Skip to main content
JAMA Network logoLink to JAMA Network
. 2018 Sep 27;5(1):67–73. doi: 10.1001/jamaoncol.2018.4051

Combining Immune Checkpoint Blockade and Tumor-Specific Vaccine for Patients With Incurable Human Papillomavirus 16–Related Cancer

A Phase 2 Clinical Trial

Erminia Massarelli 1, William William 2, Faye Johnson 2, Merrill Kies 2, Renata Ferrarotto 2, Ming Guo 3, Lei Feng 4, J Jack Lee 4, Hai Tran 2, Young Uk Kim 5, Cara Haymaker 6, Chantale Bernatchez 5, Michael Curran 7, Tomas Zecchini Barrese 6, Jaime Rodriguez Canales 6, Ignacio Wistuba 6, Lerong Li 8, Jing Wang 8, Sjoerd H van der Burg 9, Cornelis J Melief 10,11, Bonnie Glisson 2,
PMCID: PMC6439768  PMID: 30267032

Abstract

Importance

In recurrent human papilloma virus (HPV)–driven cancer, immune checkpoint blockade with anti–programmed cell death 1 (PD-1) antibodies produces tumor regression in only a minority of patients. Therapeutic HPV vaccines have produced strong immune responses to HPV-16, but vaccination alone has been ineffective for invasive cancer.

Objective

To determine whether the efficacy of nivolumab, an anti–PD-1 immune checkpoint antibody, is amplified through treatment with ISA 101, a synthetic long-peptide HPV-16 vaccine inducing HPV-specific T cells, in patients with incurable HPV-16–positive cancer.

Design, Setting, and Participants

In this single-arm, single-center phase 2 clinical trial, 24 patients with incurable HPV-16–positive cancer were enrolled from December 23, 2015, to December 12, 2016. Duration of follow-up for censored patients was 12.2 months through August 31, 2017.

Interventions

The vaccine ISA101, 100 μg/peptide, was given subcutaneously on days 1, 22, and 50. Nivolumab, 3 mg/kg, was given intravenously every 2 weeks beginning day 8 for up to 1 year.

Main Outcomes and Measures

Assessment of efficacy reflected in the overall response rate (per Response Evaluation Criteria in Solid Tumors, version 1.1).

Results

Of the 24 patients (4 women and 20 men; 22 with oropharyngeal cancer; median age, 60 years [range, 36-73 years]), the overall response rate was 33% (8 patients; 90% CI, 19%-50%). Median duration of response was 10.3 months (95% CI, 10.3 months to inestimable). Five of 8 patients remain in response. Median progression-free survival was 2.7 months (95% CI, 2.5-9.4 months). Median overall survival was 17.5 months (95% CI, 17.5 months to inestimable). Grades 3 to 4 toxicity occurred in 2 patients (asymptomatic grade 3 transaminase level elevation in 1 patient and grade 4 lipase elevation in 1 patient), requiring discontinuation of nivolumab therapy.

Conclusions and Relevance

The overall response rate of 33% and median overall survival of 17.5 months is promising compared with PD-1 inhibition alone in similar patients. A randomized clinical trial to confirm the contribution of HPV-16 vaccination to tumoricidal effects of PD-1 inhibition is warranted for further study.

Trial Registration

ClinicalTrials.gov identifier: NCT02426892


This phase 2 clinical trial examines whether the efficacy of nivolumab is amplified through treatment with ISA 101, a synthetic long-peptide human papillomavirus 16 vaccine, in patients with incurable human papillomavirus 16–positive cancer.

Key Points

Question

Is the efficacy of programmed cell death 1 immune checkpoint inhibition increased by a tumor-specific vaccine in patients with incurable human papillomavirus 16–positive cancer?

Findings

In this phase 2 clinical trial of nivolumab and human papillomavirus 16 vaccine ISA101, the primary end point was met, with a 33% overall response rate (8 of 24 patients), compared with response rates of 16% to 22% with programmed cell death 1 inhibitors alone in similar patients. Survival data were also encouraging, with a median survival of 17.5 months.

Meaning

These data indicate that HPV-16 vaccination may augment the efficacy of programmed cell death 1 checkpoint inhibition and merit confirmation in a randomized trial.

Introduction

Human papillomavirus (HPV) is the cause of nearly all cervical cancers and most oropharyngeal, anal, penile, vulvar, and vaginal cancers. Although many cancers are cured with initial treatment, recurrent cancer is frequently incurable and associated with relatively short survival. The E6 and E7 viral proteins, critical in driving HPV oncogenesis and foreign to the human immune system, represent ideal targets for therapeutic cancer vaccination. Recent data indicate that, at initial diagnosis, most patients with HPV-positive oropharyngeal cancer (OPC) exhibit a strong spontaneous immune response to HPV antigens that is associated with substantial infiltration of the cancer with HPV-specific T cells and an excellent prognosis1. However, in recurrent HPV-positive OPC, immune checkpoint blockade with anti–programmed cell death 1 (PD-1) antibodies pembrolizumab and nivolumab produces tumor regression in only a minority of patients.2,3,4,5 Thus, we hypothesized that augmentation of the HPV-specific T-cell population by a therapeutic vaccine could increase the proportion of patients benefiting from anti–PD-1 therapy.

