Purpose:

Patients with HER2+ breast cancer benefit from trastuzumab-containing regimens with improved survival. Adaptive immunity, including cytotoxic T-cell and antibody immunity, is critical to clinical efficacy of trastuzumab. Because Th cells are central to the activation of these antitumor effectors, we reason that HER2 patients treated with trastuzumab may benefit by administering vaccines that are designed to stimulate Th-cell immunity.

Patients and Methods:

We developed a degenerate HER2 epitope–based vaccine consisting of four HLA class II–restricted epitopes mixed with GM-CSF that should immunize most (≥84%) patients. The vaccine was tested in a phase I trial. Eligible women had resectable HER2+ breast cancer and had completed standard treatment prior to enrollment and were disease free. Patients were vaccinated monthly for six doses and monitored for safety and immunogenicity.

Results:

Twenty-two subjects were enrolled and 20 completed all six vaccines. The vaccine was well tolerated. All patients were alive at analysis with a median follow-up of 2.3 years and only two experienced disease recurrence. The percent of patients that responded with augmented T-cell immunity was high for each peptide ranging from 68% to 88%, which led to 90% of the patients generating T cells that recognized naturally processed HER2 antigen. The vaccine also augmented HER2-specific antibody. Immunity was sustained in patients with little sign of diminishing at 2 years following the vaccination.

Conclusions:

Degenerate HLA-DR–based HER2 vaccines induce sustainable HER2-specific T cells and antibodies. Future studies, could evaluate whether vaccination during adjuvant treatment with trastuzumab-containing regimens improves patient outcomes.

Translational Relevance

The generation of tumor-specific adaptive immunity is important to the clinical efficacy of trastuzumab-containing regimens. Thus vaccination to augment immunity during or after trastuzumab-based therapy treatment could benefit patients. In this first-in-human trial report, we demonstrate that T-cell immune responses to HER2 can be safely generated with a novel vaccine in nearly all patients previously diagnosed with HER2 breast cancer. Furthermore, we show that these immune responses are persistent in the blood for up to 2 years following vaccination. Potential uses included neoadjuvant, adjuvant, and settings in conjunction with trastuzumab-, pertuzumab-, and/or TDM-1–based therapies in breast and other HER2+ cancers.

Breast cancer is a common disease with approximately 2,000,000 new cases and approximately 600,000 deaths annually, worldwide. HER2 overexpression defines a subset (15%–20%) of breast cancers (1). HER2 overexpression results in an aggressive tumor that, without treatment, is pathologic displaying rapid progression and frequent recurrence (2). The implementation of antibody-based therapeutics that target HER2, such as trastuzumab, has significantly impacted mortality in individuals with HER2+ breast cancer (3–5). While many mechanisms of action have been ascribed to the clinical activity of trastuzumab, recent studies have demonstrated an important role for the adaptive immune response. Foremost among immune effectors, endogenously generated HER2-specific antibodies, cytotoxic T cells, and helper CD4 T (Th) cells are important in disease protection mediated by trastuzumab-containing regimens and have been associated with greater objective tumor responses and significantly improved disease-free, progression-free, and overall survival in both humans and murine models (6–9).

Induction of an efficient coordinated adaptive immune response against infectious diseases and cancer is dependent on the generation of Th cell responses (10–13). They are critical to immune surveillance and when antigen is encountered they produce numerous cytokines that coordinate activation of various adaptive and innate immune effectors such as cytotoxic T cells, B cells, macrophages, and NK cells (10–13). On the basis of these important roles, we envision that the addition of Th cell vaccination to trastuzumab-based therapy would result in improved disease-free survival (DFS). Consistent with this rationale, we developed a vaccine that consists of four naturally presented tumor-associated epitopes derived from HER2 that bind to at least 12 of 15 common HLA-DR molecules (14). The vaccine represents a significant novel advance over prior HER2 vaccines. Notably, the vaccine was formulated on the basis of actual binding affinities of peptides to 15 different HLA-DR variants that broadly represented the North American population, including Caucasians, Asians, and African Americans (15). This is in contrast to our prior studies vaccines which were selected based solely on prediction to a single HLA-DR molecule resulting in selection of several epitopes with limited binding to most HLA-DR variants (16). Thus, the new vaccine was predicted to generate in vivo HER2-specific T-cell immunity to naturally processed and presented antigen in the vast majority of people regardless of HLA-DR background, which contrasts with other advanced HER2 peptide vaccines which are designed for use specifically in patients with select HLA backgrounds (17–19). Therefore, the primary advantage of this new vaccine is its broad applicability for individuals of all HLA-DR subtypes. In this study, this new vaccine was tested in a phase I clinical trial for safety and immunogenicity in patients with HER2+ breast cancer. In accordance with instructions from the FDA, the vaccine was tested following completion of trastuzumab-based regimens as a safety precaution with the ultimate goal of testing the vaccine concurrently with trastuzumab-based regimens.

Eligibility and enrollment

This study enrolled women ≥18 years old with resected HER2+ stage II–III breast cancer that had completed HER2-directed systemic therapy. Eligibility criteria included an Eastern Cooperative Oncology Group performance status of 0–1, and adequate hematologic, renal, and hepatic function. Exclusion criteria included autoimmunity, systemic steroid use within 90 days of registration, baseline left ventricular ejection fraction (LVEF) < 55%, history of trastuzumab-related cardiac toxicity, pregnancy, or breast feeding. Patients received a tetanus booster prior to HER2 vaccination unless they had one within 1 year of registration. Written, informed consent was obtained from all patients. The study was conducted in accordance with recognized ethical guidelines including the U.S. Common Rule and approval by institutional review board was obtained in accordance with a filed assurance approved by the Department of Health and Human Services (Clinicaltrials.gov #: NCT01632332).

Preparation of the vaccine

The HER2-derived peptides (Polypeptide Systems) that constituted the vaccine included NLELTYLPTNASLSF [p59, amino acid (aa) positions 59–73], HNQVRQVPLQRLRIV (p88, aa 88–102), LSVFQNLQVIRGRIL (p422, aa 422–435), and PIKWMALESILRRRF (p885, aa 885–899; ref. 14).

