MedPath

Human papillomavirus 9-valent vaccine, recombinant (Merck & CO.) Advanced Drug Monograph

Published:Aug 29, 2025

A Comprehensive Clinical and Public Health Review of the Human Papillomavirus 9-valent Vaccine, Recombinant (GARDASIL 9)

I. Introduction and Product Characterization

Overview of Human Papillomavirus (HPV) as a Public Health Concern

Human Papillomavirus (HPV) represents the most prevalent sexually transmitted viral infection globally and is the etiological agent for a significant spectrum of human diseases, ranging from benign lesions to invasive malignancies.[1] While the majority of HPV infections are transient and immunologically cleared by cell-mediated immune responses within several years, a subset of infections with high-risk oncogenic HPV types can persist.[2] This persistence is the primary causal factor for the development of precancerous intraepithelial neoplasias and subsequent invasive cancers.[2] The public health burden of HPV is substantial, encompassing cervical, anal, oropharyngeal, vulvar, vaginal, and penile cancers.[3] In the United States, HPV-attributable oropharyngeal cancer has now surpassed cervical cancer as the most common HPV-related malignancy, underscoring the evolving epidemiology of the disease and the need for comprehensive prevention strategies that address multiple anatomical sites in both sexes.[6] Beyond malignancy, low-risk HPV types, primarily 6 and 11, are responsible for approximately 90% of cases of anogenital warts (condyloma acuminata) and recurrent respiratory papillomatosis, conditions that cause significant morbidity and psychological distress.[4] The management of HPV-related diseases also imposes a considerable economic burden on healthcare systems, involving costs associated with screening, diagnostic procedures like colposcopy, and treatments for precancerous and cancerous lesions.[2]

Development and Composition of the 9-valent Recombinant Vaccine

In response to this global health challenge, Merck & Co., through its subsidiary Merck Sharp & Dohme LLC, developed the Human Papillomavirus 9-valent Vaccine, Recombinant, marketed under the tradename GARDASIL 9.[3] This vaccine represents a significant advancement in prophylactic biotechnology. It is a non-infectious, sterile preparation composed of highly purified virus-like particles (VLPs) derived from the major capsid protein (L1) of nine distinct HPV types.[1]

The production of GARDASIL 9 utilizes recombinant DNA technology. The L1 gene for each of the nine HPV types is cloned into the yeast Saccharomyces cerevisiae. Following fermentation and protein expression, the L1 proteins self-assemble into VLPs, which are structurally and immunologically analogous to the native HPV virion but are devoid of any viral genetic material.[1] This composition ensures that the vaccine is incapable of causing infection, replication, or oncogenic transformation. The nine HPV types targeted by the vaccine were selected based on their global prevalence and disease association:

  • High-Risk (Oncogenic) Types: HPV 16, 18, 31, 33, 45, 52, and 58. These seven types are responsible for approximately 90% of cervical cancers and a high proportion of other HPV-related anogenital and oropharyngeal cancers.[4]
  • Low-Risk (Wart-Causing) Types: HPV 6 and 11. These two types cause the vast majority of anogenital warts.[4]

Each 0.5-mL dose of the vaccine contains a specific amount of L1 protein for each HPV type and is adjuvanted with amorphous aluminum hydroxyphosphate sulfate, which potentiates the immune response to the VLP antigens.[2]

Comparative Profile: Evolution from Bivalent and Quadrivalent to Nonavalent Formulations

GARDASIL 9 is the culmination of a multi-generational effort in HPV vaccine development, building upon the foundations laid by its predecessors. The expansion of HPV type coverage from the first- and second-generation vaccines to the current 9-valent formulation marks a critical step towards comprehensive cancer prevention.

  • Bivalent Vaccine (2vHPV, Cervarix): Developed by GlaxoSmithKline, this vaccine targets the two most prevalent high-risk HPV types, 16 and 18, which are responsible for approximately 70% of cervical cancers.[1] While it has demonstrated high efficacy against these types, it offers no protection against genital warts. It is no longer available in the United States but remains in use in other countries.[1]
  • Quadrivalent Vaccine (4vHPV, Gardasil): The first-generation HPV vaccine from Merck, Gardasil, targeted HPV types 6, 11, 16, and 18.[12] This formulation provided protection against the two main cancer-causing types and the two main wart-causing types. Gardasil has been superseded by GARDASIL 9 in the U.S. market and is no longer distributed.[13]
  • Nonavalent Vaccine (9vHPV, GARDASIL 9): This vaccine incorporates the four original types from Gardasil and adds five additional high-risk oncogenic types: 31, 33, 45, 52, and 58.[1] These five additional types account for an estimated 20% of cervical cancers, thereby increasing the potential protection against this malignancy from approximately 70% to 90%.[10]

This evolution from quadrivalent to 9-valent coverage represents a fundamental strategic shift in the public health approach to HPV prevention. The initial goal, enabled by the 4vHPV vaccine, was to prevent the majority of HPV-related cancers. While a monumental achievement, this left a significant proportion of disease burden unaddressed. The development of GARDASIL 9, by targeting the next five most common cancer-causing types, moved the goalpost from substantial reduction to near-complete prevention. This expanded coverage is a key technological enabler of ambitious global health initiatives, such as the World Health Organization's strategy to eliminate cervical cancer as a public health problem, transforming a once-abstract goal into a tangible possibility.

