatic colorectal cancer patients (NCT03189030). Ags. There are reports of shared neoantigens, but most are not shared
across tumor types and are specific for the patient.289 Personalized
vaccines targeting the unique set of neoantigens in an individual’s
3.7 iPSC-based vaccine
tumor have been developed; the challenges facing this platform is
The concept of embryonic tissue being used for vaccination dated primarily that of immunogenicity and heterogeneity. Early examples in
back to 1906, when Georg Schöne reported that mice immunized with murine Lewis Lung carcinoma models in 1994 of mutated connexin 37
fetal tissue rejected transplanted tumors.277,278 Published reports showed the biological basis of these neoantigen based platforms, but
now indicate that embryonic cells and tumor cells share transcriptome their translational potential was spurred by advancements in the fields
profile and Ag repertoire.279,280 Induced pluripotent cell-based vac- of next generation sequencing and bioinformatics.290-295 The typical
cines circumvent the ethical roadblock and are the newest addition in workflow for the development of personalized neoantigen vaccine
the family of cancer vaccine. Reports now show that iPSCs share nearly involves whole exome and RNA sequencing of tumor and matched nor-
identical gene expression profiles with that of embryonic stem cells mal tissue, followed by bioinformatics platforms to analyze the clonal
and cancer cells and have immunogenic properties as well.281-287 The ancestry for determining the neoantigen clonality in the tumor and
concept has recently been tested by Kooreman et al. In their elegant intratumor heterogeneity. Potential vaccine neoepitopes are next pre-
study, the authors have developed an autologous vaccine from irradi- dicted based on their binding affinity to the HLA subtype determined
ated mouse pluripotent stem cells (to arrest their proliferation in vivo in the patient. NetMHC and IEDB consensus are 2 such platforms.
and prevent the formation of teratoma). Used prophylactically, the vac- Finally, the validated epitopes are selected for vaccine formulation
cine was effective in preventing tumor formation in syngeneic models and administered to the patients. In some settings, immunogenicity
of breast cancer, mesothelioma, and melanoma and showed clear indi- can be determined with ELISPOT using synthetic peptides. In one such
cations of humoral and cellular immune responses. The vaccine culmi- study, 3 melanoma patients were treated with autologous DCs that
nated in mature APCs in the draining lymph nodes (dLN) and increase were loaded with 7 HLA-A2 restricted synthetic 7-mer peptides that
in the local and systemic helper and cytotoxic T cells. In an adjuvant represented the individual antigenic mutations in the patients.179
setting, it reduced metastatic tumor burden and potentially altered In addition to the neoepitope vaccine, they also received melanoma
immune responses characterized by fewer Th17 cells and increased gp100-derived peptides G209-2M and G280-9V (as positive controls
infiltration of Gr1+ CD11b+ myeloid cells into the tumors. Interest- for vaccination). The treatment was well tolerated in the patients who
ingly, when the authors adoptively transferred T cells from vaccinated developed neoantigen specific T cell responses.
tumor bearing mice, it elicited an Ag-specific antitumor response in To date, multiple combinatorial approaches have been exploited
unvaccinated recipients.288 to deliver different neoantigen-based vaccines.294,296–304 Examples
One major advantage of autologous iPSC vaccine is that it circum- range from direct injection of unformulated Ags to DC pulsed with
vents the challenge of patient specific tumor Ag selection. Also, it will immunogenic neoantigens179 or bioengineered neoantigen delivery
be readily available from the patient skin or blood. However, there are a formulations.305-307 It is noteworthy that based on the recent devel-
few safety concerns that need to be noted. iPSCs are immature progen- opments, multiple personalized vaccines trials are now ongoing or in
itor cells that do not have growth restraint and therefore may give rise the pipeline, awaiting their evaluation in clinical settings. RNA based
to teratomas once injected. Genetic modifications with suicide gene ex personalized vaccine RO7198457 (Genentech) is currently in a Phase
vivo are potentially feasible options in the near future. Another issue Ia/Ib dose escalation study either as a single agent or in combina-
is the labor-intensive process of reprogramming iPSCs ex vivo that tion with Atezolizumab (MPDL3280A, an engineered anti-PD-L1 Ab).
introduces concerns for cost and time required to prepare the vaccine. Neovax (formulated with poly-ICLC) will be combined with Ipilimumab
Although iPSC-based vaccines showed encouraging results in preclini- in a cohort of 18–20 patients diagnosed with stage III/IV clear cell
cal settings, its translation in patients is pending. renal cell carcinoma (ccRCC; NCT02950766). NEO-PV-01, a person-
alized cancer vaccine containing up to 20 neoantigens individually
selected for each patient diagnosed with advanced melanoma, blad-
3.8 Personalized cancer vaccine der cancer, or non-small cell lung cancer (NSCLC) is also ongoing
As mentioned earlier, TAAs are often expressed in low level in normal (NCT01970358). A second Phase II study is currently ongoing to eval-
tissue and hinder immune responses to vaccine therapy through cen- uate the efficacy, safety, pharmacokinetics, and patient-reported out-
tral and peripheral tolerance or autoimmunity. Thus, the lack of enough comes (PROs) of RO7198457 plus Pembrolizumab (PD-1 blocking
ROY ET AL . 21

agent) compared with Pembrolizumab alone in patients with previ- nition and elimination of cancer cells.314 These tumor intrinsic resis-
ously untreated advanced melanoma. tance mechanisms have been extensively discussed elsewhere.315
The design and development of neoantigen based personalized
cancer vaccines involve the rapid and accurate identification of
patient’s mutanome and thus heavily rely on the robustness of in silico
4.2 T cell exclusion from solid tumors
modeling and derivation of neoantigens- working with patient sample
to sequencing and bioinformatics analysis. This is especially imperative Excluding T cells from the TME (concept of immunologic ignorance)
for tumors with lower mutation load. There are still significant gaps in is a common mechanism of immune evasion by tumors. Leaky tumor
HLA presentation prediction based solely on Kd versus actual immuno- vasculature and hypoxic regions resulting in spatial differences in
genicity that is not accounted for in the sequencing-based methods metabolites are important factors that influence T cell extravasation,
to generate these personalized vaccines. Current bioinformatics algo- survival, and function in the TME.315 Also, the endothelium of tumor
rithms face significant challenges in accurately assessing MHC binding blood vessels often downregulate the expression of leucocyte adhe-
affinity and specificity partly due to limited knowledge available about sion molecules like intercellular adhesion molecule-1 (ICAM-1) and
several HLA alleles. Recent studies showed that CD8+ T cell response vascular cell adhesion molecule-1 (VCAM-1) or increase the level of
against predicted high affinity binders were low as 29%, warranting Endothelin B receptor in the tumor microvasculature that dampen
improvements of the algorithms currently used.31 Researchers are T cell entry. TAMs and MDSC also secrete chemokines that support
currently exploiting other methods including mass spectrometry to Tregs trafficking but prevent cytotoxic T cells. Tumor cells and TAMs
identify actual neoantigens presented on HLA molecules. For example, also express Fas ligand (FasL) to induce apoptosis in infiltrating T
mass spectrometric analysis of peptide-HLA complexes revealed cells.315 Such molecular mechanisms may explain robust desmoplastic
the HLA ligandome derived from human tumors.308-312 Collectively, reaction in pancreatic cancer that can trap T cells in the stroma to
this entire process of identification of patient tumor neoantigen, prevent their migration to the tumor cell rich areas.
validating their presentation on HLA molecules, and demonstrating
their immunogenicity may take up to several weeks or months, which
may not be a feasible option if the patient is diagnosed with large
4.3 Freeing immune cells from co-inhibitory
tumor burden or an advanced stage of cancer. Last but not the least,
the platform also introduces the concern for affordability by majority
checkpoints and exhaustion
of the patients. Immune checkpoint molecules are essential for maintaining immune
homeostasis and autoimmunity. They regulate the breadth and mag-
nitude of T cell response by balancing co-stimulatory and co-inhibitory
4 HURDLES FACING SUCCESS OF CANCER signals for T cell activation. However, cancer cells, as a mechanism to
VACCINE THERAPY escape immune killing often hijack these molecules to suppress T cell
activation. Further, due to chronic Ag stimulation, tumor infiltrating T
Challenges still remain as each platform has its own limitations as cells become exhausted, that is, attain a hyporesponsive state charac-
noted above. Additionally, several tumor intrinsic factors also pose a terized by progressive loss of effector functions.316,317 In this context,
significant hurdle to the success of any type of treatment employed. combinatorial ICB involving CTLA-4, PD-1 and its ligands PDL-1/PDL-
There are multiple reviews that delineate some of these tumor- 2, and LAG3 have become attractive targets in cancer immune therapy.
intrinsic problems (Refs below), and we would like to summarize some Immune checkpoint mechanisms have been extensively reviewed else-
of these salient features. where and is beyond the scope of this article.318-321 In brief, CTLA-4
competes with CD28 for binding with CD80 and CD86 and provides a
co-inhibitory signal to prevent T cell activation by APCs. PD-1 on T cells
4.1 Development of tumor cell intrinsic resistance
can bind to PDL-1 or PDL-2 expressed by cancer cells or TAMs and this
mechanisms
can arrest T cells in a non-activated state. For example, in a clinical trial
The heterogeneity of cancer cells in a tumor results in the develop- using TG4010 Muc-1 vaccine in lung cancer, patients with lower PDL-
ment of resistance mechanisms, and clinical persistence or recurrence 1 expression showed higher progression free survival.322 Abs target-
of disease to immunotherapy are the manifestation of this important ing immune checkpoints are now widely used in clinical trials as single
mechanism. For example, epigenetic modifications often trigger alter- agent or combination therapy for boosting immune response.323 The
nate signaling pathways in cancer cells.313 Also, genetic mutations in best clinical responses have been demonstrated in melanoma where
genes following therapeutic intervention (“immuno-editing”) can ren- PD-1 and CTLA4 blocking antibodies (response rates of 45–60%) are
der immune therapy ineffective. One study investigated the genetic now standard of care. Combining cancer vaccines with checkpoint
mutations in patients that were responsible for acquired resistance to blockade may yield superior immune responses in patients. This was
Pembrolizumab. Surprisingly, 2 patients were found to have mutations observed in a phase II clinical trial with DC vaccine and Ipilimumab.
in JAK1 or JAK2 genes that interfered with IFN-𝛾 signaling. A third Patients who received combination therapy showed better response
patient had a mutation in B2M gene that was important for the recog- compared to those who received monotherapy.21
22 ROY ET AL .

In addition to checkpoint blockade, several co-stimulatory agonists cytokines.356-361 Further, lactate produced by glycolysis is detrimental
like OX40 and 4-1BB have shown synergistic effects in preclinical to T cell proliferation and IFN-𝛾 release.362 Glucose restriction on the
studies324,325 and are now being tested in the clinics.323 other hand does not affect Tregs that depend on oxidative phospho-
rylation for their function.363 In addition to glucose, glutamine addic-
tion is common for many cancer types. For example, ovarian cancer
4.4 Immunosuppressive cells in the TME
cells metabolize glutamine to glutamate. This is taken up by CAFs that
The solid TME is a highly complex structure and is often heavily infil- convert it back to glutamine and thus fosters a vicious feed forward
trated by endothelial cells, cancer-associated fibroblasts (CAF), TAMs, loop with cancer cells. Unfortunately, glutamine is also important for T
tolerogenic DC, MDSCs, suppressive regulatory B cells, 𝛾𝛿 T cells, and cell proliferation. Hence, glutamine deprivation faced by T cells in TME
Tregs. They engage in a complex cross-talk to collectively establish affects their antitumor activity.315
an environment of immune suppression315,326–332 and aid in cancer An example of amino acid dependent mechanism of immunosup-
cell survival, growth, and metastases. For instance, TAMs and MDSC pression in the TME is IDO. This enzyme converts tryptophan to
secrete suppressive cytokines like IL-10, TGF-𝛽, and IL-4 to dampen kynurenines and is highly expressed on tumor cells, DC, and sup-
T cell function, induce T cell apoptosis and favor Tregs formation. pressive myeloid population. Tryptophan is important for T cell acti-
A similar suppressive activity has been reported for regulatory B vation, and its depletion restricts tumoricidal effector function of T
and 𝛾𝛿 T cells.333–338 Additionally, TAMs up-regulate expression of cells. Additionally, IDO can directly recruit myeloid cells and Tregs and
co-inhibitory molecules like PDL-1 to drive T cells to suppression. blunt NK cell function.315 High level of Arginase-1 in myeloid cells is
They are also important for pre metastatic niche formation. Each of another contributor to T cell dysfunction in TME. Although Arginase-1
these TME members are well known contributors to therapy failure. inhibitors are in clinical development, we are not aware of any clinical
Another emerging mechanism in the human TME is the immunosup- trial using Arginase-1 inhibitor in combination with vaccines to date.
pressive process of efferocytosis. It has recently been reported that
phagocytosis of apoptotic debris in the TME drives M𝜙s toward an
4.6 Hypoxic environment of the tumor
immunosuppressive phenotype.252,339,340 This is one of the factors
that contributes to the relapse of the disease in more aggressive form. As a tumor grows, large areas within can become hypoxic. The
In addition, often circulating MDSCs increase in patients undergoing tumor compensates by producing more angiogenic factors like vas-
therapy.341 A variety of strategies are being tested to target TAMs or cular endothelial growth factor (VEGF), but most often the neoan-
MDSCs in clinics and are discussed elsewhere.323 For example, STING giogenic and lymphangiogenic blood vessels fail to induce normoxia.
agonists can reprogram suppressive MDSCs to an activated inflamma- Also, in highly desmoplastic tumors like pancreatic adenocarcinoma,
tory type.342 In a clinical trial with NY-ESO1-ISCOMATRIX vaccine in blood vessels collapse and further contribute to hypoxic TME. The lack
melanoma, poor response to the vaccine correlated with higher Tregs of oxygen within the tumor has profound effect on several aspects
number in metastatic patients compared to early stage patients.343 of tumor growth, metastases, metabolism and associated immune
Ab-mediated Tregs depletion or by adding low-dose CpG-ODN to response. Hypoxia induces the expression of Hypoxia Inducible Factor
the ISCOM formulation restored efficacy of the vaccine in preclinical 1 𝛼 (HIF1𝛼), a key mediator of hypoxia signaling, which drives angiogen-
models.344 However, selective depletion of Tregs in patients is chal- esis, procurement of epithelial to mesenchymal (EMT) phenotype, gly-
lenging. Using cyclophosphamide has shown some success, achieved colysis in cancer cells and immune suppression. Hypoxia also induces
by metronomic scheduling.345 As said in one review, “there is no partic- the expression of targetable CD47 in tumor cells that relays “do not eat
ular hierarchy in immunosuppression in solid tumors and the dynamic me” signal to M𝜙s or DCs. It can also induce the expression of HLA-G,
nature and the potential redundancy of suppressive mechanisms”315 an important negative immune modulator that dampens the function-
continue to remain one of the biggest hurdles for vaccine therapy. ality of macrophages, NK cells, T cells, and B cells by binding to cognate
receptors expressed by these cells.
Hypoxia also affects immune cells within the tumor. For example,
4.5 Immunosuppressive metabolic barrier
hypoxia increases the expression of PDL-1 in M𝜙s, MDSCs, and DCs
Another important consideration is the metabolic demands of the to promote immune suppression of T cells. One of the hallmarks of
tumor immune cells and the limited metabolic milieu of the TME cancer is aerobic glycolysis where glucose is converted to lactate
that can affect their metabolic requirements and therefore, functional instead of being shuttled to oxidative phosphorylation. This way,
polarization.346-352 For an effective vaccine, naïve T cells need to the intermediates from glycolysis can be used for biosynthesis of
mature into multifunctional effector cells for which they heavily rely other macromolecules. HIF-1𝛼 up-regulates the expression of several
on aerobic glycolysis. However, inside the TME, factors like hypoxia, glucose transporters and glycolytic genes such as glucose transporters
areas of necrosis as well as rapid proliferation of tumor cells create 1 and 3 (GLUT1 and GLUT3), pyruvate dehydrogenase, lactate dehy-
nutrient constriction. In general, the metabolic landscape is hostile and drogenase A (LDHA), phosphoglycerate kinase 1, and hexokinase 1
does not support effector T cell activation. The glycolytic cancer cells (HK1). This restricts glucose availability to effector T cells that rely
outcompete the T cells for glucose consumption.353–355 This directly on glycolysis to mature to multifunctional T cells. Also, the lactate
reduce TCR signaling, proliferation, and production of tumor cell killing produced from glycolysis dampens the activity of T cells and NK
ROY ET AL . 23