Vaccinations

Patients were vaccinated monthly six times intradermally in the forearm or the outer upper leg with 500 μg/peptide and 125 μg of GM-CSF (Sargramostim, Genzyme). Patients underwent assessments at multiple time points: prior to registration, within 14 days after registration, prior to vaccination, 4 weeks after completion of vaccinations, and 3, 6, 12, 18, and 24 months after last vaccination thereafter until disease recurrence or a maximum of 30 months. Assessments included physical examination, blood work, cardiac monitoring, toxicity assessments, and research blood draws. Peripheral blood mononuclear cells (PBMC) and sera for immune monitoring were isolated as described previously (20, 21).

Vaccinations were discontinued if patients developed a grade ≥2 allergic reaction, autoimmune reaction, injection site ulceration, neurologic difficulties, or grade ≥3 adverse events. Vaccination was discontinued if there was a (i) ≥10% LVEF decrease from pretreatment to below the institutional lower limit of normal, (ii) ≥15% LVEF decrease from pretreatment, or (iii) any LVEF decrease with symptoms of congestive heart failure.

Enzyme-linked immunosorbent spot assay

Enzyme-linked immunosorbent spot (ELISpot) was used to determine frequencies of antigen-specific T cells (21). Antigens were cyclin D1 (control) peptide (14, 22), vaccine peptides, HER2 extracellular domain protein (ECD, Sino Biological), HER2 intracellular domain protein (ICD, Cell Sciences), or tetanus toxoid (TT; List Biological Laboratories). Peptides were plated at 10 μg/mL, TT at 1 μg/mL, ECD at 25 ng/mL, and ICD at 100 ng/mL.

ELISA

Antibody (IgG) responses were assessed using a typical ELISA (21). Antigens tested included 20 ng/mL of peptides, or 100 ng/mL of TT, ECD, or ICD.

Statistical analysis

The primary endpoint was the percentage of patients who developed a severe toxicity (grade 3–5 adverse events, NCI Common Terminology Criteria for Adverse Events version 4.0). DFS was defined as time from registration to disease recurrence, second primary cancer, or death without disease recurrence or second primary cancer, and estimated with the Kaplan–Meier method (23). The laboratory endpoints for the reported analyses herein were levels of antigen-specific T cells and antibodies which were compared using either the Wilcoxon matched pairs two-tailed test or the Friedman test followed by post hoc Dunn Multiple Comparison's Test (Instat, V.3. or Prism V. 8., GraphPad). Unpaired data were compared using the Mann–Whitney two-tailed test. A patient was considered to have responded if they had developed a ≥3-fold increase in HER2-specific T cells or antibodies at any point during vaccine period (21). If T-cell immunity was undetectable, a positive response was defined as ≥50 antigen-specific T cells/million PBMCs.

Vaccination against the HER2 is safe

Twenty-two patients with HER2+ breast cancer enrolled in study (Table 1). Twenty of 22 patients completed all six immunizations. One patient withdrew after developing a grade 1 injection site reaction and another due to headaches. Twenty-two patients were evaluable for toxicity assessments. Overall, vaccination was well-tolerated with only grades 1 and 2 adverse reactions, mostly injection site reactions and fatigue (Table 2). One patient developed a grade 3 increased international normalized ratio (INR), deemed unlikely related to treatment. Two patients had LVEF drops of 10% or more but remained in the normal range.

Table 1.

Patient and tumor characteristics.

 
Median age (range) 49 (42–54) 
Race, n (%) 
 White 19 (86.4%) 
 Black 1 (4.5%) 
 Unknown/not reported 2 (9.1%) 
Ethnicity, n (%) 
 Non-Hispanic 20 (90.9%) 
 Unknown/not reported 2 (9.1%) 
Nottingham grade, n (%) 
 1 (Low) 0 (0%) 
 2 (Moderate) 3 (13.6%) 
 3 (Poor) 18 (81.8%) 
 Unknown 1 (4.5%) 
Stage, n (%) 
 T1N0 4 (18.2%) 
 T1N1 2 (9.1%) 
 T2N0  
 T2N1 9 (40.9%) 
 T3 2 (9.1%) 
 T4 2 (9.1%) 
Histology (ER/PR/HER2), n (%) 
 pos/pos/pos 4 (18.2%) 
 pos/neg/pos 3 (13.6%) 
 neg/pos/pos 1 (4.6%) 
 neg/neg/pos 14 (63.6%) 
 
Median age (range) 49 (42–54) 
Race, n (%) 
 White 19 (86.4%) 
 Black 1 (4.5%) 
 Unknown/not reported 2 (9.1%) 
Ethnicity, n (%) 
 Non-Hispanic 20 (90.9%) 
 Unknown/not reported 2 (9.1%) 
Nottingham grade, n (%) 
 1 (Low) 0 (0%) 
 2 (Moderate) 3 (13.6%) 
 3 (Poor) 18 (81.8%) 
 Unknown 1 (4.5%) 
Stage, n (%) 
 T1N0 4 (18.2%) 
 T1N1 2 (9.1%) 
 T2N0  
 T2N1 9 (40.9%) 
 T3 2 (9.1%) 
 T4 2 (9.1%) 
Histology (ER/PR/HER2), n (%) 
 pos/pos/pos 4 (18.2%) 
 pos/neg/pos 3 (13.6%) 
 neg/pos/pos 1 (4.6%) 
 neg/neg/pos 14 (63.6%) 

Abbreviations: neg, negative; pos, positive.

Table 2.

Adverse events regardless of attribution to study treatment.