CharacteristicBivalent (2vHPV)Quadrivalent (4vHPV)9-valent (9vHPV)
Brand NameCervarixGardasilGARDASIL 9
ManufacturerGlaxoSmithKlineMerck & Co., Inc.Merck & Co., Inc.
HPV Types Covered16, 186, 11, 16, 186, 11, 16, 18, 31, 33, 45, 52, 58
Primary IndicationsCancers caused by HPV 16, 18Cancers caused by HPV 16, 18; Genital warts caused by HPV 6, 11Cancers caused by HPV 16, 18, 31, 33, 45, 52, 58; Genital warts caused by HPV 6, 11
AdjuvantAluminum hydroxide + ASO4Amorphous aluminum hydroxyphosphate sulfateAmorphous aluminum hydroxyphosphate sulfate
Current U.S. AvailabilityNoNo (Replaced by GARDASIL 9)Yes
Data compiled from.1

II. Immunological Mechanism of Prophylactic Protection

The Role of L1 Virus-Like Particles (VLPs) in Eliciting an Immune Response

The prophylactic efficacy of GARDASIL 9 is predicated on a sophisticated immunological mechanism centered on its L1 virus-like particle (VLP) technology. The VLPs are engineered to be structurally identical to the native HPV capsid, presenting the same conformational epitopes to the host immune system that would be encountered during a natural infection.[1] However, as these particles are composed solely of protein and contain no viral DNA, they are non-infectious and non-oncogenic.[1] When administered intramuscularly, the VLPs are recognized by antigen-presenting cells as foreign pathogens, initiating a cascade of immune events that leads to the generation of a robust and specific protective response.[1] This process effectively mimics the immunogenic properties of a live virus without posing any risk of disease.

Induction of a High-Titer, Durable Humoral Response

While the precise mechanism of action is still under investigation, it is widely hypothesized that the protection conferred by GARDASIL 9 is primarily mediated by the humoral immune system.[1] The introduction of VLPs into the body triggers a strong B-cell response, leading to the production of high titers of neutralizing immunoglobulin G (IgG) antibodies specific to the L1 proteins of the nine HPV types contained in the vaccine.[1] Clinical studies have demonstrated that more than 98% of vaccine recipients develop a detectable antibody response to all nine HPV types one month after completing the vaccination series.[8]

The magnitude of this response is a critical factor in the vaccine's success. Immunogenicity studies have shown that the peak antibody titers induced by the vaccine are 10- to 100-fold higher than those generated following a natural HPV infection.[1] This supraphysiological antibody level is believed to be essential for long-term protection. The prevailing model suggests that these high concentrations of serum IgG antibodies transudate from dermal capillaries across the basement membrane to the surface of the genital epithelium.[2] HPV infection occurs when virions gain access to the basal cells of the epithelium, typically through micro-abrasions that occur during sexual activity. In a vaccinated individual, the transudated antibodies are present at the site of potential infection and can bind to and neutralize the HPV virions, preventing them from attaching to and entering the basal cells, thereby blocking the initiation of infection.[2]

Immunological Superiority Compared to Natural Infection-Acquired Immunity

The mechanism of vaccine-induced immunity stands in stark contrast to the often inadequate immune response generated by natural HPV infection. Natural infections are typically restricted to the intraepithelial layer and are highly effective at evading systemic immune surveillance.[2] The virus does not cause viremia or significant cell lysis, resulting in a muted inflammatory response that often fails to trigger robust, systemic immunity. Consequently, only about half of individuals with a natural HPV infection develop detectable serum antibodies, and these titers are generally low and may wane over time, failing to confer reliable long-term protection against reinfection with the same HPV type.[2]

GARDASIL 9 overcomes these limitations. By delivering the VLP antigens intramuscularly along with an aluminum adjuvant, the vaccine bypasses the localized immune evasion strategies of the virus and forces a direct and powerful engagement with the systemic immune system.[2] This process effectively "unmasks" the viral antigens, leading to the generation of a high-titer, systemic antibody response and the formation of long-lived immunological memory. This fundamental difference explains why vaccination provides a level of protection that is qualitatively and quantitatively superior to that acquired through natural infection. It also provides the strong scientific rationale for recommending vaccination for all individuals within the approved age range, regardless of their prior sexual history or potential HPV exposure, as the vaccine-induced immunity is far more reliable and potent than any immunity that may have been naturally acquired.

III. Clinical Efficacy and Long-Term Durability of Protection

The clinical development program for GARDASIL 9 was designed to rigorously establish its safety, immunogenicity, and efficacy in preventing HPV-related diseases. The evidence from pivotal trials and long-term follow-up studies provides a robust foundation for its use in public health programs worldwide.