cells. All this collectively pose a significant hurdle to rewiring vaccine tensin homolog) with disease progression and treatment.368 Similarly,
mediated anti-tumor immunity. Currently, a variety of metabolic analysis of non-small-cell lung cancers (NSCLCs) patients showed a
modulators are being studied or in clinical developmental stage. complete loss of Retinoblastoma (RB) gene in course of treatment
Combing them carefully with existing cancer vaccines may improve in cases where the disease transformed to small-cell lung cancer
patient responses.315 (SCLC) in a subset of patients, also acquiring elevated expression
of neuroendrocrine markers.369 An extensive genomic characteri-
zation carried out in glioblastoma patients identified differences in
4.7 Suboptimal Ag selection
mutations in responders versus non-responders to immune check-
Since the discovery of the first tumor Ag MAGE-1 in 1911, extensive point inhibitors.370 These tumor heterogeneity issues have been
efforts have been undertaken to design and develop efficient vaccines extensively discussed elsewhere371–379 and highlighted in several
to eradicate tumor. For reliable activation of immune responses, selec- recent research articles366,380–383 and are beyond the scope of this
tion of appropriate immunogenic Ag is essential. TAAs like EGFR or review. Unless carefully detected and characterized, these factors can
gp100 that are overexpressed on cancer cells and often shared across have detrimental effects by misdirecting treatment decisions. Collec-
multiple cancer types have been targeted for vaccine development. tively, these form the basis for precision medicine targeting specific
However, they are also expressed in low levels in non-cancerous tis- genetic alterations present in a tumor instead of a “one size fits all”
sues resulting in a weaker immune response due to central and periph- treatment strategy.
eral tolerance.91 Thus, more recent approaches have focused on Ags Therefore, one of the most important factors that affects the
that are mutated in individual tumors. These neoantigens or tumor- efficacy of vaccine or immune therapy is the selection of patient
specific Ags are identified through next generation sequencing and are population who are most likely to respond to a treatment modality.
unique to the tumor and patient. As discussed above, their expression For example, in a recent study, the peripheral blood TCR repertoire
levels can vary within different tumor clones in the same individual. was sequenced from pancreatic cancer patient cohorts: in one cohort
Also, among the identified neoepitopes, not all will be immunogenic patients received checkpoint blockade molecule Ipilimumab with or
enough to stimulate T cell responses. Therefore, the current person- without GVAX; in the other cohort, patients were given GVAX and
alized vaccines are designed to target multiple neoepitopes, usually up a bacterial cancer vaccine with or without Nivolumab. Higher TCR
to 20.91,364,365 clonality in the GVAX plus Ipilimumab arm correlated with long-term
survival.384 Cancers like melanoma, NSCLC, bladder cancer, gastric
cancer, and squamous cell carcinoma of the head and neck (SCCHN)
4.8 Selection of patients, vaccine platform, time,
that carry heavier mutational load and more inflamed TME compared
dose, and route of delivery to “cold” cancers like pancreatic adenocarcinoma or prostate cancer,
It is now appreciated that there co-exist distinct genetic and pheno- may be better targets for vaccine development.385,386 Another recent
typic populations within the same tumor, between tumors in the same study showed that patients with maximal heterozygosity of HLA class
patient and in tumors of a particular type in different patients. Even I locus (HLA-A, B, and C) showed greater survival following treatment
for a tumor of the same histological type, patient specific factors such with either anti PD-1 or anti CTLA-4.383
as germline genetic and somatic mutation profile (point mutations, Other factors include the type of vaccine administered for a type
single nucleotide variation, insertion, deletion, copy number variation, of cancer, the combination therapy employed, dose, and timing of
allelic loss, karyotype aberrations) either present before/at the time therapy. For many cancers, the standard of care is either radiation
of treatment or acquired at progression (temporal heterogeneity), or chemotherapy. Chemotherapeutic agents have profound effect on
epigenetic changes, and environmental factors contribute to the non-malignant cells as well. It is possible when the patient receives a
development of intratumor and inter-patient heterogeneity. Germline vaccine or any type of immune therapy after radiation or chemother-
genetic polymorphism analysis across multiple cancer types has been apy, the immune cells are already in a compromised state and too
shown to influence lymphoid and myeloid cell infiltration into the weak to mount a robust response. Similarly, when using vaccines and
tumor, which would affect clinical response since inflamed tumors are checkpoint blockade as combinatorial therapy, timing could play a very
more likely to respond to checkpoint therapies like antiPD-1 or anti important role. For example, CTLA-4 and PD-1 blockade had bet-
PDL-1.366 Another example comes from the loss of heterozygosity at ter efficacy in preclinical models when administered after GVAX or
the HLA (HLA-C*08:02) locus in the resistant clones during treatment TG4010 (Muc-1-targeted MVA vaccine) treatment, respectively,18,387
with T cell clones engineered to recognize KRASG12D . Since this whereas a PSA-targeted DNA vaccine worked better when used con-
allele was necessary to present KRASG12D neoantigenic epitope, currently with PD-1 blockade.388
its loss enabled immune escape.367 In a recent study, rapid autopsy Another concern is selection of route of delivery of the drug. Follow-
performed on breast cancer patients with metastases harboring ing parenteral administration, vaccines quickly spread in circulation
activating PIK3CA mutations (phosphatidylinositol-4,5-bisphosphate and very small amounts home to lymph nodes for T cell activation.
3-kinase, catalytic subunit alpha, PI(3)K𝛼) revealed intertumoral Hence, researchers are now focusing their efforts to develop other
variations where some patients lost the original PIK3CA mutation, effective methods of vaccine delivery, like nanoparticles, bioscaffolds,
whereas some developed bi-allelic loss of PTEN (phosphatase and and so on.
24 ROY ET AL .

F I G U R E 3 Integrating next generation cancer vaccine platforms with combinatorial standard of care therapies, biomarker-based patient
selection and ICB to maximize clinical response and vaccine efficacy

4.9 Effect of microbiome have yielded unsatisfactory results in clinical setting. The past few
years have seen an enormous advancement in the field of immune
In recent times, there is a renewed interest in the >100 trillion
therapy, like immune checkpoint blockade and adoptive cell therapy,
microbes in each person. Emerging evidences suggest host micro-
both of which have shown clinical benefits in a fraction of patients.
biome can not only increase one’s susceptibility to developing a
However, many patients do not respond to immune therapies alone
particular type of cancer but also affect response to therapy by reg-
and often relapse with more aggressive form of the disease. Ideally, a
ulating the immune system. Gut microbes have been shown to affect
“hot,” “altered,” or “cold” tumor characterization based on parameters
therapeutic efficacy in preclinical models and patient cohorts.389
like tumor foreignness, immunome status, presence or absence of
For example, patients who respond to anti-PD-1 based therapy have
inhibitory metabolites, checkpoint expression, and soluble inhibitors
a higher diversity of gut bacteria and differences in composition
needs to be performed on the resected tumor (primary as well as
that correlated with prolonged progression-free survival.390,391 The
metastatic) to best direct the course of treatment. Unfortunately, the
mechanisms for these correlative findings are still early and their use
genetic and immunologic composition of the tumor may vary within a
as a clinical biomarker is yet to be validated.
tumor, between different tumors, and over time, even within the same
patient. Enormous efforts are being exhausted to develop alternative
strategies like liquid biopsies that capture circulating tumor cells and
5 CONCLUSIONS AND immuno-positron-emission tomography (PET) imaging to visualize
FUTURE PERSPECTIVE T cells in the tumor. The numerous hurdles in the TME indicate the
requirement for multi-pronged approach based on patient history
Cancer remains the second leading cause of death worldwide. The as well as cancer type, stage, and grade and most importantly, pre-
ability of the host immune system to recognize and attack trans- existing immune landscape. Lessons learnt from failed attempts are
formed cells, and their ability to generate durable clinical responses now driving researchers to look for more immunogenic TAAs as well
to immunotherapy has revolutionized oncology and cancer research. as tumor-specific neoantigens, improved vaccine platforms, optimum
However, heterogeneity of the tumor cells and tumor-associated dose, and route of delivery to patients. Although combination with
immune suppression produce substantial barriers. Fortunately, checkpoint blockade or CAR T cell therapy has shown promising
extensive research in the last decade has dramatically increased our results in a handful of clinical trials, we are still a long way from estab-
understanding of tumor-immune cell interactions. This has resulted lishing the perfect treatment regime and dose that will be optimum
in breakthroughs in cancer immune therapy treatment modalities. to elicit clinically relevant immune response. Finally, with the advent
Cancer therapeutic vaccines have emerged as an attractive treatment of tumor specific neoantigen personalized vaccines, cancer vaccine
strategy, mainly because of their ability to elicit long lasting memory therapy has come of age. With the advancement in the field of high
against cancer Ags. Successful cancer vaccines rely on careful identi- throughput sequencing and bioinformatics (like CIBERSORT, xCELL,
fication and selection of immunogenic Ags and appropriate delivery and TIMER that can deconvolute bulk sequencing data from tumor
vectors that can act in synergy and trigger effector and memory T cell and reveal the presence of enriched immune cell types in the TME),
responses against transformed cells. Unfortunately, most vaccines we can now identify the elusive immunogenic unique Ag epitopes with
ROY ET AL . 25