Grade, n (%)
Adverse event123
Injection site reaction 21 (95.5%) 1 (4.5%) — 
Fatigue 15 (63.6%) 2 (9.1%) — 
Fever 5 (22.7%) 1 (4.5%) — 
Alkaline phosphatase increased 4 (18.2%) — — 
Neutrophil count decreased 1 (4.5%) 3 (13.6%) — 
White blood cell decreased 2 (9.1%) 2 (9.1%) — 
Lymphocyte count decreased — 2 (9.1%) — 
Platelet count decreased 2 (9.1%) — — 
Aspartate aminotransferase increased 1 (4.5%) 1 (4.5%) — 
Headache — 1 (4.5%) — 
INR increased — — 1 (4.5%) 
Lymphedema — 1 (4.5%) — 
Sinusitis — 1 (4.5%) — 
Grade, n (%)
Adverse event123
Injection site reaction 21 (95.5%) 1 (4.5%) — 
Fatigue 15 (63.6%) 2 (9.1%) — 
Fever 5 (22.7%) 1 (4.5%) — 
Alkaline phosphatase increased 4 (18.2%) — — 
Neutrophil count decreased 1 (4.5%) 3 (13.6%) — 
White blood cell decreased 2 (9.1%) 2 (9.1%) — 
Lymphocyte count decreased — 2 (9.1%) — 
Platelet count decreased 2 (9.1%) — — 
Aspartate aminotransferase increased 1 (4.5%) 1 (4.5%) — 
Headache — 1 (4.5%) — 
INR increased — — 1 (4.5%) 
Lymphedema — 1 (4.5%) — 
Sinusitis — 1 (4.5%) — 

Note: The maximum grade during treatment per adverse event per patient is tabulated.

Vaccination-generated T-cell and antibody immunity to HER2 following immunization

The mean frequencies of T cells specific for HER2 were compared between pretreatment and posttreatment samples. PBMCs were available from 21 of 22 patients. In Fig. 1A the preimmunization and highest postimmunization T-cell frequencies are plotted as described previously (21, 24, 25). The median number of postvaccination period samples was six (range, 4–6). The preimmunization (baseline) mean T-cell frequency to p59 was 80 ± 21 (±SEM, n = 21) T cells/million PBMCs which increased to a mean of 552 ± 63 T cells/million PBMCs. T cells increased from 70 ± 20 to 443 ± 54 T cells/million PBMCs for p88, from 80 ± 22 to 380 ± 55 for p422, and from 102 ± 31 to 356 ± 50 for p885.

Figure 1.

Vaccination generates HER2-specific T-cell and antibody immunity following immunization. A, Bars show the mean (n = 21) preimmunization (Pre) and highest postvaccination (Post) frequency of antigen-specific T-cells frequencies (per million PBMCs plated) that recognize vaccine antigens, HER2 p59, p88, p422, and p885. Also shown are frequencies to control cyclin D1 peptide, HER2 ICD protein, HER2 ECD protein, and TT. Combined paired ICD and ECD responses are also shown to compare directly with the T-cell immunity to TT. B, Shows the mean preimmunization and highest postvaccination frequency of antigen-specific antibodies (μg/mL) that recognize the same antigens. For both panels, each line traces the pre- and postantigen-specific T-cell levels for a single patient. P values are calculated using the Wilcoxon matched pairs test.

Figure 1.

Vaccination generates HER2-specific T-cell and antibody immunity following immunization. A, Bars show the mean (n = 21) preimmunization (Pre) and highest postvaccination (Post) frequency of antigen-specific T-cells frequencies (per million PBMCs plated) that recognize vaccine antigens, HER2 p59, p88, p422, and p885. Also shown are frequencies to control cyclin D1 peptide, HER2 ICD protein, HER2 ECD protein, and TT. Combined paired ICD and ECD responses are also shown to compare directly with the T-cell immunity to TT. B, Shows the mean preimmunization and highest postvaccination frequency of antigen-specific antibodies (μg/mL) that recognize the same antigens. For both panels, each line traces the pre- and postantigen-specific T-cell levels for a single patient. P values are calculated using the Wilcoxon matched pairs test.

Close modal

The mean T-cell frequency to ICD increased significantly from 72 ± 28 to 347 ± 56 T cells and increased from 98 ± 29 to 398 ± 62 T cells for the ECD, demonstrating that vaccine-elicited T cells are recognizing naturally processed antigens. Consistent with tetanus boosters given prior to HER2 vaccination, the mean TT-specific T-cell frequency increased from 695 ± 62 to 891 ± 61 TT-specific T cells/million PBMCs. The HER2 T-cell frequency, which is the paired sum of the ICD- and ECD-specific T-cell responses, is also reported for comparison with TT. As shown, the mean T-cell frequency to HER2 increased significantly from 170 ± 50 to 754 ± 120 antigen-specific T cells/million PBMCs. It is notable that this mean postvaccination HER2 T-cell frequency is approximately 85% of the magnitude of the postvaccination response to TT, which is a large 150 kDa protein with many foreign epitopes. Reactivity to control cyclin D1 peptide, in contrast, did not significantly increase during the vaccinations.

Increased mean antibody responses to the epitopes were generally small (Fig. 1B). Sera for antibody assessments were available from 21 of 22 patients. The median number of vaccination period serum samples was six (range, 5–6). The preimmunization mean antibody concentration to p59 was 1.1 ± 0.15 μg/mL (± SEM) and increased to 1.7 ± 0.21 μg/mL, postimmunization. To p88, antibodies increased from 1.2 ± 0.2 to 2.3 ± 0.6 μg/mL, to p422 from 1.0 ± 0.12 to 1.6 ± 0.22 μg/mL, and to p885 from 0.8 ± 0.13 to 1.1 ± 0.18 μg/mL. These increases led to an increase in antibodies that bound whole ICD (1.6 ± 0.33 to 2.8 ± 0.4 μg/mL) and ECD proteins (2.5 ± 0.40 to 4.2 ± 0.35 μg/mL). The mean TT antibody concentration increased from 6.6 ± 0.4 to 9.1 ± 0.8 μg/mL. Similar to the T-cell analysis, the paired sum of the ICD- and ECD-specific antibody revealed that HER2-specific antibody immunity increased from 4.1 ± 0.61 to 7.1 ± 0.62 μg/mL (∼77% of the mean high TT antibody response).

