Jump to content
RemedySpot.com

RE: HPV vaccine

Rate this topic


Guest guest

Recommended Posts

I do

have a personal subscription and do believe it is worth it as I use it

regularly while the patient is in the room with me. The write ups are pretty

comprehensive and seem balanced. On the negative side: these are “expert

opinions” so it may not reflect true evidence based literature. On the positive

side, it automatically tracks every time you look something up, so you get CME

for it (although the AAFP only recognizes up to 20 hours/year).

P.S. I am just a customer,

I have no financial ties to UpToDate.

Re:

HPV vaccine

Thanks!

Do you have a personal subscription?

Do you think the expense is worth it? For the CME?

I personally regard it as a superb resource, I have access through my hospitals

and wonder if I should invest in it for my personal use (the hospital access is

only through their intranet).

--

Pedro Ballester, M.D.

Warren, OH

Link to comment
Share on other sites

,

Thanks for posting this.  I read it, I thought in detail and am

very confused now.  I also am a doubting as far as the value of HPV. 

Everything says that cervical cancer is a problem in other countries so send

the vaccine there.

The numbers aren’t adding up.  I know they did studies that like

100% of college graduates have HPV by the time they graduate and Gardasil did

an excellent job preventing that.  But HPV also has a 90% spontaneous

resolution rate.  So does this mean that we are vaccinating 100 people to

prevent HPV in 10% and that’s all types?

Also, they say that cancer takes 10 years to form, but they did

studies that show decreased cancer that only lasted 5 years?  This confused

me.  And when they announce that there was no cancer found in these studies,

remember these studies only lasted 3-5 years so you wouldn’t expect to find

cervical cancer or CIN1 or CIN2 in this time period in someone who was HPV naïve

at the start of the study.

Are we going to block the body’s ability to get spontaneous

resolution?  It looks that way.  I don’t know.  But what adverse outcomes were

recently released?

Kathy Saradarian, MD

Branchville, NJ

www.qualityfamilypractice.com

Solo 4/03, Practicing since 9/90

Practice Partner 5/03

Low staffing

From:

[mailto: ] On Behalf Of Brady

Sent: Monday, December 15, 2008 9:21 PM

To: practiceimprovement1

Subject: Brady has sent you an UpToDate

topic

Below is a topic taken from UpToDate

that I thought you might find interesting.

UpToDate is a clinical information service that has thousands of topics

such as this one, designed to give immediate answers to clinical questions.

Visit them on the web at www.uptodate.com.

©2008

UpToDate ®

Official topic review from UpToDate®, the clinical

information service on the web, desktop, and PDA devices. To subscribe to UpToDate®, visit us online at http://www.uptodate.com/home/store/index.do.

Human

papillomavirus vaccines

Author

Diane M Harper, MD, MPH, MS

Section Editor

S Hirsch, MD

Deputy Editor

Barbara H McGovern, MD

INTRODUCTION — Human

papillomaviruses (HPV) are highly prevalent, tissue-specific, DNA viruses that

infect epithelial cells [1]

.. Persistent viral infection with oncogenic types of HPV leads to cancer of the

cervix, anus, vagina, vulva, penis, mouth, and sinuses [2-4]

.. Cervical cancer is the second most common malignant disease in women

worldwide and has a bimodal onset in the third and sixth decades of life [2]

.. (See " Epidemiology; clinical features; and diagnosis of invasive

cervical cancer " ).

Use of cervical cytology has reduced the incidence of cervical cancer by 70

percent. However, cervical cancer remains a leading cause of death in countries

without screening programs. Vaccines that prevent these persistent HPV infections

have the potential to further reduce the burden of disease [5-8]

..

Clinical data on the use of these candidate vaccines will be reviewed here.

The epidemiology, virology, pathogenesis, clinical manifestations, and

treatment of HPV are discussed separately. (See " Clinical presentation and

diagnosis of human papillomavirus infections " and see " Epidemiology

of human papillomavirus infections " and see " Virology of human

papillomavirus infections and the link to cancer " and see " Treatment

and prevention of human papillomavirus infections " ).

HPV TYPES — Specific HPV types

are associated with squamous intraepithelial lesions. HPV 16, 18, 45, and 31

cause over half of the low grade lesions, and about 65 percent of the high

grade lesions. HPV 6 and 11 cause 12 percent of low grade lesions [9]

and approximately 80 percent of genital warts [9-15]

.. Up to 40 percent of patients are infected with more than one HPV type. (See

" Epidemiology of human papillomavirus infections " ).

HPV 16 and HPV 18 are associated with approximately 50 and 20 percent of

cervical cancers, respectively [16,17]

.. HPV 45 and 31 are the next most common HPV types associated with cervical

cancer, accounting for another five percent each. The first peak of oncogenic

HPV infection occurs between the ages of 15 to 25 years, with a secondary peak

in the sixth decade of life [18]

..

It is estimated that targeted HPV protection with the bivalent or

quadrivalent vaccine would prevent half of the high-grade precancerous lesions

(CIN 2 or 3) and two thirds of the invasive cancers. The quadrivalent vaccine

would also prevent most genital warts [19]

..

LIFE CYCLE AND THE IMMUNE RESPONSE — Human

papillomaviruses are small, nonenveloped viruses, whose circular genome encodes

the encapsulating structural proteins, L1 and L2 [20]

.. The life cycle of the virus is integrally linked to the maturation of the

keratinocyte. Initial infection occurs in the basal stem cell. Specific gene

products are transcribed at every level of differentiation of the squamous

keratinocyte [20]

.. At the most superficial level, the L1 protein coat is transcribed, and upon

desquamation of this short-lived cell, HPV virions are infective for

transmission to the next basal stem cell in the adjacent mucosal epithelium.

(See " Virology of human papillomavirus infections and the link to

cancer " ).