the potential to unleash the full power of the immune system against 11. Spranger S, Spaapen RM, Zha Y, Williams J, Meng Y, Ha TT, Gajew-
the tumor. Additionally, integrating multiple platforms including tumor ski TF. Up-regulation of PD-L1, IDO, and T(regs) in the melanoma
tumor microenvironment is driven by CD8(+) T cells. Sci Transl Med.
genomics, immunoscore assay, multiplex immunohistochemistry,
2013;5(200):200ra116.
immune gene signature analysis, single cell sequencing, or bar code 12. Badoual C, Hans S, Merillon N, et al. PD-1-expressing tumor-
sequencing could now help reveal multiple prognostic and predictive infiltrating T cells are a favorable prognostic biomarker in HPV-
biomarkers and direct the best possible combination treatment associated head and neck cancer. Cancer Res. 2013;73(1):128-138.
13. Duraiswamy J, Kaluza KM, Freeman GJ, Coukos G. Dual block-
modality for an individual patient.
ade of PD-1 and CTLA-4 combined with tumor vaccine effectively
In summary, armed with improved understanding and advanced restores T-cell rejection function in tumors. Cancer Res. 2013;73(12):
technological resources, the future looks promising for therapeutic 3591-603.
cancer vaccines. One cannot deny that there are still pending questions 14. Hurwitz AA, Foster BA, Kwon ED, et al. Combination immunotherapy
of primary prostate cancer in a transgenic mouse model using CTLA-
to be addressed. However, with combinatorial therapy of checkpoint
4 blockade. Cancer Res. 2000;60(9):2444-2448.
blockade or CAR T cell adoptive transfer and neoantigen personalized 15. Rice AE, Latchman YE, Balint JP, Lee JH, Gabitzsch ES, Jones FR. An
vaccine (Fig. 3), it is plausible that substantial progress may await in the HPV-E6/E7 immunotherapy plus PD-1 checkpoint inhibition results
near future. in tumor regression and reduction in PD-L1 expression. Cancer Gene
Ther. 2015;22(9):454-62.
16. Soares KC, Rucki AA, Wu AA, et al. PD-1/PD-L1 blockade together
DISCLOSURES with vaccine therapy facilitates effector T-cell infiltration into pan-
creatic tumors. J Immunother. 2015;38(1):1-11.
D.B.J. serves on advisory boards for Array Biopharma, BMS, Incyte, 17. van Elsas A, Hurwitz AA, Allison JP. Combination immunotherapy of
Merck, and Novartis, and receives research funding from BMS and B16 melanoma using anti-cytotoxic T lymphocyte-associated antigen
4 (CTLA-4) and granulocyte/macrophage colony-stimulating factor
Incyte, and travel support from Genentech. Y.J.K. serves on advisory
(GM-CSF)-producing vaccines induces rejection of subcutaneous and
boards for Dracen, Aduro, Novartis, Sanofi, and Takeda.
metastatic tumors accompanied by autoimmune depigmentation.
J Exp Med. 1999;190(3):355-66.
ORCID 18. Wada S, Jackson CM, Yoshimura K, et al. Sequencing CTLA-4 block-
ade with cell-based immunotherapy for prostate cancer. J Transl Med.
Douglas B. Johnson https://orcid.org/0000-0002-6390-773X 2013;11:89.
19. Le DT, Lutz E, Uram JN, et al. Evaluation of ipilimumab in com-
bination with allogeneic pancreatic tumor cells transfected with a
GM-CSF gene in previously treated pancreatic cancer. J Immunother.
REFERENCES 2013;36(7):382-389.
20. Ribas A, Comin-Anduix B, Chmielowski B, et al. Dendritic cell vacci-
1. Rosenberg SA, Yang JC, Restifo NP. Cancer immunotherapy: moving
nation combined with CTLA4 blockade in patients with metastatic
beyond current vaccines. Nat Med. 2004;10(9):909-915.
melanoma. Clin Cancer Res. 2009;15(19):6267-6276.
2. Guo C, Manjili MH, Subjeck JR, Sarkar D, Fisher PB, Wang XY. Ther-
21. Wilgenhof S, Corthals J, Heirman C, et al. Phase II study of autol-
apeutic cancer vaccines: past, present, and future. Adv Cancer Res.
ogous monocyte-derived mRNA electroporated dendritic cells
2013;119:421-475.
(TriMixDC-MEL) plus ipilimumab in patients with pretreated
3. Pan RY, Chung WH, Chu MT, Chen SJ, Chen HC, Zheng L, Hung SI.
advanced melanoma. J Clin Oncol. 2016;34(12):1330-1338.
Recent development and clinical application of cancer vaccine: tar-
22. Madan RA, Mohebtash M, Arlen PM, et al. Ipilimumab and a poxviral
geting neoantigens. J Immunol Res. 2018;2018:4325874.
vaccine targeting prostate-specific antigen in metastatic castration-
4. Melero I, Gaudernack G, Gerritsen W, et al. Therapeutic vac-
resistant prostate cancer: a phase 1 dose-escalation trial. Lancet
cines for cancer: an overview of clinical trials. Nat Rev Clin Oncol.
Oncol. 2012;13(5):501-518.
2014;11(9):509-524.
23. Massarelli E, William W, Johnson F, et al. Combining immune check-
5. Sylvester Richard J, van der Meijden APM, Lamm DL. Intravesical
point blockade and tumor-specific vaccine for patients with incur-
bacillus calmette-guerin reduces the risk of progression in patients
able human papillomavirus 16-related cancer: a phase 2 clinical trial.
with superficial bladder cancer: A meta-analysis of the published
JAMA Oncol. 2019;5(1):67-73.
results of randomized clinical trials. J Urol. 2002;168(5):1964-1970.
24. Chung V, Kos FJ, Hardwick N, et al. Evaluation of safety and efficacy
6. Kantoff PW, Higano CS, Shore ND, et al. Sipuleucel-T immunother-
of p53MVA vaccine combined with pembrolizumab in patients with
apy for castration-resistant prostate cancer. N Engl J Med.
advanced solid cancers. Clin Transl Oncol. 2019;21(3):363-372.
2010;363(5):411-422
25. Gibney GT, Kudchadkar RR, DeConti RC, et al. Safety, correlative
7. Andtbacka RHI, Collichio FA, Amatruda T, et al. OPTiM: A random-
markers, and clinical results of adjuvant nivolumab in combination
ized phase III trial of talimogene laherparepvec (T-VEC) versus sub-
with vaccine in resected high-risk metastatic melanoma. Clin Cancer
cutaneous (SC) granulocyte-macrophage colony-stimulating factor
Res. 2015;21(4):712-720.
(GM-CSF) for the treatment (tx) of unresected stage IIIB/C and IV
26. Weber JS, Kudchadkar RR, Yu B, et al. Safety, efficacy, and biomark-
melanoma. J Clin Oncol. 2013;31(18_suppl):LBA9008-LBA.
ers of nivolumab with vaccine in ipilimumab-refractory or -naive
8. DeMaria PJ, Bilusic M. Cancer vaccines. Hematol Oncol Clin North Am.
melanoma. J Clin Oncol. 2013;31(34):4311-4318.
2019;33(2):199-214.
27. Curran MA, Glisson BS. New hope for therapeutic cancer vac-
9. Mougel A, Terme M, Tanchot C. Therapeutic cancer vaccine and
cines in the era of immune checkpoint modulation. Annu Rev Med.
combinations with antiangiogenic therapies and immune checkpoint
2019;70:409-424.
blockade. Front Immunol. 2019;10:467.
28. Tureci O, Vormehr M, Diken M, Kreiter S, Huber C, Sahin U. Targeting
10. Fu J, Kanne DB, Leong M, et al. STING agonist formulated cancer
the heterogeneity of cancer with individualized neoepitope vaccines.
vaccines can cure established tumors resistant to PD-1 blockade. Sci
Clin Cancer Res. 2016;22(8):1885-1896.
Transl Med. 2015;7(283):283ra52.
26 ROY ET AL .

29. Yarchoan M, Johnson BA, Lutz ER 3rd, Laheru DA, Jaffee EM. Tar- 49. Hellmann MD, Rizvi NA, Goldman JW, et al. Nivolumab plus ipili-
geting neoantigens to augment antitumour immunity. Nat Rev Cancer. mumab as first-line treatment for advanced non-small-cell lung can-
2017;17(4):209-222. cer (CheckMate 012): results of an open-label, phase 1, multicohort
30. Hu Z, Ott PA, Wu CJ. Towards personalized, tumour-specific, thera- study. Lancet Oncol. 2017;18(1):31-41.
peutic vaccines for cancer. Nat Rev Immunol. 2018;18(3):168-182. 50. Hellmann MD, Ciuleanu TE, Pluzanski A, et al. Nivolumab plus ipili-
31. Sahin U, Derhovanessian E, Miller M, et al. Personalized RNA mumab in lung cancer with a high tumor mutational burden. N Engl J
mutanome vaccines mobilize poly-specific therapeutic immunity Med. 2018;378(22):2093-2104.
against cancer. Nature. 2017;547(7662):222-226. 51. Hellmann MD, Paz-Ares L. Lung cancer with a high tumor mutational
32. Ahmadzadeh M, Johnson LA, Heemskerk B, et al. Tumor antigen- burden. N Engl J Med. 2018;379(11):1093-1094.
specific CD8 T cells infiltrating the tumor express high levels of PD-1 52. Ready N, Hellmann MD, Awad MM, et al. First-line nivolumab plus
and are functionally impaired. Blood. 2009;114(8):1537-1544. ipilimumab in advanced non-small-cell lung cancer (CheckMate 568):
33. Appay V, Jandus C, Voelter V, et al. New generation vaccine induces outcomes by programmed death ligand 1 and tumor mutational bur-
effective melanoma-specific CD8+ T cells in the circulation but not in den as biomarkers. J Clin Oncol. 2019;37(12):992-1000.
the tumor site. J Immunol. 2006;177(3):1670-1678. 53. Antonia SJ, Lopez-Martin JA, Bendell J, et al. Nivolumab alone
34. Baitsch L, Baumgaertner P, Devevre E, et al. Exhaustion of tumor- and nivolumab plus ipilimumab in recurrent small-cell lung cancer
specific CD8(+) T cells in metastases from melanoma patients. J Clin (CheckMate 032): a multicentre, open-label, phase 1/2 trial. Lancet
Invest. 2011;121(6):2350-2360. Oncol. 2016;17(7):883-895.
35. Zippelius A, Batard P, Rubio-Godoy V, et al. Effector function of 54. Boudadi K, Suzman DL, Anagnostou V, et al. Ipilimumab plus
human tumor-specific CD8 T cells in melanoma lesions: a state of nivolumab and DNA-repair defects in AR-V7-expressing metastatic
local functional tolerance. Cancer Res. 2004;64(8):2865-2873. prostate cancer. Oncotarget. 2018;9(47):28561-28571.
36. Wolchok JD, Kluger H, Callahan MK, et al. Nivolumab plus ipilimumab 55. Janjigian YY, Bendell J, Calvo E, et al. CheckMate-032 study:
in advanced melanoma. N Engl J Med. 2013;369(2):122-133. efficacy and safety of nivolumab and nivolumab plus ipilimumab
37. Postow MA, Chesney J, Pavlick AC, et al. Nivolumab and ipili- in patients with metastatic esophagogastric cancer. J Clin Oncol.
mumab versus ipilimumab in untreated melanoma. N Engl J Med. 2018;36(28):2836-2844.
2015;372(21):2006-2017. 56. Carbone A, Gloghini A. Checkpoint blockade therapy resistance in
38. Larkin J, Chiarion-Sileni V, Gonzalez R, et al. Combined nivolumab Hodgkin’s lymphoma. Lancet. 2018;392(10154):1194-1196.
and ipilimumab or monotherapy in untreated melanoma. N Engl J Med. 57. Andre P, Denis C, Soulas C, et al. Anti-NKG2A mAb Is a checkpoint
2015;373(1):23-34. inhibitor that promotes anti-tumor immunity by unleashing both T
39. Larkin J, Hodi FS, Wolchok JD. Combined nivolumab and ipili- and NK cells. Cell. 2018;175(7):1731-1743 e13.
mumab or monotherapy in untreated melanoma. N Engl J Med. 58. Fayette J, Lefebvre G, Posner MR, et al. Results of a phase II study
2015;373(13):1270-1271. evaluating monalizumab in combination with cetuximab in previously
40. Wolchok JD, Chiarion-Sileni V, Gonzalez R, et al. Overall survival with treated recurrent or metastatic squamous cell carcinoma of the head
combined nivolumab and ipilimumab in advanced melanoma. N Engl J and neck (R/M SCCHN). Ann Oncol. 2018;29(Suppl 8):viii374.
Med. 2017;377(14):1345-1356. 59. Harding JJ, Patnaik A, Moreno V, et al. A phase Ia/Ib study of
41. Hodi FS, Chiarion-Sileni V, Gonzalez R,et al. Nivolumab plus ipil- an anti-TIM-3 antibody (LY3321367) monotherapy or in combina-
imumab or nivolumab alone versus ipilimumab alone in advanced tion with an anti-PD-L1 antibody (LY3300054): Interim safety, effi-
melanoma (CheckMate 067): 4-year outcomes of a multicentre, ran- cacy, and pharmacokinetic findings in advanced cancers. J Clin Oncol.
domised, phase 3 trial. Lancet Oncol. 2018;19(11):1480-1492. 2019;37(8_suppl):12.
42. Long GV, Atkinson V, Cebon JS, et al. Standard-dose pembrolizumab 60. Glisson B, Leidner R, Ferris RL, et al. Safety and clinical activity
in combination with reduced-dose ipilimumab for patients with of MEDI0562, a humanized OX40 agonist monoclonal antibody, in
advanced melanoma (KEYNOTE-029): an open-label, phase 1b trial. adult patients with advanced solid tumors. Ann Oncol. 2018;29(Suppl
Lancet Oncol. 2017;18(9):1202-1210. 8):viii410.
43. Tawbi HA, Chung C, Margolin K. Nivolumab and ipilimumab 61. Simon S, Vignard V, Varey E, et al. Emergence of high-avidity melan-A-
in melanoma metastatic to the brain. N Engl J Med. 2018;379 specific clonotypes as a reflection of anti-PD-1 clinical efficacy. Can-
(22):2178. cer Res. 2017;77(24):7083-7093.
44. Tawbi HA, Forsyth PA, Algazi A, et al. Combined nivolumab and 62. Wieland A, Kamphorst AO, Adsay NV, et al. T cell receptor
ipilimumab in melanoma metastatic to the brain. N Engl J Med. sequencing of activated CD8 T cells in the blood identifies tumor-
2018;379(8):722-730. infiltrating clones that expand after PD-1 therapy and radiation
45. Weber JS, Gibney G, Sullivan RJ, et al. Sequential administration of in a melanoma patient. Cancer Immunol Immunother. 2018;67(11):
nivolumab and ipilimumab with a planned switch in patients with 1767-1776.
advanced melanoma (CheckMate 064): an open-label, randomised, 63. Bonaventura P, Shekarian T, Alcazer V, et al. Cold tumors: a therapeu-
phase 2 trial. Lancet Oncol. 2016;17(7):943-955. tic challenge for immunotherapy. Front Immunol. 2019;10:168.
46. Hodi FS, Chesney J, Pavlick AC, et al. Combined nivolumab and 64. Pages F, Mlecnik B, Marliot F, et al. International validation of
ipilimumab versus ipilimumab alone in patients with advanced the consensus immunoscore for the classification of colon can-
melanoma: 2-year overall survival outcomes in a multicentre, cer: a prognostic and accuracy study. Lancet. 2018;391(10135):
randomised, controlled, phase 2 trial. Lancet Oncol. 2016;17(11): 2128-2139.
1558-1568. 65. Galon J, Costes A, Sanchez-Cabo F, et al. Type, density, and location
47. Meerveld-Eggink A, Rozeman EA, Lalezari F, van Thienen JV, Haanen of immune cells within human colorectal tumors predict clinical out-
J, Blank CU. Short-term CTLA-4 blockade directly followed by PD-1 come. Science. 2006;313(5795):1960-1964.
blockade in advanced melanoma patients: a single-center experience. 66. Mlecnik B, Tosolini M, Kirilovsky A, et al. Histopathologic-based prog-
Ann Oncol. 2017;28(4):862-867. nostic factors of colorectal cancers are associated with the state of
48. Kirchberger MC, Hauschild A, Schuler G, Heinzerling L. Com- the local immune reaction. J Clin Oncol. 2011;29(6):610-618.
bined low-dose ipilimumab and pembrolizumab after sequential ipil- 67. Galon J, Angell HK, Bedognetti D, Marincola FM. The continuum of
imumab and pembrolizumab failure in advanced melanoma. Eur cancer immunosurveillance: prognostic, predictive, and mechanistic
J Cancer. 2016;65:182-184. signatures. Immunity. 2013;39(1):11-26.
ROY ET AL . 27