The vaccine ISA101, which is among the most promising vaccines targeted to E6 and E7, consists of 9 overlapping long E6 peptides (five 32-mer E6 peptides and four 25-mer E6 peptides) and 4 overlapping 35-mer E7 peptides (synthetic long peptide HPV-16 vaccine), covering the complete sequence of the HPV-16 E6 and E7 oncoproteins.6 These long peptides effectively deliver antigens to dendritic cells, which then induce CD4+ and CD8+ T-cell responses by HLA classes I and II presentation of the HPV-16 E6 and E7 processed epitope peptides.7,8

In a landmark clinical trial, ISA101 demonstrated notable activity in high-grade vulvar intraepithelial neoplasia, with durable and complete remission in 9 of 19 patients at 2 years.6 Furthermore, clinical responses were directly correlated with vaccine-activated T-cell immune responses against HPV-16. Recently, these results were confirmed with the additional observation that patients with a complete histologic response had also cleared the virus from these sites.9 However, by itself the vaccine did not affect regression of advanced cervical cancer, suggesting that vaccine-activated T cells are held in check by a tumor-induced immunosuppressive environment.10,11 Thus, there should be potential to enable the cytotoxic effects of vaccine-activated T cells by inhibiting mechanisms of immunosuppression.12,13

Human papillomavirus DNA vaccines targeting E6 and E7, such as VGX-3100 and GX-188E, have also been shown to induce potent HPV-specific CD4+ and CD8+ T-cell responses and regression of high-grade premalignant cervical lesions.14,15,16 As with ISA101, the activity of VGX-3100 and GX-188E is limited to premalignant lesions. However, ISA101 is distinguished by the ability to induce HPV-specific T-cell responses in patients with endstage cervical cancer.10,17

We report the results of a single-arm phase 2 clinical trial designed to evaluate the efficacy of ISA101 combined with PD-1 immune checkpoint blockade in patients with incurable HPV-16–positive malignant neoplasms.

Methods

Patients

Patients must have had histologically or cytologically documented diagnosis of incurable HPV-16–positive solid tumors from oropharyngeal, cervical, vulvar, vaginal, penile, or anal primaries with 0 or 1 line of treatment for recurrent HPV-16–positive cancer. Patients were required to be age 18 years or older, to have an Eastern Cooperative Oncology Group performance status of 0 or 1, and to have measurable disease per Response Evaluation Criteria in Solid Tumors, version 1.1 (RECIST, version 1.1).18 Consent for a baseline biopsy was required. Major exclusion criteria included active central nervous system metastases and active autoimmune disease, except vitiligo, type 1 diabetes, and hypothyroidism. The study was conducted in accordance with the Declaration of Helsinki19 and the International Conference on Harmonization Good Clinical Practice Guidelines.20 The study protocol was approved by the University of Texas MD Anderson Cancer Center Institutional Review Board (trial protocol in Supplement 1). All patients provided informed written consent.

Study Design

This was a single-arm nonrandomized phase 2 clinical trial. The primary objective was assessment of efficacy as reflected in the overall response rate (ORR) per RECIST, version 1.1.18 Secondary objectives were assessment of safety, tolerability, HPV-specific immune response, and estimation of progression- free survival (PFS) and overall survival (OS). Exploratory biomarker analyses included the correlation of programmed death-ligand 1 (PD-L1) expression with response and survival. Patients were treated with ISA101, 100 μg/peptide in Montanide adjuvant (Seppic) subcutaneously, for a total of 3 doses on days 1, 22, and 50 and nivolumab, 3 mg/kg intravenously, starting on day 8 adminstered every 2 weeks for a total of 12 months or progression of disease, toxic effects, or withdrawal of consent. Imaging was performed at baseline, prior to cycle 6 of nivolumab, and then every 6 weeks thereafter. Tumor biopsies were mandatory before treatment and planned at the time of first restaging. Blood samples were drawn at baseline, before the second and third dose of vaccine, prior to cycle 5 and 6 of nivolumab, and then every 3 months.

