Next Article in Journal
Potential for a Combined Oral Inactivated Whole-Cell Vaccine Against ETEC and Shigella: Preclinical Studies Supporting Feasibility
Previous Article in Journal
Addressing the Underestimated Burden of RSV in Older Adults in Europe: Epidemiology, Surveillance Gaps, and Public Health Implications
Previous Article in Special Issue
A Systematic Review of Human Papillomavirus Vaccination Challenges and Strategies to Enhance Uptake
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Safety, Tolerability, and Immunogenicity of a Recombinant Nonavalent Human Papillomavirus Vaccine (Escherichia coli) in Healthy Chinese Women Aged 18–45 Years: A Phase 1 Clinical Trial

1
School of Public Health, National Vaccine Innovation Platform, Nanjing Medical University, Nanjing 211166, China
2
Department of Vaccine Clinical Evaluation, Jiangsu Provincial Center for Disease Control and Prevention (Jiangsu Provincial Academy of Preventive Medicine), Nanjing 210009, China
3
Jiangsu Provincial Medical Innovation Center, National Health Commission (NHC) Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention (Jiangsu Provincial Academy of Preventive Medicine), Nanjing 210009, China
4
Sheyang City Center for Disease Control and Prevention, Yancheng 224499, China
5
Beijing Health Guard Biotechnology Inc., Beijing 100176, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Vaccines 2025, 13(5), 511; https://doi.org/10.3390/vaccines13050511
Submission received: 31 March 2025 / Revised: 4 May 2025 / Accepted: 10 May 2025 / Published: 13 May 2025
(This article belongs to the Special Issue Prevention of Human Papillomavirus and Vaccines Strategies)

Abstract

:
Background: Prophylactic human papillomavirus (HPV) vaccination substantially alleviates cervical cancer burden. This study aimed to evaluate the safety, tolerability, and immunogenicity of an Escherichia coli-expressed recombinant nonavalent HPV vaccine. Methods: A dose-escalating phase 1 clinical trial was conducted in Sheyang County, Jiangsu Province, China. Each participant received either the test vaccine or the control vaccine (Gardasil 9) following a 0/2/6-month schedule. Adverse reactions (ARs) within 7 days after vaccination, adverse events (AEs) within 30 days, and serious adverse events (SAEs) throughout the study were recorded. Blood parameters were measured before and 3 days after each dose. Serum immunoglobulin G (IgG) and neutralizing antibodies (nAbs) against nine HPV types were analyzed at months 0, 3, and 7. Results: A total of 160 women aged 18–45 years were enrolled, and 155 participants completed the full vaccination regimen. Within 7 days following vaccination, the incidence of ARs ranged from 56.67% to 90.00%, with the low-dose group showing a significantly higher rate than the control group (p = 0.004). Most AEs were mild or moderate, and no vaccine-related SAEs occurred. No significant differences were observed among the four groups regarding the incidence of abnormal laboratory findings. Seroconversion rates for nAbs and IgG against nine HPV types exceeded 97.92% following three doses. High levels of nAbs and IgG were observed at months 3 and 7, with geometric mean titers (GMTs) showing further increases by month 7. Conclusions: This new recombinant nonavalent HPV vaccine exhibits good tolerability and strong immunogenicity among women aged 18–45 years, supporting further efficacy studies in larger populations.

1. Introduction

Human papillomavirus (HPV) is the leading etiological agent of cervical cancer, which is recognized as the fourth most prevalent malignant cancer among women globally [1]. In 2022, an estimated 662,301 new cervical cancer cases and 348,874 related deaths were reported worldwide [2]. In China alone, approximately 150,000 cervical cancer cases emerged, and 55,000 people died from this cancer in 2022 [3]. Nearly all cervical cancer cases, along with a large proportion of anogenital and oropharyngeal cancers, as well as genital warts, are caused by persistent HPV infection [4]. Over 70% of cervical cancer cases are attributable to the high-risk HPV types 16 and 18 [5], while about 90% of genital warts cases are caused by low-risk HPV types 6 and 11 [6].
Prophylactic HPV vaccination plays a crucial role in mitigating the global disease burden caused by HPV [7]. In 2020, the World Health Organization (WHO) officially introduced the Cervical Cancer Elimination Strategy, aiming to make cervical cancer no longer a public health concern by the year 2030. A key objective of this strategy is to ensure that 90% of girls have completed the HPV vaccination series by the age of 15 [8]. Currently, six licensed HPV vaccines have been approved, all of which demonstrate favorable safety profiles and effectiveness in preventing HPV infection. These include three bivalent vaccines targeting HPV types 16 and 18, two quadrivalent vaccines targeting HPV6, 11, 16, and 18, and a nonavalent vaccine covering HPV6, 11, 16, 18, 31, 33, 45, 52, and 58. Gardasil 9, the nonavalent HPV vaccine developed by Merck Sharp & Dohme Corp., Rahway, NJ, USA, incorporates five extra high-risk HPV types (31, 33, 45, 52, and 58), thereby expanding cervical cancer prevention coverage from approximately 70% to 90% [9]. However, Gardasil 9 remains largely inaccessible in low-resource settings due to limited production and high cost [10]. This presents a major obstacle for low- and middle-income countries, which bear 88.1% of the global cervical cancer burden [9].
Of the six licensed HPV vaccines, a bivalent HPV 16/18 vaccine (Cecolin; Xiamen Innovax, Xiamen, China) produced by Escherichia coli (E. coli) expression platform was launched in China in 2019 and obtained WHO prequalification in 2021 [11]. Its affordability and scalable production make it a promising solution to address vaccine shortages and expand coverage [12,13]. Our candidate recombinant nonavalent HPV vaccine, also produced using an E. coli expression system similar to Cecolin, may offer an expanded option for preventing HPV-related diseases. Here, we report the safety and immunogenicity results of it from a phase 1 clinical trial conducted in healthy Chinese women aged 18–45 years.