68. Angell H, Galon J. From the immune contexture to the Immunoscore: 87. Reinhard K, Rengstl B, Oehm P, et al. An RNA vaccine drives expan-
the role of prognostic and predictive immune markers in cancer. Curr sion and efficacy of claudin-CAR-T cells against solid tumors. Science.
Opin Immunol. 2013;25(2):261-267. 2020;367(6476):446-453.
69. Galon J, Fridman WH, Pages F. The adaptive immunologic microen- 88. Galon J, Bruni D. Approaches to treat immune hot, altered and cold
vironment in colorectal cancer: a novel perspective. Cancer Res. tumours with combination immunotherapies. Nat Rev Drug Discov.
2007;67(5):1883-1886. 2019;18(3):197-218.
70. Pages F, Kirilovsky A, Mlecnik B, et al. In situ cytotoxic and memory T 89. Zhao J, Chen Y, Ding ZY, Liu JY. Safety and efficacy of therapeu-
cells predict outcome in patients with early-stage colorectal cancer. tic cancer vaccines alone or in combination with immune checkpoint
J Clin Oncol. 2009;27(35):5944-5951. inhibitors in cancer treatment. Front Pharmacol. 2019;10:1184.
71. Galon J, Mlecnik B, Bindea G, et al. Towards the introduction 90. Foley KC, Nishimura MI, Moore TV. Combination immunotherapies
of the ‘Immunoscore’ in the classification of malignant tumours. implementing adoptive T-cell transfer for advanced-stage melanoma.
J Pathol. 2014;232(2):199-209. Melanoma Res. 2018;28(3):171-184.
72. Camus M, Tosolini M, Mlecnik B, et al. Coordination of intratumoral 91. Fennemann FL, de Vries IJM, Figdor CG, Verdoes M. Attacking
immune reaction and human colorectal cancer recurrence. Cancer tumors from all sides: personalized multiplex vaccines to tackle intra-
Res. 2009;69(6):2685-2693. tumor heterogeneity. Front Immunol. 2019;10:824.
73. Mlecnik B, Bindea G, Angell HK, et al. Integrative analyses of colorec- 92. Banchereau J, Palucka K. Immunotherapy: cancer vaccines on the
tal cancer show immunoscore is a stronger predictor of patient sur- move. Nat Rev Clin Oncol. 2018;15(1):9-10.
vival than microsatellite instability. Immunity. 2016;44(3):698-711. 93. Chiang CL, Coukos G, Kandalaft LE. Whole tumor antigen vaccines:
74. Mlecnik B, Bindea G, Angell HK, et al. Functional network pipeline where are we? Vaccines. 2015;3(2):344-372.
reveals genetic determinants associated with in situ lympho- 94. Chiang CL, Benencia F, Coukos G. Whole tumor antigen vaccines.
cyte proliferation and survival of cancer patients. Sci Transl Med. Semin Immunol. 2010;22(3):132-143.
2014;6(228):228ra37. 95. Buckwalter MR, Srivastava PK. “It is the antigen(s), stupid” and other
75. Chew V, Chen J, Lee D, et al. Chemokine-driven lymphocyte lessons from over a decade of vaccitherapy of human cancer. Semin
infiltration: an early intratumoural event determining long-term Immunol. 2008;20(5):296-300.
survival in resectable hepatocellular carcinoma. Gut. 2012;61(3): 96. Dranoff G, Jaffee E, Lazenby A, et al. Vaccination with irradiated
427-438. tumor cells engineered to secrete murine granulocyte-macrophage
76. Tahkola K, Mecklin JP, Wirta EV, Ahtiainen M, Helminen O, Bohm J, colony-stimulating factor stimulates potent, specific, and long-
Kellokumpu I. High immune cell score predicts improved survival in lasting anti-tumor immunity. Proc Natl Acad Sci USA. 1993;90(8):
pancreatic cancer. Virchows Arch. 2018;472(4):653-665. 3539-3543.
77. Bosmuller HC, Wagner P, Peper JK, et al. Combined immunoscore of 97. Armstrong CA, Botella R, Galloway TH, et al. Antitumor effects
CD103 and CD3 identifies long-term survivors in high-grade serous of granulocyte-macrophage colony-stimulating factor production by
ovarian cancer. Int J Gynecol Cancer. 2016;26(4):671-679. melanoma cells. Cancer Res. 1996;56(9):2191-2198.
78. Hao D, Liu J, Chen M, et al. Immunogenomic analyses of advanced 98. Sanda MG, Ayyagari SR, Jaffee EM, et al. Demonstration of a
serous ovarian cancer reveal immune score is a strong prognos- rational strategy for human prostate cancer gene therapy. J Urol.
tic factor and an indicator of chemosensitivity. Clin Cancer Res. 1994;151(3):622-628.
2018;24(15):3560-3571. 99. Dunussi-Joannopoulos K, Dranoff G, Weinstein HJ, Ferrara JL,
79. Li B, VanRoey M, Wang C, Chen TH, Korman A, Jooss K. Anti- Bierer BE, Croop JM. Gene immunotherapy in murine acute myeloid
programmed death-1 synergizes with granulocyte macrophage leukemia: granulocyte-macrophage colony-stimulating factor tumor
colony-stimulating factor–secreting tumor cell immunotherapy pro- cell vaccines elicit more potent antitumor immunity compared
viding therapeutic benefit to mice with established tumors. Clin Can- with B7 family and other cytokine vaccines. Blood. 1998;91(1):
cer Res. 2009;15(5):1623-1634. 222-230.
80. Duraiswamy J, Freeman GJ, Coukos G. Therapeutic PD-1 pathway 100. Dranoff G. GM-CSF-based cancer vaccines. Immunol Rev.
blockade augments with other modalities of immunotherapy T-cell 2002;188:147-154.
function to prevent immune decline in ovarian cancer. Cancer Res. 101. Soiffer R, Hodi FS, Haluska F, et al. Vaccination with irradiated,
2013;73(23):6900-6912. autologous melanoma cells engineered to secrete granulocyte-
81. Duraiswamy J, Freeman GJ, Coukos G. Dual blockade of PD-1 macrophage colony-stimulating factor by adenoviral-mediated gene
and CTLA-4 combined with tumor vaccine effectively restores T- transfer augments antitumor immunity in patients with metastatic
cell rejection function in tumors–response. Cancer Res. 2014;74(2): melanoma. J Clin Oncol. 2003;21(17):3343-3350.
633-634. 102. Soiffer R, Lynch T, Mihm M, et al. Vaccination with irradiated autol-
82. Curran MA, Montalvo W, Yagita H, Allison JP. PD-1 and CTLA-4 com- ogous melanoma cells engineered to secrete human granulocyte-
bination blockade expands infiltrating T cells and reduces regulatory macrophage colony-stimulating factor generates potent antitumor
T and myeloid cells within B16 melanoma tumors. Proc Natl Acad Sci immunity in patients with metastatic melanoma. Proc Natl Acad Sci
USA. 2010;107(9):4275-4280. USA. 1998;95(22):13141-13146.
83. Deng L, Liang H, Xu M, et al. STING-dependent cytosolic DNA sensing 103. Thomas AM, Santarsiero LM, Lutz ER, et al. Mesothelin-specific
promotes radiation-induced type I interferon-dependent antitumor CD8(+) T cell responses provide evidence of in vivo cross-priming
immunity in immunogenic tumors. Immunity. 2014;41(5):843-852. by antigen-presenting cells in vaccinated pancreatic cancer patients.
84. Woo SR, Fuertes MB, Corrales L, et al. STING-dependent cytosolic J Exp Med. 2004;200(3):297-306.
DNA sensing mediates innate immune recognition of immunogenic 104. Eager R, Nemunaitis J. GM-CSF gene-transduced tumor vaccines. Mol
tumors. Immunity. 2014;41(5):830-842. Ther. 2005;12(1):18-27.
85. Zheng W, Skowron KB, Namm JP, et al. Combination of radiotherapy 105. Hege KM, Jooss K, Pardoll D. GM-CSF gene-modifed cancer cell
and vaccination overcomes checkpoint blockade resistance. Oncotar- immunotherapies: of mice and men. Int Rev Immunol. 2006;25(5-
get. 2016;7(28):43039-43051. 6):321-352.
86. Ma L, Dichwalkar T, Chang JYH, et al. Enhanced CAR-T cell activity 106. Laheru D, Lutz E, Burke J, et al. Allogeneic granulocyte macrophage
against solid tumors by vaccine boosting through the chimeric recep- colony-stimulating factor-secreting tumor immunotherapy alone or
tor. Science. 2019;365(6449):162-168. in sequence with cyclophosphamide for metastatic pancreatic can-
28 ROY ET AL .

cer: a pilot study of safety, feasibility, and immune activation. Clin Can- 126. Nizzoli G, Krietsch J, Weick A, et al. Human CD1c+ dendritic cells
cer Res. 2008;14(5):1455-1463. secrete high levels of IL-12 and potently prime cytotoxic T-cell
107. Lipson EJ, Sharfman WH, Chen S, et al. Safety and immunologic corre- responses. Blood. 2013;122(6):932-942.
lates of Melanoma GVAX, a GM-CSF secreting allogeneic melanoma 127. Perussia B, Fanning V, Trinchieri G. A leukocyte subset bearing HLA-
cell vaccine administered in the adjuvant setting. J Transl Med. DR antigens is responsible for in vitro alpha interferon produc-
2015;13:214. tion in response to viruses. Nat Immun Cell Growth Regul. 1985;4(3):
108. Small EJ, Sacks N, Nemunaitis J, et al. Granulocyte macrophage 120-137.
colony-stimulating factor–secreting allogeneic cellular immunother- 128. Anguille S, Smits EL, Bryant C, et al. Dendritic cells as pharmaco-
apy for hormone-refractory prostate cancer. Clin Cancer Res. logical tools for cancer immunotherapy. Pharmacol Rev. 2015;67(4):
2007;13(13):3883-3891. 731-753.
109. Salgia R, Lynch T, Skarin A, et al. Vaccination with irradiated autol- 129. Anguille S, Smits EL, Lion E, van Tendeloo VF, Berneman ZN.
ogous tumor cells engineered to secrete granulocyte-macrophage Clinical use of dendritic cells for cancer therapy. Lancet Oncol.
colony-stimulating factor augments antitumor immunity in some 2014;15(7):e257-e267.
patients with metastatic non-small-cell lung carcinoma. J Clin Oncol. 130. Koski GK, Cohen PA, Roses RE, Xu S, Czerniecki BJ. Reengi-
2003;21(4):624-630. neering dendritic cell-based anti-cancer vaccines. Immunol Rev.
110. Le DT, Picozzi VJ, Ko AH, et al. Results from a phase IIb, 2008;222:256-276.
randomized, multicenter study of GVAX pancreas and CRS-207 131. Garg AD, Coulie PG, Van den Eynde BJ, Agostinis P. Integrat-
compared with chemotherapy in adults with previously treated ing next-generation dendritic cell vaccines into the current cancer
metastatic pancreatic adenocarcinoma (ECLIPSE Study). Clin Cancer immunotherapy landscape. Trends Immunol. 2017;38(8):577-593.
Res. 2019;25(18):5493-5502. 132. Inaba K, Inaba M, Romani N, et al. Generation of large numbers
111. Hoover HC, Jr., Surdyke MG, Dangel RB, Peters LC, Hanna of dendritic cells from mouse bone marrow cultures supplemented
MG, Jr. Prospectively randomized trial of adjuvant active- with granulocyte/macrophage colony-stimulating factor. J Exp Med.
specific immunotherapy for human colorectal cancer. Cancer. 1992;176(6):1693-1702.
1985;55(6):1236-1243. 133. Inaba K, Pack M, Inaba M, Sakuta H, Isdell F, Steinman RM. High
112. Hoover HC, Jr., Brandhorst JS, Peters LC, et al. Adjuvant active spe- levels of a major histocompatibility complex II-self peptide complex
cific immunotherapy for human colorectal cancer: 6.5-year median on dendritic cells from the T cell areas of lymph nodes. J Exp Med.
follow-up of a phase III prospectively randomized trial. J Clin Oncol. 1997;186(5):665-672.
1993;11(3):390-399. 134. Inaba K, Steinman RM, Pack MW, et al. Identification of pro-
113. Vermorken JB, Claessen AM, van Tinteren H, et al. Active specific liferating dendritic cell precursors in mouse blood. J Exp Med.
immunotherapy for stage II and stage III human colon cancer: a ran- 1992;175(5):1157-1167.
domised trial. Lancet. 1999;353(9150):345-350. 135. Butterfield LH. Dendritic cells in cancer immunotherapy clinical tri-
114. Van Poppel H, Joniau S, Van Gool SW. Vaccine therapy in patients als: are we making progress? Front Immunol. 2013;4:454.
with renal cell carcinoma. Eur Urol. 2009;55(6):1333-1342. 136. Butterfield LH, Ribas A, Dissette VB, et al. Determinant spreading
115. May M, Kendel F, Hoschke B, et al. [Adjuvant autologous tumour associated with clinical response in dendritic cell-based immunother-
cell vaccination in patients with renal cell carcinoma. Overall survival apy for malignant melanoma. Clin Cancer Res. 2003;9(3):998-1008.
analysis with a follow-up period in excess of more than 10 years]. Urol 137. Hsu FJ, Benike C, Fagnoni F, et al. Vaccination of patients with B-cell
A. 2009;48(9):1075-1083. lymphoma using autologous antigen-pulsed dendritic cells. Nat Med.
116. Simons JW, Jaffee EM, Weber CE, et al. Bioactivity of autolo- 1996;2(1):52-58.
gous irradiated renal cell carcinoma vaccines generated by ex vivo 138. Markowicz S, Engleman EG. Granulocyte-macrophage colony-
granulocyte-macrophage colony-stimulating factor gene transfer. stimulating factor promotes differentiation and survival of human
Cancer Res. 1997;57(8):1537-1546. peripheral blood dendritic cells in vitro. J Clin Invest. 1990;85(3):
117. Bol KF, Schreibelt G, Gerritsen WR, de Vries IJ, Figdor CG. Dendritic 955-961.
cell-based immunotherapy: state of the art and beyond. Clin Cancer 139. Mukherji B, Chakraborty NG, Yamasaki S, et al. Induction of antigen-
Res. 2016;22(8):1897-1906. specific cytolytic T cells in situ in human melanoma by immunization
118. van Willigen WW, Bloemendal M, Gerritsen WR, Schreibelt G, de with synthetic peptide-pulsed autologous antigen presenting cells.
Vries IJM, Bol KF. Dendritic cell cancer therapy: vaccinating the right Proc Natl Acad Sci USA. 1995;92(17):8078-8082.
patient at the right time. Front Immunol. 2018;9:2265. 140. Nestle FO, Alijagic S, Gilliet M, et al. Vaccination of melanoma
119. Santos PM, Butterfield LH. Dendritic cell-based cancer vaccines. patients with peptide- or tumor lysate-pulsed dendritic cells. Nat
J Immunol. 2018;200(2):443-449. Med. 1998;4(3):328-332.
120. Mastelic-Gavillet B, Balint K, Boudousquie C, Gannon PO, Kandalaft 141. Romani N, Gruner S, Brang D, et al. Proliferating dendritic cell pro-
LE. Personalized dendritic cell vaccines-recent breakthroughs and genitors in human blood. J Exp Med. 1994;180(1):83-93.
encouraging clinical results. Front Immunol. 2019;10:766. 142. Van Tendeloo VF, Van de Velde A, Van Driessche A, et al. Induction
121. Huber A, Dammeijer F, Aerts J, Vroman H. Current state of dendritic of complete and molecular remissions in acute myeloid leukemia by
cell-based immunotherapy: opportunities for in vitro antigen loading Wilms’ tumor 1 antigen-targeted dendritic cell vaccination. Proc Natl
of different DC subsets? Front Immunol. 2018;9:2804. Acad Sci USA. 2010;107(31):13824-13829.
122. Saxena M, Bhardwaj N. Re-emergence of dendritic cell vaccines for 143. Lim DS, Kim JH, Lee DS, Yoon CH, Bae YS. DC immunother-
cancer treatment. Trends Cancer. 2018;4(2):119-137. apy is highly effective for the inhibition of tumor metastasis or
123. Ahmed MS, Bae YS. Dendritic cell-based therapeutic cancer vaccines: recurrence, although it is not efficient for the eradication of
past, present and future. Clin Exp Vaccine Res. 2014;3(2):113-116. established solid tumors. Cancer Immunol Immunother. 2007;56(11):
124. Dzionek A, Fuchs A, Schmidt P, et al. BDCA-2, BDCA-3, and BDCA-4: 1817-1829.
three markers for distinct subsets of dendritic cells in human periph- 144. Germeau C, Ma W, Schiavetti F, et al. High frequency of antitumor T
eral blood. J Immunol. 2000;165(11):6037-6046. cells in the blood of melanoma patients before and after vaccination
125. Lande R, Gregorio J, Facchinetti V, et al. Plasmacytoid dendritic with tumor antigens. J Exp Med. 2005;201(2):241-248.
cells sense self-DNA coupled with antimicrobial peptide. Nature. 145. Akasaki Y, Kikuchi T, Homma S, et al. Phase I/II trial of combination
2007;449(7162):564-569. of temozolomide chemotherapy and immunotherapy with fusions
ROY ET AL . 29