Statistical Analysis

Efficacy and safety were evaluated in all patients who received at least 1 dose of ISA101 and nivolumab. The 2-stage MiniMax design by Simon21 was used, targeting an alternative hypothesis response rate of 0.3 vs a null hypothesis response rate of 0.10 with 80% power and a 1-sided .03 significance level. A response was defined as ORR (complete response plus partial response), per RECIST, version 1.1.18 This design required 2 or more responses in 15 patients in the first stage to accrue 10 additional patients in the second stage. A total of 6 or more responses in 25 patients was required to reject the null hypothesis. The ORR was based on investigators’ assessment. Independent blinded radiology review was not performed.

Toxic effects were monitored continuously in cohorts of 5 patients and graded according to National Cancer Institute Common Terminology Criteria for Adverse Events, version 4.03.22 Duration of response, PFS, and OS were estimated with Kaplan-Meier statistics. Duration of response was calculated from time of first response to progression or death, whichever occurred first. Progression-free survival was calculated from treatment start date to progression date or death date, whichever was earlier. Overall survival was calculated from treatment start date to death date or to the last follow-up date. Patients who had not reached a time-to-event end point were censored at the last follow-up date.

The nonparametric Wilcoxon rank sum test was used to evaluate correlation between PD-L1 tumor, immune, and combined scores with ORR. The log-rank test was used to evaluate correlations of PD-L1 scores with PFS and OS. P < .05 was considered statistically significant. The Welch 2-sample t test was used to evaluate enzyme-linked immunospot (ELISPOT) values at baseline and after vaccination. The analyses were performed using R, version 3.3.2, a publicly available statistical tool (https://www.r-project.org/).

HPV Genotype

Archival tumors were required to harbor HPV-16 in order to be eligible for this clinical trial. The Cervista HPV-16/18 assay was conducted according to previously published methods.23 Its use has been validated in formalin-fixed tissue and in HPV-related oropharyngeal cancer.24

Immunohistochemistry

Tumor sections from formalin-fixed, paraffin-embedded core needle biopsies were used for immunohistochemical analysis as previously described.25 Hematoxylin-eosin–stained sections were first assessed visually for evaluable viable tumor cells. An automated staining system (BOND-MAX; Leica Microsystems) with antibodies against PD-L1 (Clone 28-8, dilution1:400; Abcam, cat#ab205921) was used. Programmed death-ligand 1 was detected using a Novocastra Bond Polymer Refine Detection kit (Leica Microsystems). Immunohistochemistry slides were digitally scanned at ×200 using a ScanScope Aperio AT Turbo slide scanner (Leica Microsystems) and visualized using the ImageScope software program (Leica Microsystems). Visual scoring was also performed. Each individual core needle biopsy was divided by a pathologist (J.R.C.) into tumor cell and tumor stroma compartments using the Aperio tool box (Leica Microsystems). The tumor cell compartment was defined as groups or nests of malignant cells and the tumor stroma compartment was represented by the fibrous tissue present between tumor cells. Programmed death-ligand 1 expression was scored considering a partial or complete membranous staining at any intensity, on a scale from 0% to 100%, of malignant cells or tumor-associated immune cells, respectively.

Interferon-γ ELISPOT Assay

Interferon-γ ELISPOT assay was performed as described previously with modifications.9 Peripheral blood mononuclear cells were thawed, plated at 0.5 to 1 × 106 cells/well, and stimulated for 4 days with a pool of 9 HPV-16 E6 synthetic long peptides and 4 HPV-16 E7 synthetic long peptides. After 4 days, cells were harvested, counted, and plated at a concentration of 50 000 or 100 000 cells/well in 200 μL of complete tumor-infiltrating lymphocyte culture media in anti–IFN-γ (5 μg/mL; Mabtech, catalog 3420-3)–coated ELISPOT plates (Millipore; catalog MAHAS4510). After overnight incubation at 37°C, plates were washed with phosphate buffered saline with 0.05% of polysorbate 20 (Invitrogen) and incubated for 1 hour at 37°C with 1 μg/mL of biotin-labeled anti–IFN-γ antibody (Mabtech, catalog 3420-6). Plates were then incubated with diluted extravidin-alkaline phosphatase (1:5000 dilution), for 1 hour at room temperature. Spots were immediately developed by 5-bromo-4-chloro-3-indolyl-phosphate in conjunction with nitro blue tetrazolium and counted on an ImmunoSpot ELISPOT reader (CTL Immunospot Reader, software version 6.0.0.0). The assay was conducted in triplicates. Phorbol 12-myristate 13-acetate and ionomycin were used as positive controls and media alone as negative controls. Values were normalized for reactivity in the negative control. ISA101 was supplied by ISA Pharmaceuticals and nivolumab by Bristol-Myers Squibb.