Pivotal Phase III Trial Results in Adolescent and Adult Populations

The efficacy of GARDASIL 9 was established through a series of large-scale, randomized, controlled clinical trials involving tens of thousands of participants across various age groups and geographic regions.[15] The cornerstone of the efficacy data was a pivotal Phase III study conducted in approximately 14,000 women aged 16 to 26 years.[15] This study employed an active comparator design, with participants randomized to receive either GARDASIL 9 or the quadrivalent Gardasil vaccine. This design allowed for a direct assessment of the additional protection conferred by the five new HPV types in GARDASIL 9, while simultaneously confirming its non-inferior immunogenicity for the four shared HPV types (6, 11, 16, 18).[15]

For the primary target population of young adolescents (ages 9-15), establishing direct efficacy against cancer or high-grade precancerous lesions is not feasible, as these outcomes can take decades to develop. Therefore, regulatory approval for this age group was based on the principle of "immunobridging".[8] This approach involves demonstrating that the antibody responses generated by the vaccine in the younger population are non-inferior (as good as or better than) the responses observed in the older population (16-26 years) in whom clinical efficacy has been directly proven. Immunogenicity trials in approximately 1,200 males and 2,800 females aged 9-15 confirmed that their antibody responses to GARDASIL 9 were comparable or superior to those in the 16-26 age group, allowing for the logical inference of similar high efficacy.[8] This immunobridging strategy represents a sophisticated and pragmatic evolution in vaccine development, balancing rigorous scientific standards with the urgent public health need to protect adolescents before the onset of sexual activity, the primary window of opportunity for HPV prevention.

Efficacy Against the Five Additional HPV Types (31, 33, 45, 52, 58)

The primary objective of the pivotal efficacy trial was to determine the effectiveness of GARDASIL 9 in preventing disease caused by the five additional high-risk HPV types (31, 33, 45, 52, and 58). The results were compelling. In the per-protocol efficacy analysis, GARDASIL 9 was 96.7% effective in preventing the combined endpoint of high-grade cervical, vulvar, and vaginal disease (Cervical Intraepithelial Neoplasia [CIN] grade 2 or higher, Adenocarcinoma in situ, Vulvar Intraepithelial Neoplasia [VIN] grade 2 or higher, and Vaginal Intraepithelial Neoplasia [VaIN] grade 2 or higher) caused by these five HPV types.[19] This high efficacy was demonstrated by the observation of only one case of high-grade disease in the GARDASIL 9 group (n=6,016) compared to 30 cases in the Gardasil (4vHPV) control group (n=6,017) over the course of the study.[19]

Demonstrated Protection Against Anogenital Warts and Precancerous Lesions

GARDASIL 9's efficacy against diseases caused by the four shared HPV types (6, 11, 16, and 18) was established through immunogenicity non-inferiority relative to the quadrivalent Gardasil vaccine, for which efficacy has been extensively documented.[15] The original Gardasil clinical trials demonstrated nearly 100% efficacy in preventing high-grade cervical precancerous lesions (CIN 2/3 and AIS) related to HPV 16 and 18 in HPV-naïve populations.[8] Similarly, efficacy against HPV 6 and 11-related genital warts was approximately 99%.[8] Based on the comparable antibody responses, GARDASIL 9 is expected to provide the same high level of protection against diseases caused by these four types.[15]

The prevention of anal cancer is a key indication for the vaccine. Due to the lower incidence of anal cancer, direct efficacy data for this endpoint is more limited. The indication is supported by the demonstrated effectiveness of the quadrivalent Gardasil vaccine, which showed 78% efficacy against HPV 6/11/16/18-related high-grade anal intraepithelial neoplasia (AIN 2/3), the immediate precursor to anal cancer.[15] This is further supported by robust immunogenicity data for GARDASIL 9 in both males and females.[15]

Long-Term Follow-Up Studies: Evidence of Sustained Immunogenicity and Effectiveness

A critical question for any prophylactic vaccine is the duration of protection. Extensive long-term follow-up studies and post-marketing surveillance have provided reassuring evidence of the durability of protection conferred by HPV vaccination. Data from clinical trials and real-world monitoring show that protection lasts for more than 10 years, with no evidence of waning effectiveness over time.[5]

A landmark long-term extension study followed a cohort of individuals who were vaccinated with GARDASIL 9 at ages 9 to 15.[10] At 10 years post-vaccination (Month 126), antibody titers remained elevated, and seropositivity rates were robust, remaining at or above 95% for all nine vaccine types as measured by a sensitive immunoassay. Most importantly, the clinical effectiveness was sustained. After a median follow-up of 10 years, there were zero reported cases of high-grade intraepithelial neoplasia or condyloma related to any of the nine vaccine-targeted HPV types among the study participants.[10] These findings provide strong evidence that the immune response generated by GARDASIL 9 is both potent and highly durable, offering long-lasting protection through the period of highest risk for HPV acquisition and disease development.

IV. Regulatory Approvals and Evolving Indications

The regulatory history of GARDASIL 9 illustrates a dynamic process of evidence accumulation and indication expansion, reflecting a growing understanding of the vaccine's broad utility in cancer prevention. Its journey from initial approval for adolescents to its current indications for a wide adult population and for non-anogenital cancers highlights its increasing importance in public health.