of dendritic and glioma cells in patients with glioblastoma. Cancer monocyte-derived dendritic cells: implications for immunotherapy.
Immunol Immunother. 2016;65(12):1499-1509. Vaccine. 2002;20:(Suppl 4):A8-A22.
146. Rosenblatt J, Stone RM, Uhl L, Neuberg D, et al. Individualized vac- 164. Jongmans W, Tiemessen DM, van Vlodrop IJ, Mulders PF, Oosterwijk
cination of AML patients in remission is associated with induction E. Th1-polarizing capacity of clinical-grade dendritic cells is triggered
of antileukemia immunity and prolonged remissions. Sci Transl Med. by Ribomunyl but is compromised by PGE2: the importance of matu-
2016;8(368):368ra171. ration cocktails. J Immunother. 2005;28(5):480-487.
147. Crozat K, Guiton R, Guilliams M, et al. Comparative genomics as a tool 165. Krause P, Singer E, Darley PI, Klebensberger J, Groettrup M, Legler
to reveal functional equivalences between human and mouse den- DF. Prostaglandin E2 is a key factor for monocyte-derived dendritic
dritic cell subsets. Immunol Rev. 2010;234(1):177-198. cell maturation: enhanced T cell stimulatory capacity despite IDO.
148. Osugi Y, Vuckovic S, Hart DN. Myeloid blood CD11c(+) dendritic cells J Leukoc Biol. 2007;82(5):1106-1114.
and monocyte-derived dendritic cells differ in their ability to stimu- 166. Carreno BM, Becker-Hapak M, Huang A, et al. IL-12p70-producing
late T lymphocytes. Blood. 2002;100(8):2858-2866. patient DC vaccine elicits Tc1-polarized immunity. J Clin Invest.
149. Villani AC, Satija R, Reynolds G, et al. Single-cell RNA-seq reveals new 2013;123(8):3383-3394.
types of human blood dendritic cells, monocytes, and progenitors. Sci- 167. Kolanowski ST, Sritharan L, Lissenberg-Thunnissen SN, Van Schijndel
ence. 2017;356(6335):eaah4573. GM, Van Ham SM, ten Brinke A. Comparison of media and serum
150. Helft J, Bottcher J, Chakravarty P, et al. GM-CSF mouse bone supplementation for generation of monophosphoryl lipid
marrow cultures comprise a heterogeneous population of A/interferon-gamma-matured type I dendritic cells for immunother-
CD11c(+)MHCII(+) macrophages and dendritic cells. Immunity. apy. Cytotherapy. 2014;16(6):826-834.
2015;42(6):1197-1211. 168. Mailliard RB, Wankowicz-Kalinska A, Cai Q, et al. alpha-type-1 polar-
151. Balan S, Ollion V, Colletti N, et al. Human XCR1+ dendritic ized dendritic cells: a novel immunization tool with optimized CTL-
cells derived in vitro from CD34+ progenitors closely resemble inducing activity. Cancer Res. 2004;64(17):5934-5937.
blood dendritic cells, including their adjuvant responsiveness, con- 169. Wilgenhof S, Van Nuffel AM, Benteyn D, et al. A phase IB
trary to monocyte-derived dendritic cells. J Immunol. 2014;193(4): study on intravenous synthetic mRNA electroporated dendritic cell
1622-1635. immunotherapy in pretreated advanced melanoma patients. Ann
152. Collin M, McGovern N, Haniffa M. Human dendritic cell subsets. Oncol. 2013;24(10):2686-2693.
Immunology. 2013;140(1):22-30. 170. Massa C, Thomas C, Wang E, Marincola F, Seliger B. Different matura-
153. Lundberg K, Albrekt AS, Nelissen I, et al. Transcriptional profiling of tion cocktails provide dendritic cells with different chemoattractive
human dendritic cell populations and models—unique profiles of in properties. J Transl Med. 2015;13:175.
vitro dendritic cells and implications on functionality and applicabil- 171. Van Nuffel AM, Benteyn D, Wilgenhof S, et al. Intravenous and
ity. PLoS One. 2013;8(1):e52875. intradermal TriMix-dendritic cell therapy results in a broad T-
154. Jongbloed SL, Kassianos AJ, McDonald KJ, et al. Human CD141+ cell response and durable tumor response in a chemorefrac-
(BDCA-3)+ dendritic cells (DCs) represent a unique myeloid DC tory stage IV-M1c melanoma patient. Cancer Immunol Immunother.
subset that cross-presents necrotic cell antigens. J Exp Med. 2012;61(7):1033-1043.
2010;207(6):1247-1260. 172. Koido S. Dendritic-tumor fusion cell-based cancer vaccines. Int J Mol
155. Orsini E, Guarini A, Chiaretti S, Mauro FR, Foa R. The circulat- Sci. 2016;17(6)828.
ing dendritic cell compartment in patients with chronic lymphocytic 173. Lee JM, Lee MH, Garon E, et al. Phase I trial of intratumoral injection
leukemia is severely defective and unable to stimulate an effective T- of CCL21 gene-modified dendritic cells in lung cancer elicits tumor-
cell response. Cancer Res. 2003;63(15):4497-4506. specific immune responses and CD8(+) T-cell infiltration. Clin Cancer
156. Schreibelt G, Bol KF, Westdorp H, et al. Effective clinical responses Res. 2017;23(16):4556-4568.
in metastatic melanoma patients after vaccination with primary 174. Chiang CL, Kandalaft LE, Tanyi J, et al. A dendritic cell vaccine pulsed
myeloid dendritic cells. Clin Cancer Res. 2016;22(9):2155-2166. with autologous hypochlorous acid-oxidized ovarian cancer lysate
157. Tel J, Aarntzen EH, Baba T, et al. Natural human plasmacytoid den- primes effective broad antitumor immunity: from bench to bedside.
dritic cells induce antigen-specific T-cell responses in melanoma Clin Cancer Res. 2013;19(17):4801-4815.
patients. Cancer Res. 2013;73(3):1063-1075. 175. Zhao L, Niu C, Shi X, et al. Dendritic cells loaded with the lysate
158. Di Nicola M, Carlo-Stella C, Mortarini R, et al. Boosting T cell- of tumor cells infected with Newcastle Disease Virus trigger potent
mediated immunity to tyrosinase by vaccinia virus-transduced, anti-tumor immunity by promoting the secretion of IFN-gamma and
CD34(+)-derived dendritic cell vaccination: a phase I trial in IL-2 from T cells. Oncol Lett. 2018;16(1):1180-1188.
metastatic melanoma. Clin Cancer Res. 2004;10(16):5381-5390. 176. Tanyi JL, Bobisse S, Ophir E, et al. Personalized cancer vaccine effec-
159. Mackensen A, Herbst B, Chen JL, et al. Phase I study in melanoma tively mobilizes antitumor T cell immunity in ovarian cancer. Sci Transl
patients of a vaccine with peptide-pulsed dendritic cells generated Med. 2018;10(436):eaao5931.
in vitro from CD34(+) hematopoietic progenitor cells. Int J Cancer. 177. Liau LM, Ashkan K, Tran DD, et al. First results on survival from
2000;86(3):385-392. a large phase 3 clinical trial of an autologous dendritic cell vac-
160. Balan S, Arnold-Schrauf C, Abbas A, et al. Large-scale human den- cine in newly diagnosed glioblastoma. J Transl Med. 2018;16(1):
dritic cell differentiation revealing notch-dependent lineage bifurca- 142.
tion and heterogeneity. Cell Rep. 2018;24(7):1902-1915 e6. 178. Overwijk WW, Wang E, Marincola FM, Rammensee HG, Restifo NP.
161. Kirkling ME, Cytlak U, Lau CM, et al. Notch signaling facilitates in Mining the mutanome: developing highly personalized Immunother-
vitro generation of cross-presenting classical dendritic cells. Cell Rep. apies based on mutational analysis of tumors. J Immunother Cancer.
2018;23(12):3658-3672 e6. 2013;1:11.
162. Proietto AI, Mittag D, Roberts AW, Sprigg N, Wu L. The equivalents 179. Carreno BM, Magrini V, Becker-Hapak M, et al. Cancer immunother-
of human blood and spleen dendritic cell subtypes can be gener- apy. a dendritic cell vaccine increases the breadth and diversity
ated in vitro from human CD34(+) stem cells in the presence of fms- of melanoma neoantigen-specific T cells. Science. 2015;348(6236):
like tyrosine kinase 3 ligand and thrombopoietin. Cell Mol Immunol. 803-808.
2012;9(6):446-454. 180. Bruggen P, Traversari C, Chomez P, et al. A gene encoding an anti-
163. Lee AW, Truong T, Bickham K, et al. A clinical grade cocktail gen recognized by cytolytic T lymphocytes on a human melanoma.
of cytokines and PGE2 results in uniform maturation of human Science. 1991;254(5038):1643-1647.
30 ROY ET AL .