HPV infection evades the immune system through a variety of mechanisms. HPV

infection leads to down regulation of interferon expression and regulatory

pathways, which subsequently prohibits the activation of cytotoxic

T-lymphocytes [21]

.. No associated cytolysis or release of pro-inflammatory cytokines occurs as a

result of basal cell infection [21]

.. In the absence of the usual signals that identify a virally-infected cell,

the cellular immune system is not activated and HPV infection persists [21]

.. The importance of cellular immunity is clinically apparent in HIV-infected

patients and in renal transplant patients, who have a higher incidence of

HPV-related disease [22]

.. (See " Preinvasive and invasive cervical neoplasia in HIV-infected women " ).

Approximately 75 to 90 percent of HPV infections will clear within a year of

initial infection [17]

.. Clearance is mediated by the natural desquamation of epithelial cells,

cell-mediated immunity and in part, by low-levels of neutralizing antibody

responses to the specific HPV L1 epitope [23]

.. There is a limited antibody response from natural infection and a significant

loss of these antibodies within three years [24,25]

..

HPV infection induces a time-limited, type-specific immunity to HPV

infections [26]

.. Individuals who are infected with one HPV type may develop protective

antibodies against future infection for a period of time with that specific

type, but remain immunologically naive to other HPV types. Thus, vaccine design

strategies need to incorporate the most common disease-causing HPV types in

order to protect against the majority of cervical intraepithelial neoplasias

and cervical cancers. The interval between acquisition of incident HPV

infection and malignancy usually takes ten years or longer [27]

..

ANAMNESTIC RESPONSE — HPV

infects only the epithelium and does not have a viremic phase. HPV actively

evades immune recognition. In vaccinated women, therefore, it is unlikely there

will be any antibody response with reinfection of type specific HPV. Protective

antibody levels will be dependent on initial and sustained vaccine responses or

requisite booster shots.

At this time, most alum-based vaccinations have booster shots recommended at

7 to 10 year intervals, to maintain disease protection. This concept is

important for HPV disease since the benefits of vaccination are to prevent long

term cancer development, and not immediate epidemic protection as has been the

standard for infectious diseases, such as pertussis, meningitis, or polio.

Sustained antibody titers have been reported with use of an ASO4 adjuvant

system in the bivalent vaccine, which specifically is designed to prime antigen

presenting cells and may lengthen B cell memory response [28]

..

TOOLS FOR ASSESSMENT OF VACCINE EFFICACY — Vaccine

efficacy is measured only by the prevention of virologic infection or clinical

events. The immunogenicity of the vaccines are not equated with vaccine

efficacy for three reasons:

The antibody titers are unique for each HPV type

and not comparable across types.

The antibody titers for each vaccine are in

different noncomparable assay units (milli-ELISA units/mL and milli-Merck

units/mL).

There is no established correlate between a titer

level and protection against HPV infection or the development of

CIN/cancer.

Absolute values of antibody titers are not comparable within or between

studies. Therefore, immunogenicity is measured by comparing type-specific,

vaccine-induced titers to natural infection titers and then expressed as a

ratio at a specific point in time after vaccination. HPV antibody titer levels

at or above infection titers are generally considered to be protective.

Vaccine efficacy for cervical cancer is defined for several natural history

events that all serve as surrogate markers for the development of cervical

cancer:

Incident HPV infection

Persistent type-specific HPV infection (>6 or

>12 months)

Abnormal cytology

Abnormal histologic changes

VACCINE DEVELOPMENT — The HPV

L1 major capsid protein self-assembles into virus-like particles (VLPs) for

each HPV type [29]

.. Because the VLPs are hollow, without inner HPV DNA cores, the vaccine itself

is not infectious. Preclinical research demonstrated that L1 is highly

immunogenic with and without an adjuvant [29]

.. The major capsid protein, L1, is the only portion of HPV used in vaccine

design [30]

..

The proof-of-concept vaccine trial with HPV 16 VLP [31]

demonstrated that HPV infection could be prevented. This vaccine, which is no

longer being used in clinical trials, led to the development of the following

immunizations: the bivalent HPV 16/18 VLP vaccine, and the quadrivalent HPV

6/11/16/18 VLP vaccine.

BIVALENT HPV 16/18 L1 VIRUS-LIKE PARTICLE

VACCINE

Safety and efficacy — The

efficacy of a bivalent HPV 16/18 L1 virus-like particle vaccine was

demonstrated in a double blind, randomized, placebo-controlled trial in 1113

women, 15 to 25 years of age [32,33]

.. Study participants were randomly assigned to receive 20 micrograms each of

HPV type 16 and 18 VLPs with ASO4 adjuvant or placebo at months 0, 1 and 6 with

follow-up to 5.5 years [34]

.. The trial results were significant for the following observations:

Protection against incident HPV 16/18 infection

occurred in 96 percent of women who received vaccination according to

protocol and in 91 percent of all participants, including those who

received vaccine doses at irregular times.

Protection against persistent HPV 16 and 18

infections (≥ 12 months) occurred in 100 percent of women who received

vaccination and follow up visits according to protocol; and in 96 percent

of all participants.

The vaccine was generally safe and well

tolerated; no new chronic diseases occurred during follow up.

Greater than 98 percent of the vaccine

recipients sustained seroconversion for more than 5.5 years for both HPV

types.

No cases of CIN caused by HPV 16/18 were

reported in the vaccine group, compared to 11 cases in the placebo group.

Protection against incident HPV 45 and HPV 31

infections occurred in 88 and 53 percent of all women enrolled in the

trial, respectively.

In another study in women 15 to 55 years of age, the bivalent HPV vaccine

was safe and immunogenic for at least two years, including those women in the

oldest age group (46 to 55 years of age) [35]

..

An interim analysis of the phase III efficacy trial of the bivalent HPV vaccine

was conducted when 23 cases of CIN2 or greater with HPV 16 or 18 DNA were

identified among 18,644 women who had been randomly assigned to HPV vaccine or

control vaccine (hepatitis A vaccine) [36]

.. Cases of CIN 2 or greater were noted in two participants in the HPV vaccine

group and in 21 participants in the control group. Based on this interim

analysis with a mean length of follow-up of 14.8 months, HPV vaccine efficacy

was estimated to be greater than 90 percent among women who did not have

evidence of HPV 16/18 infection at baseline.

