A pool of long synthetic peptides derived from HPV16 proteins induce objective partial or complete histologic regression of lesions in more than 50% of patients with high-grade vulvar (VuVIN3) and vaginal intraepithelial neoplasia (VaIN3). The intensity of T-cell response induced by the vaccine was correlated with clinical response. Clin Cancer Res; 22(10); 2317–9. ©2016 AACR.

See related article by van Poelgeest et al., p. 2342

In this issue of Clinical Cancer Research, van Poelgeest and colleagues report the clinical results of a therapeutic human papillomavirus (HPV) vaccine for the treatment of premalignant anogenital lesions (1). Currently, two prophylactic vaccines against oncogenic HPV16 and HPV18, the genotypes predominantly found in cancers, and a new vaccine directed against 9 HPV serotypes have demonstrated their efficacy in non-HPV–infected individuals. However, these vaccines failed to demonstrate any clinical activity in subjects already infected with HPV with 14 million new HPV infections each year in the world. In a subgroup of patients who do not spontaneously clear their infection, premalignant lesions will ultimately be followed by the development of invasive cancer of the cervix, anus, vagina, penis, and oropharynx. In addition, only a few countries have achieved vaccine coverage exceeding 50%. Therapeutic HPV vaccines (THV), therefore, correspond to a currently unmet medical need. van Poelgeest and colleagues report the ability of ISA 101 vaccine [13 HPV16 E6 and E7 synthetic long peptides mixed with the Montanide adjuvant] without additional treatment to induce objective partial or complete histologic regression of the lesions in more than 50% of patients with high-grade vulvar (VuVIN3) and vaginal intraepithelial neoplasia (VaIN3) with a follow-up of 12 months (1). Interestingly, 7 of 29 patients displayed complete response, which coincided with clearance of HPV16 likely to indicate complete cure. The only potential bias of this clinical trial was the absence of a control arm, but, as recalled by the authors, spontaneous regression of the lesions is observed in less than 1.5% of cases, a much lower rate than the clinical response observed after vaccination (>50%). This report therefore confirms the efficacy of ISA 101 to elicit durable complete regression in vulvar intraepithelial neoplasia (2). These results extend recent findings from a randomized control trial based on an HPV DNA vaccine, which induced regression of HPV16- or HPV18-positive CIN2/3 in 48.2% of cases compared with a spontaneous regression rate of 30% in the placebo arm (3).

A different HPV DNA vaccine administered in 9 CIN3 patients also induced complete regression of lesions and viral clearance in 7 patients (4). A recombinant Modified Vaccinia Ankara (MVA) HPV16 E6 and E7 cDNA vaccine achieved cytologic clearance of CIN2/3 lesions and clearance of HPV16 virus in previously infected tissue in 7 of 10 patients (5).

The first administration of THV was 1996 and, after many clinical failures, encouraging clinical results have now been observed in recent clinical trials. Many THV formats have been developed and the most advanced formats include long synthetic peptides, DNA vaccine, recombinant E6/E7 protein fused to Bordetella pertussis cyclase A, HSP65, B subunit of Shiga toxin, Pseudomonas aeruginosa exotoxin, as well as attenuated bacteria (Salmonella, Listeria monocytogenes) or viral vectors (alphavirus replicon particles, adenovirus, MVA; ref. 5). Most of these vaccines were designed to target the vaccine in dendritic cells and/or promote cross-presentation of HPV antigen to CD8+ T cells. The vaccine itself did not appear to be the critical element to explain the differential clinical activity of THV, as the clinical impact of HPV vaccines was only observed in patients with premalignant HPV-associated lesions, whereas none of the vaccines displayed any clinical activity in established cancer. In addition, the same vaccines (long synthetic peptide and MVA) which failed in HPV-associated cancer-induced significant clinical responses in premalignant lesions (5). It has been hypothesized that higher levels of immunosuppression and immune effector-evading mechanisms of tumor cells may explain the resistance of cancer to HPV vaccines.

In contrast to prophylactic vaccines, based on a humoral response, preclinical models support the main role of cytotoxic CD8+ T cells for THV. van Poelgeest and colleagues clearly demonstrated a positive correlation between the clinical effects of the HPV vaccine and the strength of the induction of IFNγ- and TNFα-producing T cells in the blood of vaccinated patients (1). Surprisingly, they did not observe a correlation between the ex vivo detectable HPV16-specific CD8+ T-cell response and clinical outcome. This absence of correlation of CD8+ T-cell response against HPV in the blood and clinical response has already been reported (5). It may mean that both CD4+ and CD8+ T-cell responses are required to achieve regression of the lesions. Other studies strongly support that intratumoral CD8+ T-cell infiltration is correlated with spontaneous regression of HPV lesions and the efficacy of the cancer vaccine. CIN2/3 lesions with CD8+ T-cell infiltrates in the dysplastic epithelium are more prone to clinical regression. In murine models and in an approved prostate cancer vaccine, only the induction of intratumoral CD8+ T cells was correlated with vaccine efficacy and not with CD8+ T-cell counts in blood (6, 7).