181. Kallen K-J, Gnad-Vogt U, Scheel B, Rippin G, Stenzl A. A phase I/IIa 200. Rahma OE, Hamilton JM, Wojtowicz M, et al. The immunological and
study of the mRNA based cancer vaccine CV9103 prepared with the clinical effects of mutated ras peptide vaccine in combination with
RNActive technology results in distinctly longer survival than pre- IL-2, GM-CSF, or both in patients with solid tumors. J Transl Med.
dicted by the Halabi Nomogram which correlates with the induc- 2014;12(1):55.
tion of antigen-specific immune responses. J Immunother Cancer. 201. Toubaji A, Achtar M, Provenzano M, et al. Pilot study of mutant ras
2013;1(1):P219. peptide-based vaccine as an adjuvant treatment in pancreatic and
182. Shariat S, Badiee A, Jalali SA, et al. P5 HER2/neu-derived pep- colorectal cancers. Cancer Immunol Immunother. 2008;57(9):1413-
tide conjugated to liposomes containing MPL adjuvant as an effec- 1420.
tive prophylactic vaccine formulation for breast cancer. Cancer Lett. 202. Suzuki E, Kapoor V, Jassar AS, Kaiser LR, Albelda SM. Gemcitabine
2014;355(1):54-60. selectively eliminates splenic Gr-1+/CD11b+ myeloid suppressor
183. Schmidt SM, Schag K, Müller MR, et al. Survivin is a shared tumor- cells in tumor-bearing animals and enhances antitumor immune
associated antigen expressed in a broad variety of malignancies and activity. Clin Cancer Res. 2005;11(18):6713-6721.
recognized by specific cytotoxic T cells. Blood. 2003;102(2):571-576. 203. Wang Z, Till B, Gao Q. Chemotherapeutic agent-mediated elim-
184. Bakker AB, Schreurs MW, de Boer AJ, et al. Melanocyte lineage- ination of myeloid-derived suppressor cells. Oncoimmunology.
specific antigen gp100 is recognized by melanoma-derived tumor- 2017;6(7):e1331807.
infiltrating lymphocytes. J Exp Med. 1994;179(3):1005-1009. 204. Lutsiak ME, Semnani RT, De Pascalis R, Kashmiri SV, Schlom J, Sabze-
185. Mikolajczyk SD, Rittenhouse HG. Tumor-associated forms of vari H. Inhibition of CD4(+)25+ T regulatory cell function implicated
prostate specific antigen improve the discrimination of prostate in enhanced immune response by low-dose cyclophosphamide. Blood.
cancer from benign disease. Rinsho Byori. 2004;52(3):223-230. 2005;105(7):2862-2868.
186. Chi DD, Merchant RE, Rand R, et al. Molecular detection of tumor- 205. Le HK, Graham L, Cha E, Morales JK, Manjili MH, Bear HD. Gemc-
associated antigens shared by human cutaneous melanomas and itabine directly inhibits myeloid derived suppressor cells in BALB/c
gliomas. Am J Pathol. 1997;150(6):2143-2152. mice bearing 4T1 mammary carcinoma and augments expansion of
187. Jungbluth AA, Busam KJ, Kolb D, et al. Expression of MAGE-antigens T cells from tumor-bearing mice. Int Immunopharmacol. 2009;9(7-
in normal tissues and cancer. Int J Cancer. 2000;85(4):460-465. 8):900-909.
188. Schultz-Thater E, Noppen C, Gudat F, et al. NY-ESO-1 tumour asso- 206. Vincent J, Mignot G, Chalmin F, et al. 5-Fluorouracil selectively
ciated antigen is a cytoplasmic protein detectable by specific mon- kills tumor-associated myeloid-derived suppressor cells resulting
oclonal antibodies in cell lines and clinical specimens. Br J Cancer. in enhanced T cell-dependent antitumor immunity. Cancer Res.
2000;83(2):204-208. 2010;70(8):3052-3061.
189. Williams RR, McIntire KR, Waldmann TA, et al. Tumor-associated 207. Qu X, Felder MA, Perez Horta Z, Sondel PM, Rakhmilevich AL. Antitu-
antigen levels (carcinoembryonic antigen, human chorionic mor effects of anti-CD40/CpG immunotherapy combined with gemc-
gonadotropin, and alpha-fetoprotein) antedating the diagno- itabine or 5-fluorouracil chemotherapy in the B16 melanoma model.
sis of cancer in the Framingham Study 1. J Natl Cancer Inst. Int Immunopharmacol. 2013;17(4):1141-1147.
1977;58(6):1547-1551. 208. Sawant A, Schafer CC, Jin TH, et al. Enhancement of antitumor immu-
190. Kelderman S, Heemskerk B, Fanchi L, et al. Antigen-specific TIL nity in lung cancer by targeting myeloid-derived suppressor cell path-
therapy for melanoma: a flexible platform for personalized cancer ways. Cancer Res. 2013;73(22):6609-6620.
immunotherapy. Eur J Immunol. 2016;46(6):1351-1360. 209. Otsubo D, Yamashita K, Fujita M, et al. Early-phase treatment
191. Kelderman S, Kvistborg P. Tumor antigens in human cancer control. by low-dose 5-fluorouracil or primary tumor resection inhibits
Biochim Biophys Acta. 2016;1865(1):83-89. MDSC-mediated lung metastasis formation. Anticancer Res.
192. Pleasance ED, Cheetham RK, Stephens PJ, et al. A comprehensive cat- 2015;35(8):4425-4431.
alogue of somatic mutations from a human cancer genome. Nature. 210. Annels NE, Shaw VE, Gabitass RF, et al. The effects of gemc-
2010;463(7278):191-196. itabine and capecitabine combination chemotherapy and of low-
193. Marchand M, Weynants P, Rankin E, et al. Tumor regression dose adjuvant GM-CSF on the levels of myeloid-derived suppressor
responses in melanoma patients treated with a peptide encoded by cells in patients with advanced pancreatic cancer. Cancer Immunol
gene MAGES-3. Int J Cancer. 1995;63(6):883-885. Immunother. 2014;63(2):175-183.
194. Parmiani G, Russo V, Maccalli C, Parolini D, Rizzo N, Maio M. 211. Chaudhary B, Elkord E. Regulatory T cells in the tumor microenvi-
Peptide-based vaccines for cancer therapy. Hum Vaccin Immunother. ronment and cancer progression: role and therapeutic targeting. Vac-
2014;10(11):3175-3178. cines. 2016;4(3):28.
195. Habjanec L, Halassy B, Tomašić J. Immunomodulatory activity 212. Ghiringhelli F, Larmonier N, Schmitt E, et al. CD4+CD25+ regula-
of novel adjuvant formulations based on Montanide ISA oil- tory T cells suppress tumor immunity but are sensitive to cyclophos-
based adjuvants and peptidoglycan monomer. Int Immunopharmacol. phamide which allows immunotherapy of established tumors to be
2008;8(5):717-424. curative. Eur J Immunol. 2004;34(2):336-344.
196. Sabbatini P, Tsuji T, Ferran L, et al. Phase I trial of overlapping long 213. Li JY, Duan XF, Wang LP, et al. Selective depletion of regulatory T cell
peptides from a tumor self-antigen and poly-ICLC shows rapid induc- subsets by docetaxel treatment in patients with nonsmall cell lung
tion of integrated immune response in ovarian cancer patients. Clin cancer. J Immunol Res. 2014;2014:286170.
Cancer Res. 2012;18(23):6497-6508. 214. Taniguchi H, Iwasa S, Yamazaki K, et al. Phase 1 study of OCV-C02,
197. Tsuji T, Sabbatini P, Jungbluth AA, et al. Effect of Montanide and poly- a peptide vaccine consisting of two peptide epitopes for refractory
ICLC adjuvant on human self/tumor antigen-specific CD4+ T cells in metastatic colorectal cancer. Cancer Sci. 2017;108(5):1013-1021.
phase I overlapping long peptide vaccine trial. Cancer Immunol Res. 215. Restifo NP, Ying H, Hwang L, Leitner WW. The promise of nucleic acid
2013;1(5):340-350. vaccines. Gene Ther. 2000;7(2):89-92.
198. Chamani R, Ranji P, Hadji M, Nahvijou A, Esmati E, Alizadeh AM. 216. Hobernik D, Bros M. DNA vaccines-how far from clinical use? Int J Mol
Application of E75 peptide vaccine in breast cancer patients: a sys- Sci. 2018;19(11):3605.
tematic review and meta-analysis. Eur J Pharmacol. 2018;831:87-93. 217. Pastor F, Berraondo P, Etxeberria I, et al. An RNA toolbox for cancer
199. Khleif SN, Abrams SI, Hamilton JM, et al. A phase I vaccine trial immunotherapy. Nat Rev Drug Discov. 2018;17(10):751-767.
with peptides reflecting ras oncogene mutations of solid tumors. 218. Rodriguez-Ruiz ME, Perez-Gracia JL, Rodriguez I, et al. Combined
J Immunother. 1999;22(2):155-165. immunotherapy encompassing intratumoral poly-ICLC, dendritic-cell
ROY ET AL . 31

vaccination and radiotherapy in advanced cancer patients. Ann Oncol. cinoma (RCC): phase 2 study results. J Immunother Cancer. 2015;
2018;29(5):1312-1319. 3:14.
219. Pardi N, Muramatsu H, Weissman D, Kariko K. In vitro transcrip- 237. Dorfel D, Appel S, Grunebach F, et al. Processing and presentation of
tion of long RNA containing modified nucleosides. Methods Mol Biol. HLA class I and II epitopes by dendritic cells after transfection with in
2013;969:29-42. vitro-transcribed MUC1 RNA. Blood. 2005;105(8):3199-3205.
220. Pardi N, Hogan MJ, Porter FW, Weissman D. mRNA vaccines - a new 238. Grunebach F, Kayser K, Weck MM, Muller MR, Appel S, Brossart P.
era in vaccinology. Nat Rev Drug Discov. 2018;17(4):261-279. Cotransfection of dendritic cells with RNA coding for HER-2/neu and
221. Conry RM, LoBuglio AF, Wright M, et al. Characterization of 4-1BBL increases the induction of tumor antigen specific cytotoxic T
a messenger RNA polynucleotide vaccine vector. Cancer Res. lymphocytes. Cancer Gene Ther. 2005;12(9):749-756.
1995;55(7):1397-1400. 239. Heiser A, Coleman D, Dannull J, et al. Autologous dendritic
222. Dileo J, Miller TE, Jr., Chesnoy S, Huang L. Gene transfer to subdermal cells transfected with prostate-specific antigen RNA stimulate
tissues via a new gene gun design. Hum Gene Ther. 2003;14(1):79-87. CTL responses against metastatic prostate tumors. J Clin Invest.
223. Qiu P, Ziegelhoffer P, Sun J, Yang NS. Gene gun delivery of mRNA in 2002;109(3):409-417.
situ results in efficient transgene expression and genetic immuniza- 240. Morse MA, Nair SK, Boczkowski D, et al. The feasibility and safety of
tion. Gene Ther. 1996;3(3):262-268. immunotherapy with dendritic cells loaded with CEA mRNA follow-
224. Steitz J, Britten CM, Wolfel T, Tuting T. Effective induction of anti- ing neoadjuvant chemoradiotherapy and resection of pancreatic can-
melanoma immunity following genetic vaccination with synthetic cer. Int J Gastrointest Cancer. 2002;32(1):1-6.
mRNA coding for the fusion protein EGFP.TRP2. Cancer Immunol 241. Morse MA, Nair SK, Mosca PJ, et al. Immunotherapy with autolo-
Immunother. 2006;55(3):246-253. gous, human dendritic cells transfected with carcinoembryonic anti-
225. Weide B, Pascolo S, Scheel B, et al. Direct injection of protamine- gen mRNA. Cancer Invest. 2003;21(3):341-349.
protected mRNA: results of a phase 1/2 vaccination trial in metastatic 242. Van Lint S, Wilgenhof S, Heirman C, et al. Optimized dendritic cell-
melanoma patients. J Immunother. 2009;32(5):498-507. based immunotherapy for melanoma: the TriMix-formula. Cancer
226. Stenzl A, Feyerabend S, Syndikus I, et al. 1149PResults of the Immunol Immunother. 2014;63(9):959-967.
randomized, placebo-controlled phase I/IIB trial of CV9104, an 243. Wilgenhof S, Corthals J, Van Nuffel AM, et al. Long-term clini-
mRNA based cancer immunotherapy, in patients with metastatic cal outcome of melanoma patients treated with messenger RNA-
castration-resistant prostate cancer (mCRPC). Ann Oncol. electroporated dendritic cell therapy following complete resection of
2017;28(suppl_5):v403-v407. metastases. Cancer Immunol Immunother. 2015;64(3):381-388.
227. Sebastian M, Schroder A, Scheel B, et al. A phase I/IIa study of 244. Lopes A, Vandermeulen G, Preat V. Cancer DNA vaccines: current
the mRNA-based cancer immunotherapy CV9201 in patients with preclinical and clinical developments and future perspectives. J Exp
stage IIIB/IV non-small cell lung cancer. Cancer Immunol Immunother. Clin Cancer Res. 2019;38(1):146.
2019;68(5):799-812. 245. Tiptiri-Kourpeti A, Spyridopoulou K, Pappa A, Chlichlia K. DNA vac-
228. Hong HS, Koch SD, Scheel B, et al. Distinct transcriptional changes cines to attack cancer: strategies for improving immunogenicity and
in non-small cell lung cancer patients associated with multi- efficacy. Pharmacol Ther. 2016;165:32-49.
antigenic RNActive(R) CV9201 immunotherapy. Oncoimmunology. 246. Aghi M, Visted T, Depinho RA, Chiocca EA. Oncolytic herpes
2016;5(12):e1249560. virus with defective ICP6 specifically replicates in quiescent cells
229. Papachristofilou A, Hipp MM, Klinkhardt U, et al. Phase Ib evaluation with homozygous genetic mutations in p16. Oncogene. 2008;27(30):
of a self-adjuvanted protamine formulated mRNA-based active can- 4249-4254.
cer immunotherapy, BI1361849 (CV9202), combined with local radi- 247. Howells A, Marelli G, Lemoine NR, Wang Y. Oncolytic viruses-
ation treatment in patients with stage IV non-small cell lung cancer. interaction of virus and tumor cells in the battle to eliminate cancer.
J Immunother Cancer. 2019;7(1):38. Front Oncol. 2017;7:195.
230. Burris HA, Patel MR, Cho DC, et al. A phase I multicenter study to 248. Russell SJ, Barber GN. Oncolytic viruses as antigen-agnostic cancer
assess the safety, tolerability, and immunogenicity of mRNA-4157 vaccines. Cancer Cell. 2018;33(4):599-605.
alone in patients with resected solid tumors and in combination with 249. Twumasi-Boateng K, Pettigrew JL, Kwok YYE, Bell JC, Nelson
pembrolizumab in patients with unresectable solid tumors. J Clin BH. Oncolytic viruses as engineering platforms for combination
Oncol. 2019;37(15_suppl):2523. immunotherapy. Nat Rev Cancer. 2018;18(7):419-432.
231. Bonehill A, Van Nuffel AM, Corthals J, et al. Single-step antigen load- 250. Bommareddy PK, Shettigar M, Kaufman HL. Integrating oncolytic
ing and activation of dendritic cells by mRNA electroporation for the viruses in combination cancer immunotherapy. Nat Rev Immunol.
purpose of therapeutic vaccination in melanoma patients. Clin Cancer 2018;18(8):498-513.
Res. 2009;15(10):3366-3375. 251. Coffey MC, Strong JE, Forsyth PA, Lee PW. Reovirus therapy
232. Kyte JA, Kvalheim G, Aamdal S, Saeboe-Larssen S, Gaudernack G. of tumors with activated Ras pathway. Science. 1998;282(5392):
Preclinical full-scale evaluation of dendritic cells transfected with 1332-1334.
autologous tumor-mRNA for melanoma vaccination. Cancer Gene 252. Roy S, Bag AK, Dutta S, et al. Macrophage-derived neuropilin-
Ther. 2005;12(6):579-591. 2 exhibits novel tumor-promoting functions. Cancer Res.
233. McNamara MA, Nair SK, Holl EK. RNA-based vaccines in cancer 2018;78(19):5600-5617.
immunotherapy. J Immunol Res. 2015;2015:794528. 253. Barber GN. Cytoplasmic DNA innate immune pathways. Immunol Rev.
234. Nair SK, Morse M, Boczkowski D, et al. Induction of tumor-specific 2011;243(1):99-108.
cytotoxic T lymphocytes in cancer patients by autologous tumor 254. Barber GN. Innate immune DNA sensing pathways: STING, AIMII and
RNA-transfected dendritic cells. Ann Surg. 2002;235(4):540-549. the regulation of interferon production and inflammatory responses.
235. Su Z, Dannull J, Heiser A, et al. Immunological and clinical Curr Opin Immunol. 2011;23(1):10-20.
responses in metastatic renal cancer patients vaccinated with 255. Franz KM, Kagan JC. Innate immune receptors as competitive deter-
tumor RNA-transfected dendritic cells. Cancer Res. 2003;63(9): minants of cell fate. Mol Cell. 2017;66(6):750-760.
2127-2133. 256. Takeuchi O, Akira S. Innate immunity to virus infection. Immunol Rev.
236. Amin A, Dudek AZ, Logan TF, et al. Survival with AGS-003, an 2009;227(1):75-86.
autologous dendritic cell-based immunotherapy, in combination with 257. Kaminskyy V, Zhivotovsky B. To kill or be killed: how viruses interact
sunitinib in unfavorable risk patients with advanced renal cell car- with the cell death machinery. J Intern Med. 2010;267(5):473-482.
32 ROY ET AL .