QUADRIVALENT HPV 6/11/16/18 L1 VIRUS-LIKE

PARTICLE VACCINE

Safety and efficacy — The

efficacy of a quadrivalent HPV 6/11/16/18 L1 VLP vaccine has been demonstrated

in a number of trials (show

table 1A-1D), including a double blind, placebo-controlled trial in 552

women (mean age 20 years) [23]

.. Participants were given either alum placebo or vaccine, composed of 20

micrograms each of HPV types 6 and 18 VLPs and 40 micrograms each of HPV types

11 and 16 VLPs with alum, at months, 0, 2, and 6 with follow up through three

years. The trial results were significant for the following results:

Overall protection against persistent HPV 16/18

infection (>4 months) occurred in 89 percent of women who received

vaccination and follow up visits according to protocol, with 100 percent,

86 percent, and 89 percent efficacy for HPV 6, 16, and 18, respectively.

There were no cases of HPV 11 infection reported.

Protection against CIN caused by HPV 6/11/16/18

occurred in 100 percent of all participants who were not infected with

these types at first vaccination while seven cases occurred in the placebo

group.

Protection against all external genital warts

caused by HPV 6/11 occurred in 100 percent of all participants with four

cases occurring in the placebo group.

The vaccine was generally safe and well

tolerated.

After three vaccine doses, seroconversion rates

were 94, 96, 100, and 76 percent for HPV types 6, 11, 16, and 18 at month

36, respectively [37]

..

Two hundred forty-one of these study participants were subsequently enrolled

in an extension trial to obtain an additional two years of follow-up data [38]

.. The combined incidence of HPV 6/11/16/18-related persistent infection or

disease was reduced by 96 percent in vaccine recipients (two cases in the

vaccine group versus 46 in the placebo group).

FUTURE II trial — In a phase

III, multinational prospective, double-blind, placebo-controlled trial (FUTURE

II), more than 12,000 women, aged 15 to 26 years (with a lifetime number of no

more than four sexual partners), were randomly assigned to receive a three-dose

regimen of vaccine or placebo [39,40]

.. The majority of the study participants were from Europe (65 percent) and

Latin America (26 percent). At baseline, cervical cytology was abnormal in 11

percent of both vaccine and placebo groups; approximately 16 and 7 percent had

evidence of HPV 16 and 18 infection, respectively.

The primary efficacy analysis was performed in those subjects who did not

have evidence of either HPV-16 or 18 infection (by DNA or serologic testing)

through one month after the third dose of vaccine; these patients were referred

to as " HPV susceptible " per protocol. The primary composite end point

was CIN 2 or 3, adenocarcinoma in situ, or cervical cancer related to HPV-16 or

HPV-18. The mean duration of follow-up was three years. The study demonstrated

the following results:

Vaccine efficacy for the prevention of the

primary composite end point was 98 percent in study participants who were

" HPV susceptible " .

Vaccine efficacy remained high (95 percent) in

those HPV-negative participants who did not receive all doses of vaccine according

to protocol, suggesting some flexibility in the timing of the vaccine

schedule.

Seroconversion rates at 24 months among 1512

vaccinated women in the immunogenicity substudy were 96, 97, 99, and 68

percent for HPV types 6, 11, 16, and 18, respectively.

The vaccine was well-tolerated and there were

no serious adverse events.

However, vaccine efficacy for CIN2 or 3 disease due to all HPV types was

significantly lower (44 percent) in the overall population of women who had

undergone randomization, which included participants with baseline or incident

HPV infection by one month after the last dose of vaccine.

FUTURE 1 trial — A similarly

designed phase III placebo controlled trial was conducted in 5455 women aged 16

to 24 years to assess the efficacy of quadrivalent vaccine to prevent

HPV-related anogenital disease (FUTURE I) [41]

.. The primary aim of the trial was to determine vaccine efficacy in reducing

the combined incidence of anogenital warts, vulvar or vaginal intraepithelial

neoplasia grades 1 to 3 or cancer associated with HPV vaccine types 6,11,16, or

18. A secondary aim was to observe whether the administration of vaccine

reduced the combined incidence of CIN grades 1 to 3, adenocarcinoma in situ, or

cancer associated with vaccine-type HPV.

The following results were demonstrated after a mean follow-up of three

years:

The vaccine was 100 percent effective in

preventing anogenital disease in women who were " HPV

susceptible " (ie, no cases were identified in the vaccine group

versus 60 cases in the placebo group).

Vaccine efficacy was 100 percent in preventing

CIN grades 1 to 3 or adenocarcinoma in situ with vaccine type HPV in those

women who were " HPV susceptible " (ie, no cases were diagnosed in

the vaccine group, whereas 65 cases were diagnosed in placebo group).

As noted in the FUTURE II trial, vaccine

efficacy was lower when women with baseline HPV infection were included in

the intent-to-treat analysis (ie, 73 percent efficacy in prevention of

external anogenital or vaginal lesions and 55 percent efficacy in the

prevention of CIN).

There was no clear evidence that vaccination altered the course of disease

or infection present before administration of the first dose of vaccine in both

FUTURE I and II trials. These data reinforce the use of the HPV vaccine as a

prophylactic, and not a therapeutic immunization. (See " Vaccine

utilization " below).

A summary of the efficacy data from four placebo controlled, double-blind,

randomized Phase II and III clinical studies of the quadrivalent vaccine may

also be viewed on the website for the US Food and Drug Administration (www.fda.gov).

Recommendations — The

Advisory Committee on Immunization Practices (ACIP) and the American College of

Obstetricians and Gynecologists (ACOG) have recommended that quadrivalent HPV

6/11/16/18 L1 VLP vaccine should be administered in girls and women 9 to 26

years of age [42,43]

.. (See " Human papillomavirus quadrivalent (types 6, 11, 16, 18)

recombinant vaccine: Drug information " ).

Other vaccine approaches have been considered for immunotherapy of cervical

cancer as well as prevention of the acquisition of HPV infection. (See

" Virology of human papillomavirus infections and the link to

cancer " ).