Results from the van der Brug group in this current issue of Clinical Cancer Research and from the Trimble group convincingly demonstrate the clinical efficacy of THV (1, 3). However, only a complete response would prevent surgical mutilation and such a response is observed in only about one-quarter of patients exhibiting vulvar/genital CIN3 lesions in the van Poelgeest report. These results may not be sufficient enough to modify clinical practice, except if predictive factors of response can be identified to allow better selection of patients likely to benefit from the vaccine. Prior to clinical implementation of these vaccines, strategies need to be developed to improve their clinical efficacy (Fig. 1). Cutaneous administration of imiquimod by van Poelgeest and colleagues did not increase the T-cell immune response elicited by the vaccine, but its impact on clinical response was not described (1). In murine models, peripheral vaccination with HPV16 E7 long peptides, followed by intravaginal administration of imiquimod, induced a 5-fold increase in the infiltration of vaccine-induced CD8+ T cells in the vaginal mucosa (8). The Wu group reported that cancer vaccine boosts via the cervicovaginal rather than the intramuscular route of immunization appears to be crucial to induce genital CD8+ T cells and tumor regression (9). In humans, a DNA HPV vaccine administered by cervical intralesional injections induced more intense recruitment of intraepithelial CD8+ T-cell infiltrates than the other routes of administration (10). These examples emphasize the critical role of mucosal administration of adjuvant or vaccine to specifically recruit resident memory T cells at the mucosal site of the lesions (11).

Figure 1.

Strategies to improve therapeutic HPV vaccine against HPV-associated genital lesions. A, the first approach is designed to block immunosuppression in the tumor microenvironment. Off-target effects of low-dose irradiation, chemotherapy, or VEGF blockade may be used to inhibit various inhibitory pathways in the tumor microenvironment. Combination of HPV vaccines with antagonist antibodies against inhibitory receptors (e.g., CTLA-4, PD-1, Tim-3) or agonist antibodies against costimulatory molecules (e.g., OX40 and CD137), although the optimal combination and the clinical sequence of administration need to be defined. B, the objective of the second approach is to promote homing of anti-HPV T cells in HPV-associated lesions. To simultaneously treat lesions of the skin and of mucosal epithelium (vagina, cervix), a primeboost strategy could be based on systemic (intramuscular, subcutaneous) or mucosal administration (cervicovaginal or nasal). MDSC, myeloid-derived suppressive cell; Treg, regulatory T cell.

Figure 1.

Strategies to improve therapeutic HPV vaccine against HPV-associated genital lesions. A, the first approach is designed to block immunosuppression in the tumor microenvironment. Off-target effects of low-dose irradiation, chemotherapy, or VEGF blockade may be used to inhibit various inhibitory pathways in the tumor microenvironment. Combination of HPV vaccines with antagonist antibodies against inhibitory receptors (e.g., CTLA-4, PD-1, Tim-3) or agonist antibodies against costimulatory molecules (e.g., OX40 and CD137), although the optimal combination and the clinical sequence of administration need to be defined. B, the objective of the second approach is to promote homing of anti-HPV T cells in HPV-associated lesions. To simultaneously treat lesions of the skin and of mucosal epithelium (vagina, cervix), a primeboost strategy could be based on systemic (intramuscular, subcutaneous) or mucosal administration (cervicovaginal or nasal). MDSC, myeloid-derived suppressive cell; Treg, regulatory T cell.

Close modal

Strategies designed to counteract immunosuppressive mechanisms such as regulatory T cells, myeloid-derived suppressive cells or checkpoint inhibitors expressed by T cells in the tumor microenvironment may overcome clinical resistance to THV. HPV-positive tumors often present upregulated levels of PD-1 ligand on their surface and decreased levels of costimulatory molecules such as OX40 and 4-1BB on T cells. In preclinical models, blockade of these cells or molecules synergized with HPV vaccine, but it was not always the same antagonist or agonist of costimulatory molecules that optimally synergized with selected HPV vaccines (12–14). Several clinical trials are currently examining the effects of checkpoint inhibitors such as anti–CTLA-4 and/or anti–PD-1/PD-L1 in advanced stage HPV-associated disease (NCT01711515, NCT01693783, NCT01975831). Second-generation THV integrating the requirement of mucosal route of immunization to elicit protective resident memory T cells and combined with an antagonist of costimulatory inhibitory molecules or an agonist of positive cosignaling pathways may finally lead to the clinical success of THV after a long, winding road.

No potential conflicts of interest were disclosed.

Conception and design: S. Karaki, E. Tartour

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): S. Karaki, C. Badoual, E. Tartour

Writing, review, and/or revision of the manuscript: S. Karaki, H. Pere, C. Badoual, E. Tartour

Study supervision: S. Karaki, E. Tartour

E. Tartour was supported by grants from Institut National du Cancer (INCA), Ligue Contre le Cancer, Université Sorbonne Paris Cité, LabEx Immuno-Oncology, Site Intégré de Recherche en cancerologie (SIRIC CARPEM), and Agence Nationale de la Recherche (ANR).