Chronology of U.S. Food and Drug Administration (FDA) Approvals

The U.S. FDA has granted a series of key approvals that have progressively broadened the scope of GARDASIL 9's use:

  • Initial Approval (December 10, 2014): The FDA first approved GARDASIL 9 for the prevention of certain cancers and diseases caused by the nine targeted HPV types. The initial indication was for use in females aged 9 through 26 and males aged 9 through 15.[14]
  • Expanded Male Indication (December 15, 2015): The FDA expanded the age indication for males to include those aged 16 through 26, aligning the recommended age range for both sexes.[14]
  • Age Expansion to 45 Years (October 5, 2018): In a landmark decision, the FDA approved a supplemental Biologics License Application (sBLA) extending the use of GARDASIL 9 to include women and men aged 27 through 45 years.[14] This approval was based on long-term follow-up efficacy and effectiveness data from studies of the quadrivalent Gardasil vaccine, which demonstrated significant protection in this older age group.[23]
  • Accelerated Approval for Oropharyngeal and Head and Neck Cancers (June 12, 2020): Recognizing the rising incidence of HPV-related head and neck cancers, the FDA granted accelerated approval for GARDASIL 9 for the prevention of oropharyngeal and other head and neck cancers caused by HPV types 16, 18, 31, 33, 45, 52, and 58.[6] This approval was based on the vaccine's demonstrated effectiveness in preventing HPV-related anogenital disease, with the understanding that the underlying mechanism of protection is likely similar across different mucosal sites. Continued approval for this indication is contingent upon verification of clinical benefit in an ongoing confirmatory trial.[24]

This regulatory trajectory, particularly the expansion to age 45 and the inclusion of head and neck cancers, signifies a paradigm shift in the vaccine's application. It has evolved from being perceived primarily as a pediatric or adolescent intervention to prevent cervical cancer into a comprehensive cancer prevention tool applicable across a much wider segment of the adult population. This reflects a more nuanced understanding of HPV transmission dynamics, which can continue throughout life, and acknowledges the significant and growing burden of non-cervical HPV-related malignancies.

European Medicines Agency (EMA) Marketing Authorisation

GARDASIL 9 also underwent a thorough review process in the European Union:

  • Positive Opinion (March 27, 2015): The EMA's Committee for Medicinal Products for Human Use (CHMP) issued a positive opinion, recommending the vaccine for marketing authorisation.[27]
  • Marketing Authorisation (June 10, 2015): The European Commission granted a marketing authorisation for GARDASIL 9, valid throughout the European Union. The initial approval was for a 3-dose schedule for individuals from the age of 9 years.[28]
  • Two-Dose Schedule Approval (April 7, 2016): Based on new immunogenicity data, the EMA approved a 2-dose schedule for adolescents aged 9 to 14 years, aligning the vaccine's label with the public health recommendations in several European countries and facilitating its integration into national immunization programs.[30]

Detailed Breakdown of Approved Indications

The FDA-approved indications for GARDASIL 9 are extensive, reflecting its broad-spectrum activity against multiple HPV-related diseases. The specific indications differ slightly for females and males but cover a wide range of cancers, precancerous lesions, and genital warts.

Indication for Females (Ages 9-45)Indication for Males (Ages 9-45)
Cancers PreventedCancers Prevented
• Cervical, vulvar, vaginal, anal, oropharyngeal and other head and neck cancers caused by HPV types 16, 18, 31, 33, 45, 52, and 58.• Anal, oropharyngeal and other head and neck cancers caused by HPV types 16, 18, 31, 33, 45, 52, and 58.
Precancerous or Dysplastic Lesions PreventedPrecancerous or Dysplastic Lesions Prevented
• Cervical intraepithelial neoplasia (CIN) grades 1, 2, and 3• Anal intraepithelial neoplasia (AIN) grades 1, 2, and 3
• Cervical adenocarcinoma in situ (AIS)
• Vulvar intraepithelial neoplasia (VIN) grades 2 and 3
• Vaginal intraepithelial neoplasia (VaIN) grades 2 and 3
• Anal intraepithelial neoplasia (AIN) grades 1, 2, and 3
(All lesions caused by HPV types 6, 11, 16, 18, 31, 33, 45, 52, and 58)(All lesions caused by HPV types 6, 11, 16, 18, 31, 33, 45, 52, and 58)
Genital Warts PreventedGenital Warts Prevented
• Genital warts (condyloma acuminata) caused by HPV types 6 and 11.• Genital warts (condyloma acuminata) caused by HPV types 6 and 11.
Data compiled from U.S. FDA prescribing information.4

V. Comprehensive Safety and Tolerability Profile

The safety and tolerability of GARDASIL 9 have been extensively evaluated in pre-licensure clinical trials and through robust post-marketing surveillance systems. The cumulative evidence from these sources confirms a favorable safety profile.

Analysis of Adverse Events from Pre-Licensure Clinical Trials

The safety of GARDASIL 9 was a primary endpoint in clinical trials that enrolled more than 15,000 males and females.[17] The data from these studies consistently demonstrated that the vaccine is well-tolerated. The most frequently reported adverse reactions were mild-to-moderate, transient local injection-site events and systemic effects.

  • Local Reactions: The most common adverse events were related to the injection site, which is expected for an adjuvanted, intramuscular vaccine. These included:
  • Pain: Reported by approximately 89-90% of recipients.[20]
  • Swelling: Reported by approximately 48% of recipients.[24]
  • Erythema (redness): Reported by approximately 34% of recipients.[24]
  • Other less common local reactions included pruritus (itching), bruising, bleeding, and the formation of a lump at the injection site.[33]
  • Systemic Reactions: Systemic adverse events were generally mild and self-limiting. The most common included:
  • Headache: Reported by approximately 11-12% of recipients.[13]
  • Fever: Reported in a similar proportion of vaccine and placebo recipients.[17]
  • Other reported systemic effects included nausea, dizziness, fatigue, diarrhea, abdominal pain, and sore throat.[33]

Post-Marketing Surveillance Data and Long-Term Safety Monitoring

Following licensure, the safety of GARDASIL 9 has been continuously monitored through passive and active surveillance systems, such as the Vaccine Adverse Event Reporting System (VAERS) in the United States. With over 135 million doses of HPV vaccines distributed in the U.S. alone, the post-marketing data provides a vast real-world dataset confirming the safety profile observed in clinical trials.[18] Over more than 15 years of monitoring, no new or unexpected serious adverse events have been causally linked to HPV vaccination.[18]

Contraindications, Warnings, and Precautions

The prescribing information for GARDASIL 9 includes specific contraindications, warnings, and precautions to guide safe administration.