SPECIFIC ISSUES

Vaccine safety — Since

virus-like particles (VLPs) are recombinant proteins, manufactured in benign

biologic systems (yeast and Baculovirus), they have no known oncogenic or

disease-causing potential [19]

..

The clinical trials of the bivalent and quadrivalent vaccines have

demonstrated mild injection site reactions [23,32]

; no serious adverse events have been recorded, including one study with more

than five years of follow-up [34]

.. Furthermore, HPV vaccine has been studied in diverse populations of women in

both developed and developing countries [39,41]

..

The vaccine adverse events reporting system (VAERS) has registered five

cases and confirmed two cases of Guillain-Barre syndrome (GBS) after

quadrivalent vaccine administration. Ongoing reporting continues for adverse events

from coadministration of the quadrivalent vaccine with other vaccines, as there

has been limited data available prior to regulatory approval.

Anaphylaxis has been reported following administration of the quadrivalent

vaccine and the incidence appears to be higher than that of most other

routinely-administered vaccines, although this complication is still rare. (See

" Anaphylactic reactions to vaccines " ).

Neither HPV vaccine contains live virus: the quadrivalent vaccine has been

classified as a category B drug by the FDA. However, use in pregnancy is not

recommended because of limited data on safety in this setting. In the FUTURE II

trial, pregnancy occurred in 1053 women in the vaccine group and 1106 in the

placebo group; no obvious anomalies attributable to vaccine were observed [39]

..

The manufacturer maintains a pregnancy registry to monitor fetal outcomes of

pregnant women exposed to the vaccine. Exposures can be reported by calling

[44]

.. However, lactating women can receive the immunization series since subunit

vaccines do not affect the safety of infant breast-feeding. (See

" Immunizations in pregnant women " ).

Cost-effectiveness — Mathematical

models have examined the cost-effectiveness of HPV vaccination [45-48]

.. One study suggested that vaccination of the entire United States population

of 12 year-old girls would prevent more than 200,000 HPV infections, 100,000

abnormal cervical cytology examinations, and 3300 cases of cervical cancer if

cervical cancer screening continued as currently recommended [45]

.. Studies have demonstrated that the effectiveness of HPV vaccination will

depend on the duration of vaccine immunity, including one study which supported

continued cost-effectiveness when the vaccine was given to 12 to 26 year-old

females [49]

..

Vaccinating both men and women is predicted to be more beneficial in

reducing HPV infection and disease than by vaccinating only women, but at a

higher cost [50,51]

.. However, models of cost-effectiveness are plagued by substantial uncertainty

regarding major issues such as duration of protection [52]

, the effect of herd immunity, and the prevalence of vaccine-specific HPV types

circulating in the population [53]

.. (See " Epidemiology of human papillomavirus infections " ).

Vaccine utilization — Clinical

trials of the HPV vaccines suggest high efficacy and an excellent safety

profile in women who do not have abnormal cervical cytology or HPV infection at

the time of immunization.

Several questions remain regarding vaccine utilization including [19]

:

The duration of protection is unknown;

protective antibodies persist for at least five years. Duration of

protection is important in determining when booster vaccines will be

needed [54]

..

The precise level of antibody needed for

protection against infection is also unknown.

HPV exposure occurs at all ages of life, starting in infancy [55,56]

.. The risk of HPV exposure increases with the number of sexual partners,

regardless of the age at which new partners are acquired [57]

.. Decisions about the age at which to initiate HPV immunization have been

guided by epidemiologic data regarding ages of peak HPV acquisition and

estimated duration of vaccine protection [4,18,26]

.. HPV vaccination has been recommended by the ACIP in girls and women 9 to 26

years of age.

The American Cancer Society (ACS) guidelines are in general agreement with the

ACIP recommendations about the need for vaccination in young females [57]

..

Effective vaccination strategies will require further education of the

public on the importance of HPV infection and its consequences [58]

.. A study of 400 women in a university setting demonstrated that awareness and

knowledge of HPV was very limited [59]

.. Vaccination against cervical cancer will be especially important in

developing countries, where nearly 80 percent of cancer cases are reported, and

where screening cytologies are rarely used [60]

..

Molecular-based testing for HPV detection is NOT recommended prior to

immunization [61]

.. (See " Clinical presentation and diagnosis of human papillomavirus

infections " , section on Diagnosis).

Efficacy in established infection — Since

there are many HPV types that can infect the cervix, there are important

distinctions that need to be made about the efficacy of the HPV vaccine in

women with prior versus current HPV infection.

The HPV vaccine does not clear HPV infection

that is present at the time of vaccination [61]

..

The absence of therapeutic efficacy for established infection was

illustrated in a trial of 2189 women with cervical HPV DNA who were randomly

assigned to receive either three doses of bivalent HPV-16/18 L1 virus-like

particle vaccine or a control (hepatitis A) vaccine [62]

.. There was no evidence of increased viral clearance of established infection

at 6 or 12 months in the group that received HPV vaccine.

The HPV vaccine can still protect women with

current HPV infections from acquisition of additional HPV types (ie, HPV

6,11,16,18) [63]

..

The vaccine was effective in preventing CIN 2/3 or adenocarcinoma in situ in

a subgroup of 4722 women from the FUTURE 1 and FUTURE 2 studies who had

evidence of current infection with at least one cancer-causing HPV vaccine type

(16 or 18) at the time of vaccination, but were naive to the other vaccine type

[63]

.. These data suggest that vaccinating a woman currently infected with 16 or 18

can still confer benefit in preventing infection with the other oncogenic HPV

types.

IMPORTANCE OF CERVICAL SCREENING — Appropriate

use of cervical cytology has reduced the death rate from cervical cancer by 70

percent in the past five decades [64]

.. However, there are limitations to cervical screening:

Many women do not seek or receive cervical

screening, especially minority groups and recent immigrants.

The results of cervical cytology smears cannot

be used to make a definitive diagnosis or to initiate treatment (See

" The cervical cytology report " ).

Despite optimal cytologic screening strategies,

cervical cancers still occur [65]

..