All of the peptides in the vaccine appear to be immunogenic in most patients

The percentage of patients that responded in the vaccine period was analyzed. Responses to all the peptides were frequent with 71%, 86%, 67%, and 76% to p59, p88, p422, and p885, respectively, in 19 of 21 evaluable patients (Fig. 2A). Although mean levels of antibodies increased significantly, antibody response rates (n = 20 evaluable) were minimal at 10%, 5%, 5%, and 0% for p59, p88, p422, and p885, respectively, (Fig. 2B), using the threefold criteria. Nineteen of 21 evaluable (90%) patients responded to vaccine with a T-cell response, all responding to multiple epitopes (Fig. 2C). The median and mean numbers of epitopes that patients responded to with T cells were 3 and 3, respectively. Four of 20 evaluable patients responded with antibody responses (Fig. 2D). Those patients who did not respond to vaccine peptides had preexistent T-cell immunity (Fig. 2E). Responders (n = 15–18; depending on the peptide) had low preexisting mean immunity ranging from 31 T cells/million PBMCs for p422 to 42 T cells/million PBMCs for p88. In contrast, nonresponders (n = 3–7) had higher preexisting immunity ranging from 180 million T cells/million PBMCs for p422 to 316 T cells/million PBMCs for p885 (Fig. 2E). Thus, at the end of vaccination, 100% of patients demonstrated immunity, the majority through vaccine augmentation. High preexisting immunity in a fraction of patients is consistent with prior findings (14). Significant correlations between peptide-specific responses and protein-specific responses demonstrate that the epitopes are naturally processed antigen (Fig. 2FI).

Figure 2.

All of the vaccine epitopes are immunogenic in most patients. The percentage of patients that responded to vaccine epitopes with either T-cell responses (A) or antibody responses (B). C and D, Show the distribution of T-cell and antibody responses to the individual epitopes. E, Shows the mean (±SEM) T-cell frequencies of the peptide-specific T cells in the responders and nonresponders. P values are from the Mann–Whitney test. F–I, Show dot plots of the correlations of the peptide- and protein-specific T-cell responses for each constituent peptide. Line, r value and P value are derived from linear regression analysis.

Figure 2.

All of the vaccine epitopes are immunogenic in most patients. The percentage of patients that responded to vaccine epitopes with either T-cell responses (A) or antibody responses (B). C and D, Show the distribution of T-cell and antibody responses to the individual epitopes. E, Shows the mean (±SEM) T-cell frequencies of the peptide-specific T cells in the responders and nonresponders. P values are from the Mann–Whitney test. F–I, Show dot plots of the correlations of the peptide- and protein-specific T-cell responses for each constituent peptide. Line, r value and P value are derived from linear regression analysis.

Close modal

T-cell immunity rapidly developed following immunization

The T-cell data were also examined at each individual timepoint over the course of vaccination, enabling an analysis of the time to maximal immunity (Fig. 3AH). For this analysis, there were samples from 21, 20, 21, 20, 21, 21, and 20 patients for time points 0, 1, 2, 3, 4, 5, and 6, respectively. Time to maximal immunity was estimated to be 2, 2, 2, and 3 months for p59, p88, p422, and p885, respectively, with a median time of 2 months. This coincided with maximal immunity at 2 months to the ICD and ECD (Fig 3EF). T-cell immunity to the cyclin D1 peptide remained low (Fig. 3G), whereas T-cell immunity to TT was high at baseline and maintained over the course of vaccinations (Fig. 3H). As a comparison, the mean levels of total HER2-specific T cells (red line, Fig 3H) were calculated by paired summing of the ICD- and ECD-specific T-cell frequencies. Levels reached maximal levels at 2 months and remained on average at 57% ± 2% of the level of T-cell immunity to TT.

Figure 3.

Mean T-cell immunity increases rapidly over the vaccine course. A–H, Shows times courses of antigen-specific T-cell frequencies (T cells/million PBMCs, mean ± SEM) for HER2 p59, p88, p422, and p885, HER2 proteins (ICD and ECD), TT, and control cyclin D1 peptide, respectively, in 21 evaluable patients. I–P, Show times courses of antigen-specific antibody frequencies (μg/mL, mean ± SEM) for 21 evaluable patients. *, P < 0.05 significance by Wilcoxon matched pairs test. Combined paired ICD and ECD responses are shown in red in TT graph.

Figure 3.

Mean T-cell immunity increases rapidly over the vaccine course. A–H, Shows times courses of antigen-specific T-cell frequencies (T cells/million PBMCs, mean ± SEM) for HER2 p59, p88, p422, and p885, HER2 proteins (ICD and ECD), TT, and control cyclin D1 peptide, respectively, in 21 evaluable patients. I–P, Show times courses of antigen-specific antibody frequencies (μg/mL, mean ± SEM) for 21 evaluable patients. *, P < 0.05 significance by Wilcoxon matched pairs test. Combined paired ICD and ECD responses are shown in red in TT graph.

Close modal

In contrast, levels of HER2 peptide–specific antibodies tended to increase only slightly or remain stable over the vaccine period (Fig. 3IP). There were samples from 20, 21, 21, 20, 21, 20, and 18 patients for time points 0, 1, 2, 3, 4, 5, and 6, respectively, for longitudinal assessment of antibody levels. Time to maximal immunity was 5, 4, 4, and 6 months for p59, p88, p422, and p885 with a median of 4.5 months. Paired analysis showed significant increases in the means for p59, p88, and p422 but not for p885 or the cyclin D1 peptide. Despite lack of antibody immunity to p885, there were significant increases in the levels of antibodies to the ICD of HER2 which increased from 1.5 ± 0.3 μg/mL (±SEM) to a maximum of 2.6 ± 0.4 μg/mL (Fig. 3M). Mean levels of antibodies to ECD protein also rose in parallel, increasing from 2.5 ± 0.4 μg/mL (±SEM) to a maximum of 3.4 ± 0.3 μg/mL (Fig. 3N). Mean levels of TT-specific antibodies increased from 6.6 ± 0.4 to 8.3 ± 0.9 μg/mL (Fig. 3P). For comparison, Fig. 3P shows the combined ICD and ECD mean antibody levels which increased throughout the course of immunization and ranged from 59% to 72% of the TT-specific antibody levels.