  • Contraindications: The vaccine is strictly contraindicated for any individual with a history of a hypersensitivity reaction to any vaccine component, including a severe allergic reaction (e.g., anaphylaxis) to yeast, as the L1 VLP antigens are produced in Saccharomyces cerevisiae.[3] It is also contraindicated in individuals who have had a hypersensitivity reaction to a previous dose of GARDASIL 9 or the quadrivalent Gardasil vaccine.[3]
  • Warnings and Precautions (Syncope): One of the most prominent warnings associated with GARDASIL 9 is the potential for syncope (fainting). Post-marketing reports have included cases of syncope, sometimes associated with tonic-clonic (seizure-like) movements and other vasovagal responses, following vaccination.[25] It is critical to recognize that this phenomenon is not a pharmacological reaction to the vaccine's components but rather a well-documented stress-related physiological response to the process of injection itself (needle-related vasovagal syncope).[17] This response is particularly common among adolescents receiving any vaccine, not just HPV vaccine.[18] To mitigate the risk of injury from a fall during a syncopal event, official recommendations from the CDC and the manufacturer state that vaccine recipients should be observed while seated or lying down for 15 minutes after administration.[3] This simple procedural safeguard effectively manages the risk and is crucial for maintaining public confidence by correctly attributing the event to the injection process rather than the vaccine's intrinsic safety.
  • Other Precautions: Vaccination should be deferred for individuals experiencing a moderate or severe acute illness. However, a minor illness, such as a mild upper respiratory infection, is not a contraindication to vaccination.[17]

Considerations for Special Populations

  • Pregnancy: GARDASIL 9 has not been studied in pregnant women, and therefore, its use is not recommended during pregnancy.[3] If a woman initiates the vaccination series and subsequently discovers she is pregnant, the completion of the series should be postponed until after the pregnancy. No intervention is needed if a dose is inadvertently administered during pregnancy.[9]
  • Immunocompromised Individuals: Individuals with compromised immune systems due to disease (e.g., HIV infection) or immunosuppressive therapy may have a diminished antibody response to the vaccine. For this reason, a 3-dose schedule is recommended for all immunocompromised persons aged 9 through 26 years to maximize their potential for a protective immune response.[17]

VI. Public Health Framework: Recommendations and Real-World Impact

The translation of GARDASIL 9's robust clinical trial data into effective public health programs is guided by recommendations from national and international health bodies. The real-world impact of these programs, now evident after more than a decade of use, validates the vaccine's role as a transformative tool in cancer prevention.

U.S. Centers for Disease Control and Prevention (CDC) / ACIP Vaccination Guidelines

In the United States, the Advisory Committee on Immunization Practices (ACIP) provides evidence-based recommendations for vaccine use. The guidelines for HPV vaccination are designed to maximize prevention by targeting individuals before they are likely to be exposed to the virus.

  • Routine Vaccination: The ACIP routinely recommends HPV vaccination for all adolescents at age 11 or 12 years. The vaccination series can be initiated as early as age 9.[17] This timing aligns with other adolescent vaccines and aims to ensure protection well before the onset of sexual activity for most individuals.
  • Catch-Up Vaccination: For individuals who were not vaccinated at the recommended age, catch-up vaccination is recommended for everyone through age 26.[17]
  • Shared Clinical Decision-Making (Ages 27-45): For adults aged 27 through 45 years, the ACIP does not recommend routine vaccination for everyone, as many individuals in this age group will have already been exposed to HPV, thus deriving less benefit. However, it recommends shared clinical decision-making, wherein a patient and their clinician can discuss individual risks and potential benefits to decide if vaccination is appropriate.[17]
  • Dosing Schedules: The recommended schedule is age-dependent, reflecting immunogenicity data showing that younger adolescents mount a more robust immune response:
  • 2-Dose Schedule: For individuals initiating the series before their 15th birthday, two doses are recommended, with the second dose administered 6 to 12 months after the first.[3]
  • 3-Dose Schedule: For individuals initiating the series on or after their 15th birthday, and for all immunocompromised individuals, a 3-dose schedule is recommended at 0, 1-2, and 6 months.[3]
Age at Series InitiationRecommended ScheduleNotes / Special Considerations
9 through 14 years2-dose series at 0, 6-12 monthsIf the second dose is given less than 5 months after the first, a third dose is required.
15 through 45 years3-dose series at 0, 1-2, 6 monthsThis schedule applies to all individuals starting the series at age 15 or older.
Immunocompromised Persons (9-26 years)3-dose series at 0, 1-2, 6 monthsA 3-dose series is recommended to ensure an adequate immune response in this population.
Data compiled from CDC/ACIP recommendations.3

World Health Organization (WHO) Vaccination Guidelines

The WHO's recommendations are tailored for a global audience, with a focus on maximizing impact in diverse healthcare settings, including those with limited resources.