258. Schock SN, Chandra NV, Sun Y, et al. Induction of necroptotic cell 279. Ben-Porath I, Thomson MW, Carey VJ, et al. An embryonic stem cell-
death by viral activation of the RIG-I or STING pathway. Cell Death like gene expression signature in poorly differentiated aggressive
Differ. 2017;24(4):615-625. human tumors. Nat Genet. 2008;40(5):499-507.
259. Raja J, Ludwig JM, Gettinger SN, Schalper KA, Kim HS. Oncolytic virus 280. Ghosh Z, Huang M, Hu S, Wilson KD, Dey D, Wu JC. Dissecting
immunotherapy: future prospects for oncology. J Immunother Cancer. the oncogenic and tumorigenic potential of differentiated human
2018;6(1):140. induced pluripotent stem cells and human embryonic stem cells. Can-
260. Lundstrom K. New frontiers in oncolytic viruses: optimizing cer Res. 2011;71(14):5030-5039.
and selecting for virus strains with improved efficacy. Biologics. 281. de Almeida PE, Meyer EH, Kooreman NG, et al. Transplanted ter-
2018;12:43-60. minally differentiated induced pluripotent stem cells are accepted
261. Andtbacka RH, Kaufman HL, Collichio F, et al. Talimogene laher- by immune mechanisms similar to self-tolerance. Nat Commun.
parepvec improves durable response rate in patients with advanced 2014;5:3903.
melanoma. J Clin Oncol. 2015;33(25):2780-2788. 282. Cao J, Li X, Lu X, Zhang C, Yu H, Zhao T. Cells derived from iPSC can
262. Chesney J, Puzanov I, Collichio F, et al. Randomized, open-label be immunogenic - yes or no? Protein Cell. 2014;5(1):1-3.
phase II study evaluating the efficacy and safety of talimogene laher- 283. Zhao T, Zhang ZN, Rong Z, Xu Y. Immunogenicity of induced pluripo-
parepvec in combination with ipilimumab versus ipilimumab alone tent stem cells. Nature. 2011;474(7350):212-215.
in patients with advanced, unresectable melanoma. J Clin Oncol. 284. Bock C, Kiskinis E, Verstappen G, et al. Reference Maps of human
2018;36(17):1658-1667. ES and iPS cell variation enable high-throughput characterization of
263. Ribas A, Dummer R, Puzanov I, et al. Oncolytic virotherapy promotes pluripotent cell lines. Cell. 2011;144(3):439-52.
intratumoral T cell infiltration and improves anti-PD-1 immunother- 285. Mallon BS, Chenoweth JG, Johnson KR, et al. StemCellDB: the human
apy. Cell. 2017;170(6):1109-1119 e10. pluripotent stem cell database at the National Institutes of Health.
264. Desjardins A, Gromeier M, Herndon JE, et al. Recurrent glioblastoma Stem Cell Res. 2013;10(1):57-66.
treated with recombinant poliovirus. N Engl J Med. 2018;379(2):150- 286. Mallon BS, Hamilton RS, Kozhich OA, et al. Comparison of the molec-
161. ular profiles of human embryonic and induced pluripotent stem cells
265. Brown MC, Holl EK, Boczkowski D, et al. Cancer immunother- of isogenic origin. Stem Cell Res. 2014;12(2):376-386.
apy with recombinant poliovirus induces IFN-dominant activation 287. Soldner F, Hockemeyer D, Beard C, et al. Parkinson’s disease patient-
of dendritic cells and tumor antigen-specific CTLs. Sci Transl Med. derived induced pluripotent stem cells free of viral reprogramming
2017;9(408):eaan4220. factors. Cell. 2009;136(5):964-977.
266. Samson A, Scott KJ, Taggart D, et al. Intravenous delivery of oncolytic 288. Kooreman NG, Kim Y, de Almeida PE, et al. Autologous iPSC-
reovirus to brain tumor patients immunologically primes for subse- based vaccines elicit anti-tumor responses in vivo. Cell Stem Cell.
quent checkpoint blockade. Sci Transl Med. 2018;10(422):eaam7577. 2018;22(4):501-513 e7.
267. Bourgeois-Daigneault MC, Roy DG, Aitken AS, et al. Neoadju- 289. Klebanoff CA, Wolchok JD. Shared cancer neoantigens: making pri-
vant oncolytic virotherapy before surgery sensitizes triple-negative vate matters public. J Exp Med. 2018;215(1):5-7.
breast cancer to immune checkpoint therapy. Sci Transl Med. 290. Castle JC, Kreiter S, Diekmann J, et al. Exploiting the mutanome for
2018;10(422):eaao1641. tumor vaccination. Cancer Res. 2012;72(5):1081-1091.
268. Lawler SE, Speranza MC, Cho CF, Chiocca EA. Oncolytic viruses in 291. Kreiter S, Vormehr M, van de Roemer N, et al. Mutant MHC class
cancer treatment: a review. JAMA Oncol. 2017;3(6):841-849. II epitopes drive therapeutic immune responses to cancer. Nature.
269. Pol J, Bloy N, Obrist F, et al. Trial watch: oncolytic viruses for cancer 2015;520(7549):692-696.
therapy. Oncoimmunology. 2014;3:e28694. 292. Matsushita H, Vesely MD, Koboldt DC, et al. Cancer exome analy-
270. Hoption Cann SA, van Netten JP, van Netten C. Dr William Coley and sis reveals a T-cell-dependent mechanism of cancer immunoediting.
tumour regression: a place in history or in the future. Postgrad Med J. Nature. 2012;482(7385):400-404.
2003;79(938):672-680. 293. Gubin MM, Zhang X, Schuster H, et al. Checkpoint blockade can-
271. Richardson MA, Ramirez T, Russell NC, Moye LA. Coley toxins cer immunotherapy targets tumour-specific mutant antigens. Nature.
immunotherapy: a retrospective review. Altern Ther Health Med. 2014;515(7528):577-581.
1999;5(3):42-47. 294. Yadav M, Jhunjhunwala S, Phung QT, et al. Predicting immuno-
272. Felgner S, Pawar V, Kocijancic D, Erhardt M, Weiss S. Tumour- genic tumour mutations by combining mass spectrometry and exome
targeting bacteria-based cancer therapies for increased specificity sequencing. Nature. 2014;515(7528):572-576.
and improved outcome. Microb Biotechnol. 2017;10(5):1074-1078. 295. Duan F, Duitama J, Al Seesi S, et al. Genomic and bioinformatic profil-
273. Theys J, Lambin P. Clostridium to treat cancer: dream or reality? Ann ing of mutational neoepitopes reveals new rules to predict anticancer
Transl Med. 2015;3(Suppl 1):S21. immunogenicity. J Exp Med. 2014;211(11):2231-2248.
274. Zhang YL, Lu R, Chang ZS, et al. Clostridium sporogenes deliv- 296. Li L, Goedegebuure SP, Gillanders WE. Preclinical and clinical
ers interleukin-12 to hypoxic tumours, producing antitumour activ- development of neoantigen vaccines. Ann Oncol. 2017;28(suppl_12):
ity without significant toxicity. Lett Appl Microbiol. 2014;59(6): xii11-xii7.
580-586. 297. Tran E, Robbins PF, Rosenberg SA. ‘Final common pathway’ of human
275. Chowdhury S, Castro S, Coker C, Hinchliffe TE, Arpaia N, Danino cancer immunotherapy: targeting random somatic mutations. Nat
T. Programmable bacteria induce durable tumor regression and sys- Immunol. 2017;18(3):255-262.
temic antitumor immunity. Nat Med. 2019;25(7):1057-1063. 298. van der Burg SH, Arens R, Ossendorp F, van Hall T, Melief CJ. Vac-
276. Le DT, Brockstedt DG, Nir-Paz R, et al. A live-attenuated Listeria cines for established cancer: overcoming the challenges posed by
vaccine (ANZ-100) and a live-attenuated Listeria vaccine expressing immune evasion. Nat Rev Cancer. 2016;16(4):219-233.
mesothelin (CRS-207) for advanced cancers: phase I studies of safety 299. Fifis T, Gamvrellis A, Crimeen-Irwin B, et al. Size-dependent immuno-
and immune induction. Clin Cancer Res. 2012;18(3):858-868. genicity: therapeutic and protective properties of nano-vaccines
277. Goyvaerts C, Breckpot K. Towards a personalized iPSC-based vac- against tumors. J Immunol. 2004;173(5):3148-3154.
cine. Nat Biomed Eng. 2018;2(5):277-278. 300. Liu H, Moynihan KD, Zheng Y, et al. Structure-based program-
278. Brewer BG, Mitchell RA, Harandi A, Eaton JW. Embryonic vaccines ming of lymph-node targeting in molecular vaccines. Nature.
against cancer: an early history. Exp Mol Pathol. 2009;86(3):192-197. 2014;507(7493):519-522.
ROY ET AL . 33