Cytologic screening is of paramount importance since HPV immunization is NOT

effective in clearing cytologically evident disease or infection. Furthermore,

one large US-based prevalence study found that although HPV infection was

highly prevalent, specific HPV vaccine types were not [66]

.. Strategic linking of immunization initiatives to cervical screening programs

may be effective in reducing the number of abnormal cytology results and in

reducing the number of cervical cancers not detected by cervical cytology [10]

.. (See " The cervical cytology report " and see " Epidemiology of

human papillomavirus infections " , section on Prevalence).

Cytologic screening is recommended for any woman who has been sexually

active for three or more years or is 21 years of age. A preventive health care

visit in which vaccination is discussed or offered represents an ideal

opportunity to offer Pap screening to sexually active patients as well [57]

..

In contrast, in the young child or adolescent, the pubertal, physiologic,

and psychological development of the young child or adolescent needs to be

considered; thus cervical screening should not be considered until at least

three years have passed after the onset of sexual activity.

PREVENTION OF VULVAL AND VAGINAL LESIONS — Vulvar

and vaginal cancers are uncommon complications of HPV infection, which are

preceded by vulva intraepithelial neoplasia (VIN2-3) and vaginal

intraepithelial neoplasia (VaIN2-3). Data from three placebo-controlled

clinical trials evaluating the quadrivalent HPV vaccine for cervical cancer

were combined to assess the potential effect of immunization on the prevention

of moderate to severe vulvar intraepithelial neoplasia (VIN-2 and VIN-3) [36]

.. Among 15,596 women who were naive to HPV 16 or 18 infection through one month

after the final dose of vaccine, immunization was 100 percent effective against

the development of VaIN-2/3 and VIN-2/3 caused by HPV 16/18 over a mean

follow-up time of three years (ie, no cases demonstrated in vaccinated group;

13 in the placebo group). In the intention to treat population of 18,174 women,

vaccine efficacy was 49 percent for VaIN or VIN grades 2 or 3, irrespective of HPV

type (27 cases in the vaccinated group; 53 cases in the placebo group).

VACCINATION IN MEN — The

potential strategy of using HPV vaccine in men to prevent cervical cancer in

women is being planned [30]

.. Studies of male vaccination to prevent HPV-associated genital warts and

cancers occurring in men are underway [19,67]

..

Excellent immunogenicity of the HPV vaccine has been demonstrated in male

adolescents [68,69]

.. In one randomized, double-blind trial 1781 children, ranging in age from 9 to

15 years, were assigned 2:1 to quadrivalent HPV vaccine or placebo.

Approximately half of the study participants were young boys. The trial was

significant for the following results:

At month seven, seroconversion rates were ≥

99.5 percent for the four vaccine types in both genders.

Although geometric mean titers declined by

month 18, antibody levels were comparable for each HPV type when comparing

responses between genders.

The vaccine was well-tolerated.

Mathematical models suggest that male vaccination may not be cost-effective

for the prevention of cervical cancer in women [45,57,70]

..

FUTURE IMPACT OF HPV VACCINE — The

impact of HPV vaccination programs on reduction of cervical cancer in the

future will be dependent on multiple issues, such as the amount of circulating

HPV types in the population, the percent of patients vaccinated, and the

duration of protection [57]

.. Another important factor will be whether patients and providers continue to

adhere to recommendations for cervical screening, since premature cessation of

these preventive interventions could lead to an increase in cervical cancer

rates.

INFORMATION FOR PATIENTS — Educational

materials on this topic are available for patients. (See " Patient

information: Human papillomavirus (HPV) vaccine " , see " Patient

information: Condyloma (genital warts) in women " , and see " Patient

information: Cervical cancer screening " ). We encourage you to print or

e-mail these topic reviews, or to refer patients to our public web site, www.uptodate.com/patients, which

includes these and other topics.

SUMMARY AND RECOMMENDATIONS

Persistent infection with high-risk oncogenic

HPV types leads to cervical cancer, the second most common malignant

disease in women worldwide. (See " Introduction " above).

Multicenter, double-blind, placebo-controlled

trials for both quadrivalent and bivalent HPV vaccines demonstrate

efficacy against incident and persistent HPV infection and abnormal

cervical cytology and histology. Bivalent HPV 16/18 L1 VLP AS04 adjuvanted

vaccine has been submitted to the FDA for approval; quadrivalent HPV

6/11/16/18 L1 VLP vaccine is available for use and distribution. The

quadrivalent vaccine also prevents most genital warts. (See " Vaccine

development " above).

Both vaccines have an excellent safety profile.

(See " Vaccine safety " above).

We recommend immunization with HPV vaccine, as

suggested by the ACIP and ACOG (Grade

1A). The currently suggested immunization schedule is in girls and

women 9 to 26 years of age. Quadrivalent HPV 6/11/16/18 L1 VLP vaccine

(Gardasilâ„¢ ) is administered in three doses at 0, 2, and 6 months. The

duration of immunity is unknown. (See " Vaccine development "

above).

Cytologic screening is recommended for any

woman who has been sexually active for three or more years or is 21 years

of age. Clinicians need to be aware that HPV immunization is NOT effective

in clearing cytologically evident disease or infection. A preventive

health care visit in which vaccination is discussed or offered represents

an ideal opportunity to offer Pap screening to sexually active patients as

well. (See " Vaccine utilization " above).

Vaccination can be offered to females as young

as nine years of age. Cervical screening is not appropriate until

pubertal, physiologic, and psychological development has been established

and at least three years from onset of sexual activity has passed.

HPV vaccine is a major advance in the

prevention of cervical cancer, but it will not replace the need for other

preventive strategies, such as cervical screening. (See " Importance

of cervical screening " above).

Use of UpToDate is subject to

the Subscription and

License Agreement .

REFERENCES

1

,

JT. Introduction. Curr Opin Obstet Gynecol 2006; 18 Suppl 1:s3.

2

Parkin,

DM, Bray, F, Ferlay, J, Pisani, P. Global cancer statistics, 2002. CA Cancer

J Clin 2005; 55:74.