T-cell immunity persisted for at least 2 years following the last vaccination

A critical outcome of an efficacious vaccine is persistent immunity (26). To determine whether vaccination resulted in the generation of immune memory, blood specimens were collected over 24 months after the last vaccination. Similar to Fig. 3, we determined the frequency of IFNγ-producing T cells specific for the HER2 peptides, HER2 protein, and control peptide which were compared between pretreatment samples and multiple posttreatment observation period samples (Fig. 4A). PBMCs were available from 20 of 21 patients. Specifically, there were samples from 20, 20, 19, 17, 15, and 10 patients for time points 0, 9, 12, 18, 24, and 30 months (time following start of vaccinations), respectively.

Figure 4.

T-cell immunity persisted for at least 2 years following the last vaccination. A–F, Shows time courses of antigen-specific T-cell frequencies (T cells/million PBMCs, mean ± SEM) for HER2 p59, p88, p422, and p885, HER2 proteins (ICD and ECD), TT, and control cyclin D1 peptide for the observation points. G–L, Show times courses of antigen-specific antibody frequencies (μg/mL, mean ± SEM). Each line traces the pre- and highest observation period (Post) antigen-specific T-cell levels for a unique patient measured during the vaccine period. Assays were done separately from the vaccine period. *, P < 0.05 significance by Wilcoxon matched pairs test (AL). M and N, Show the highest observation period frequency of antigen-specific T-cells frequencies (per million PBMCs plated) and antibodies that recognize vaccine antigens, HER2 p59, p88, p422, and p885, respectively. Also shown are frequencies to control cyclin D1 peptide, and HER2 protein (combined ICD and ECD). Each symbol represents a unique patient and the blue bars represent the means. CD1 = cyclin D1. P values in M and N compare the means of each antigen and the mean of the CD1 responses using the Friedman's Nonparametric Repeated Measures ANOVA followed by the post hoc Dunn multiple comparisons test.

Figure 4.

T-cell immunity persisted for at least 2 years following the last vaccination. A–F, Shows time courses of antigen-specific T-cell frequencies (T cells/million PBMCs, mean ± SEM) for HER2 p59, p88, p422, and p885, HER2 proteins (ICD and ECD), TT, and control cyclin D1 peptide for the observation points. G–L, Show times courses of antigen-specific antibody frequencies (μg/mL, mean ± SEM). Each line traces the pre- and highest observation period (Post) antigen-specific T-cell levels for a unique patient measured during the vaccine period. Assays were done separately from the vaccine period. *, P < 0.05 significance by Wilcoxon matched pairs test (AL). M and N, Show the highest observation period frequency of antigen-specific T-cells frequencies (per million PBMCs plated) and antibodies that recognize vaccine antigens, HER2 p59, p88, p422, and p885, respectively. Also shown are frequencies to control cyclin D1 peptide, and HER2 protein (combined ICD and ECD). Each symbol represents a unique patient and the blue bars represent the means. CD1 = cyclin D1. P values in M and N compare the means of each antigen and the mean of the CD1 responses using the Friedman's Nonparametric Repeated Measures ANOVA followed by the post hoc Dunn multiple comparisons test.

Close modal

T-cell immunity was retained to all of the vaccine epitopes and HER2 protein at a consistently elevated level over a period of 2 years following the final vaccination. In contrast to the vaccination period, antibodies to the peptides appeared to robustly increase after the vaccinations were stopped, measurable at month 9 following start of vaccination (i.e., 3 months after last vaccine; Fig. 4GL). Elevated antibody responses to both p59 and p88 did not increase after month 9 but appeared to persist over the observation period. In contrast, antibody levels to p422 and p885, despite the initial increase, returned to baseline levels at 2 years. Elevated mean levels of antibodies to HER2 protein, which were increased during the vaccine period, were also stably elevated out to 30 months without signs of diminishing.

The highest observation period T-cell frequencies and antibody concentrations are depicted in Fig 4M and N, respectively. The mean of the highest observation period T-cell frequencies for p59 was 389 ± 44 (SEM) T cells/million PBMCs, 322 ± 47 T cells/million PBMCs for p88, 197 ± 24 T cells/million PBMCs for p422, and 195 ± 49 T cells/million PBMCs for p885 (Fig. 4M). The mean of the highest observation period T-cell frequencies for HER2 protein was 379 ± 46 T cells/million PBMCs. All the means were significantly higher than the mean of the highest observation period T-cell frequencies for control cyclin D1 peptide, which was only 22 ± 5 T cells/million PBMCs. The mean of the highest observation period antibody concentration for p59 was 2.2 ± 0.3 (SEM), 2.3 ± 0.3 for p88, 1.8 ± 0.2 for p422, and 2.4 ± 0.3 μg/mL serum for p885 (Fig. 4N). The mean of the highest observation period antibody concentration for HER2 protein was 8 ± 1 μg/mL serum. All the means were significantly higher than the mean of the highest observation period antibody levels for control cyclin D1 peptide, which was only 0.57 ± 0.07 μg/mL serum.

Finally, the fraction of patients demonstrating T-cell immunity to whole HER2 protein remained high into the observation years. Although the numbers of evaluable patients in each period (year 1 observation and year 2 observation periods) varied, T-cell response rates to ICD remained above 75%, to ECD above 72%, and to either ICD or ECD above 85%. In the final period (i.e., year 2), 12 of 12 (100%) of evaluable patients demonstrated elevated immunity to HER2.

Clinical outcomes

Although the trial was not designed to evaluate clinical outcomes, all patients remain alive at last vital status follow-up with a median of 2.3 years following trial enrollment (range, 0.1–3.4 years). With a median of 26.8 months of disease assessment follow-up, only two patients developed recurrence (one nodal recurrence 11.9 months and one in-breast recurrence 26.6 months, after trial enrollment). This translated into a 2-year DFS rate of 94.7% [95% confidence interval (CI), 68.1%–99.2%]. The in-breast recurrence was deemed likely a new primary rather than a local recurrence as the new lesion occurred in the opposite quadrant of the breast (9 vs. 2 o'clock position). The patient who developed nodal recurrence did not complete the vaccination treatment and only received four vaccinations. There were no distant recurrences.