2. Materials and Methods

2.1. Study Design and Participants

This single-center, dose-escalating phase 1 clinical trial was conducted to evaluate the safety, tolerability, and immunogenicity of a recombinant nonavalent HPV vaccine (E. coli) in healthy Chinese women from September 2019 to May 2020. The trial received approval from the Independent Ethics Committee of the Jiangsu Provincial Center for Disease Control and Prevention (JSJK2019-A003-01) and was performed in accordance with the principles of the Declaration of Helsinki and Good Clinical Practice guidelines. The study was registered at ClinicalTrials.gov (NCT05680454).
An overview of the study design is shown in Figure 1. The study was conducted in two stages, following a sequential and dose-escalating approach. Based on a 7-day safety follow-up, dose escalation could be paused if any safety risks were observed. The first stage was an open-label trial. The medium-dose group enrolled 20 participants in two steps (5 and 15 participants) with a 3-day interval. The high-dose group followed the same enrollment process after confirming safety. In total, 40 women aged 27–45 years received either medium-dose or high-dose test vaccine. The second stage was a randomized, blinded, and positive-controlled trial in which 120 women aged 18–26 years were divided into low-dose, medium-dose, and high-dose groups (40 per group). Participants in each dose group received either the test or control vaccine (Gardasil 9) in a 3:1 ratio. Enrollment for the low-dose group was carried out in four steps (5, 10, 10, and 15 participants, respectively) with a 3-day interval between each step. Subsequently, the same enrollment process was adopted for the medium-dose and high-dose groups. Each participant was under follow-up for approximately 7 months.
Healthy women aged 18–45 years residing in Sheyang County, Yancheng, Jiangsu, China, were recruited for the study. Written informed consent was obtained from all enrolled individuals prior to the initiation of any study procedures. Eligibility criteria included an age range of 18 to 45 years, normal axillary temperature (≤37.0 °C), a negative result on a urine pregnancy test, and either no childbearing potential or childbearing potential with a requirement to abstain from sexual activity. Women who were pregnant or breastfeeding, had received any HPV vaccine previously, had a history of severe allergic reactions with vaccines, or had an autoimmune disease or immunodeficiency were excluded. A complete list of the inclusion and exclusion criteria is provided in the Supplementary Materials.

2.2. Randomization and Masking

The first stage was an open-label trial. The second stage used stratified block randomization to ensure balance across different dose groups (low-dose, medium-dose, and high-dose groups). Eligible participants were stratified by dose level, and within each stratum, randomization lists were generated within each stratum using SAS version 9.4 software. In the second stage, participants were randomly assigned in a 3:1 ratio to receive three doses of either the test vaccine or the positive control vaccine. Each participant was assigned a unique vaccine code sequentially, based on their enrollment order. To maintain the blinding of the study, both the test and control vaccines were indistinguishable in terms of outer packaging, labeling, and volume. Due to differences in the inner packaging, the personnel responsible for vaccine retrieval and administration were aware of the subject allocation, but they were not permitted to participate in any other aspects of the study, including follow-up. The allocation of participants to study groups and vaccine codes remained blinded to all participants, investigators, and study center personnel.

2.3. Procedures

The recombinant nonavalent HPV vaccine (types 6/11/16/18/31/33/45/52/58), derived from E. coli, was manufactured by Beijing Health Guard Biotechnology Inc., Beijing, China. The HPV L1 proteins for each type were produced in E. coli and purified to obtain high-purity HPV L1 pentamer proteins. Partial truncations were made at both the N- and C-termini of the L1 protein to improve the solubility of the L1 pentamer. This modification enhanced the expression of L1 pentamer proteins in the bacterial expression system and prevented degradation of the terminal amino acids that could affect the integrity and stability of the virus-like particles (VLPs). Each 0.5 mL dose of the vaccine contains a specific amount of HPV L1 VLPs, with the low-dose group, medium-dose group, and high-dose group containing a total of 220 µg, 270 µg, and 360 µg, respectively. The control vaccine used in this study was Gardasil 9 (Merck Sharp & Dohme Corp., Rahway, NJ, USA), which was first marketed in China in 2018. Both vaccines were administered by intramuscular injection into the deltoid muscle of the upper arm at 0, 2, and 6 months.
Following each vaccination, participants were monitored on site for 30 min to detect any immediate adverse events (AEs). They were also instructed on the use of diary cards to document solicited local and systemic adverse reactions (ARs) for 7 days, as well as any AEs occurring within 30 days following each vaccination. Serious adverse events (SAEs) were recorded throughout the entire study duration. Blood samples were collected prior to and on day 3 after each vaccination for routine hematological and biochemical analyses. Laboratory indices included two routine blood tests, white blood cell count (WBC) and hemoglobin (HGB), three serum biochemical tests, creatinine (CREA), alanine aminotransferase (ALT), and aspartate aminotransferase (AST), and two blood coagulation function tests, prothrombin time (PT) and activated partial thromboplastin time (APTT). The severity grading of both AEs and abnormal lab findings was carried out by investigators according to the Guidelines for Adverse Event Classification Standards for Clinical Trials of Preventive Vaccines (2019) issued by the National Medical Products Administration (NMPA).
Serum samples were collected from all participants on day 0, month 3 (30 days after the second vaccination), and month 7 (30 days after the third vaccination) to evaluate the levels of HPV 6/11/16/18/31/33/45/52/58 type-specific immunoglobulin G (IgG) and neutralizing antibodies (nAbs). These samples were also tested for anti-glutathione-S-transferase (GST) and 3C protease-related antibodies. Detection of IgG antibodies was performed using an enzyme-linked immunosorbent assay (ELISA) based on HPV VLPs produced in E. coli, while neutralizing antibodies were assessed using a pseudovirion-based neutralization assay (PBNA). The IgG and neutralizing antibody tests in this study were carried out at the China National Institute for Food and Drug Control (NIFDC), and the anti-GST and 3C protease-related antibody tests were conducted at Beijing Health Guard Biotechnology Inc.

2.4. Outcomes

The primary outcome was the incidence of ARs within 7 days post each vaccination. Secondary safety outcomes included the incidence of AEs within 30 days, the percentage of abnormal blood routine indexes, blood biochemical indexes, and coagulation time within 3 days after each dose, as well as the overall rate of SAEs recorded throughout the study. Secondary immunogenicity outcomes included the seroconversion rates and levels of HPV 6/11/16/18/31/33/45/52/58 type-specific nAbs and IgG antibodies at months 3 and 7 post-vaccination. Subgroup analyses were conducted on participants with negative pre-immunization antibodies against nine HPV subtypes, evaluating their seroconversion rates and antibody responses at months 3 and 7 post-vaccination. Exploratory outcomes included the levels of anti-GST and 3C protease-related antibodies at baseline and at months 3 and 7 post-vaccination.