301. Manolova V, Flace A, Bauer M, Schwarz K, Saudan P, Bachmann MF. placebo-controlled, phase 2b/3 trial. Lancet Oncol. 2016;17(2):212-
Nanoparticles target distinct dendritic cell populations according to 223.
their size. Eur J Immunol. 2008;38(5):1404-1413. 323. Vermaelen K. Vaccine strategies to improve anti-cancer cellular
302. Hanson MC, Crespo MP, Abraham W, et al. Nanoparticulate STING immune responses. Front Immunol. 2019;10:8.
agonists are potent lymph node-targeted vaccine adjuvants. J Clin 324. Cuadros C, Dominguez AL, Lollini PL, et al. Vaccination with den-
Invest. 2015;125(6):2532-2546. dritic cells pulsed with apoptotic tumors in combination with anti-
303. Nuhn L, Vanparijs N, De Beuckelaer A, et al. pH-degradable OX40 and anti-4-1BB monoclonal antibodies induces T cell-mediated
imidazoquinoline-ligated nanogels for lymph node-focused immune protective immunity in Her-2/neu transgenic mice. Int J Cancer.
activation. Proc Natl Acad Sci USA. 2016;113(29):8098-8103. 2005;116(6):934-943.
304. Ott PA, Hu Z, Keskin DB, et al. An immunogenic personal neoanti- 325. Ito F, Li Q, Shreiner AB, et al. Anti-CD137 monoclonal antibody
gen vaccine for patients with melanoma. Nature. 2017;547(7662): administration augments the antitumor efficacy of dendritic cell-
217-221. based vaccines. Cancer Res. 2004;64(22):8411-8419.
305. Guo Y, Lei K, Tang L. Neoantigen vaccine delivery for personalized 326. Becker JC, Andersen MH, Schrama D, Thor Straten P. Immune-
anticancer immunotherapy. Front Immunol. 2018;9:1499. suppressive properties of the tumor microenvironment. Cancer
306. Luo M, Wang H, Wang Z, et al. A STING-activating nanovaccine for Immunol Immunother. 2013;62(7):1137-1148.
cancer immunotherapy. Nat Nanotechnol. 2017;12(7):648-654. 327. Hanahan D, Coussens LM. Accessories to the crime: functions
307. Li AW, Sobral MC, Badrinath S, et al. A facile approach to enhance of cells recruited to the tumor microenvironment. Cancer Cell.
antigen response for personalized cancer vaccination. Nat Mater. 2012;21(3):309-322.
2018;17(6):528-534. 328. Kumar V, Patel S, Tcyganov E, Gabrilovich DI. The nature of myeloid-
308. Rammensee HG, Singh-Jasuja H. HLA ligandome tumor antigen derived suppressor cells in the tumor microenvironment. Trends
discovery for personalized vaccine approach. Expert Rev Vaccines. Immunol. 2016;37(3):208-220.
2013;12(10):1211-1217. 329. Stromnes IM, Brockenbrough JS, Izeradjene K, et al. Targeted deple-
309. Singh-Jasuja H, Emmerich NP, Rammensee HG. The Tubingen tion of an MDSC subset unmasks pancreatic ductal adenocarcinoma
approach: identification, selection, and validation of tumor- to adaptive immunity. Gut. 2014;63(11):1769-1781.
associated HLA peptides for cancer therapy. Cancer Immunol 330. Stromnes IM, DelGiorno KE, Greenberg PD, Hingorani SR. Stro-
Immunother. 2004;53(3):187-195. mal reengineering to treat pancreas cancer. Carcinogenesis.
310. Walter S, Weinschenk T, Stenzl A, et al. Multipeptide immune 2014;35(7):1451-1460.
response to cancer vaccine IMA901 after single-dose cyclophos- 331. Stromnes IM, Greenberg PD, Hingorani SR. Molecular path-
phamide associates with longer patient survival. Nat Med. ways: myeloid complicity in cancer. Clin Cancer Res. 2014;20(20):
2012;18(8):1254-1261. 5157-5170.
311. Bentzen AK, Marquard AM, Lyngaa R, et al. Large-scale detection of 332. Whiteside TL. The tumor microenvironment and its role in promoting
antigen-specific T cells using peptide-MHC-I multimers labeled with tumor growth. Oncogene. 2008;27(45):5904-5912.
DNA barcodes. Nat Biotechnol. 2016;34(10):1037-1045. 333. Gunderson AJ, Kaneda MM, Tsujikawa T, et al. Bruton tyrosine
312. Bentzen AK, Such L, Jensen KK, et al. T cell receptor finger- kinase-dependent immune cell cross-talk drives pancreas cancer.
printing enables in-depth characterization of the interactions gov- Cancer Discov. 2016;6(3):270-285.
erning recognition of peptide-MHC complexes. Nat Biotechnol. 334. Lee KE, Spata M, Bayne LJ, et al. Hif1a deletion reveals pro-
2018;36:1191-1196. neoplastic function of B cells in pancreatic neoplasia. Cancer Discov.
313. Zugazagoitia J, Guedes C, Ponce S, Ferrer I, Molina-Pinelo S, 2016;6(3):256-269.
Paz-Ares L. Current challenges in cancer treatment. Clin Ther. 335. Schwartz M, Zhang Y, Rosenblatt JD. B cell regulation of the anti-
2016;38(7):1551-1566. tumor response and role in carcinogenesis. J Immunother Cancer.
314. Zaretsky JM, Garcia-Diaz A, Shin DS, et al. Mutations associated with 2016;4:40.
acquired resistance to PD-1 blockade in melanoma. N Engl J Med. 336. Pylayeva-Gupta Y, Das S, Handler JS, et al. IL35-producing B cells
2016;375(9):819-829. promote the development of pancreatic neoplasia. Cancer Discov.
315. Anderson KG, Stromnes IM, Greenberg PD. Obstacles posed by the 2016;6(3):247-255.
tumor microenvironment to T cell activity: a case for synergistic ther- 337. Oberg HH, Kellner C, Peipp M, et al. Monitoring circulating gam-
apies. Cancer Cell. 2017;31(3):311-325. madelta T cells in cancer patients to optimize gammadelta T cell-
316. Blank CU, Haining WN, Held W, et al. Defining ‘T cell exhaustion’. Nat based immunotherapy. Front Immunol. 2014;5:643
Rev Immunol. 2019;19(11):665-674. 338. Wesch D, Peters C, Siegers GM. Human gamma delta T regulatory
317. Philip M, Schietinger A. Heterogeneity and fate choice: T cell exhaus- cells in cancer: fact or fiction? Front Immunol. 2014;5:598.
tion in cancer and chronic infections. Curr Opin Immunol. 2019;58: 339. Stanford JC, Young C, Hicks D, et al. Efferocytosis produces a
98-103. prometastatic landscape during postpartum mammary gland involu-
318. Wei SC, Duffy CR, Allison JP. Fundamental mechanisms of tion. J Clin Invest. 2014;124(11):4737-1452.
immune checkpoint blockade therapy. Cancer Discov. 2018;8(9): 340. Vaught DB, Cook RS. Clearance of dying cells accelerates malignancy.
1069-1086. Oncotarget. 2015;6(28):24590-24591.
319. He QF, Xu Y, Li J, Huang ZM, Li XH, Wang X. CD8+ T-cell exhaustion 341. Wesolowski R, Duggan MC, Stiff A, et al. Circulating myeloid-derived
in cancer: mechanisms and new area for cancer immunotherapy. Brief suppressor cells increase in patients undergoing neo-adjuvant
Funct Genomics. 2019;18(2):99-106. chemotherapy for breast cancer. Cancer Immunol Immunother.
320. Jiang Y, Li Y, Zhu B. T-cell exhaustion in the tumor microenvironment. 2017;66(11):1437-1447.
Cell Death Dis. 2015;6:e1792. 342. Chandra D, Quispe-Tintaya W, Jahangir A, et al. STING ligand c-di-
321. Wang JC, Xu Y, Huang ZM, Lu XJ. T cell exhaustion in cancer: GMP improves cancer vaccination against metastatic breast cancer.
mechanisms and clinical implications. J Cell Biochem. 2018;119(6): Cancer Immunol Res. 2014;2(9):901-910.
4279-4286. 343. Nicholaou T, Ebert LM, Davis ID, et al. Regulatory T-cell-mediated
322. Quoix E, Lena H, Losonczy G, et al. TG4010 immunotherapy and attenuation of T-cell responses to the NY-ESO-1 ISCOMATRIX vac-
first-line chemotherapy for advanced non-small-cell lung cancer cine in patients with advanced malignant melanoma. Clin Cancer Res.
(TIME): results from the phase 2b part of a randomised, double-blind, 2009;15(6):2166-2173.
34 ROY ET AL .

344. Jacobs C, Duewell P, Heckelsmiller K, et al. An ISCOM vaccine com- 366. Lim YW, Chen-Harris H, Mayba O, et al. Germline genetic polymor-
bined with a TLR9 agonist breaks immune evasion mediated by reg- phisms influence tumor gene expression and immune cell infiltration.
ulatory T cells in an orthotopic model of pancreatic carcinoma. Int J Proc Natl Acad Sci USA. 2018;115(50):E11701-E11710.
Cancer. 2011;128(4):897-907. 367. Tran E, Robbins PF, Lu YC, et al. T-cell transfer therapy targeting
345. Ghiringhelli F, Menard C, Puig PE, et al. Metronomic cyclophos- mutant KRAS in cancer. N Engl J Med. 2016;375(23):2255-2262.
phamide regimen selectively depletes CD4+CD25+ regulatory T 368. Juric D, Castel P, Griffith M, et al. Convergent loss of PTEN
cells and restores T and NK effector functions in end stage cancer leads to clinical resistance to a PI(3)Kalpha inhibitor. Nature.
patients. Cancer Immunol Immunother. 2007;56(5):641-648. 2015;518(7538):240-244.
346. O’Sullivan D, Sanin DE, Pearce EJ, Pearce EL. Metabolic interventions 369. Niederst MJ, Sequist LV, Poirier JT, et al. RB loss in resistant EGFR
in the immune response to cancer. Nat Rev Immunol. 2019;19(5):324- mutant lung adenocarcinomas that transform to small-cell lung can-
335. cer. Nat Commun. 2015;6:6377.
347. Porporato PE, Filigheddu N, Pedro JMB, Kroemer G, Galluzzi L. Mito- 370. Zhao J, Chen AX, Gartrell RD, et al. Immune and genomic correlates
chondrial metabolism and cancer. Cell Res. 2018;28(3):265-280. of response to anti-PD-1 immunotherapy in glioblastoma. Nat Med.
348. Gardiner CM. NK cell metabolism. J Leukoc Biol. 2019;105(6): 2019;25(3):462-469.
1235-1242. 371. Stanta G, Bonin S. Overview on clinical relevance of intra-tumor het-
349. Terren I, Orrantia A, Vitalle J, Zenarruzabeitia O, Borrego F. erogeneity. Front Med. 2018;5:85.
NK cell metabolism and tumor microenvironment. Front Immunol. 372. Meacham CE, Morrison SJ. Tumour heterogeneity and cancer cell
2019;10:2278. plasticity. Nature. 2013;501(7467):328-337.
350. Gabrilovich DI. Myeloid-derived suppressor cells. Cancer Immunol 373. Reiter JG, Baretti M, Gerold JM, et al. An analysis of genetic hetero-
Res. 2017;5(1):3-8. geneity in untreated cancers. Nat Rev Cancer. 2019;19(11):639-650.
351. Kishton RJ, Sukumar M, Restifo NP. Metabolic regulation of T 374. Jamal-Hanjani M, Quezada SA, Larkin J, Swanton C. Trans-
cell longevity and function in tumor immunotherapy. Cell Metab. lational implications of tumor heterogeneity. Clin Cancer Res.
2017;26(1):94-109. 2015;21(6):1258-1266.
352. O’Neill LA, Pearce EJ. Immunometabolism governs dendritic cell and 375. Cusnir M, Cavalcante L. Inter-tumor heterogeneity. Hum Vaccin
macrophage function. J Exp Med. 2016;213(1):15-23. Immunother. 2012;8(8):1143-1145.
353. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. 376. de Sousa VML, Carvalho L. Heterogeneity in Lung Cancer. Pathobiol-
Cell. 2011;144(5):646-674. ogy. 2018;85(1-2):96-107.
354. Cascone T, McKenzie JA, Mbofung RM, et al. Increased tumor glycol- 377. Liu J, Dang H, Wang XW. The significance of intertumor and intratu-
ysis characterizes immune resistance to adoptive T cell therapy. Cell mor heterogeneity in liver cancer. Exp Mol Med. 2018;50(1):e416.
Metab. 2018;27(5):977-87 e4. 378. Finotello F, Eduati F. Multi-omics profiling of the tumor microenvi-
355. Cao Y, Rathmell JC, Macintyre AN. Metabolic reprogramming ronment: paving the way to precision immuno-oncology. Front Oncol.
towards aerobic glycolysis correlates with greater proliferative abil- 2018;8:430.
ity and resistance to metabolic inhibition in CD8 versus CD4 T cells. 379. Janiszewska M. The microcosmos of intratumor heterogeneity: the
PLoS One. 2014;9(8):e104104. space-time of cancer evolution. Oncogene. 2019;39:2031-2039.
356. Chang CH, Pearce EL. Emerging concepts of T cell metabolism as a 380. Losic B, Craig AJ, Villacorta-Martin C, et al. Intratumoral heterogene-
target of immunotherapy. Nat Immunol. 2016;17(4):364-368. ity and clonal evolution in liver cancer. Nat Commun. 2020;11(1):291.
357. Chang CH, Qiu J, O’Sullivan D, et al. Metabolic competition in the 381. Kemper K, Krijgsman O, Cornelissen-Steijger P, et al. Intra- and inter-
tumor microenvironment is a driver of cancer progression. Cell. tumor heterogeneity in a vemurafenib-resistant melanoma patient
2015;162(6):1229-1241. and derived xenografts. EMBO Mol Med. 2015;7(9):1104-1118.
358. Ho PC, Bihuniak JD, Macintyre AN, et al. Phosphoenolpyruvate 382. Charoentong P, Finotello F, Angelova M, et al. Pan-cancer
is a metabolic checkpoint of anti-tumor T cell responses. Cell. immunogenomic analyses reveal genotype-immunophenotype
2015;162(6):1217-1228. relationships and predictors of response to checkpoint blockade. Cell
359. Jacobs SR, Herman CE, Maciver NJ, et al. Glucose uptake is limiting Rep. 2017;18(1):248-262.
in T cell activation and requires CD28-mediated Akt-dependent and 383. Chowell D, Morris LGT, Grigg CM, et al. Patient HLA class I genotype
independent pathways. J Immunol. 2008;180(7):4476-4486. influences cancer response to checkpoint blockade immunotherapy.
360. Renner K, Geiselhoringer AL, Fante M, et al. Metabolic plasticity of Science. 2018;359(6375):582-587.
human T cells: preserved cytokine production under glucose depriva- 384. Hopkins AC, Yarchoan M, Durham JN, et al. T cell receptor repertoire
tion or mitochondrial restriction, but 2-deoxy-glucose affects effec- features associated with survival in immunotherapy-treated pancre-
tor functions. Eur J Immunol. 2015;45(9):2504-2516. atic ductal adenocarcinoma. JCI Insight. 2018;3(13):e122092.
361. Ota Y, Ishihara S, Otani K, et al. Effect of nutrient starvation on prolif- 385. Thorsson V, Gibbs DL, Brown SD, et al. The immune landscape of can-
eration and cytokine secretion of peripheral blood lymphocytes. Mol cer. Immunity. 2018;48(4):812-30 e14.
Clin Oncol. 2016;4(4):607-310. 386. Wellenstein MD, de Visser KE. Cancer-cell-intrinsic mechanisms
362. Scharping NE, Delgoffe GM. Tumor microenvironment metabolism: shaping the tumor immune landscape. Immunity. 2018;48(3):
a new checkpoint for anti-tumor immunity. Vaccines. 2016;4 399-416.
(4):46. 387. Remy-Ziller C, Thioudellet C, Hortelano J, et al. Sequential adminis-
363. Pearce EL, Walsh MC, Cejas PJ, et al. Enhancing CD8 T-cell mem- tration of MVA-based vaccines and PD-1/PD-L1-blocking antibodies
ory by modulating fatty acid metabolism. Nature. 2009;460(7251): confers measurable benefits on tumor growth and survival: Preclini-
103-137. cal studies with MVA-betaGal and MVA-MUC1 (TG4010) in a murine
364. Heemskerk B, Kvistborg P, Schumacher TN. The cancer antigenome. tumor model. Hum Vaccin Immunother. 2018;14(1):140-145.
EMBO J. 2013;32(2):194-203. 388. McNeel DG, Eickhoff JC, Wargowski E, et al. Concurrent, but not
365. Yarchoan M, Johnson BA, 3rd, Lutz ER, Laheru DA, Jaffee EM. Tar- sequential, PD-1 blockade with a DNA vaccine elicits anti-tumor
geting neoantigens to augment antitumour immunity. Nat Rev Cancer. responses in patients with metastatic, castration-resistant prostate
2017;17(9):569 cancer. Oncotarget. 2018;9(39):25586-25596.
ROY ET AL . 35

389. Helmink BA, Khan MAW, Hermann A, Gopalakrishnan V, Wargo


JA. The microbiome, cancer, and cancer therapy. Nat Med. How to cite this article: Roy S, Sethi TK, Taylor D, Kim YJ,
2019;25(3):377-388.
Johnson DB. Breakthrough concepts in immune-oncology: Can-
390. Gopalakrishnan V, Spencer CN, Nezi L, et al. Gut microbiome mod-
ulates response to anti-PD-1 immunotherapy in melanoma patients.
cer vaccines at the bedside. J Leukoc Biol. 2020;1–35. https://
Science. 2018;359(6371):97-103. doi.org/10.1002/JLB.5BT0420-585RR
391. Routy B, Le Chatelier E, Derosa L, et al. Gut microbiome influences
efficacy of PD-1-based immunotherapy against epithelial tumors. Sci-
ence. 2018;359(6371):91-97.

You might also like