The goals of this trial were to assess the safety and immunogenicity of a novel broad coverage HER2-targeted Th-cell vaccine. The vaccine was found to be safe overall. Furthermore, the vaccine was highly immunogenic and rapidly expanded T cells that recognized naturally processed HER2 antigens in 90% of patients. Patients with weaker vaccine-mediated responses were those that demonstrated elevated immunity prior to vaccine administration. The vaccine also elicited HER2-specific antibodies. Both elevated T-cell and antibody immunity were stably elevated at least 2 years after last vaccine.

One potential reason for the good safety profile is that the vaccine targets CD4 T cells whose activity is regulated by HLA class II, which is typically only expressed on immune and stromal cells (21). Because normal tissues shed very little HER2, it is likely that there is very little presentation of HER2 on HLA class II in normal tissue that would be targeted by vaccine-induced T cells (27). Historically, we have found very few HER2-specific T-cell responses to the vaccine HLA class II peptides in normal healthy individuals, a finding which is indicative of either a lack of prior exposure to the epitopes or to tolerance. A lack of prior exposure is most likely because the possibility of tolerance is not supported by data that the epitopes are highly immunogenic in the majority of patients (14).

A key finding is that immune responses were rapidly induced with a median time to maximal immunity of 2 months. This finding contrasts our previously published study using a degenerate FRα-based HLA class II vaccine in which we observed a median time to maximal immunity of 5 months in healthy disease-free patients previously diagnosed with either ovarian (n = 14) or breast cancer (n = 8). One potential reason for the differential response could be prior exposure to antigen, which could have resulted in higher levels of immune memory. Patients in this trial all demonstrated expression of HER2 in their cancers, whereas expression of FRα was not required in the FRα vaccine trial due to prior observations that FRα can be expressed on recurrent lesions even if absent on the primary lesion (21). Indeed, in that prior study, only 12 of 20 evaluable patients demonstrated FRα expression, which could have skewed the time to maximal immunity. In addition, in the FRα trial, patients were not previously exposed to immune therapy, whereas the patients in this trial received trastuzumab, which has been shown to induce endogenous immunity including activation of helper CD4 T cells as previously described above (7–9).

While the epitopes were designed primarily to stimulate T cells, antibody responses were also assessed. While increased levels of antibodies to the individual peptides were observed during the vaccine period, the responses were minor and did not meet response criteria. Considering that small increases of even 25% are associated with improved survival, perhaps the response criteria in this study were too rigid (7). Nonetheless, despite the low level response, levels of antibodies to the whole HER2 protein steadily and significantly increased during the vaccination period. Interestingly, we did observe stronger, but brief, increases in peptide-specific antibodies after vaccination ended, some of which persisted for reasons that were unclear but could have been due to the Th1-skewing nature of GM-CSF (28–30). Th1 T cells are known to favor the development of cell-mediated immunity and do not release high levels of cytokines (IL4 and IL5) that activate B cells and augment antibody responses. GM-CSF is a key regulator of Th2-mediated antibody-based immunity and is known to suppress both IgG and IgA responses in infectious disease models (31). In addition to suppressing Th2 induction, GM-CSF also induces subsets of regulatory T-cells that suppress production of autoantibodies (32). Thus, the immune system has multiple mechanisms that restrict antibody development that when modulated or removed could have resulted in increased antibody production that we observed in the trial.

An important finding from this trial is that vaccination resulted in HER2 protein–specific T-cell and antibody immunity that persisted well into the observation period. Specifically, time course analysis showed that the mean T-cell immune responses were stabilized at 15 months following the first immunization and continued unabated at significantly elevated levels at least to 2 years after the final vaccination. This is an ideal outcome for a vaccine for prevention of relapse (33). Despite the observation of persistence, we do not yet know whether the immune responses are sustainable over longer periods of time that are commensurate with the patterns of relapse that are observed in patients with HER2+ breast cancer, which can range from several months to several years (34). If the immune response is not sustainable, one possible remediation is the inclusion of boosters. In other previously reported HER2 vaccine trials, boosters are required to sustain HER2-specific immunity long term, and in at least one trial, there was some evidence to suggest that boosters may impact disease recurrence (19, 35).

The vaccine may synergize with trastuzumab or other HER2-specific antibody-based therapies to improve survival, as has been suggested previously (24). While designed for safety and given after trastuzumab therapy, a secondary objective was patient survival. Given the observation that only two of 22 patients recurred within the 24 months following last vaccination, 2-year DFS (94.7%, measured from after completion of adjuvant treatment and prior to vaccination initiation) compared favorably with historical data from the adjuvant trastuzumab trials. For example, in the combined analysis of trials NCCTG N9831 and NSABP B-31, 3-year recurrence-free survival (measured from just prior to start of all adjuvant therapy) of similar patients with stage II and III HER2+ breast cancer was 89.22% (95% CI, 87.72%–90.76%; personal communication with study team for applicable subset of patients reported by Perez and colleagues; ref. 3). Importantly, there was no distant recurrence observed in our cohort. This vaccine, given its broad patient coverage and ability to induce immune memory, should be evaluated for its ability to increase the cure rate for women with HER2 breast cancer, a disease controlled by the immune system (7–9, 25, 36). Our group has recently developed a phase II trial in which this vaccine will be coadministered with standard-of-care trastuzumab-based therapy in the adjuvant setting in patients with HER2+ breast cancer. The trial will ask whether the inclusion of vaccinations with trastuzumab-based regimens, along with periodic boosters, extends the invasive DFS period in patients at high risk for relapse.

K.L. Knutson reports receiving commercial research grants from Marker Therapeutics. M.S. Block reports receiving commercial research grants from Merck, Genentech, Bristol-Myers Squibb, Pharmacyclics, and Marker Therapeutics. G. Wilson is an employee/paid consultant for Marker Therapeutics. No potential conflicts of interest were disclosed by the other authors.