2.5. Statistical Analysis

The sample size was determined in accordance with the Technical Guidelines for Vaccine Clinical Trials issued by the NMPA, which recommends a sample size of 20 to 30 participants for Phase 1 clinical trials.
The safety set (SS) included all individuals who received at least one vaccine dose. The full analysis set (FAS), closely aligned with the intention-to-treat principle, comprised participants who received at least one dose and provided at least one serum sample. Immunogenicity assessments were performed using the per-protocol set (PPS), consisting of participants who satisfied all eligibility criteria, completed the three-dose vaccination schedule, and submitted blood samples as per the study protocol. Seroconversion was defined as either a change from seronegative at baseline to seropositive after vaccination, or at least a four-fold increase in antibody titers over baseline. The geometric mean increase (GMI) refers to the geometric mean of fold increase in the antibody titers.
All AEs were summarized by dose group as frequencies and percentages. Differences in AE incidence rates across groups were calculated using Pearson’s Chi-squared test or Fisher’s exact test. The geometric mean titer (GMT) and GMI for both IgG and nAbs, along with their 95% confidence intervals (CIs), were calculated based on Student’s t distribution of the log-transformed antibody titers. The Wilcoxon rank-sum test was used to analyze data that did not follow a normal distribution. Seroconversion rates and antibody titers were compared across the four groups using Pearson’s Chi-squared test and an ANOVA, respectively. For multiple comparisons, the significance level was adjusted using the Bonferroni correction, with an adjusted α value of 0.008. Differences between groups were considered statistically significant if p < 0.008 for multiple comparisons. For all other analyses, p < 0.05 was considered statistically significant. Data were analyzed using SAS version 9.4 (SAS Institute Inc., Cary, NC, USA), and GraphPad Prism 9.5 (GraphPad Software, Inc., San Diego, CA, USA) was used for graphing.

3. Results

3.1. Baseline Demographic Characteristics

From 1 September 2019 to 10 May 2020, a total of 167 women aged 18–45 years were screened for eligibility, with 160 healthy women ultimately included in the study. In Stage 1, 40 women aged 27–45 years were included, while in Stage 2, 120 women aged 18–26 years were recruited (Figure 2). Participants in Stage 2 were only included after confirming the absence of vaccine-related SAEs within 7 days following the first dose administered to the participants in Stage 1. Among the enrolled participants, five did not complete the full vaccination schedule, and six failed to provide serum samples at all three scheduled time points (months 0, 3, and 7). The low-dose and control groups each had 30 participants, while the medium-dose and high-dose groups each had 50 participants. Baseline demographic characteristics were comparable across all groups (Table 1).

3.2. Safety and Tolerability

All 160 participants enrolled in the study received at least one dose of the vaccine and were included in the safety analysis set. Within 7 days following each vaccination, a total of 338 ARs were recorded (Table 2). The overall incidence of ARs was 90.00% in the low-dose group, 74.00% in the medium-dose group, 74.00% in the high-dose group, and 56.67% in the control group. A significant difference was observed between the low-dose and control groups (p = 0.004). Similarly, the rate of solicited ARs was also higher in the low-dose group compared to the control group (90.00% vs. 56.67%, p = 0.004). The incidences of unsolicited AEs remained comparable across the groups, with rates of 6.67%, 8.00%, 8.00%, and 6.67%, respectively. Most reported ARs were mild or moderate. Two participants reported Grade 3 ARs, one being induration and the other being fever, both of which occurred in the high-dose group (Table S3). No serious adverse reaction occurred during the study.
The most frequently reported local AR within 7 days post-vaccination was pain at the injection site, occurring in 59.38% of participants. A statistically significant difference in incidence was found between the low-dose and control groups (80.00% vs. 40.00%, p = 0.006) (Figure 3). Erythema was also commonly reported, with incidences of 20.00%, 8.00%, 16.00%, and 20.00% in the low-dose, medium-dose, high-dose, and control groups, respectively. Induration was reported in 30.00%, 8.00%, 8.00%, and 3.33% of participants in the four groups, with the low-dose group showing a significantly higher incidence than the control group (p = 0.006). The most common systemic ARs were fatigue (13.75%), cough (7.50%), and headache (6.88%), with no significant differences observed among the groups.
The overall incidence of AEs within 30 days post-vaccination was as follows: 90.00% in the low-dose group, 80.00% in the medium-dose group, 82.00% in the high-dose group, and 66.67% in the control group (Table 3). The incidence of total ARs ranged from 56.67% to 90.00%, with a significant difference found between the low-dose and control groups (p = 0.004). Notably, the rate of solicited AEs was significantly higher in the low-dose group compared to the control group (90.00% vs. 56.67%, p = 0.004). Most reported AEs were mild or moderate. As described before, two Grade 3 AEs occurred within 7 days post-vaccination in the high-dose group (Table S3). During the entire study period, one serious adverse event (SAE) occurred more than 30 days after vaccination. The investigators ultimately determined that it was unrelated to the vaccination.
When stratified by age, a significant difference was observed in the incidence of total AEs within 30 days post-vaccination in women aged 27–45 years, with the medium-dose group showing a lower incidence compared to the high-dose group (65.00% vs. 95.00%, p = 0.048) (Table S1). For women aged 18–26 years, another significant difference was found in the incidence of total ARs between the low-dose and control groups (90.00% vs. 56.67%, p = 0.004) (Table S2).

3.3. Laboratory Parameters

A total of seven hematological parameters were analyzed to assess changes before and 3 days after each dose of vaccination. Table 4 summarizes the post-vaccination abnormalities, defined as cases where the test result was “normal” prior to vaccination but became “abnormal” afterward. The incidence of abnormalities in various indicators following vaccination was generally low. Among these, the highest incidence was observed in abnormal WBC and CREA levels after the third dose, with both showing an incidence of 5.19%. No significant differences were observed among the four groups regarding the incidence of abnormal laboratory findings. The majority of post-vaccination abnormalities were classified as mild or moderate and were deemed clinically insignificant by clinicians.