  • The primary target population for HPV vaccination programs globally is adolescent girls aged 9-14 years, prior to sexual debut.[11]
  • In a significant policy update in December 2022, the WHO's Strategic Advisory Group of Experts on Immunization (SAGE) endorsed a flexible schedule of one or two doses for the primary target group of girls aged 9-14 and for young women aged 15-20.[11] This change was based on emerging evidence suggesting that a single dose provides a high degree of protection and can greatly simplify logistical challenges, thereby increasing vaccination coverage.
  • For women older than 21, two doses are recommended. For individuals who are immunocompromised or living with HIV, a minimum of two, and preferably three, doses are recommended to ensure a sufficient immune response.[11]

The differing recommendations between the CDC and WHO highlight a key principle in public health strategy: the balancing of optimal individual protection with maximal population-level impact. The CDC's recommendations, designed for a high-resource setting, are based on the 2- and 3-dose regimens that demonstrated the highest immunogenicity in clinical trials. The WHO's more flexible approach, including a single-dose option, is a pragmatic strategy designed to overcome the significant cost and logistical barriers that hinder multi-dose vaccine programs in many low- and middle-income countries.[37] This approach recognizes that vaccinating a larger proportion of the population with one dose may ultimately prevent more cancers globally than vaccinating a smaller proportion with two or three doses.

Evidence of Population-Level Effectiveness

The ultimate measure of a vaccine's success is its real-world impact on disease. More than a decade of post-licensure data from countries with established HPV vaccination programs has provided powerful evidence of GARDASIL 9's population-level effectiveness.

  • Decline in HPV Prevalence: In the United States, surveillance data has shown dramatic reductions in the prevalence of vaccine-targeted HPV types. Within 12 years of vaccine introduction, infections with the four HPV types included in the original Gardasil vaccine had decreased by 88% among females aged 14-19 and by 81% among females aged 20-24.[18]
  • Decline in Anogenital Warts: The impact on clinical disease was observed relatively quickly. Between 2006 and 2014, the prevalence of anogenital warts in the U.S. decreased by 61% among females aged 15-19.[18]
  • Decline in Cervical Precancers: As vaccinated cohorts have aged into the screening population, a steep decline in cervical precancers has been documented. Data from the CDC's HPV-IMPACT project showed that between 2008 and 2022, the incidence of high-grade cervical precancers (CIN2+) among screened women aged 20-24 years plummeted by 79%.[38] A significant 37% decrease was also observed for the first time in the 25-29 age group, demonstrating the expanding impact of the program.[38]
  • Decline in Cancer Incidence: The final proof of efficacy—prevention of invasive cancer—is now emerging. A landmark population-based study from Sweden, which followed nearly 1.7 million women, found that HPV vaccination was associated with a 63% reduced risk of cervical cancer overall. The reduction was most profound among women vaccinated before age 17, who experienced a nearly 90% decrease in cervical cancer incidence compared to their unvaccinated peers.[39] More recently, a 2024 retrospective study found a 56% reduction in the risk of developing HPV-associated head and neck cancers among vaccinated men, providing the first real-world evidence of the vaccine's impact on these malignancies.[40]

VII. Synthesis and Concluding Remarks

Summary of the Vaccine's Strengths and Limitations

The Human Papillomavirus 9-valent Vaccine, Recombinant (GARDASIL 9) represents a pinnacle of achievement in modern preventive medicine. Its development and implementation have fundamentally altered the landscape of HPV-related disease control.

  • Strengths: The vaccine's primary strength lies in its high and durable efficacy, combined with its broad-spectrum coverage. By targeting nine HPV types, it has the potential to prevent approximately 90% of cervical cancers and a similarly high proportion of other HPV-related malignancies and genital warts.[15] This efficacy is supported by a robust and long-lasting immune response that is far superior to naturally acquired immunity.[1] Furthermore, its safety profile is excellent, established through rigorous clinical trials and extensive post-marketing surveillance of hundreds of millions of doses worldwide.[18] Its broad indications for both sexes across a wide age range (9-45 years) maximize its potential public health utility.[4]
  • Limitations: It is essential to recognize the vaccine's limitations. GARDASIL 9 is a prophylactic vaccine; it is not a treatment for existing HPV infections, anogenital warts, or HPV-related precancerous or cancerous lesions.[3] Its efficacy is highest in individuals who are vaccinated before exposure to the vaccine-targeted HPV types. The vaccine does not protect against diseases caused by HPV types not contained within the vaccine, nor does it prevent all cases of the cancers for which it is indicated, as some are caused by non-HPV etiologies.[24] Consequently, vaccination does not eliminate the need for routine cancer screening, such as cervical cytology (Pap tests), which should continue according to standard public health recommendations.[3] Finally, as with any vaccine, protection may not be achieved in all recipients.[13]

Future Directions: Ongoing Research and Investigational Regimens

The science of HPV prevention continues to evolve. Merck is actively conducting clinical trials to further optimize the use of GARDASIL 9, including studies to assess the efficacy and durability of a single-dose regimen.[26] Success in this area could dramatically improve vaccine uptake and program feasibility, particularly in resource-constrained settings. Research is also underway to develop next-generation, multi-valent HPV vaccines with the potential to protect against an even broader array of HPV types.[26] Additionally, studies are exploring the immunogenicity of the vaccine in specific populations, such as transgender individuals undergoing gender-affirming hormone therapy, to ensure equitable protection for all groups.[42]