3

Munoz,

N, Bosch, FX, de Sanjose, S, et al. Epidemiologic classification of human

papillomavirus types associated with cervical cancer. N Engl J Med 2003;

348:518.

4

Dunne,

EF, Markowitz, LE. Genital human papillomavirus infection. Clin Infect Dis

2006; 43:624.

5

Yoon,

H, Chung, MK, Min, SS, et al. Synthetic peptides of human papillomavirustype

18 E6 harboring HLA-A2.1 motif can induce peptide specific cytotoxic T-cells

from peripheral blood mononuclear cells of healthy donors. Virus Res 1998;

54:23.

6

Lowy,

DR, Schiller, JT. Papillomaviruses and cervical cancer: pathogenesis and

vaccine development. J Natl Cancer Inst Monogr 1998; :27.

7

Rose,

RC, White, WI, Suzich, JA, et al. Human Papillomavirus type 11 recombinant

capsomeres induce virus-neutralising antibodies. J Virol 1998; 72:6151.

8

Hines,

JF, Ghim, SJ, Jenson, AB, et al. Prospects for human papillomavirus vaccine

development: Emerging HPV vaccines. Curr Opin Obstet Gynecol 1998; 10:15.

9

Clifford,

GM, Gallus, S, Herrero, R, et al. Worldwide distribution of huma

papillomavirus types in cytologically normal women in the International

Agency for Research on Cancer HPV prevalence surveys: a pooled analysis.

Lancet 2005; 366:991.

10

Clifford,

GM, Rana, RK, Franceschi, S, et al. Human papillomavirus genotype

distribution in low-grade cervical lesions: comparison by geographic region

and with cervical cancer. Cancer Epidemiol Biomarkers Prev 2005; 14:1157.

11

Clifford,

GM, , JS, Plummer, M, et al. Human papillomavirus types in invasive

cervical cancer worldwide: a meta-analysis. Br J Cancer 2003; 88:63.

12

Clifford,

GM, , JS, Aguado, T, Franceschi, S. Comparison of HPV type distribution

in high-grade cervical lesions and cervical cancer: a meta-analysis. Br J

Cancer 2003; 89:101.

13

Li,

HX, Zhu, WY, Xia, MY. Detection with the polymerase chain reaction of human

papillomavirus DNA in condylomata acuminata treated with CO2 laser and

microwave. Int J Dermatol 1995; 34:209.

14

,

N, Birley, HD, Renton, AM, et al. Immunological events in regressing genital

warts. Am J Clin Pathol 1994; 102:768.

15

Greer,

CE, Wheeler, CM, Ladner, MB, et al. Human papillomavirus (HPV) type

distribution and serological response to HPV type 6 virus-like particles in

patients with genital warts. J Clin Microbiol 1995; 33:2058.

16

Munoz,

N. Against which human papillomavirus types shall we vaccinate and screen?

The international perspective. Int J Cancer 2004; 111:278.

17

,

JT. The development of cervical cancer and its precursors: what is the role

of human papillomavirus infection?. Curr Opin Obstet Gynecol 2006; 18 Suppl

1:s5.

18

Schiffman,

M, Kjaer, SK. Chapter 2: Natural history of anogenital human papillomavirus

infection and neoplasia. J Natl Cancer Inst Monogr 2003; :14.

19

Frazer,

IH, , JT, Mayeaux, EJ Jr, et al. Advances in prevention of cervical cancer

and other human papillomavirus-related diseases. Pediatr Infect Dis J 2006;

25:S65.

20

Munger,

K, Howley, PM. Human papillomavirus immortalization and transformation

functions. Virus Res 2002; 89:213.

21

Stanley,

M. Immune responses to human papillomavirus. Vaccine 2006; 24 Suppl 1:S16.

22

Ozsaran,

AA, Ates, T, Dikmen, Y, et al. Evaluation of the risk of cervical

intrepithelial neoplasia and human papilloma virus infection in renal

transplant patients receiving immunosuppressive therapy. Eur J Gynaecol Oncol

1999; 20:127.

23

Villa,

LL, Costa, RL, Petta, CA, et al. Prophylactic quadrivalent human

papillomavirus (types 6, 11, 16, and 18) L1 virus-like particle vaccine in

young women: a randomised double-blind placebo-controlled multicentre phase

II efficacy trial. Lancet Oncol 2005; 6:271.

24

Ho,

GY, Studentsov, YY, Bierman, R, Burk, RD. Natural history of human

papillomavirus type 16 virus-like particle antibodies in young women. Cancer

Epidemiol Biomarkers Prev 2004; 13:110.

25

,

J. Comparison of human papillomavirus types 16,18, and 6 capsid antibody

responses following incident infection. J Infect Dis 2000; 181:1911.

26

Mayeaux,

EJ Jr. Harnessing the power of prevention: human papillomavirus vaccines.

Curr Opin Obstet Gynecol 2006; 18 Suppl 1:s15.

27

Snijders,

PJ, Steenbergen, RD, Heideman, DA, Meijer, CJ. HPV-mediated cervical

carcinogenesis: concepts and clinical implications. J Pathol 2006; 208:152.

28

Giannini,

SL, Hanon, E, Moris, P, et al. Enhanced humoral and memory B cellular

immunity using HPV16/18 L1 VLP vaccine formulated with the MPL/aluminium salt

combination (AS04) compared to aluminium salt only. Vaccine 2006; 24:5937.

29

Kirnbauer,

R, Booy, F, Cheng, N, et al. Papillomavirus L1 major capsid protein

self-assembles into virus-like particles that are highly immunogenic. Proc

Natl Acad Sci U S A 1992; 89:12180.

30

Lowy,

DR, Schiller, JT. Prophylactic human papillomavirus vaccines. J Clin Invest

2006; 116:1167.

31

Koutsky,

LA, Ault, KA, Wheeler, CM, et al. A controlled trial of a human

papillomavirus type 16 vaccine. N Engl J Med 2002; 347:1645.