Results

Patients and Treatment

From December 23, 2015, to December 12, 2016, 34 patients were screened, and 24 patients (22 with oropharyngeal cancer, 1 with anal cancer, and 1 with cervical cancer) were enrolled. Enrollment closed prior to accrual of the 25th patient owing to impending expiration of the ISA101 vaccine lot. Patient characteristics are summarized in Table 1. A flowchart of the patients is provided in Figure 1.

Table 1. Characteristics of the 24 Study Patients.

Characteristic Patients, No. (%)
Age, median (range), y 60 (36-73)
Male sex 20 (83)
Race/ethnicity
White 21 (88)
Black 2 (8)
Hispanic 1 (4)
ECOG performance status
0 12 (50)
1 12 (50)
Primary site
Oropharynx 22 (92)
Cervix 1 (4)
Anus 1 (4)
Platin status
Exposed 23 (96)
Resistant, 6 mo 19 (79)
Treatment setting
First line 10 (42)
Second line 14 (58)
Baseline PD-L1 expression
Evaluability
Tumor 18 (75)
Immune 20 (83)
≥1%, No./total No.
Tumor 7/18 (39)
Tumor and immune 13/18 (72)

Abbreviations: ECOG, Eastern Cooperative Oncology Group; PD-L1, programmed death-ligand 1.

Figure 1. Patient Flowchart.

Figure 1.

Efficacy

Response data are detailed in Table 2. Best percentage change in target lesions (per RECIST, version 1.1) from baseline and duration of response are depicted in Figure 2 and eFigure 1 in Supplement 2. There were 4 responders in the first 15 patients, directing accrual to the second stage. Of the total 24 patients accrued, 8 patients, all with OPC, achieved a response: 2 complete responses and 6 partial responses, for an ORR of 33% (90% CI, 19%-50%).26 Three patients achieved their best overall response of partial response subsequent to the first restaging at 11 weeks. To provide a perspective in comparison with monotherapy effects of PD-1 inhibition, response data for patients with OPC and subsets of that group are presented in Table 2, including patients refractory to platins and cetuximab (progression within 6 months of treatment) and for patients treated second-line for recurrence. The ORR in these admittedly small subsets confirms efficacy similar to the less heavily treated overall population. Median duration of response was 10.3 months (95% CI, 10.3 months to inestimable), with 5 of 8 responses ongoing at the time of analysis (August 25, 2017).

Table 2. Response Overall, in Oropharyngeal Cancer, and by Treatment Historya.

Response per RECIST, version 1.1 No. (%)
All Patients (N = 24) Patients With Oropharyngeal Cancer
All (n = 22) Platin-Refractory Disease (n = 17) Cetuximab-Refractory Disease (n = 8) Platin- and Cetuximab-Refractory Disease (n = 6) Received Second-line Treatment (n = 12)
Overall response rate 8 (33) 8 (36) 6 (35) 5 (63) 3 (50) 5 (42)
Complete response 2 (8) 2 (9) 2 (12) 1 (13) 0 2 (17)
Partial response 6 (25) 6 (27) 4 (24) 4 (50) 3 (50) 3 (25)
Stable disease 3 (13) 2 (9) 1 (6) 0 1 (17) 1 (8)
Disease control rateb 11 (46) 10 (45) 7 (41) 5 (63) 4 (67) 6 (50)
Progression of disease 13 (54) 12 (55) 10 (59) 3 (38) 2 (33) 6 (50)

Abbreviation: RECIST, Response Evaluation Criteria in Solid Tumors.

a

Refractory indicates progression of disease within 6 months of treatment.

b

Disease control rate is calculated by adding the percentages of patients whose tumors exhibited complete response, partial response, and stable disease and indicates lack of progression per RECIST.

Figure 2. Efficacy of ISA101 and Nivolumab.

Figure 2.

A, Best percentage change in sum of target lesions from baseline and postbaseline assessments per Response Evaluation Criteria in Solid Tumors, version 1.1 (blue horizontal line). The orange horizontal line indicates a 20% increase, considered progressive disease per Response Evaluation Criteria in Solid Tumors, version 1.1. B, Duration of response. OPC indicates oropharyngeal cancer.

Median PFS was 2.7 months (95% CI, 2.5-9.4 months) and median OS was 17.5 months (95% CI, 17.5 to inestimable), with median follow-up time among censored patients of 12.2 months (eFigures 2 and 3 in Supplement 2). The rate of PFS at 6 months was 37% (95% CI, 22%-63%) and at 12 months was 25% (95% CI, 12%-50%). The rate of OS at 6 months was 75% (95% CI, 59%-94%) and at 12 months was 70% (95% CI, 54%-91%). Estimates of PFS and OS in the 22 patients with OPC were identical to those in the overall population.