3.4. Immunogenicity

The seroconversion rates for nAbs and IgG against HPV types 6/11/16/18/31/33/45/52/58 at month 3 (one month after the second dose) in the PPS cohorts are presented in Table S4. In the low-dose group, all participants achieved seroconversion for both nAbs and IgG against all nine HPV types. Within the medium-dose group, one participant did not seroconvert for nAbs of HPV 45 (seroconversion rate: 97.92%), and two did not seroconvert for nAbs of HPV 58 (seroconversion rate: 95.83%). Similarly, in the high-dose group, one participant failed to seroconvert for nAbs against HPV 45 and HPV 58, with seroconversion rates of 97.96% for each. In the control group, one participant did not achieve seroconversion for nAbs against HPV 45 (seroconversion rate: 96.30%), three failed to seroconvert for nAbs of HPV 58 (seroconversion rate: 88.89%), and one failed to seroconvert for IgG against HPV 6 and HPV 11 (seroconversion rate: 96.30% for both).
At month 7, all participants in the PPS cohort achieved seroconversion for IgG against all nine HPV types, while one participant in the medium-dose group and another in the control group failed to seroconvert for nAbs against HPV 45 (seroconversion rates: 97.92% and 96.30%, respectively) (Table S5). Additionally, one participant in the high-dose group did not achieve seroconversion for nAbs against HPV 31 and HPV 58 (seroconversion rate: 97.96% for both).
The GMTs of nAbs for all nine HPV types showed substantial increases one month after the administration of two vaccine doses. A further increase in nAb titers was observed one month after the third dose, which was administered at month 6 (Figure 4). Notably, no clear dose-dependent escalation in antibody responses was observed among the three dose groups. Overall, the GMTs of antibodies were comparable across the different dose groups and the control group, except for the GMT of HPV 6 nAbs, which was significantly higher in the medium-dose group compared to the other groups. The GMT of HPV 33 nAbs was significantly higher in the high-dose group, while the GMT of HPV 45 nAbs was higher in the low-dose group compared to the other groups.
Similar trends were observed in the IgG antibody data (Figure 5). Significant differences in GMTs were noted among the groups for all nine HPV types. Specifically, the GMT of HPV 11 IgG was significantly higher in the medium-dose group compared to the others, while the low-dose group showed elevated IgG GMTs for the remaining HPV types.
All participants who were seronegative for nine HPV types at baseline successfully seroconverted for both nAbs and IgG against all HPV types at month 7. The GMI and GMTs for both nAbs and IgG at months 3 and 7 in these participants are presented in Tables S6 and S7, and Figures S1 and S2.
The exploratory analysis revealed that the levels of antibodies against GST and 3C protease at baseline and at months 3 and 7 post-vaccination were negative across all groups, with no statistically significant differences observed between the groups.

4. Discussion

This phase 1 clinical trial preliminarily indicated that the recombinant nonavalent HPV vaccine (E. coli) is safe and well tolerated in healthy women aged 18–45 years. Our findings show that the incidence of total ARs within 7 days after vaccination ranged from 56.67% to 90.00%, with a higher incidence in the low-dose group compared to the control group. Most reported AEs were mild or moderate, and no vaccine-related SAEs were reported. Laboratory findings further corroborated the favorable safety profile of the test vaccine. No significant differences were found in the incidence of abnormal laboratory parameters within 3 days of each dose across all four groups. Additionally, the exploratory analysis revealed that antibody levels against GST and 3C protease were negative at baseline and at months 3 and 7 post-vaccination across all groups, further confirming the safety of the trial vaccine.
Within 30 days following vaccination, the reported incidence of AEs was 90.00% in the low-dose group, 80.00% in the medium-dose group, and 82.00% in the high-dose group, which is comparable to the 83.30% observed with the E. coli-expressed 9vHPV vaccine (Cecolin9, Xiamen Innovax, Xiamen, China) [14]. Previous studies on the 9-valent HPV vaccine showed AR incidence rates of 60.00–70.00% in adults, with injection-site pain reported in 60.00% of participants [15]. In comparison to Gardasil 9, the candidate vaccine demonstrated a higher incidence of ARs within 7 days post-vaccination, which may be attributed to differences between the adjuvants and the production systems used [16]. The recombinant nonavalent HPV vaccine uses E. coli-derived VLPs (a prokaryotic system), whereas Gardasil 9 employs a yeast-derived system (eukaryotic). Prokaryotic systems like E. coli may introduce higher levels of endotoxins and bacterial proteins, which can lead to stronger local inflammatory responses, thus increasing the occurrence of ARs [17].
Furthermore, the low-dose group exhibited a significantly higher incidence of solicited reactions than the control group (90.00% vs. 56.67%, p = 0.004), such as injection-site pain (80.00% vs. 40.00%, p = 0.002) and induration (30.00% vs. 3.33%, p = 0.006). This elevated reactogenicity may be attributed to a stronger immune response triggered by the lower antigen dose, particularly at the injection site. These findings indicate that with similarly good immunogenicity, the medium-dose and high-dose options may be more suitable for further evaluation.
Robust immunogenicity was observed after three doses of the vaccine. The seroconversion rates for nAbs and IgG against all nine HPV types were over 97.92% at 7 months post-vaccination, which is similar to the findings from the phase 3 study of Gardasil 9 (99.60–100.00%) [18]. The GMTs for all nine HPV types showed a marked rise as early as one month after the second dose (month 3), supporting the WHO’s 2022 recommendation for two-dose schedules of HPV vaccine, especially in resource-limited settings [7]. Our findings suggest the potential for a two-dose nonavalent HPV vaccine, laying the groundwork for future trials assessing optimized two-dose regimens for women [10]. A further increase in nAb and IgG titers was observed one month after the third dose, which was administered at month 6. The robust immunogenic responses observed in this trial are consistent with results from early-phase studies of other E. coli-derived HPV vaccines. For instance, a phase 1 study evaluating an E. coli-based bivalent HPV vaccine reported similarly high seroconversion rates and GMTs for HPV 6/11 [19]. Similarly, a phase 2 trial of Cecolin9, also manufactured using E. coli, demonstrated a comparable rise in GMTs for all nine HPV types one month after both the second and third doses [10].
Given that HPV infects through the mucosal epithelium, mucosal immunity plays an important role in protection, with secretory immunoglobulin A (sIgA) serving as a key neutralizing antibody at mucosal surfaces. Previous studies have shown that intramuscularly administered HPV vaccines, such as Gardasil, can induce detectable levels of mucosal IgA [20]. As the candidate vaccine is produced using an E. coli expression system, its ability to induce mucosal immune responses, particularly sIgA at the cervicovaginal site, should be further investigated to provide a more comprehensive understanding of its protective potential.
The nAb levels against the nine HPV types across three dose groups were comparable to those in the control group. However, IgG GMTs varied significantly among the groups for all nine HPV types. Specifically, the medium-dose group exhibited higher GMTs for HPV 11 IgG, whereas the low-dose group showed significantly higher IgG levels for the remaining HPV types compared to the other groups, suggesting no dose-dependent immune responses across the dose groups. This may be attributed to the early saturation of immune responses, where even lower doses of the vaccine are sufficient to induce a strong immune reaction [21].
This study has several limitations. First, the study did not evaluate the immunization schedules of a single dose or two doses with a 6-month interval in parallel. Since the WHO recently recommended a two-dose regimen for adults, further research on this schedule is needed to provide additional evidence for policy-making. Second, in the analysis of the primary outcome, a per-protocol analysis was adopted, excluding those who did not complete the full vaccination and missed serum antibody results, which could potentially introduce selection bias. Additionally, the trial only includes women aged 18–45 years, which may not fully represent the broader population who could benefit from the vaccine. Other age groups, particularly adolescents aged 9–14 years (the target population for HPV vaccination), should be included in future studies.