The Integral Role of GARDASIL 9 in the Oncologic Prevention Paradigm

In conclusion, GARDASIL 9 is more than just a vaccine against an infectious agent; it is a validated and powerful tool for cancer prevention. Its success story serves as a compelling model of translational medicine, demonstrating a seamless progression from fundamental molecular biology and virology to innovative vaccine engineering, rigorous clinical validation, strategic regulatory approval, and ultimately, transformative public health impact. The remarkable real-world data showing dramatic declines in HPV prevalence, precancerous lesions, and now, invasive cancers, provides unequivocal evidence of its value. GARDASIL 9 stands as a cornerstone of the modern oncologic prevention paradigm, shifting the approach to several major cancers from a reliance on secondary prevention (screening) and treatment to the far more desirable state of primary prevention. Its continued and expanded global implementation is critical to the ongoing effort to reduce the burden of HPV-associated malignancies and is an indispensable component of the global strategy to eliminate cervical cancer as a public health threat.

Works cited

  1. Human Papilloma Virus Vaccine - StatPearls - NCBI Bookshelf, accessed August 29, 2025, https://www.ncbi.nlm.nih.gov/books/NBK562186/
  2. Review of Gardasil - PMC, accessed August 29, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3690661/
  3. GARDASIL®9 (Human Papillomavirus 9-valent Vaccine, Recombinant) | Official Site, accessed August 29, 2025, https://www.gardasil9.com/
  4. GARDASIL 9 | FDA, accessed August 29, 2025, https://www.fda.gov/vaccines-blood-biologics/vaccines/gardasil-9
  5. Long-Term Effects of Human Papillomavirus Vaccination in Clinical Trials and Real-World Data: A Systematic Review - MDPI, accessed August 29, 2025, https://www.mdpi.com/2076-393X/10/2/256
  6. FDA Approves Nine-Valent HPV Vaccine for the Prevention of Certain HPV-Related Head and Neck Cancers - The ASCO Post, accessed August 29, 2025, https://ascopost.com/issues/july-10-2020/fda-approves-nine-valent-hpv-vaccine-for-the-prevention-of-certain-hpv-related-head-and-neck-cancers/
  7. Update on Safety and Efficacy of HPV Vaccines: Focus on Gardasil - PMC, accessed August 29, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8496244/
  8. HPV Vaccine Efficacy - CDC, accessed August 29, 2025, https://www.cdc.gov/vaccines/vpd/hpv/hcp/vaccines.html
  9. Use of 9-Valent Human Papillomavirus (HPV) Vaccine: Updated HPV Vaccination Recommendations of the Advisory Committee on Immunization Practices - PMC, accessed August 29, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4584883/
  10. Ten-Year Follow-up of 9-Valent Human Papillomavirus Vaccine: Immunogenicity, Effectiveness, and Safety - AAP Publications, accessed August 29, 2025, https://publications.aap.org/pediatrics/article/152/4/e2022060993/193886/Ten-Year-Follow-up-of-9-Valent-Human
  11. Human papillomavirus (HPV) - Immunization, Vaccines and Biologicals, accessed August 29, 2025, https://www.who.int/teams/immunization-vaccines-and-biologicals/diseases/human-papillomavirus-vaccines-(HPV)
  12. GARDASIL | FDA, accessed August 29, 2025, https://www.fda.gov/vaccines-blood-biologics/vaccines/gardasil
  13. Merck Statement on a Potential Reduced Dosing Recommendation for GARDASIL®9, accessed August 29, 2025, https://www.merck.com/news/merck-statement-on-a-potential-reduced-dosing-recommendation-for-gardasil9/
  14. Gardasil 9 (human papillomavirus 9-valent vaccine, recombinant) FDA Approval History, accessed August 29, 2025, https://www.drugs.com/history/gardasil-9.html
  15. FDA Approves Gardasil 9 for Prevention of Certain Cancers Caused by Five Additional Types of HPV - Archive | ESMO, accessed August 29, 2025, https://www.esmo.org/oncology-news/archive/fda-approves-gardasil-9-for-prevention-of-certain-cancers-caused-by-five-additional-types-of-hpv
  16. Gardasil 9 (human papillomavirus 9-valent vaccine) - Uses ... - WebMD, accessed August 29, 2025, https://www.webmd.com/drugs/2/drug-144456/gardasil-pf-intramuscular/details
  17. HPV Vaccination Recommendations - CDC, accessed August 29, 2025, https://www.cdc.gov/vaccines/vpd/hpv/hcp/recommendations.html
  18. HPV Vaccine Safety and Effectiveness Data - CDC, accessed August 29, 2025, https://www.cdc.gov/hpv/hcp/vaccination-considerations/safety-and-effectiveness-data.html
  19. Efficacy of GARDASIL®9 (Human Papillomavirus 9-valent Vaccine ..., accessed August 29, 2025, https://www.merckvaccines.com/gardasil9/efficacy/