Safety

Treatment-related adverse events are listed in eTable 1 in Supplement 2. The toxic effects profile was additive for expected reactions to ISA101, namely, injection site reactions and fever, and those predicted from nivolumab, such as fatigue, diarrhea, and hepatoxicity. Two patients discontinued treatment owing to asymptomatic grade 3 immune adverse events (asymptomatic grade 3 transaminase level elevation in 1 patient and grade 4 lipase and amylase elevation level in 1 patient). There were no other dose-limiting toxic effects.

Efficacy by PD-L1 Status

Data on the evaluability of baseline core needle biopsies for PD-L1 expression and on the incidence of PD-L1 expression of 1% or more are provided in Table 1. The correlation of clinical response to baseline PD-L1 expression score in tumor, immune, and combined compartments is shown in eFigure 4 in Supplement 2. Distribution of PD-L1 expression, medians, and interquartile ranges are in eTable 2 in Supplement 2. In view of the bimodal, nonnormal distribution observed in both tumor and immune cells, data were subjected to the Wilcoxon rank-sum test, using a score of 1% or more as threshold for positivity. A significant correlation of PD-L1 expression with response was demonstrated for tumor score, with an ORR of 43% (3 of 7) compared with 18% (2 of 11) in PD-L1–negative tumors (P = .04). Neither the immune scores nor the combined scores were correlated with response. Furthermore, tumor score, immune score, and their combined scores for PD-L1 less than 1% vs 1% or more were not correlated with PFS or OS.

Efficacy by HPV-16–Specific Immune Response

Interferon-γ release data from cultured peripheral blood lymphocytes in response to pooled HPV-16 E6 and E7 peptides, segregated by clinical response, are shown in eFigure 5 in Supplement 2. Of those with baseline data, there was no or minimal reactivity, consistent with what was previously observed in patients with vulvar intraepithelial neoplasia or cervical cancer.6,9,11 After vaccination, a variable increased number of HPV-specific T cells was observed in both responders and nonresponders. The immune response did not correlate with any efficacy end points, suggesting that local factors in the tumor environment exert preeminent influences on vaccine effect.

Discussion

ISA101, an HPV-16 synthetic long peptide vaccine combined with nivolumab, a PD-1 immune checkpoint inhibitor, exhibited promising efficacy outcomes in patients with recurrent HPV-16–positive cancer. With a total of 24 patients treated instead of the 25 planned, the statistical power was reduced to 77.6%. Despite that, the primary end point was met, with an ORR of 33% (8 of 24 patients) and responses were durable with 63% (5 of 8 patients) ongoing. Furthermore, the 12-month OS rate of 70% and median OS of 17.5 months are encouraging for this population. The combination of ISA101 and nivolumab was very well tolerated, with only additive effects from each agent apparent without increased immune adverse events, relative to nivolumab monotherapy. The absence of synergistic toxic effects is integral to building rational combination immunotherapy on the anti–PD-1 platform, and combined with the efficacy outcomes, supports further investigation of this approach in a randomized trial. Although combining therapeutic cancer vaccination with immune checkpoint blockade is the focus of much ongoing research, our results are among the first to be reported, to our knowledge.

During the trial’s design in 2014, we used preliminary data from Keynote-012, which showed an ORR of 20% (4 of 20 patients) in patients with p16-positive OPC treated with pembrolizumab27 as a historical reference. Given the accrual of patients with predominantly OPC in our trial, the most appropriate historical references available now are the subsets with p16-positive OPC treated with nivolumab in CheckMate 141 and pembrolizumab in Keynote-012 and Keynote-055 (Table 3).2,3,4,5,28 Eligibility differed among these trials, particularly in regard to prior treatment. Nevertheless, the ORR of 36% among patients in our trial with OPC is numerically higher than that observed in the reference trials, and higher ORRs were confirmed in more comparable subsets of the patients with OPC in this trial, albeit with very small denominators. The median OS of 17.5 months (95% CI, 17.5 months to inestimable) and the 12-month OS rate of 70% are approximately double that observed for the CheckMate 141, Keynote-012, and Keynote-055 trials, from reported rates and inspection of the published survival curves.2,3,4,5 Given the lower bound on the confidence interval of 17.5 months, the survival outcome appears to more clearly distinguish our trial, compared with the ORRs from these previous trials with anti–PD-1 monotherapy. However, owing to heterogeneity in eligibility and the small number of patients, judgments from this unplanned subset analysis are appropriately limited. Clear demonstration of the contribution of therapeutic HPV vaccine to anti–PD-1 therapy awaits testing in a larger randomized trial with more homogenous eligibility.

Table 3. Efficacy of PD-L1 Inhibition in Patients With Recurrent Human Papillomavirus and OPC.