1.
van Poelgeest
MIE
,
Welters
MJP
,
Vermeij
R
,
Stynenbosch
LFM
,
Loof
NM
,
Berends-van der Meer
DMA
, et al
Vaccination against oncoproteins of HPV16 for noninvasive vulvar/vaginal lesions: lesion clearance is related to the strength of the T-cell response
.
Clin Cancer Res
2016
;
22
:
2342
50
.
2.
Kenter
GG
,
Welters
MJ
,
Valentijn
AR
,
Lowik
MJ
,
Berends-van der Meer
DM
,
Vloon
AP
, et al
Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia
.
N Engl J Med
2009
;
361
:
1838
47
.
3.
Trimble
CL
,
Morrow
MP
,
Kraynyak
KA
,
Shen
X
,
Dallas
M
,
Yan
J
, et al
Safety, efficacy, and immunogenicity of VGX-3100, a therapeutic synthetic DNA vaccine targeting human papillomavirus 16 and 18 E6 and E7 proteins for cervical intraepithelial neoplasia 2/3: a randomised, double-blind, placebo-controlled phase 2b trial
.
Lancet
2015
;
386
:
2078
88
.
4.
Kim
TJ
,
Jin
HT
,
Hur
SY
,
Yang
HG
,
Seo
YB
,
Hong
SR
, et al
Clearance of persistent HPV infection and cervical lesion by therapeutic DNA vaccine in CIN3 patients
.
Nat Commun
2014
;
5
:
5317
.
5.
Skeate
JG
,
Woodham
AW
,
Einstein
MH
,
Da Silva
DM
,
Kast
WM
. 
Current therapeutic vaccination and immunotherapy strategies for HPV-related diseases
.
Hum Vaccin Immunother.
2016 Feb 2
.
[Epub ahead of print]
.
6.
Sandoval
F
,
Terme
M
,
Nizard
M
,
Badoual
C
,
Bureau
MF
,
Freyburger
L
, et al
Mucosal imprinting of vaccine-induced CD8+ T cells is crucial to inhibit the growth of mucosal tumors
.
Sci Transl Med
2013
;
5
:
172ra20
.
7.
Fong
L
,
Carroll
P
,
Weinberg
V
,
Chan
S
,
Lewis
J
,
Corman
J
, et al
Activated lymphocyte recruitment into the tumor microenvironment following preoperative sipuleucel-T for localized prostate cancer
.
J Natl Cancer Inst
2014
;
106
:
dju268
.
8.
Domingos-Pereira
S
,
Decrausaz
L
,
Derre
L
,
Bobst
M
,
Romero
P
,
Schiller
JT
, et al
Intravaginal TLR agonists increase local vaccine-specific CD8 T cells and human papillomavirus-associated genital-tumor regression in mice
.
Mucosal Immunol
2013
;
6
:
393
404
.
9.
Sun
YY
,
Peng
S
,
Han
L
,
Qiu
J
,
Song
L
,
Tsai
Y
, et al
Local HPV recombinant vaccinia boost following priming with an HPV DNA vaccine enhances local HPV-specific CD8+ T-cell-mediated tumor control in the genital tract
.
Clin Cancer Res
2016
;
22
:
657
69
.
10.
Alvarez
RD
,
Huh
WK
,
Bae
S
,
Lamb
LS
 Jr
,
Conner
MG
,
Boyer
J
, et al
A pilot study of pNGVL4a-CRT/E7(detox) for the treatment of patients with HPV16+ cervical intraepithelial neoplasia 2/3 (CIN2/3)
.
Gynecol Oncol
2016
;
140
:
245
52
.
11.
Nizard
M
,
Roussel
H
,
Tartour
E
. 
Resident memory T cells as surrogate markers of the efficacy of cancer vaccines
.
Clin Cancer Res
2016
;
22
:
530
2
.
12.
Badoual
C
,
Hans
S
,
Merillon
N
,
Van Ryswick
C
,
Ravel
P
,
Benhamouda
N
, et al
PD-1-expressing tumor-infiltrating T cells are a favorable prognostic biomarker in HPV-associated head and neck cancer
.
Cancer Res
2013
;
73
:
128
38
.
13.
Bartkowiak
T
,
Singh
S
,
Yang
G
,
Galvan
G
,
Haria
D
,
Ai
M
, et al
Unique potential of 4-1BB agonist antibody to promote durable regression of HPV+ tumors when combined with an E6/E7 peptide vaccine
.
Proc Natl Acad Sci U S A
2015
;
112
:
E5290
9
.
14.
Liu
Z
,
Zhou
H
,
Wang
W
,
Fu
Y-X
,
Zhu
M
. 
A novel dendritic cell targeting HPV16 E7 synthetic vaccine in combination with PD-L1 blockade elicits therapeutic antitumor immunity in mice
.
Oncoimmunology
.
In press
.