Conception and design: K.L. Knutson, L. Karyampudi, G. Wilson, A.C. Degnim

Development of methodology: K.L. Knutson, M.S. Block, C.L. Erskine, A.B. Dietz, A.C. Degnim

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): K.L. Knutson, M.S. Block, C.L. Erskine, T.J. Hobday, D. Padley, T.K. Mangskau, S. Chumsri

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): K.L. Knutson, M.S. Block, N. Norton, S. Chumsri, A.C. Dueck, G. Wilson, A.C. Degnim

Writing, review, and/or revision of the manuscript: K.L. Knutson, N. Norton, T.J. Hobday, A.B. Dietz, M.P. Gustafson, S. Chumsri, A.C. Dueck, L. Karyampudi, G. Wilson, A.C. Degnim

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): K.L. Knutson, M.P. Gustafson, D. Puglisi-Knutson, T.K. Mangskau, A.C. Degnim

Study supervision: K.L. Knutson

This work was supported by grants from the U.S. NIH (P50CA116201-Mayo Clinic Breast Cancer SPORE) and P30CA015083-Mayo Comprehensive Cancer Center Grant, Marker Therapeutics, Inc., and the U.S. Department of Defense (W81XWH-18-1-0563, W81XWH-18-1-0564, W81XWH-16-1-0265 and W81XWH-16-1-0266, all to K.L. Knutson and A.C. Degnim). The authors are grateful to the administrative and clinical staff in the Mayo Clinic Comprehensive Cancer Center for their assistance throughout the trial. The authors are grateful to the patients with breast cancer who participated in this study.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Bray
F
,
Ferlay
J
,
Soerjomataram
I
,
Siegel
RL
,
Torre
LA
,
Jemal
A
. 
Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries
.
CA Cancer J Clin
2018
;
68
:
394
424
.
2.
Slamon
DJ
,
Clark
GM
,
Wong
SG
,
Levin
WJ
,
Ullrich
A
,
McGuire
WL
. 
Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene
.
Science
1987
;
235
:
177
82
.
3.
Perez
EA
,
Romond
EH
,
Suman
VJ
,
Jeong
JH
,
Sledge
G
,
Geyer
CE
 Jr
, et al
Trastuzumab plus adjuvant chemotherapy for human epidermal growth factor receptor 2-positive breast cancer: planned joint analysis of overall survival from NSABP B-31 and NCCTG N9831
.
J Clin Oncol
2014
;
32
:
3744
52
.
4.
Baselga
J
,
Cortes
J
,
Kim
SB
,
Im
SA
,
Hegg
R
,
Im
YH
, et al
Pertuzumab plus trastuzumab plus docetaxel for metastatic breast cancer
.
N Engl J Med
2012
;
366
:
109
19
.
5.
von Minckwitz
G
,
Huang
CS
,
Mano
MS
,
Loibl
S
,
Mamounas
EP
,
Untch
M
, et al
Trastuzumab emtansine for residual invasive HER2-positive breast cancer
.
N Engl J Med
2019
;
380
:
617
28
.
6.
Park
S
,
Jiang
Z
,
Mortenson
ED
,
Deng
L
,
Radkevich-Brown
O
,
Yang
X
, et al
The therapeutic effect of anti-HER2/neu antibody depends on both innate and adaptive immunity
.
Cancer Cell
2010
;
18
:
160
70
.
7.
Knutson
KL
,
Clynes
R
,
Shreeder
B
,
Yeramian
P
,
Kemp
KP
,
Ballman
K
, et al
Improved survival of HER2+ breast cancer patients treated with trastuzumab and chemotherapy is associated with host antibody immunity against the HER2 intracellular domain
.
Cancer Res
2016
;
76
:
3702
10
.
8.
Norton
N
,
Fox
N
,
McCarl
CA
,
Tenner
KS
,
Ballman
K
,
Erskine
CL
, et al
Generation of HER2-specific antibody immunity during trastuzumab adjuvant therapy associates with reduced relapse in resected HER2 breast cancer
.
Breast Cancer Res
2018
;
20
:
52
.
9.
Taylor
C
,
Hershman
D
,
Shah
N
,
Suciu-Foca
N
,
Petrylak
DP
,
Taub
R
, et al
Augmented HER-2 specific immunity during treatment with trastuzumab and chemotherapy
.
Clin Cancer Res
2007
;
13
:
5133
43
.
10.
Knutson
KL
,
Disis
ML
. 
Tumor antigen-specific T helper cells in cancer immunity and immunotherapy
.
Cancer Immunol Immunother
2005
;
54
:
721
8
.
11.
Crotty
S
. 
A brief history of T cell help to B cells
.
Nat Rev Immunol
2015
;
15
:
185
9
.
12.
Melssen
M
,
Slingluff
CL
 Jr
. 
Vaccines targeting helper T cells for cancer immunotherapy
.
Curr Opin Immunol
2017
;
47
:
85
92
.
13.
Bedoui
S
,
Heath
WR
,
Mueller
SN
. 
CD4(+) T-cell help amplifies innate signals for primary CD8(+) T-cell immunity
.
Immunol Rev
2016
;
272
:
52
64
.
14.
Karyampudi
L
,
Formicola
C
,
Erskine
CL
,
Maurer
MJ
,
Ingle
JN
,
Krco
CJ
, et al
A degenerate HLA-DR epitope pool of HER-2/neu reveals a novel in vivo immunodominant epitope, HER-2/neu88–102
.
Clin Cancer Res
2010
;
16
:
825
34
.
15.
Southwood
S
,
Sidney
J
,
Kondo
A
,
del Guercio
MF
,
Appella
E
,
Hoffman
S
, et al
Several common HLA-DR types share largely overlapping peptide binding repertoires
.
J Immunol
1998
;
160
:
3363
73
.
16.
Salazar
LG
,
Fikes
J
,
Southwood
S
,
Ishioka
G
,
Knutson
KL
,
Gooley
TA
, et al