Source Treatment Eligibility RR, % (95% CI) Overall Survival, Median (95% CI), mo
Mehra et al,28 2016 (Keynote-012 study) Pembrolizumab Treated with mixed platins (n = 28) 22 (12-34) NR in p16-positive OPC
Bauml et al,4 2017 (Keynote-055 study) Pembrolizumab Platin-treated and cetuximab-resistant (n = 37) 16 (6-32) 8 (6-11)
Ferris et al,5 2016 (CheckMate 141 study) Nivolumab Platin-resistant (n = 63) 16 (NR) 9.1 (7.2-10)
Current trial Nivolumab and ISA101 Up to 1 prior regimen for recurrence (n = 22) 36 (17-59) 17.5 (17.5 to inestimable)

Abbreviations: NR, not reported; OPC, oropharyngeal cancer; PD-L1, programmed death-ligand 1; RR, response rate.

Similar to data from CheckMate 141 with nivolumab, we found that PD-L1 expression of 1% or more on tumor cells increased the chance of tumor response to 43% (3 of 7); however, response was also observed in 18% (2 of 11) of patients with PD-L1 expression less than 1%. Although these rates are higher than the 17% (PD-L1 expression ≥1%) and 12% (PD-L1 expression <1%) rates reported for the HPV-positive patients with OPC treated with nivolumab alone,5 small patient numbers and heterogeneity limit firm conclusions. It can be speculated, however, that the outcome of vaccination on tumor regression in the setting of PD-1 inhibition is predominantly in the PD-L1–positive subset for whom ORR was more than doubled compared with nivolumab alone (43% vs 18%). Neither PD-L1 expression in stromal inflammatory cells nor the combined score with tumor and immune cell expression was associated with response, discordant with what has been reported with both nivolumab and pembrolizumab, for which the combined tumor plus immune cell scores provided an increased association with response vs tumor score alone.3,29 The method we used differed from those analyses in many ways, including that immune cells were scored only in peritumoral stroma and not in the intratumoral region in our analysis. Scoring of the immune cells, whether intratumoral or stromal, for PD-L1 expression is highly discordant among observers, and has not been validated.30 The bimodal distribution of PD-L1 expression we observed is also notable. Relevant previous trials with PD-1 inhibition alone have not displayed PD-L1 expression data graphically, although tabular data suggest that PD-L1 expression represents a continuous, more normally distributed variable.4,5 The bimodal distribution and low number of evaluable biopsies in this trial mandate that this analysis must be viewed as exploratory. More comprehensive immunophenotyping and gene expression profiling are ongoing and may provide further insight as to biomarkers associated with benefit from combined HPV vaccination and PD-1 inhibition.

Limitations

This study has some limitations. This is a small single-arm trial including patients with heterogeneous treatment backgrounds. A larger randomized trial is necessary to confirm the benefit of vaccination added to PD-1 checkpoint inhibition.

Conclusions

Because only a subset of patients could be evaluated for HPV-16–specific immune responses with IFN-γ release, these results do not allow firm conclusions. Although the immune response is encouraging and consistent with earlier studies that demonstrated increased HPV-16–specific T cells after vaccination, it could be that these vaccine-induced T-cell populations are necessary, but not sufficient, for increased ORR in combination with nivolumab, perhaps owing to additional immunosuppressive pathways.13,31

The results of our trial are among the first clinical data to support the general concept of combining cancer vaccination with immune checkpoint blockade to enhance efficacy of vaccine-activated T cells in the immunosuppressive tumor environment. A randomized clinical trial testing the contribution of ISA101 to PD-1 inhibition, in patients with platin-resistant HPV-16–positive recurrent OPC is planned.

Supplement 1.

Trial Protocol

Supplement 2.

eFigure 1. Percentage Change in Sum of Target Lesions Over Time by Patient Accession Number

eFigure 2. Kaplan-Meier Curve for PFS

eFigure 3. Kaplan-Meier Curve for OS

eFigure 4. Box Plots Displaying Medians and Interquartile Ranges of PD-L1 Tumor, Immune, and Combined Score by Response

eFigure 5. IFN-γ Release in Response to Pooled HPV-16 E6 and E7 Peptides (ELISPOT) by Response, Patient Number, and Nivolumab Cycle Number, Color Coded

eTable 1. Treatment-Related Adverse Events

eTable 2. Distribution of PD-L1 Expression (%)