32

Harper,

DM, Franco, EL, Wheeler, C, et al. Efficacy of a bivalent L1 virus-like

particle vaccine in prevention of infection with human papillomavirus types

16 and 18 in young women: a randomised controlled trial. Lancet 2004;

364:1757.

33

Harper,

DM, Franco, EL, Wheeler, CM, et al. Sustained efficacy up to 4.5 years of a

bivalent L1 virus-like particle vaccine against human papillomavirus types 16

and 18: follow-up from a randomised control trial. Lancet 2006; 367:1247.

34

Gall, SA, Teixeira, J, Naud, P et al. Substantial impact

on precancerous lesions and HPV infections through 5.5 years in women

vaccinated with the HPV-16/18 L1 VLP AS04 candidate vaccine. Annual American

Assoication for Cancer Research Meeting, Los Angeles, CA. April 14-18, 2007.

Abstract #4900.

35

Schwarz, T, Wurzburg, J. An ASO4-containing human

papillomavirus (HPV) 16/18 vaccine for prevention of cervical cancer is

immunogenic and well-tolerated in women 15 to 55 years old. Presented at the

42nd annual meeting of the American Society of Cancer Oncology, June 2-6,

2006, Atlanta, GA [abstract #1008].

36

Joura,

EA, Leodolter, S, -Avila, M, et al. Efficacy of a quadrivalent

prophylactic human papillomavirus (types 6, 11, 16, and 18) L1

virus-like-particle vaccine against high-grade vulval and vaginal lesions: a

combined analysis of three randomised clinical trials. Lancet 2007; 369:1693.

37

Villa,

LL, Ault, KA, Giuliano, AR, et al. Immunologic responses following

administration of a vaccine targeting human papillomavirus Types 6, 11, 16,

and 18. Vaccine 2006; 24:5571.

38

Villa,

LL, Costa, RL, Petta, CA, et al. High sustained efficacy of a prophylactic

quadrivalent human papillomavirus types 6/11/16/18 L1 virus-like particle

vaccine through 5 years of follow-up. Br J Cancer 2006; 95:1459.

39

Quadrivalent

vaccine against human papillomavirus to prevent high-grade cervical lesions.

N Engl J Med 2007; 356:1915.

40

Ault,

KA. Effect of prophylactic human papillomavirus L1 virus-like-particle

vaccine on risk of cervical intraepithelial neoplasia grade 2, grade 3, and

adenocarcinoma in situ: a combined analysis of four randomised clinical

trials. Lancet 2007; 369:1861.

41

Garland,

S, -Avila, M, Wheeler, C, et al. Quadrivalent vaccine against human

papillomavirus to prevent anogenital diseases. N Engl J Med 2007; 356:1928.

42

Adult Immunization. Treat Guidel Med Lett 2006; 4:47.

43

Markowitz,

LE, Dunne, EF, Saraiya, M, et al. Quadrivalent Human Papillomavirus Vaccine:

Recommendations of the Advisory Committee on Immunization Practices (ACIP).

MMWR Recomm Rep 2007; 56:1.

44

ACOG

Committee Opinion No. 343: psychosocial risk factors: perinatal screening and

intervention. Obstet Gynecol 2006; 108:469.

45

,

GD, Taira, AV. Cost-effectiveness of a potential vaccine for human

papillomavirus. Emerg Infect Dis 2003; 9:37.

46

Goldie,

SJ, Kohli, M, Grima, D, et al. Projected clinical benefits and

cost-effectiveness of a human papillomavirus 16/18 vaccine. J Natl Cancer

Inst 2004; 96:604.

47

Kulasingam,

SL, Myers, ER. Potential health and economic impact of adding a human

papillomavirus vaccine to screening programs. JAMA 2003; 290:781.

48

Chesson, HW, et al. Cost effectiveness models of HPV

vaccines. May 9, 2006 - 2006 National STD Prevention Conference.

49

Kim,

JJ, Goldie, SJ. Health and economic implications of HPV vaccination in the

United States. N Engl J Med 2008; 359:821.

50

,

JP, Garnett, GP, Koutsky, L. The theoretical population-level impact of a

prophylactic human papilloma virus vaccine. Epidemiology 2002; 13:631.

51

Taira,

AV, Neukermans, CP, , GD. Evaluating human papillomavirus vaccination

programs. Emerg Infect Dis 2004; 10:1915.

52

Stanley,

M. Immunobiology of HPV and HPV vaccines. Gynecol Oncol 2008; 109:S15.

53

Newall,

AT, Beutels, P, Wood, JG, et al. Cost-effectiveness analyses of human

papillomavirus vaccination. Lancet Infect Dis 2007; 7:289.

54

Gunther,

OP, Ogilvie, G, Naus, M, et al. Protecting the Next Generation: What Is the

Role of the Duration of Human Papillomavirus Vaccine-Related Immunity?. J

Infect Dis 2008; 197:1653.

55

Rintala,

MA, Grenman, SE, Jarvenkyla, ME, et al. High-risk types of human

papillomavirus (HPV) DNA in oral and genital mucosa of infants during their

first 3 years of life: experience from the Finnish HPV Family Study. Clin

Infect Dis 2005; 41:1728.

56

Dunne,

EF, Karem, KL, Sternberg, MR, et al. Seroprevalence of human papillomavirus

type 16 in children. J Infect Dis 2005; 191:1817.

57

Saslow,

D, Castle, PE, , JT, et al. American Cancer Society Guideline for human

papillomavirus (HPV) vaccine use to prevent cervical cancer and its

precursors. CA Cancer J Clin 2007; 57:7.

58

Zimet,

GD. Understanding and overcoming barriers to human papillomavirus vaccine

acceptance. Curr Opin Obstet Gynecol 2006; 18 Suppl 1:s23.

59

Pitts,

M, e, T. Human papillomavirus infections and risks of cervical cancer:

what do women know?. Health Educ Res 2002; 17:706.

60

Biddlecom,

A, Bankole, A, , K. Vaccine for cervical cancer: reaching

adolescents in sub-Saharan Africa. Lancet 2006; 367:1299.

61

Markowitz, LE. HPV vaccines prophylactic, not therapeutic.

JAMA 2007; 298:805.