Immunization of cancer patients with HER-2/neu-derived peptides demonstrating high-affinity binding to multiple class II alleles
.
Clin Cancer Res
2003
;
9
:
5559
65
.
17.
Mittendorf
EA
,
Lu
B
,
Melisko
M
,
Price Hiller
J
,
Bondarenko
I
,
Brunt
AM
, et al
Efficacy and safety analysis of Nelipepimut-S vaccine to prevent breast cancer recurrence: a randomized, multicenter, phase III clinical trial
.
Clin Cancer Res
2019
;
25
:
4248
54
.
18.
Clifton
GT
,
Litton
JK
,
Arrington
K
,
Ponniah
S
,
Ibrahim
NK
,
Gall
V
, et al
Results of a phase Ib trial of combination immunotherapy with a CD8+ T Cell eliciting vaccine and trastuzumab in breast cancer patients
.
Ann Surg Oncol
2017
;
24
:
2161
7
.
19.
Mittendorf
EA
,
Clifton
GT
,
Holmes
JP
,
Schneble
E
,
van Echo
D
,
Ponniah
S
, et al
Final report of the phase I/II clinical trial of the E75 (nelipepimut-S) vaccine with booster inoculations to prevent disease recurrence in high-risk breast cancer patients
.
Ann Oncol
2014
;
25
:
1735
42
.
20.
Disis
ML
,
Grabstein
KH
,
Sleath
PR
,
Cheever
MA
. 
Generation of immunity to the HER-2/neu oncogenic protein in patients with breast and ovarian cancer using a peptide-based vaccine
.
Clin Cancer Res
1999
;
5
:
1289
97
.
21.
Kalli
KR
,
Block
MS
,
Kasi
PM
,
Erskine
CL
,
Hobday
TJ
,
Dietz
A
, et al
Folate receptor alpha peptide vaccine generates immunity in breast and ovarian cancer patients
.
Clin Cancer Res
2018
;
24
:
3014
25
.
22.
Karyampudi
L
,
Krco
CJ
,
Kalli
KR
,
Erskine
CL
,
Hartmann
LC
,
Goodman
K
, et al
Identification of a broad coverage HLA-DR degenerate epitope pool derived from carcinoembryonic antigen
.
Cancer Immunol Immunother
2010
;
59
:
161
71
.
23.
Kaplan
EL
,
Meier
P
. 
Nonparametric estimation from incomplete observations
.
J Am Stat Assoc
1958
;
53
:
457
81
.
24.
Disis
ML
,
Wallace
DR
,
Gooley
TA
,
Dang
Y
,
Slota
M
,
Lu
H
, et al
Concurrent trastuzumab and HER2/neu-specific vaccination in patients with metastatic breast cancer
.
J Clin Oncol
2009
;
27
:
4685
92
.
25.
Peoples
GE
,
Holmes
JP
,
Hueman
MT
,
Mittendorf
EA
,
Amin
A
,
Khoo
S
, et al
Combined clinical trial results of a HER2/neu (E75) vaccine for the prevention of recurrence in high-risk breast cancer patients: U.S. Military Cancer Institute Clinical Trials Group Study I-01 and I-02
.
Clin Cancer Res
2008
;
14
:
797
803
.
26.
Pulendran
B
,
Ahmed
R
. 
Immunological mechanisms of vaccination
.
Nat Immunol
2011
;
12
:
509
17
.
27.
Dittadi
R
,
Zancan
M
,
Perasole
A
,
Gion
M
. 
Evaluation of HER-2/neu in serum and tissue of primary and metastatic breast cancer patients using an automated enzyme immunoassay
.
Int J Biol Markers
2001
;
16
:
255
61
.
28.
Yen
HH
,
Scheerlinck
JP
. 
Co-delivery of plasmid-encoded cytokines modulates the immune response to a DNA vaccine delivered by in vivo electroporation
.
Vaccine
2007
;
25
:
2575
82
.
29.
Romagnani
S
. 
T-cell subsets (Th1 versus Th2)
.
Ann Allergy Asthma Immunol
2000
;
85
:
9
18
.
30.
Yoon
HA
,
Aleyas
AG
,
George
JA
,
Park
SO
,
Han
YW
,
Lee
JH
, et al
Cytokine GM-CSF genetic adjuvant facilitates prophylactic DNA vaccine against pseudorabies virus through enhanced immune responses
.
Microbiol Immunol
2006
;
50
:
83
92
.
31.
Kannanganat
S
,
Wyatt
LS
,
Gangadhara
S
,
Chamcha
V
,
Chea
LS
,
Kozlowski
PA
, et al
High doses of GM-CSF inhibit antibody responses in rectal secretions and diminish modified vaccinia Ankara/simian immunodeficiency virus vaccine protection in TRIM5alpha-restrictive macaques
.
J Immunol
2016
;
197
:
3586
96
.
32.
Sheng
JR
,
Li
LC
,
Ganesh
BB
,
Prabhakar
BS
,
Meriggioli
MN
. 
Regulatory T cells induced by GM-CSF suppress ongoing experimental myasthenia gravis
.
Clin Immunol
2008
;
128
:
172
80
.
33.
Sallusto
F
,
Lanzavecchia
A
,
Araki
K
,
Ahmed
R
. 
From vaccines to memory and back
.
Immunity
2010
;
33
:
451
63
.
34.
Ignatov
A
,
Eggemann
H
,
Burger
E
,
Ignatov
T
. 
Patterns of breast cancer relapse in accordance to biological subtype
.
J Cancer Res Clin Oncol
2018
;
144
:
1347
55
.
35.
Holmes
JP
,
Clifton
GT
,
Patil
R
,
Benavides
LC
,
Gates
JD
,
Stojadinovic
A
, et al
Use of booster inoculations to sustain the clinical effect of an adjuvant breast cancer vaccine: from US Military Cancer Institute Clinical Trials Group Study I-01 and I-02
.
Cancer
2011
;
117
:
463
71
.
36.
Gu-Trantien
C
,
Loi
S
,
Garaud
S
,
Equeter
C
,
Libin
M
,
de Wind
A
, et al
CD4(+) follicular helper T cell infiltration predicts breast cancer survival
.
J Clin Invest
2013
;
123
:
2873
92
.