5. Conclusions

In conclusion, this phase 1 clinical study demonstrated that the new recombinant nonavalent HPV vaccine (Escherichia coli) exhibits good tolerability and strong immunogenicity in healthy Chinese women aged 18–45 years. These findings indicate that further investigation is warranted in broader age groups and larger cohorts to assess its safety, immunogenicity, and protective efficacy.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/vaccines13050511/s1, Table S1: Adverse events within 30 days after each vaccination in women aged 27–45 years. Table S2: Adverse events within 30 days after each vaccination in women aged 18–26 years. Table S3: List of adverse events grade ≥ 3 that occurred within 30 days after vaccination. Table S4: Seroconversion rates of HPV type-specific neutralizing and IgG antibodies at month 3 in the PPS cohort. Table S5: Seroconversion rates of HPV type-specific neutralizing and IgG antibodies at month 7 in the PPS cohort. Table S6: Geometric mean increases of HPV type-specific neutralizing and IgG antibodies at month 3 in the participants who were seronegative for HPV types at baseline. Table S7: Geometric mean increases of HPV type-specific neutralizing and IgG antibodies at month 7 in the participants who were seronegative for HPV types at baseline. Figure S1: Neutralizing antibody titers at months 3 and 7 for HPV types 6/11/16/18/31/33/45/52/58 in the participants who were seronegative for HPV types at baseline. Figure S2: IgG antibody titers at months 3 and 7 for HPV types 6/11/16/18/31/33/45/52/58 in the participants who were seronegative for HPV types at baseline.

Author Contributions

Conceptualization, W.W.; Data curation, M.W., J.L., and W.H.; Formal analysis, Y.L., H.Y., and W.H.; Funding acquisition, J.L.; Investigation, M.W., J.Z., W.W. and J.L.; Methodology, Y.L., H.Y., and W.W.; Resources: Y.L. and H.Y.; Supervision, W.W.; Project administration, M.W., J.Z., and W.W.; Writing—original draft, M.W. and W.H.; Writing—review and editing, W.W., M.W. and J.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Natural Science Foundation of China (NSFC) and the Bill & Melinda Gates Foundation joint program (grant 7231101367/2023YFVA1002).

Institutional Review Board Statement

This study (NCT05680454) was conducted according to the guidelines of the Declaration of Helsinki and Good Clinical Practice guidelines and approved by the Ethics Committee of Jiangsu Provincial Center for Disease Prevention and Control (Approval code: JSJK2019-A003-01, approved on 1 April 2019).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data presented in this study are available upon request from the corresponding author.

Acknowledgments

The authors acknowledge Beijing Health Guard Biotechnology Inc. for providing the vaccines for this trial.

Conflicts of Interest

Yongjiang Liu and Hongyang Yu are employees of Beijing Health Guard Biotechnology Inc. All the other authors declare no competing interests.

Abbreviations

The following abbreviations are used in this manuscript:
AbbreviationFull Term
AEadverse event
ALTalanine aminotransferase
ARadverse reaction
APTTactivated partial thromboplastin time
ASTaspartate aminotransferase
CIconfidential interval
CREAcreatinine
E. coliEscherichia coli
ELISAenzyme-linked immunosorbent assay
FASfull analysis set
GMIgeometric mean increase
GMTgeometric mean titer
GSTglutathione-S-transferase
HBGhemoglobin
HPVhuman papillomavirus
IgGimmunoglobulin G
nAbneutralizing antibody
NMPANational Medical Products Administration
NIFDCChina National Institute for Food and Drug Control
PBNApseudovirion-based neutralization assay
PPSper-protocol set
PTprothrombin time
SAEserious adverse event
sIgAsecretory immunoglobulin A
SSsafety set
VLPvirus-like particle
WBCwhite blood cell count
WHOWorld Health Organization