62

Hildesheim,

A, Herrero, R, Wacholder, S, et al. Effect of human papillomavirus 16/18 L1

viruslike particle vaccine among young women with preexisting infection: a

randomized trial. JAMA 2007; 298:743.

63

Prophylactic

efficacy of a quadrivalent human papillomavirus (HPV) vaccine in women with

virological evidence of HPV infection. J Infect Dis 2007; 196:1438.

64

American Cancer Society. Cancer Facts and Figures 2004.

Available online at: www.cancer.org/downloads/STT/CAFF_FinalPWSecured.pdf.

Accessed on March 27, 2006.

65

Sawaya,

GF, Kerlikowske, K, Lee, NC, et al. Frequency of cervical smear abnormalities

within three years of normal cytology. Obstet Gynecol 2000; 96:219.

66

Dunne,

EF, Unger, ER, Sternberg, M, et al. Prevalence of HPV infection among females

in the United States. JAMA 2007; 297:813.

67

Barr,

E, Tamms, G. Quadrivalent human papillomavirus vaccine. Clin Infect Dis 2007;

45:609.

68

Block,

SL, Nolan, T, Sattler, C, et al. Comparison of the immunogenicity and

reactogenicity of a prophylactic quadrivalent human papillomavirus (types 6,

11, 16, and 18) L1 virus-like particle vaccine in male and female adolescents

and young adult women. Pediatrics 2006; 118:2135.

69

Reisinger,

K. Safety and persistent immunogenicity of a quadrivalent human

papillomavirus types 6,11,16,18 L1 virus-like particle vaccine in

preadolescents and adolescents. Pediatr Infect Dis J 2007; 26:201.

70

Barnabas,

RV, Laukkanen, P, Koskela, P, et al. Epidemiology of HPV 16 and cervical

cancer in Finland and the potential impact of vaccination: mathematical modelling

analyses. PLoS Med 2006; 3:e138.

©2008 UpToDate® • customerservice@...

www.uptodate.com

This e-mail message may contain confidential and/or privileged information. If you are not an addressee or otherwise authorized to receive this message, you should not use, copy, disclose or take any action based on this e-mail or any information contained in the message. If you have received this material in error, please advise the sender immediately by reply e-mail and delete this message. Thank you.

Link to comment
Share on other sites

My understanding is, per 3 year AAFP reporting cycle:

60 of required 75 Prescribed credits can be UTD (max 20 per year)

0 of required 20 Group credits can be UTD

55 of 55 Elective credits can be UTD-there is no limit in this

category (i.e. apply any extra to this category)

At least that's how I just reported it this month for the last 3 year

cycle. It involved a conversation with staff there, and they

accepted it as above without question.

FYI-I'd never bothered to use the UTD CME before, but it gave me over

240 credits over those 3 years, and saved me from scrambling to find

some other credits last minute.

Jeff Huotari

BlueSky Health

>

> I do have a personal subscription and do believe it is worth it as

I use

> it regularly while the patient is in the room with me. The write

ups are

> pretty comprehensive and seem balanced. On the negative side: these

are

> " expert opinions " so it may not reflect true evidence based

literature.

> On the positive side, it automatically tracks every time you look

> something up, so you get CME for it (although the AAFP only

recognizes

> up to 20 hours/year).

>

> P.S. I am just a customer, I have no financial ties to UpToDate.

>

> Re: HPV vaccine

>

> Thanks!

>

> Do you have a personal subscription?

>

> Do you think the expense is worth it? For the CME?

>

> I personally regard it as a superb resource, I have access through

my

> hospitals and wonder if I should invest in it for my personal use

(the

> hospital access is only through their intranet).

>

> --

> Pedro Ballester, M.D.

> Warren, OH

>

Link to comment
Share on other sites

> Thanks Jeff,

I will use the extra as " elective credits. " Cool to know.

>

> Date: 2008/12/20 Sat PM 05:29:48 EST

> To:

> Subject: Re: HPV vaccine

>

> My understanding is, per 3 year AAFP reporting cycle:

>

> 60 of required 75 Prescribed credits can be UTD (max 20 per year)

> 0 of required 20 Group credits can be UTD

> 55 of 55 Elective credits can be UTD-there is no limit in this

> category (i.e. apply any extra to this category)

>

> At least that's how I just reported it this month for the last 3 year

> cycle. It involved a conversation with staff there, and they

> accepted it as above without question.

>

> FYI-I'd never bothered to use the UTD CME before, but it gave me over

> 240 credits over those 3 years, and saved me from scrambling to find

> some other credits last minute.

>

> Jeff Huotari

> BlueSky Health

>

>

>

> >

> > I do have a personal subscription and do believe it is worth it as

> I use

> > it regularly while the patient is in the room with me. The write

> ups are

> > pretty comprehensive and seem balanced. On the negative side: these

> are

> > " expert opinions " so it may not reflect true evidence based

> literature.

> > On the positive side, it automatically tracks every time you look

> > something up, so you get CME for it (although the AAFP only

> recognizes

> > up to 20 hours/year).

> >

> > P.S. I am just a customer, I have no financial ties to UpToDate.

> >

> > Re: HPV vaccine

> >

> > Thanks!

> >

> > Do you have a personal subscription?

> >

> > Do you think the expense is worth it? For the CME?

> >

> > I personally regard it as a superb resource, I have access through

> my

> > hospitals and wonder if I should invest in it for my personal use

> (the

> > hospital access is only through their intranet).

> >

> > --

> > Pedro Ballester, M.D.

> > Warren, OH

> >

>

>

>

>

Link to comment
Share on other sites

Join the conversation

You are posting as a guest. If you have an account, sign in now to post with your account.
Note: Your post will require moderator approval before it will be visible.

Guest
Reply to this topic...

×   Pasted as rich text.   Paste as plain text instead

  Only 75 emoji are allowed.

×   Your link has been automatically embedded.   Display as a link instead

×   Your previous content has been restored.   Clear editor

×   You cannot paste images directly. Upload or insert images from URL.

Loading...
×
×
  • Create New...