References

  • 1.Welters MJP, Ma W, Santegoets SJAM, et al. . Intratumoral HPV16-specific T cells constitute a type i-oriented tumor microenvironment to improve survival in HPV16-driven oropharyngeal cancer. Clin Cancer Res. 2018;24(3):634-647. doi: 10.1158/1078-0432.CCR-17-2140 [DOI] [PubMed] [Google Scholar]
  • 2.Seiwert TY, Burtness B, Mehra R, et al. . Safety and clinical activity of pembrolizumab for treatment of recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-012): an open-label, multicentre, phase 1b trial. Lancet Oncol. 2016;17(7):956-965. doi: 10.1016/S1470-2045(16)30066-3 [DOI] [PubMed] [Google Scholar]
  • 3.Chow LQM, Haddad R, Gupta S, et al. . Antitumor activity of pembrolizumab in biomarker-unselected patients with recurrent and/or metastatic head and neck squamous cell carcinoma: results from the phase Ib KEYNOTE-012 expansion cohort. J Clin Oncol. 2016;34(32):3838-3845. doi: 10.1200/JCO.2016.68.1478 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Bauml J, Seiwert TY, Pfister DG, et al. . Pembrolizumab for platinum- and cetuximab-refractory head and neck cancer: results from a single-arm, phase II study. J Clin Oncol. 2017;35(14):1542-1549. doi: 10.1200/JCO.2016.70.1524 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Ferris RL, Blumenschein G Jr, Fayette J, et al. . Nivolumab for recurrent squamous-cell carcinoma of the head and neck. N Engl J Med. 2016;375(19):1856-1867. doi: 10.1056/NEJMoa1602252 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Kenter GG, Welters MJ, Valentijn AR, et al. . Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia. N Engl J Med. 2009;361(19):1838-1847. doi: 10.1056/NEJMoa0810097 [DOI] [PubMed] [Google Scholar]
  • 7.Bijker MS, van den Eeden SJ, Franken KL, Melief CJ, van der Burg SH, Offringa R. Superior induction of anti-tumor CTL immunity by extended peptide vaccines involves prolonged, DC-focused antigen presentation. Eur J Immunol. 2008;38(4):1033-1042. doi: 10.1002/eji.200737995 [DOI] [PubMed] [Google Scholar]
  • 8.Rosalia RA, Quakkelaar ED, Redeker A, et al. . Dendritic cells process synthetic long peptides better than whole protein, improving antigen presentation and T-cell activation. Eur J Immunol. 2013;43(10):2554-2565. doi: 10.1002/eji.201343324 [DOI] [PubMed] [Google Scholar]
  • 9.van Poelgeest MI, Welters MJ, Vermeij R, 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(10):2342-2350. doi: 10.1158/1078-0432.CCR-15-2594 [DOI] [PubMed] [Google Scholar]
  • 10.Kenter GG, Welters MJ, Valentijn AR, et al. . Phase I immunotherapeutic trial with long peptides spanning the E6 and E7 sequences of high-risk human papillomavirus 16 in end-stage cervical cancer patients shows low toxicity and robust immunogenicity. Clin Cancer Res. 2008;14(1):169-177. doi: 10.1158/1078-0432.CCR-07-1881 [DOI] [PubMed] [Google Scholar]
  • 11.van Poelgeest MI, Welters MJ, van Esch EM, et al. . HPV16 synthetic long peptide (HPV16-SLP) vaccination therapy of patients with advanced or recurrent HPV16-induced gynecological carcinoma, a phase II trial. J Transl Med. 2013;11:88. doi: 10.1186/1479-5876-11-88 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Welters MJ, van der Sluis TC, van Meir H, et al. . Vaccination during myeloid cell depletion by cancer chemotherapy fosters robust T cell responses. Sci Transl Med. 2016;8(334):334ra52. doi: 10.1126/scitranslmed.aad8307 [DOI] [PubMed] [Google Scholar]
  • 13.van der Burg SH, Arens R, Ossendorp F, van Hall T, Melief CJ. Vaccines for established cancer: overcoming the challenges posed by immune evasion. Nat Rev Cancer. 2016;16(4):219-233. doi: 10.1038/nrc.2016.16 [DOI] [PubMed] [Google Scholar]
  • 14.Bagarazzi ML, Yan J, Morrow MP, et al. . Immunotherapy against HPV16/18 generates potent TH1 and cytotoxic cellular immune responses. Sci Transl Med. 2012;4(155):155ra138. doi: 10.1126/scitranslmed.3004414 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Trimble CL, Morrow MP, Kraynyak KA, 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(10008):2078-2088. doi: 10.1016/S0140-6736(15)00239-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Kim TJ, Jin HT, Hur SY, et al. . Clearance of persistent HPV infection and cervical lesion by therapeutic DNA vaccine in CIN3 patients. Nat Commun. 2014;5:5317. doi: 10.1038/ncomms6317 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Welters MJ, Kenter GG, Piersma SJ, et al. . Induction of tumor-specific CD4+ and CD8+ T-cell immunity in cervical cancer patients by a human papillomavirus type 16 E6 and E7 long peptides vaccine. Clin Cancer Res. 2008;14(1):178-187. doi: 10.1158/1078-0432.CCR-07-1880 [DOI] [PubMed] [Google Scholar]