References

  1. Wu, S.; Jiao, J.; Yue, X.; Wang, Y. Cervical cancer incidence, mortality, and burden in China: A time-trend analysis and comparison with England and India based on the global burden of disease study 2019. Front. Public. Health 2024, 12, 1358433. [Google Scholar] [CrossRef] [PubMed]
  2. World Health Organization. Cancer Today-World. 2024. Available online: https://gco.iarc.who.int/media/globocan/factsheets/populations/900-world-fact-sheet.pdf (accessed on 25 February 2025).
  3. World Health Organization. Cancer Today-China. 2024. Available online: https://gco.iarc.who.int/media/globocan/factsheets/populations/160-china-fact-sheet.pdf (accessed on 25 February 2025).
  4. National Cancer Institute. HPV and Cancer. 31 January 2025. Available online: https://www.cancer.gov/about-cancer/causes-prevention/risk/infectious-agents/hpv-and-cancer (accessed on 25 February 2025).
  5. De Martel, C.; Plummer, M.; Vignat, J.; Franceschi, S. Worldwide burden of cancer attributable to HPV by site, country and HPV type. Int. J. Cancer 2017, 141, 664–670. [Google Scholar] [CrossRef] [PubMed]
  6. de Sanjose, S.; Quint, W.G.; Alemany, L.; Geraets, D.T.; Klaustermeier, J.E.; Lloveras, B.; Tous, S.; Felix, A.; Bravo, L.E.; Shin, H.R.; et al. Human papillomavirus genotype attribution in invasive cervical cancer: A retrospective cross-sectional worldwide study. Lancet Oncol. 2010, 11, 1048–1056. [Google Scholar] [CrossRef] [PubMed]
  7. World Health Organization. Human Papillomavirus Vaccines: WHO Position Paper, December 2022. 2022. Available online: https://www.who.int/publications/i/item/who-wer9750-645-672 (accessed on 25 February 2025).
  8. Global Strategy to Accelerate the Elimination of Cervical Cancer as a Public Health Problem. 2020. Available online: https://www.who.int/publications/i/item/9789240014107 (accessed on 25 February 2025).
  9. ICO/IARC Information Centre on HPV and Cancer (HPV Information Centre). Human Papillomavirus and Related Diseases in the World. 2023. Available online: https://hpvcentre.net/statistics/reports/XWX.pdf?t=1693896342213 (accessed on 25 February 2025).
  10. Hu, Y.M.; Bi, Z.F.; Zheng, Y.; Zhang, L.; Zheng, F.Z.; Chu, K.; Li, Y.F.; Chen, Q.; Quan, J.L.; Hu, X.W.; et al. Immunogenicity and safety of an Escherichia coli-produced human papillomavirus (types 6/11/16/18/31/33/45/52/58) L1 virus-like-particle vaccine: A phase 2 double-blind, randomized, controlled trial. Sci. Bull. 2023, 68, 2448–2455. [Google Scholar] [CrossRef] [PubMed]
  11. Zhao, F.H.; Wu, T.; Hu, Y.M.; Wei, L.H.; Li, M.Q.; Huang, W.J.; Chen, W.; Huang, S.J.; Pan, Q.J.; Zhang, X.; et al. Efficacy, safety, and immunogenicity of an Escherichia coli-produced Human Papillomavirus (16 and 18) L1 virus-like-particle vaccine: End-of-study analysis of a phase 3, double-blind, randomised, controlled trial. Lancet Infect. Dis. 2022, 22, 1756–1768. [Google Scholar] [CrossRef] [PubMed]
  12. Qiao, Y.L.; Wu, T.; Li, R.C.; Hu, Y.M.; Wei, L.H.; Li, C.G.; Chen, W.; Huang, S.J.; Zhao, F.H.; Li, M.Q.; et al. Efficacy, Safety, and Immunogenicity of an Escherichia coli-Produced Bivalent Human Papillomavirus Vaccine: An Interim Analysis of a Randomized Clinical Trial. J. Natl. Cancer Inst. 2020, 112, 145–153. [Google Scholar] [CrossRef] [PubMed]
  13. Huang, X.; Wang, X.; Zhang, J.; Xia, N.; Zhao, Q. Escherichia coli-derived virus-like particles in vaccine development. NPJ Vaccines 2017, 2, 3. [Google Scholar] [CrossRef] [PubMed]
  14. Chu, K.; Bi, Z.F.; Huang, W.J.; Li, Y.F.; Zhang, L.; Yang, C.L.; Jiang, H.M.; Zang, X.; Chen, Q.; Liu, D.L.; et al. Safety and immunogenicity of an Escherichia coli-produced 9-valent human papillomavirus L1 virus-like particle vaccine (types 6/11/16/18/31/33/45/52/58) in healthy adults: An open-label, dose-escalation phase 1 clinical trial. Lancet Reg. Health West. Pac. 2023, 34, 100731. [Google Scholar] [CrossRef]
  15. Moreira Jr, E.D.; Block, S.L.; Ferris, D.; Giuliano, A.R.; Iversen, O.E.; Joura, E.A.; Kosalaraksa, P.; Schilling, A.; Van Damme, P.; Bornstein, J.; et al. Safety Profile of the 9-Valent HPV Vaccine: A Combined Analysis of 7 Phase III Clinical Trials. Pediatrics 2016, 138, e20154387. [Google Scholar] [CrossRef] [PubMed]
  16. Li, J.; Shi, L.W.; Yu, B.W.; Huang, L.R.; Zhou, L.Y.; Shi, L.; Jiang, Z.W.; Xia, J.L.; Wang, X.Y.; Li, R.C.; et al. Safety and immunogenicity of a pichia pastoris-expressed bivalent human papillomavirus (types 16 and 18) L1 virus-like particle vaccine in healthy Chinese women aged 9-45 years: A randomized, double-blind, placebo-controlled phase 1 clinical trial. Vaccine 2023, 41, 3141–3149. [Google Scholar] [CrossRef] [PubMed]
  17. Zhao, Q.; Potter, C.S.; Carragher, B.; Lander, G.; Sworen, J.; Towne, V.; Abraham, D.; Duncan, P.; Washabaugh, M.W.; Sitrin, R.D. Characterization of virus-like particles in GARDASIL® by cryo transmission electron microscopy. Hum. Vaccin. Immunother. 2014, 10, 734–739. [Google Scholar] [CrossRef] [PubMed]
  18. Huh, W.K.; Joura, E.A.; Giuliano, A.R.; Iversen, O.E.; de Andrade, R.P.; Ault, K.A.; Bartholomew, D.; Cestero, R.M.; Fedrizzi, E.N.; Hirschberg, A.L.; et al. Final efficacy, immunogenicity, and safety analyses of a nine-valent human papillomavirus vaccine in women aged 16-26 years: A randomised, double-blind trial. Lancet 2017, 390, 2143–2159. [Google Scholar] [CrossRef] [PubMed]
  19. Mo, Z.J.; Bi, Z.F.; Sheng, W.; Chen, Q.; Huang, T.; Li, M.Q.; Cui, X.L.; Wangjiang, Y.H.; Lin, B.Z.; Zheng, F.Z.; et al. Safety and immunogenicity of an Escherichia coli-produced bivalent human papillomavirus type 6/11 vaccine: A dose-escalation, randomized, double-blind, placebo-controlled phase 1 trial. Hum. Vaccin. Immunother. 2022, 18, 2092363. [Google Scholar] [CrossRef] [PubMed]
  20. Pattyn, J.; Van Keer, S.; Tjalma, W.; Matheeussen, V.; Van Damme, P.; Vorsters, A. Infection and vaccine-induced HPV-specific antibodies in cervicovaginal secretions. A review of the literature. Papillomavirus Res. 2019, 8, 100185. [Google Scholar] [CrossRef] [PubMed]
  21. Heldens, J.; Hulskotte, E.; Voeten, T.; Breedveld, B.; Verweij, P.; van Duijnhoven, W.; Rudenko, L.; van Damme, P.; van den Bosch, H. Safety and immunogenicity in man of a cell culture derived trivalent live attenuated seasonal influenza vaccine: A Phase I dose escalating study in healthy volunteers. Vaccine 2014, 32, 5118–5124. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Overview of study design.
Figure 1. Overview of study design.
Vaccines 13 00511 g001
Figure 2. Trial profile. The dose-escalation phase 1 study was carried out in two stages. Recruitment for Stage 2 commenced only after confirming the absence of vaccine-related serious adverse events within 7 days following the first dose administered in Stage 1. All the participants received three doses of the allocated vaccine according to the protocol. SS: safety analysis set; FAS: full analysis set; PPS: per-protocol set.
Figure 2. Trial profile. The dose-escalation phase 1 study was carried out in two stages. Recruitment for Stage 2 commenced only after confirming the absence of vaccine-related serious adverse events within 7 days following the first dose administered in Stage 1. All the participants received three doses of the allocated vaccine according to the protocol. SS: safety analysis set; FAS: full analysis set; PPS: per-protocol set.
Vaccines 13 00511 g002
Figure 3. The incidences of solicited adverse reactions within 7 days after each vaccination. The multiple comparisons were adjusted using the Bonferroni method, with αadjusted = α/6 = 0.008. Differences were considered statistically significant if p < 0.008. * The significance is at the p < 0.008 level.
Figure 3. The incidences of solicited adverse reactions within 7 days after each vaccination. The multiple comparisons were adjusted using the Bonferroni method, with αadjusted = α/6 = 0.008. Differences were considered statistically significant if p < 0.008. * The significance is at the p < 0.008 level.
Vaccines 13 00511 g003
Figure 4. Neutralizing antibody titers at months 0, 3, 7 for HPV types 6/11/16/18/31/33/45/52/58 in the PPS cohort. PPS: per-protocol set, which included participants who received all three vaccine doses and donated serum samples at months 0, 3, and 7, and had no major deviation of the protocol; The antibody level of the seronegative sample was artificially set as half of the cutoff value; The black lines indicate the GMTs and 95% CI; CI: confidence interval; GMTs: geometric mean titers; nAb: neutralizing antibody. The multiple comparisons were adjusted using the Bonferroni method, with αadjusted = α/6 = 0.008. Differences were considered statistically significant if p < 0.008. * The significance is at the p < 0.008 level. ** The significance is at the p < 0.001 level.