  20. FDA Approves Merck's HPV Vaccine, GARDASIL®9, to Prevent Cancers and Other Diseases Caused by Nine HPV types – Including Types that Cause About 90% of Cervical Cancer Cases, accessed August 29, 2025, https://www.merck.com/news/fda-approves-mercks-hpv-vaccine-gardasil9-to-prevent-cancers-and-other-diseases-caused-by-nine-hpv-types-including-types-that-cause-about-90-of-cervical-cancer-cases/
  21. Gardasil - Wikipedia, accessed August 29, 2025, https://en.wikipedia.org/wiki/Gardasil
  22. FDA Approves Gardasil 9 HPV Vaccine for Use Up to Age 45 - OncLive, accessed August 29, 2025, https://www.onclive.com/view/fda-approves-gardasil-9-hpv-vaccine-for-use-up-to-age-45
  23. Gardasil Now FDA Approved for Individuals 27 to 45 Years Old - PcMED Project, accessed August 29, 2025, https://pcmedproject.com/alerts/gardasil-now-fda-approved-for-individuals-27-to-45-years-old/
  24. Package Insert - GARDASIL 9 - FDA, accessed August 29, 2025, https://www.fda.gov/media/90064/download
  25. FDA Approves Merck's GARDASIL 9 for the Prevention of Certain HPV-Related Head and Neck Cancers, accessed August 29, 2025, https://www.merck.com/news/fda-approves-mercks-gardasil-9-for-the-prevention-of-certain-hpv-related-head-and-neck-cancers/
  26. Merck Announces Plans to Conduct Clinical Trials of a Novel Investigational Multi-Valent Human Papillomavirus (HPV) Vaccine and Single-Dose Regimen for GARDASIL®9, accessed August 29, 2025, https://www.merck.com/news/merck-announces-plans-to-conduct-clinical-trials-of-a-novel-investigational-multi-valent-human-papillomavirus-hpv-vaccine-and-single-dose-regimen-for-gardasil-9/
  27. HPV vaccine Gardasil 9 is approved by European drug regulator, accessed August 29, 2025, https://pharmaceutical-journal.com/article/news/hpv-vaccine-gardasil-9-is-approved-by-european-drug-regulator
  28. GARDASIL 9 HPV vaccine approved in the European Union, accessed August 29, 2025, https://www.europeanpharmaceuticalreview.com/news/32521/gardasil-9-hpv-vaccine-approved-in-the-european-union/
  29. Gardasil 9 | European Medicines Agency (EMA), accessed August 29, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/gardasil-9
  30. GARDASIL® 9: 2-dose schedule approved in Europe - Infovac, accessed August 29, 2025, https://www.infovac.fr/docman-marc/public/bulletins/2016/1241-lien-2-gardasil-9-2dose-schedule-approved-in-europe
  31. GARDASIL® 9: 2-dose schedule approved in Europe - MultiVu, accessed August 29, 2025, http://www.multivu.com/players/uk/7805051-gardasil-9-2-dose-approved-in-europe/
  32. Human Papillomavirus Vaccine (HPV) - AAFP, accessed August 29, 2025, https://www.aafp.org/family-physician/patient-care/prevention-wellness/immunizations-vaccines/disease-pop-immunization/human-papillomavirus-vaccine-hpv.html
  33. Safety Profile and Possible Side Effects of GARDASIL®9 (Human Papillomavirus 9-valent Vaccine, Recombinant), accessed August 29, 2025, https://www.gardasil9.com/patient-a/what-is-gardasil-9/safety-and-side-effects/
  34. GARDASIL®9 (Human Papillomavirus 9-valent Vaccine, Recombinant) for Health Care Professionals, accessed August 29, 2025, https://www.merckvaccines.com/gardasil9/
  35. Gardasil Package Insert - FDA, accessed August 29, 2025, https://www.fda.gov/files/vaccines,%20blood%20%26%20biologics/published/Package-Insert---Gardasil.pdf
  36. Discussing HPV-related Cancers & Diseases for Appropriate Patients - Merck Vaccines, accessed August 29, 2025, https://www.merckvaccines.com/gardasil9/resources/patient-pd/
  37. Estimating the Impact of Human Papillomavirus (HPV) Vaccination on HPV Prevalence and Cervical Cancer Incidence in Mali | Clinical Infectious Diseases | Oxford Academic, accessed August 29, 2025, https://academic.oup.com/cid/article/52/5/641/384642
  38. Trends in Cervical Precancers Identified Through Population-Based Surveillance — Human Papillomavirus Vaccine Impact Monitoring Project, Five Sites, United States, 2008–2022 | MMWR - CDC, accessed August 29, 2025, https://www.cdc.gov/mmwr/volumes/74/wr/mm7406a4.htm
  39. Study Confirms HPV Vaccine Prevents Cervical Cancer - NCI, accessed August 29, 2025, https://www.cancer.gov/news-events/cancer-currents-blog/2020/hpv-vaccine-prevents-cervical-cancer-sweden-study
  40. 2023-24 HPV Vaccination Impact Report, accessed August 29, 2025, https://hpvroundtable.org/wp-content/uploads/2024/07/2023-24-HPV-Vaccination-Impact-Report-2.pdf
  41. Gardasil 9, INN-Human Papillomavirus 9 valent Vaccine (Recombinant, adsorbed) - European Medicines Agency, accessed August 29, 2025, https://www.ema.europa.eu/en/documents/product-information/gardasil-9-epar-product-information_en.pdf
  42. Evaluating Gardasil HPV Vaccine Humoral and Cellular Immune Responses in People With and Without HIV | ClinicalTrials.gov, accessed August 29, 2025, https://clinicaltrials.gov/study/NCT06624839

Published at: August 29, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.