  • 18.Eisenhauer EA, Therasse P, Bogaerts J, et al. . New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45(2):228-247. doi: 10.1016/j.ejca.2008.10.026 [DOI] [PubMed] [Google Scholar]
  • 19.World Medical Association World Medical Association Declaration of Helsinki: ethical principles for medical research involving human subjects. JAMA. 2013;310(20):2191-2194. doi: 10.1001/jama.2013.281053 [DOI] [PubMed] [Google Scholar]
  • 20.US Food and Drug Administration Clinical trials and human subject protection. https://www.fda.gov/ScienceResearch/SpecialTopics/RunningClinicalTrials/default.htm. Accessed August 20, 2018.
  • 21.Simon R. Optimal two-stage designs for phase II clinical trials. Control Clin Trials. 1989;10(1):1-10. doi: 10.1016/0197-2456(89)90015-9 [DOI] [PubMed] [Google Scholar]
  • 22.National Cancer Institute, National Institutes of Health CTCAE files. https://evs.nci.nih.gov/ftp1/CTCAE/CTCAE_4.03/. Accessed August 20, 2018.
  • 23.Einstein MH, Martens MG, Garcia FA, et al. . Clinical validation of the Cervista HPV HR and 16/18 genotyping tests for use in women with ASC-US cytology. Gynecol Oncol. 2010;118(2):116-122. doi: 10.1016/j.ygyno.2010.04.013 [DOI] [PubMed] [Google Scholar]
  • 24.Guo M, Khanna A, Dhillon J, et al. . Cervista HPV assays for fine-needle aspiration specimens are a valid option for human papillomavirus testing in patients with oropharyngeal carcinoma. Cancer Cytopathol. 2014;122(2):96-103. doi: 10.1002/cncy.21375 [DOI] [PubMed] [Google Scholar]
  • 25.Parra ER, Behrens C, Rodriguez-Canales J, et al. . Image analysis–based assessment of PD-L1 and tumor-associated immune cells density supports distinct intratumoral microenvironment groups in non-small cell lung carcinoma patients. Clin Cancer Res. 2016;22(24):6278-6289. doi: 10.1158/1078-0432.CCR-15-2443 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Zhao J, Yu M, Feng XP. Statistical inference for extended or shortened phase II studies based on Simon’s two-stage designs. BMC Med Res Methodol. 2015;15:48. doi: 10.1186/s12874-015-0039-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Seiwert TY, Burtness B, Weiss J, et al. . A phase 1b study of MK-3475 in patients with human papillomavirus (HPV)-associated and non-HPV-associated head and neck (H/N) cancer. J Clin Oncol. 2014;32(suppl):6011. [Google Scholar]
  • 28.Mehra R, Seiwert TY, Gupta S, et al. . Efficacy and safety of pembrolizumab in recurrent/metastatic head and neck squamous cell carcinoma: pooled analyses after long-term follow-up in KEYNOTE-012. Br J Cancer. 2018;119(2):153-159. doi: 10.1038/s41416-018-0131-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Ferris RL, Blumenschein G, Harrington K, et al. . Abstract CT021: tumor-associated immune cell PD-L1 expression and peripheral immune profiling: analyses from CheckMate 141. Cancer Res. 2017;77:CT021. doi: 10.1158/1538-7445.AM2017-CT021 [DOI] [Google Scholar]
  • 30.Büttner R, Gosney JR, Skov BG, et al. . Programmed death-ligand 1 immunohistochemistry testing: a review of analytical assays and clinical implementation in non-small-cell lung cancer. J Clin Oncol. 2017;35(34):3867-3876. doi: 10.1200/JCO.2017.74.7642 [DOI] [PubMed] [Google Scholar]
  • 31.Melief CJ, van Hall T, Arens R, Ossendorp F, van der Burg SH. Therapeutic cancer vaccines. J Clin Invest. 2015;125(9):3401-3412. doi: 10.1172/JCI80009 [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplement 1.

Trial Protocol

Supplement 2.

eFigure 1. Percentage Change in Sum of Target Lesions Over Time by Patient Accession Number

eFigure 2. Kaplan-Meier Curve for PFS

eFigure 3. Kaplan-Meier Curve for OS

eFigure 4. Box Plots Displaying Medians and Interquartile Ranges of PD-L1 Tumor, Immune, and Combined Score by Response

eFigure 5. IFN-γ Release in Response to Pooled HPV-16 E6 and E7 Peptides (ELISPOT) by Response, Patient Number, and Nivolumab Cycle Number, Color Coded

eTable 1. Treatment-Related Adverse Events

eTable 2. Distribution of PD-L1 Expression (%)


Articles from JAMA Oncology are provided here courtesy of American Medical Association

RESOURCES