Figure 4. Neutralizing antibody titers at months 0, 3, 7 for HPV types 6/11/16/18/31/33/45/52/58 in the PPS cohort. PPS: per-protocol set, which included participants who received all three vaccine doses and donated serum samples at months 0, 3, and 7, and had no major deviation of the protocol; The antibody level of the seronegative sample was artificially set as half of the cutoff value; The black lines indicate the GMTs and 95% CI; CI: confidence interval; GMTs: geometric mean titers; nAb: neutralizing antibody. The multiple comparisons were adjusted using the Bonferroni method, with αadjusted = α/6 = 0.008. Differences were considered statistically significant if p < 0.008. * The significance is at the p < 0.008 level. ** The significance is at the p < 0.001 level.
Vaccines 13 00511 g004
Figure 5. IgG antibody titers at months 0, 3, 7 for HPV types 6/11/16/18/31/33/45/52/58 in the PPS cohort. PPS: per-protocol set, which included participants who received all three vaccine doses and donated serum samples at months 0, 3, and 7, and had no major deviation of the protocol; The antibody level of the seronegative sample was artificially set as half of the cutoff value; The black lines indicate the GMTs and 95% CI; CI: confidence interval; GMTs: geometric mean titers. The multiple comparisons were adjusted using the Bonferroni method, with αadjusted = α/6 = 0.008. Differences were considered statistically significant if p < 0.008. * The significance is at the p < 0.008 level. ** The significance is at the p < 0.001 level.
Figure 5. IgG antibody titers at months 0, 3, 7 for HPV types 6/11/16/18/31/33/45/52/58 in the PPS cohort. PPS: per-protocol set, which included participants who received all three vaccine doses and donated serum samples at months 0, 3, and 7, and had no major deviation of the protocol; The antibody level of the seronegative sample was artificially set as half of the cutoff value; The black lines indicate the GMTs and 95% CI; CI: confidence interval; GMTs: geometric mean titers. The multiple comparisons were adjusted using the Bonferroni method, with αadjusted = α/6 = 0.008. Differences were considered statistically significant if p < 0.008. * The significance is at the p < 0.008 level. ** The significance is at the p < 0.001 level.
Vaccines 13 00511 g005
Table 1. Baseline demographic characteristics of the participants.
Table 1. Baseline demographic characteristics of the participants.
CharacteristicsLow-Dose
(N = 30)
Medium-Dose
(N = 50)
High-Dose
(N = 50)
Control
(N = 30)
Total
(N = 160)
p Value
Age (years)      
Mean (SD)22.60 (2.18)28.90 (8.28)28.52 (8.10)22.73 (2.41)26.44 (7.20)<0.001
Median23.0025.5024.5023.0024.00 
Min, Max18.00, 26.0018.00, 45.0019.00, 45.0018.00, 26.0018.00, 45.00 
Height (cm)      
Mean (SD)160.07 (5.12)160.90 (5.46)161.14 (5.67)161.63 (7.65)160.96 (5.90)0.748
Median160.50161.00161.00162.00161.00 
Min, Max151.00, 171.00151.00, 172.00146.00, 173.00143.00, 181.00143.00, 181.00 
Weight (kg)      
Mean (SD)57.97 (9.61)59.54 (12.32)60.54 (10.39)58.57 (11.20)59.38 (10.99)0.678
Median55.5058.5058.5055.0058.00 
Min, Max44.00, 80.0041.00, 115.0045.00, 92.0041.00, 97.0041.00, 115.00 
BMI (kg/m2)      
Mean (SD)22.63 (3.62)22.94 (4.23)23.34 (3.99)22.33 (3.20)22.89 (3.85)0.693
Median21.7722.7422.8121.7522.40 
Min, Max16.94, 31.6415.06, 40.7515.57, 35.9416.63, 29.6115.06, 40.75 
SD: standard deviation. N: the number of people in each dataset. Age = (date of informed consent − date of birth)/365.25. p values were calculated using Pearson’s Chi-squared test or Fisher’s exact test.
Table 2. Adverse reactions within 7 days after each vaccination.
Table 2. Adverse reactions within 7 days after each vaccination.
Low-Dose
(N = 30)
Medium-Dose
(N = 50)
High-Dose
(N = 50)
Control
(N = 30)
Total
(N = 160)
p Value
EventsParticipants (%)EventsParticipants (%)EventsParticipants (%)EventsParticipants (%)EventsParticipants (%)
Total adverse reactions a9227 (90.00)8637 (74.00)10837 (74.00)5217 (56.67)338118 (73.75)0.035 b
Solicited adverse reactions9027 (90.00)8235 (70.00)10436 (72.00)4917 (56.67)325115 (71.88)0.039 b
Local solicited adverse reactions6525 (83.33)5930 (60.00)7234 (68.00)3616 (53.33)232105 (65.63)0.072
Systemic solicited adverse reactions2513 (43.33)2315 (30.00)3216 (32.00)137 (23.33)9351 (31.88)0.407
Unsolicited adverse reactions22 (6.67)44 (8.00)44 (8.00)32 (6.67)1312 (7.50)>0.999
Adverse reactions ≥ grade 3000022 (4.00)0022 (1.25)0.332
a Any vaccine-related adverse events. b The low-dose group showed a higher incidence than did the control group (p = 0.004). Except for the occurrences of ARs, each subject in each trial group was counted only once per row. p values were calculated using Pearson’s Chi-squared test or Fisher’s exact test. The multiple comparisons were adjusted using the Bonferroni method, with αadjusted = α/6 = 0.008. Differences were considered statistically significant if p < 0.008.
Table 3. Adverse events within 30 days after each vaccination.
Table 3. Adverse events within 30 days after each vaccination.
Low-Dose
(N = 30)
Medium-Dose
(N = 50)
High-Dose
(N = 50)
Control
(N = 30)
Total
(N = 160)
p Value
EventsParticipants (%)EventsParticipants (%)EventsParticipants (%)EventsParticipants (%)EventsParticipants (%)
Total adverse events11227 (90.00)10640 (80.00)13141 (82.00)6320 (66.67)412128 (80.00)0.149
Adverse reactions a9327 (90.00)8637 (74.00)11137 (74.00)5217 (56.67)342118 (73.75)0.035 b
Solicited adverse reactions9027 (90.00)8235 (70.00)10436 (72.00)4917 (56.67)325115 (71.88)0.039 b
Local solicited adverse reactions6525 (83.33)5930 (60.00)7234 (68.00)3616 (53.33)232105 (65.63)0.072
Systemic solicited adverse reactions2513 (43.33)2315 (30.00)3216 (32.00)137 (23.33)9351 (31.88)0.407
Unsolicited adverse reactions33 (10.00)44 (8.00)75 (10.00)32 (6.67)1714 (8.75)0.981
Solicited adverse events9027 (90.00)8235 (70.00)10936 (72.00)4917 (56.67)330115 (71.88)0.039 b
Unsolicited adverse events2212 (40.00)2414 (28.00)2214 (28.00)149 (30.00)8249 (30.63)0.666
Adverse events ≥ grade 3000022 (4.00)0022 (1.25)0.332
Serious adverse events0000000000-
Discontinuation due to adverse events0000000000-
a Any vaccine-related adverse events. b The low-dose group showed a higher incidence than did the control group (p = 0.004). Except for the occurrences of AEs, each subject in each trial group was counted only once per row. p values were calculated using Pearson’s Chi-squared test or Fisher’s exact test. The multiple comparisons were adjusted using the Bonferroni method, with αadjusted = α/6 = 0.008. Differences were considered statistically significant if p < 0.008.
Table 4. Summary of abnormal laboratory parameters after each dose of vaccination.
Table 4. Summary of abnormal laboratory parameters after each dose of vaccination.
IndicesLow-Dose
(N = 30)
Medium-Dose
(N = 50)
High-Dose
(N = 50)
Control
(N = 30)
Total
(N = 160)
p Value
Routine blood indicesWBC      
1st vaccination03 (6.00)2 (4.00)1 (3.33)6 (3.75)0.702
2nd vaccination001 (2.04)01 (0.63)>0.999
3rd vaccination3 (10.00)2 (4.17)3 (6.12)08 (5.19)0.421
HGB      
1st vaccination00000-
2nd vaccination01 (2.04)2 (4.08)03 (1.90)0.777
3rd vaccination01 (2.08)001 (0.65)0.682
Serum biochemical indicesALT      
1st vaccination1 (3.33)01 (2.00)02 (1.25)0.803
2nd vaccination1 (3.33)1 (2.04)1 (2.04)03 (1.90)>0.999
3rd vaccination1 (3.33)2 (4.17)003 (1.95)0.398
AST      
1st vaccination00000-
2nd vaccination01 (2.04)1 (2.04)02 (1.27)>0.999
3rd vaccination1 (3.33)1 (2.08)002 (1.30)0.676
CREA      
1st vaccination1 (3.33)01 (2.00)02 (1.25)0.803
2nd vaccination004 (8.16)04 (2.53)0.068
3rd vaccination2 (6.67)3 (6.25)1 (2.04)2 (7.41)8 (5.19)0.667
Blood coagulation function indicesAPTT      
1st vaccination01 (2.00)1 (2.00)02 (1.25)>0.999
2nd vaccination02 (4.08)2 (4.08)2 (6.67)6 (3.80)0.568
3rd vaccination02 (4.17)01 (3.70)3 (1.95)0.288
PT      
1st vaccination1 (3.33)001 (3.33)2 (1.25)0.139
2nd vaccination02 (4.08)002 (1.27)0.332
3rd vaccination1 (3.33)1 (2.08)1 (2.04)1 (3.70)4 (2.60)>0.999
WBC: white blood cell count; HGB: hemoglobin; ALT: alanine aminotransferase; AST: aspartate aminotransferase; CREA: creatinine; APTT: activated partial thromboplastin time; PT: prothrombin time. Post-vaccination abnormality: The test result before vaccination was “normal”, while the test result after vaccination was “abnormal”. Data for the 1st, 2nd, and 3rd dose analyses were based on SS1, SS2, and SS3, respectively. Data are n (%), unless stated otherwise. p values were calculated using Pearson’s Chi-squared test or Fisher’s exact test.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wei, M.; Han, W.; Zhang, J.; Liu, Y.; Yu, H.; Li, J.; Wang, W. Safety, Tolerability, and Immunogenicity of a Recombinant Nonavalent Human Papillomavirus Vaccine (Escherichia coli) in Healthy Chinese Women Aged 18–45 Years: A Phase 1 Clinical Trial. Vaccines 2025, 13, 511. https://doi.org/10.3390/vaccines13050511

AMA Style

Wei M, Han W, Zhang J, Liu Y, Yu H, Li J, Wang W. Safety, Tolerability, and Immunogenicity of a Recombinant Nonavalent Human Papillomavirus Vaccine (Escherichia coli) in Healthy Chinese Women Aged 18–45 Years: A Phase 1 Clinical Trial. Vaccines. 2025; 13(5):511. https://doi.org/10.3390/vaccines13050511

Chicago/Turabian Style

Wei, Mingwei, Weiwei Han, Jing Zhang, Yongjiang Liu, Hongyang Yu, Jingxin Li, and Wenjuan Wang. 2025. "Safety, Tolerability, and Immunogenicity of a Recombinant Nonavalent Human Papillomavirus Vaccine (Escherichia coli) in Healthy Chinese Women Aged 18–45 Years: A Phase 1 Clinical Trial" Vaccines 13, no. 5: 511. https://doi.org/10.3390/vaccines13050511

APA Style

Wei, M., Han, W., Zhang, J., Liu, Y., Yu, H., Li, J., & Wang, W. (2025). Safety, Tolerability, and Immunogenicity of a Recombinant Nonavalent Human Papillomavirus Vaccine (Escherichia coli) in Healthy Chinese Women Aged 18–45 Years: A Phase 1 Clinical Trial. Vaccines, 13(5), 511. https://doi.org/10.3390/vaccines13050511

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop