Sie sind auf Seite 1von 17

Human Papillomavirus

EILEEN M. BURD1,2 and CHRISTINA L. DEAN1


1
Emory University School of Medicine, Department of Pathology and Laboratory Medicine,
Atlanta, GA 30322; 2Emory University School of Medicine, Department of Medicine,
Division of Infectious Diseases, Atlanta, GA 30322

ABSTRACT Individuals with inherited immunodeficiencies, tissue tropism and subsequent host pathology (2, 7).
autoimmune disorders, organ or bone marrow transplantation, HPV types within the alpha group have been the focus
or infection with human immunodeficiency virus (HIV) are at
of a considerable amount of research due to their ability
increased risk of infection with both low-risk and high-risk
human papillomavirus (HPV) types. Chronic immunosuppression
to cause both cutaneous and mucosal pathology (2, 4).
provides an environment for persistent HPV infection which Individual types within this genus are further classified
carries a higher risk of malignant transformation. Screening into high-risk HPV and low-risk HPV, depending on
guidelines have been developed or advocated for processes their oncogenic ability. HPV6 and HPV11 are low-risk
that have detectable premalignant lesions, such as anal cancer types and are most commonly associated with benign
or cervical cancer. For other anatomic locations, such as anogenital warts (condylomata acuminata) (1, 8). Al-
cutaneous, penile, and oropharyngeal, a biopsy of suspicious though HPV16 and HPV18 are the most prevalent types
lesions is necessary for diagnosis. HPV cannot be cultured
causing HSIL and cervical carcinoma, at least 12 high-
from clinical specimens in the laboratory, and diagnosis relies
on cytologic, histologic, or molecular methods. risk types have been identified (9, 10). Of note, not all
individuals infected with high-risk HPV will go on to
develop cancer (2, 4). Elucidation of the life cycle and
INTRODUCTION pathogenesis of HPV has largely come from studying
Human papillomavirus (HPV), a member of the Papil- HPV16 infections of the cervical epithelium (4).
lomaviridae family, is a small (8kb), nonenveloped, Within the circular genome of HPV, eight genes are
double-stranded DNA virus. HPV has a predilection for encoded, including genes expressed in both early and
infecting cutaneous and mucosal epithelial cells (1). In- late stages in the life cycle. The infection produced by
fection with HPV is associated with a wide range of HPV can follow one of two pathways and is either pro-
pathology. HPV is the etiological factor in benign cu- ductive or nonproductive (abortive or transforming).
taneous warts and juvenile respiratory papillomatosis, Regardless of the pathway, HPV requires access to the
as well as low-grade squamous intraepithelial lesions basal lamina, and such contact is likely achieved through
(LSIL), high-grade squamous intraepithelial lesions small areas of damage to the epithelium (7). Capsid
(HSIL), a precursor to cancer, and invasive carcinoma proteins, L1 (major coat protein) and L2 (minor coat
(2). Harald zur Hausen, a German virologist, was the protein), facilitate entry into the basal layer keratino-
first to describe the association of HPV and cervical
Received: 2 March 2015, Accepted: 8 February 2016,
cancer in the 1970s (3, 4). It is now understood that Published: 15 July 2016
HPV is necessary in the development of cervical cancer Editors: Randall T. Hayden, St. Jude Children’s Research Hospital,
and is also associated with carcinoma of the vulva, va- Memphis, TN; Donna M. Wolk, Geisinger Clinic, Danville, PA;
gina, penis, anus, and oropharynx (5, 6). Karen C. Carroll, Johns Hopkins University Hospital, Baltimore, MD;
Yi-Wei Tang, Memorial Sloan-Kettering Institute, New York, NY
HPV does not grow well in tissue culture and is clas- Citation: Burd EM, Dean CL. 2016. Human papillomavirus. Microbiol
sified based on its DNA sequence. Currently, more than Spectrum 4(4):DMIH2-0001-2015. doi:10.1128/microbiolspec
200 types of HPV have been fully sequenced (1, 7). .DMIH2-0001-2015.
Correspondence: Eileen M. Burd, eburd@emory.edu
Human papillomaviruses are generally grouped into five
© 2016 American Society for Microbiology. All rights reserved.
genera (alpha, beta, gamma, mu, and nu) based upon

ASMscience.org/MicrobiolSpectrum 1
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Sun, 27 Nov 2016 18:41:47
Burd and Dean

cytes (7, 11). L2 is thought to also be involved in allow- search on Cancer (IARC) are HPV16, HPV18, HPV31,
ing the viral genome to enter the host cell nucleus where HPV33, HPV35, HPV39, HPV45, HPV51, HPV52,
replication ensues via proteins E1 and E2 as well as the HPV56, HPV58, and HPV59 (9). HPV68 is classified as
host cell’s replication machinery (7, 12). The E1 gene probably carcinogenic, and HPV26, HPV30, HPV34,
product is a viral DNA helicase, which is recruited by the HPV53, HPV66, HPV67, HPV 69, HPV70, HPV73,
binding of the viral transcription factor encoded by the HPV82, HPV85, and HPV97 have been associated with
E2 gene (2). E2 is also involved in anchoring the viral rare cases of cervical cancer and are considered probable
episome to the host cell genome (13). carcinogens (9). The E6 and E7 proteins in high-risk
Initial viral replication in the basal cells seems to be HPV types are functionally different from low-risk types
independent of the host cell cycle, and viral genomes are and serve to stimulate proliferation of infected basal
amplified to densities of approximately 50 to 200 copies and suprabasal cells, allowing enlargement of the lesion.
per cell (2, 8). In the productive pathway, viral DNA Long-term persistence of HPV is a necessary foundation
copies are maintained at low copy number until the cell for nonproductive infection. Persistence facilitates inte-
moves through the epithelium in the process of differ- gration of the viral gene into the host cell chromosome.
entiation. Virus replication and assembly occurs in post- Although some cancers continue to harbor episomal
mitotic differentiated squamous epithelial cells as they HPV DNA, integration of viral DNA into the host cell
move toward the epithelial surface. E6 and E7 are reg- genome is a common step associated with further dereg-
ulated by E2, and while their precise role is somewhat ulation and overexpression of the E6 and E7 oncogenes
uncertain, they are associated with driving cell cycle and carcinogenic progression (12). Integration occurs by
progression to allow HPV DNA replication in the mid- chance, and E6/E7 overexpression typically results when
layers of the epithelium. Virus gene expression is tightly HPV DNA integration occurs in the E1/E2 region and
controlled, and viral DNA is amplified as extrachro- causes disruption or deletion of E2 gene sequences. This
mosomal nuclear plasmids with viral copies increasing leads to loss of feedback control and overexpression of
about two to four logs (2, 7, 8, 14). No viremia is pro- the E6 and E7 oncogenes as well as disruption or silencing
duced and replication takes place entirely within the of the downstream early genes that are required for reg-
cell, thus producing little immune response from the ulated viral replication. The E6 protein prevents cellular
host (15). At the epithelial surface, keratinocytes are apoptosis by degrading p53 and contributes to the ac-
considered terminally differentiated, and they die and cumulation of genetic mutations that lead to immortali-
are removed from the body by natural processes. Large zation. The E7 protein stimulates cell proliferation when
amounts of virus are released from the epithelial sur- it binds with the retinoblastoma (Rb) protein and also
face for transmission to other individuals. At this point, stimulates host genome instability (17). The E5 protein
HPV DNA may be found intracellularly as well as ex- appears to interfere with apoptosis and enhances the
tracellularly (1). This has important implications for transforming ability of the E6 and E7 proteins (2, 12, 18).
diagnostic testing. These steps that lead to productive After the initial replication, viral DNA is stably main-
virus synthesis in the superficial epithelial layers can be tained at an almost constant copy number during suc-
followed by either low- or high-risk HPV types, al- cessive divisions of the basal cells (19). This is thought to
though productive infections never lead to cancer be- occur because the viral genomes replicate once along with
cause the cell cycle is halted and infected cells leave the cellular DNA during the S-phase of the host cell cycle and
body. Most HPV infections become undetectable after are divided equally into the two daughter cells (19).
several months (16). Current thinking is that productive Even in this setting, most nonproductive infections
infection is supported in the cellular environment at the are eventually cleared (as measured by current molecular
ectocervix but is inefficiently supported by the cells at the tests), 50% within one year and 90% within two years,
endocervix, where nonproductive infection is favored by way of a T-cell-mediated immune response (2, 8).
(7). High-grade lesions and cancers usually occur in individ-
In nonproductive infection, HPV gene expression uals who do not resolve their infection and who main-
becomes deregulated and the normal life cycle of the tain oncogene expression for years or decades.
virus cannot be completed. The HPV types associated There is growing evidence supporting the ability of
with nonproductive infections are designated as high- some HPV types to persist in a long-term latent state
risk because of their association with progression to in basal epithelial cells after apparent resolution of in-
cancer. The high-risk carcinogenic types of HPV cur- fection (20). Latent infection has been shown to be a risk
rently designated by the International Agency for Re- factor for the development of disease at the same site at a

2 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Sun, 27 Nov 2016 18:41:47
Human Papillomavirus

later point in time. Studies in animal models suggest that both viruses (23). Hence, this patient population is at
HPV genome replication can continue even though gene an increased risk for developing HPV cutaneous and
expression is suppressed by the activity of memory T cells mucosal disease. Cutaneous disease including common
(20). Since this type of latency is immune-mediated, sub- warts, epidermodysplasia verruciformis-like lesions,
sequent immunosuppression can lead to reactivation of squamous cell carcinoma (SCC), and basal cell carci-
infection in these experimental systems. Although the noma is more prevalent in HIV-positive individuals (27,
duration of the latent state in humans is not known, it is 28). These cutaneous lesions tend to be associated with
a possible explanation for new HPV detection as a result HPV types usually found in the genital tract, and they
of aging immune systems in older women (20). It appears are more likely to be aggressive and difficult to treat (28).
that similar silent infections can result from low-titer in- Although the role of HPV in the oncogenesis of skin
fections or infections in epithelial cells that do not com- cancer has not been fully elucidated, there have been
pletely support the HPV life cycle (20). In this situation, many reported cases of high-risk HPV types associated
changes in the local environment that occur with me- with SCC of the distal digits and periungual skin in HIV-
chanical irritation, wounding, or exposure to ultraviolet positive individuals (28). These carcinomas require ag-
light can initiate reactivation (20). gressive treatment and tend to recur.
Benign anogenital warts, or condylomata acuminata,
are the most common clinical manifestation of HPV
infection in both immune-competent and immunosup-
EPIDEMIOLOGY AND RISK FACTORS
pressed individuals. Roughly 90% of anogenital warts
Immune-Competent Patients are attributed to low-risk types, HPV6 and HPV11 (6).
HPV is the most common sexually transmitted infection Similar to nongenital cutaneous lesions, HIV-positive
(STI) worldwide. The majority of individuals who are patients often present with widespread lesions that can
sexually active will be infected with HPV at some point be more aggressive and difficult to treat (28, 29).
in their lifetime (6). Close to 80 million people in the HPV-induced cutaneous warts and SCC also pose a
United States are currently infected with HPV, and an- major health risk for SOT recipients who are receiving
other 14 million become newly infected every year (21). immunosuppressive therapy. In fact, it has been reported
Anogenital warts are a common clinical manifestation that SOT recipients are at a higher risk for developing
of HPV infection, and there are roughly 350,000 new skin cancer than those with HIV or AIDS (30). This
cases in the United States every year (6, 22). Persis- phenomenon has not been fully elucidated, but research
tent infection with high-risk HPV is the most significant suggests that certain immunosuppressive drugs, as well
risk factor for developing carcinoma (2, 6). Cervical and as an individual’s humoral immune response, may play a
oropharyngeal carcinoma comprise the majority of the role (30, 31). The 10-year cumulative incidence of skin
newly identified HPV-associated carcinomas, and most cancer in this population reaches 10 to 40% (32). Renal
of these cases are due to infection with high-risk HPV transplant recipients seem to be at a particularly high
types, HPV16 and HPV18 (6, 23). Immune-competent risk for developing HPV-related cutaneous warts and
persons infected with HPV generally have a slow progres- skin cancers (32, 33). Immunosuppression, ultraviolet
sion to high-grade, precancerous lesions and subsequent light, and infection with high-risk HPV types are all
carcinoma (24). However, most people with competent important risk factors in patients with a history of SOT
immune systems are able to clear HPV infections with (33, 34).
no sequelae. Individuals who have compromised cell- Hematopoietic stem cell transplant (HSCT) recipients,
mediated immunity, such as those with human immu- especially those with chronic graft-versus-host disease
nodeficiency virus (HIV), AIDS, or solid-organ transplant (cGVHD), are at an overall increased risk for developing
(SOT) recipients, have shown accelerated progression to a secondary malignancy, with cutaneous SCC being the
high-grade lesions and, thus, are at an increased risk to most frequent lesion identified (35). However, the asso-
develop HPV-associated carcinomas (24–26). ciation with cutaneous SCC and HPV in this patient
population has not been well studied.
Immunocompromised Patients
External skin Epidermodysplasia verruciformis
Cutaneous warts Epidermodysplasia verruciformis (EV) is an autosomal
HPV infection is very common among HIV-positive recessive dermatologic condition characterized by per-
individuals, due to the sexually transmitted nature of sistent HPV infections of the skin that can transform into

ASMscience.org/MicrobiolSpectrum 3
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Sun, 27 Nov 2016 18:41:47
Burd and Dean

carcinoma (25, 34). In fact, nearly 50% of patients with Anal cancer
EV will develop skin cancer by the time they reach their Anal cancer is a rare malignancy, with an incidence rate
fifth decade of life (34). There are specific HPV types of around 2 per 100,000 for both men and women (44).
associated with EV (EV-HPV), but HPV5, HPV8, and A significant risk factor for anal SCC is coinfection with
HPV14 are most commonly associated with malignancy high-risk HPV and HIV (45). More than 90% of anal
in these patients (34). Whether EV-HPV is a causal fac- SCCs are associated with persistent HPV infection, and
tor in the development of cutaneous malignancy is still the majority of these cases are attributed to HPV16 and
debated, and the oncogenesis is poorly understood (36). HPV18 (45, 46). The prevalence of anal HPV detected
Recently, an acquired EV, or EV-like, syndrome has been when screening men and women with HIV surpasses
described in immunosuppressed patients infected with 90%, though not all will have abnormal pathology.
EV-HPV, specifically those with HIV and SOT recipients The incidence of anal SCC is increased 30-fold in HIV-
(37). Case reports for this entity are rare and progression positive individuals and 80-fold in HIV-positive men
to malignancy has not yet been documented. who have sex with men, compared to HIV-uninfected
individuals (45, 47). A notable increase in the incidence
Oropharyngeal cancer of anal SCC was reported with the introduction of ART,
Individuals with HIV are at an increased risk of devel- which was attributed to the increased lifespan of HIV-
oping HPV-related cancers, including head and neck positive individuals. However, a recent study suggests
cancer (HNC). Over 400,000 new cases of HNC occur that these numbers may be stabilizing (47).
every year, making it the sixth most common cancer Epidemiological studies have shown that 20 to 50%
worldwide (38). HPV is detected in roughly 25% of of SOT recipients have detectable anal HPV and a
cancers arising in the oropharynx, and in the general 10-fold increase in the relative risk of developing anal
population, detection is directly correlated with the num- SCC (48, 49). Similar to HIV-positive patients on ART,
ber of recent oral sex partners (39). More than 80% the incidence of HPV-associated anal cancer has in-
of HNCs are caused by oncogenic HPV16, and this is creased in SOT recipients who remain on immunosup-
the most frequently detected HPV type in HIV-positive pression for long periods of time, likely due to increased
patients (40, 41). Up to a 3-fold increase in the incidence survival (48).
of oral HPV infection has been demonstrated in individ-
uals infected with HIV (40). Contrary to HIV-negative Penile cancer
individuals, oral HPV infection in those with HIV is as- Penile SCC and penile HSIL are rarely encountered dis-
sociated with increased numbers of lifetime oral sex eases in the general population. In the United States,
partners (40). It is still not clear whether CD4 count and/ penile SCC accounts for less than 0.5% of all cancers in
or AIDS diagnosis has an effect on the prevalence of oral men (50). HPV is associated with roughly 50% of penile
HPV infection (39). Additionally, the effect of antiretro- cancer cases, and oncogenic HPV16 is the most common
viral therapy (ART) on oral HPV prevalence has not type detected (51, 52). Individuals infected with HIV
been fully elucidated, but some studies suggest that there have a 2- to 3-fold increase in risk of developing penile
may be an increase in oral lesions and HPV persistence in SCC (28). Up to 4% of the HIV-infected population
individuals receiving ART (39). may have precursor lesions (HSIL), but the progression
SOT recipients are also at an increased risk for devel- to cancer only occurs in about 5 to 30% of cases (28).
oping HPV-related cancers. Oropharyngeal carcinoma is Other risk factors include tobacco use, poor hygiene,
the third most common HPV-related cancer identified phimosis, and lack of circumcision (28).
in SOT recipients, after vulvar and anal carcinomas (42).
Factors such as age, race, transplanted organ, and certain Cervical cancer
immunosuppressive drugs may affect the incidence of With over 500,000 cases every year, cervical carcinoma
HPV-related oropharyngeal carcinoma (42). is the fourth leading cause of cancer in women world-
Oropharyngeal carcinoma is frequently found in wide (53). It is now known that HPV is necessary for
long-term survivors of HSCT. As in cutaneous SCC, cervical cancer to occur, and its pathogenesis has been
these lesions are strongly associated with cGVHD (35). investigated extensively. Risk factors for cervical carci-
Again, the association between HNC and HPV has not noma include tobacco use, young age of sexual debut,
been fully elucidated in HSCT recipients; however, sev- multiple sexual partners, high-risk sexual partners, his-
eral case reports suggest that these lesions may be HPV- tory of other STIs, and a history of genital SIL or carci-
driven (43). noma (54). Women with HIV/AIDS and SOT recipients,

4 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Sun, 27 Nov 2016 18:41:47
Human Papillomavirus

in particular renal transplant, have an increased inci- compared to those found in immune-competent patients
dence of persistent HPV infection and, thus, are at an (64). These differences include higher levels of immu-
increased risk for developing cervical carcinoma (25, nohistochemical staining for p53 and transforming
55). Of note, some studies suggest that SOT recipients growth factor-beta (TGFβ) proteins, along with low
on immunosuppressive therapy do not have an increased levels of phosphorylated mTOR and P70S6K, and a
incidence of cervical cancer (42, 55). HIV-infected higher incidence of a spindle cell component, indicating
women have up to a 22-fold increased incidence of epithelial-to-mesenchymal transition (64, 65).
cervical cancer, and CD4 count is inversely proportional
to abnormal cervical pathology (24, 25, 56). Because of Oropharyngeal Cancer
the high prevalence, cervical carcinoma was classified as Screening
an AIDS-defining disease in 1993 (25). It is still unclear Because some parts of the oropharynx are difficult to
whether implementation of ART in HIV-positive women visualize, oropharyngeal squamous cell cancers are often
decreases the incidence of cervical cancer in this risk detected in later stages. A number of efforts have been
group (57). made toward early detection but they have achieved
Cervical carcinoma is the third most common sec- only limited success. Current guidelines from the U.S.
ondary malignancy in HSCT recipients (35, 58). Women Preventive Services Task Force and the American Dental
who receive long-term treatment for cGVHD and those Association suggest that screening using conventional
with an unrelated HLA-matched donor are at greatest visual and tactile examination may facilitate early di-
risk for developing SIL after transplantation (58, 59). agnosis of oropharyngeal and other oral cancers, but
Whether this is due to reactivation of HPV or to infec- there is not sufficient evidence to support the use of
tion posttransplant needs to be further investigated. additional screening tests (66, 67).

Diagnosis
LABORATORY TESTING The diagnosis of oropharyngeal cancer (OPSCC) is
Skin Lesions made by microscopic examination of biopsy tissues or
Diagnosis exfoliated cells. Biopsy is often preferred since exfolia-
Immunosuppressed patients should be vigilantly fol- tive cytology does not detect all cancers.
lowed for early diagnosis of skin cancer. Lesions in im- The histopathologic terminology used to describe
munosuppressed patients tend to grow more rapidly HPV-related OPSCC has been inconsistent. Rather than
and can be more invasive (60, 61). In addition, there describing specific histologic features, OPSCC tumors
is a strong association for development of additional are often described by the presence of HPV and/or
squamous cell carcinoma lesions in patients who have cellular changes due to HPV. The National Compre-
been previously diagnosed (62). hensive Cancer Network and the College of American
Cutaneous warts can generally be diagnosed by ex- Pathologists have recommended routine testing for HPV
amining the affected area of skin, which may be exten- in all OPSCC (68). It is essential for accurate diagnosis
sive in immunocompromised patients. Warts do not that only specimens obtained from the oropharynx are
generally need to be biopsied to make the diagnosis or to tested for HPV, since involvement of HPV with oral
determine what treatment may be necessary, unless there tumors outside of the oropharynx is negligible and may
is concern that the changes could be cancerous. There not be specific to the tumor. Since HPV-positive tumors
is some suggestion that persistent low-risk beta-HPV- tend to have better clinical outcomes, HPV testing can be
induced warts may progress to actinic keratosis and skin helpful as a prognostic indicator (69). While metastatic
cancer in immunocompromised patients since squamous disease is not common with OPSCC, and metastases in
cell carcinoma lesions and warts can be found jointly HPV-positive tumors are not preceded by bulky or ad-
in this population (61, 63). For this reason, biopsy or vanced oropharyngeal disease, HPV testing may help
surgical excision specimens for histopathologic assess- indicate the site of the primary tumor (70). In the future,
ment should be obtained from all suspicious lesions. The since prognosis is good, HPV testing may also allow for
squamous cell carcinomas that develop in immunocom- more directed and less aggressive therapy for OPSCC
promised individuals have characteristic morphological than therapy for smoking-related oral tumors. Other
features of HPV-induced lesions (63). Some pheno- future uses of HPV testing could potentially include the
typic differences suggestive of a more aggressive process assessment of response to treatment and monitoring for
have been noted in skin cancers of transplant recipients recurrence of disease following treatment.

ASMscience.org/MicrobiolSpectrum 5
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Sun, 27 Nov 2016 18:41:47
Burd and Dean

While testing is recommended, the types of test(s) that are negative for p16 (77). Along with HPV DNA ISH or
should be used to detect HPV-associated OPSCC are not HPV PCR, p16 IHC is recommended to help resolve
specified. The FDA-approved nucleic acid amplification focal or weak p16 staining, p16-negative tumors with
HPV tests for cervical cytology are not approved for typical HPV histologic morphology, and p16-positive
oropharyngeal specimens. Furthermore, tests that assess tumors that do not have typical HPV histologic mor-
transcriptional activity or specifically localize HPV to phology (76) (Fig. 1). In patients with a known primary
the tumor may be more relevant since HPV can occa- OPSCC, p16 IHC may be used instead of HPV testing of
sionally be detected in the absence of disease (71, 72). fine-needle aspiration biopsies to determine if the cancer
Numerous tests, such as RT-PCR for E6/E7 mRNA, has spread to the local lymph nodes (76).
HPV DNA-based in situ hybridization (ISH), and im-
munohistochemistry (IHC) for p16, p53, Ki67, prolif-
erating cell nuclear antigen, and other markers have Penile Cancer
been investigated (73–76). HPV DNA ISH and p16 Diagnosis
IHC have emerged as the most reliable of the methods. Early recognition of penile cancer is based on visual
HPV DNA ISH allows for visualization of the virus in examination, and changes in penile skin color, thick-
tumor cells and has been found to be highly specific, but ening, ulcers, bumps, flat growths, or other lesions may
not as highly sensitive. Modifications that include the warrant further investigation. Flat penile lesions, in par-
use of nonfluorescent chromogens and changes in sig- ticular, seem to be associated with HPV (78). A biopsy is
nal amplification steps have resulted in increased sensi- needed to make an accurate diagnosis of cancer. Squa-
tivity (76). Because there will be some p16 staining in mous cell carcinoma is the most common type of penile
nearly all squamous cell carcinomas, the use of tumor- cancer, and most squamous cell lesions have distinctive
suppressor protein p16 IHC has been found to be highly features that allow their classification as HPV-related
sensitive as a surrogate marker of transcriptionally ac- or non-HPV-related. Most penile cancers are not related
tive HPV infection; unfortunately the marker is not en- to HPV and are usually classified as keratinizing squa-
tirely specific (76). When a positive p16 IHC stain is mous cell carcinomas (78, 79). Others are associated
defined as strong nuclear staining plus cytoplasmic with high-risk HPV infection and are warty, basaloid,
staining, which is present in at least 50% of tumor cells, or mixed (basaloid/usual, warty/basaloid, warty/usual)
then IHC is sufficiently sensitive and specific (76). Re- histologic types that are HPV ISH-positive and show
gardless of HPV status, p16 IHC staining alone is con- strong diffuse p16 IHC staining and the presence of
sidered to be the most useful prognostic indicator for lymphovascular invasion (78, 79). Although PCR is
patients with known OPSCC (76, 77). Outcomes for often considered to be the gold standard for detection
p16 IHC-positive tumors are not significantly different of HPV, none of the commercially available tests are
whether they are HPV-positive or negative, but out- FDA-approved for use in men. A recent study showed
comes are significantly better compared to tumors that good correlation between PCR results and a combina-

FIGURE 1 Algorithm for diagnosis of oropharyngeal squamous cell carcinoma.

6 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Sun, 27 Nov 2016 18:41:47
Human Papillomavirus

tion of morphology, p16 IHC, and/or HPV DNA ISH of screening and prevention programs. Systematic anal
(80). The authors recommend that in order to achieve cancer screening is recommended by some groups, while
the highest diagnostic specificity, morphology, p16 IHC, others suggest that more information is needed before
and HPV DNA ISH should be considered together and screening processes other than visual and digital exam-
all should be positive (80). ination can be recommended.
The importance of determining the HPV status in There are currently no formal recommendations for
penile lesions is often stressed because of the impact on routine anal cancer screening in the United States;
preventing transmission to sexual partners or to identify however, some programs, such as the New York State
partners who are already infected and at increased risk Department of Public Health AIDS Institute, have estab-
for HPV-associated malignancies (80). HPV status may lished screening algorithms (86). Screening is proposed
also be helpful in the differential diagnosis of primary only for high-risk groups, including SOT recipients, men
high-grade urothelial carcinoma versus HPV-related who have sex with men, women with abnormal cervical
basaloid or warty/basaloid squamous cell cancer of the or vulvar histology, all HIV-infected men and women,
distal penile urethra (80). Both tumors are found with and any patient with a history of anogenital condylo-
nearly equal frequency and can look histologically sim- mas. It has become clear from multiple studies that
ilar, especially when biopsies are small (80). The use of testing for HPV is not useful for anal cancer screening
both HPV DNA ISH and p16 IHC is recommended since because the prevalence of HPV and frequency of mixed
although typically associated with HPV, overexpression infections are unusually high in at-risk populations, and
of p16 has been found in up to one-half of urothelial the consequences of infection are not well understood
carcinomas (80). (87). Cytology, however, may be useful; preliminary re-
Determining HPV status as a prognostic factor in commendations for screening men who have sex with
penile cancer remains unclear. There are few reported men include obtaining anal cytology at baseline and
studies that have examined HPV-positive precursor annually for those who are HIV-positive and biennially
lesions and their potential to progress to cancer, and for those who are HIV-negative (88) (Fig. 2). Either con-
results have been inconsistent (78). Several studies from ventional Papanicolaou-stained smears or liquid-based
Europe have concluded that HPV status as determined cytology can be used. Anal cytology smears are evalu-
by PCR and/or p16 IHC positivity in penile squamous ated using the same Bethesda nomenclature as for cer-
cell carcinoma is favorably associated with cancer- vical cytology, with abnormalities designated as atypical
specific survival (81–83). In a cohort of patients in North squamous cells of undetermined significance (ASC-US);
America, HPV status as determined by high-risk HPV atypical squamous cells, cannot exclude a high-grade
ISH and/or p16 IHC was not predictive of outcome squamous intraepithelial lesion (ASC-H); and LSIL and
(recurrence, progression, overall mortality, or disease- HSIL (Table 1). The koilocytic changes associated with
specific survival) (84). A trend toward delayed progres- HPV may not be as pronounced in anal cytology, al-
sion was associated with high-risk HPV ISH positivity though binucleation, multinucleation, and cytoplasmic
in this study, although the association did not reach keratinization may be more prominent than in cervical
statistical significance (84). In another North American lesions (86). There is also a higher incidence and lower
study, p16 IHC positivity was not associated with specificity of atypical squamous cells of undetermined
stage, histologic grade, lymph node status, lymphovas- significance in the anal canal (86).
cular invasion, lymph node metastases, or overall sur-
vival in patients with penile SCC (85). Lack of p16, Diagnosis
however, was significantly associated with recurrence, Similar to cervical cytology, there is substantial inter-
especially in patients who had lymph node involvement observer variation in interpretation of anal cytology,
at diagnosis (85). Additional studies are needed to es- and histological confirmation is important. Histologi-
tablish the role of HPV status as a potential prognostic cal grading is also subject to interobserver variability.
indicator. Standardized terminology for epithelial lesions in all
anatomic locations of the lower anogenital tract was
Anal Cancer developed in 2012 by the Lower Anogenital Squamous
Screening Terminology Standardization (LAST) Project to replace
Analogous to cervical cancer, the ability to detect HPV older nomenclature and eliminate some of the subjec-
and identify precursor dysplastic lesions in the transition tivity in interpretation (89). According to older nomen-
zone of the anal canal could allow for the development clature, precancerous anal lesions were termed anal

ASMscience.org/MicrobiolSpectrum 7
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Sun, 27 Nov 2016 18:41:47
Burd and Dean

FIGURE 2 Algorithm for screening and diagnosis of anal cancer in high-risk groups.
There are currently no formal recommendations for routine anal cancer screening in the
United States.

intraepithelial neoplasia (AIN) and were categorized classifying the lesion as HSIL (89). Absence of p16 stain-
as grade 1, 2, or 3 (mild, moderate, or severe), based ing, or presence of minimal patchy staining, supports
roughly on the distribution of the abnormalities within reducing the classification to LSIL (89).
the epithelium. The LAST terminology is LSIL and HSIL Opinions differ as to the optimal management of
and may include subgrading using the older –IN termi- precursor lesions, and the success of treatment in the
nology (Table 1) (89). Clearly identifying a lesion as –IN prevention of development of anal cancer has not been
2 can be difficult, and p16 IHC is recommended to de- widely established (90, 91). Studies are needed to better
termine the appropriate classification (Fig. 2). Diffuse, characterize the natural history of HPV-associated anal
strong p16 staining in the precancerous lesion supports lesions and to provide evidence for effective treatment.

TABLE 1 Cytological and histological classification of anal dysplasia and cervical dysplasiaa

Natural LAST cervical intraepithelial LAST cervical and anal


history Bethesda 2001 cytology neoplasia histology intraepithelial neoplasia histology
Normal NILM Negative Negative
infection ASC-US atypia atypia
ASC-H
LSIL LSIL (formerly CIN 1) LSIL (formerly AIN1)
Precancer HSIL HSIL (formerly CIN 2, CIN 3, CIS) HSIL (formerly AIN2, AIN3, CIS)
Cancer Carcinoma Carcinoma Carcinoma
AGC Correlates with a variety of histologic N/A
• Not otherwise specified (NOS) outcomes including (111):
• Favor neoplasia • Negative or benign
• CIN 1
• CIN 2/3
• Microinvasive squamous cell carcinoma
• Carcinoma in situ
• Cervical adenocarcinoma
• Endometrial adenocarcinoma
• Endometrial hyperplasia
aNILM, negative for intraepithelial lesion and malignancy; ASC-US, atypical squamous cells unspecified significance; ASC-H, atypical squamous cells cannot exclude HSIL;

LSIL, low-grade squamous intraepithelial lesion; HSIL, high-grade squamous intraepithelial lesion; AGC, atypical glandular cells; CIN, cervical intraepithelial neoplasia; CIS,
carcinoma in situ; AIN, anal intraepithelial neoplasia; AIS, adenocarcinoma in situ.

8 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Sun, 27 Nov 2016 18:41:47
Human Papillomavirus

Cervical Cancer engaged in sexual intercourse should be screened using


Screening cytology (Papanicolaou-stained smear) at 6-month inter-
The current most widely recognized cervical cancer vals in the first year following diagnosis of HIV infection
screening guidelines were updated and independently and then annually after that as long as the cytology re-
published in March 2012 by the American Congress of sults remain normal (101, 102). Women with HIV should
Obstetricians and Gynecologists (ACOG), the United not begin screening until age 21 even if their HIV diag-
States Preventive Services Task Force (USPSTF), and the nosis was before age 21. There are no specific guidelines
American Cancer Society (ACS) in partnership with the regarding screening of transplant recipients for cervical
American Society for Colposcopy and Cervical Pathol- cancer. However, it is generally agreed that it is prudent
ogy (ASCCP) and the American Society for Clinical to screen annually beginning at age 21 rather than every
Pathology (ASCP), with an interim update provided 3 to 5 years as is recommended for normal-risk women
in January 2015 (92–95). These guidelines are focused (99). Screening plans may be modified for individual
on screening algorithms for immune-competent women patients, depending on the degree of immune suppression
with average risk for HPV infection and do not make and other factors (99). Some experts would recommend
recommendations for immunocompromised women. continued cytology screening every 6 months in women
Data are limited, but most studies seem to indicate with CD4 counts less than 200/mm3 or a history of HPV
that HPV infection is detected with greater frequency in infection.
immunosuppressed patients and that immune deficiency The initial biennial cytology screening is recom-
increases persistence of HPV and thereby increases the mended given concerns about the relative insensitivity
risk of cervical dysplasia and invasive cervical can- of cytology and that an abnormal lesion may be missed
cer (56, 96–99). There is some evidence to suggest that with a single cytology smear. The rate of abnormal cy-
high-grade precursor lesions progress more rapidly to tology is high among HIV-infected women and has been
cervical cancer in immune-suppressed women (56). For reported as atypical squamous cell of undetermined
HIV-infected women, no apparent reduction in HPV- significance or LSIL in 37% and as HSIL in 5%, with
associated cervical cancer is afforded by highly active normal cytology in the remaining 58% (56). Prevalence
antiretroviral therapy (100). of HPV is also high among HIV-infected women with
The Centers for Disease Control and Prevention normal cytology and has been reported as 57.4% in
(CDC), National Institutes of Health (NIH), and the In- South/Central America, 56.6% in Africa, 32.4% in
fectious Disease Society of America (IDSA) jointly pub- Europe, 31.4% in North America, and 31.1% in Asia
lished updated cervical cancer screening guidelines for (56). In contrast, HPV detection in the absence of ap-
HIV-infected women in 2009 (101, 102) (Table 2; Fig. 3). parent disease is found in about 11 to 12% of immune-
These guidelines recommend that HIV-infected women competent women. Also of note is that the rate of

TABLE 2 Cervical cancer screening guidelines

Normal-risk women (92–95) Women with HIV (101, 102)


Age at initiation Age 21, regardless of risk Age 21, even if diagnosis of HIV
infection was before age 21a
Frequency
Age 21–29 years Cytology every 3 years –or– primary HPV testing can be considered starting at Cytology at 6-month intervals in the
age 25 every 3 years first year following diagnosis of HIV
infection and then annual cytologyb,c
Age 30–65 years Cytology every 3 years –or– cytology plus HPV every 5 years – or– HPV primary
testing every 3 years starting at age 25 as above
Discontinuation Women age 65 or older who have 3 or more consecutive negative cytology tests or Never?d
of screening two consecutive negative cotests within 10 years, with the most recent test
performed within 5 years
After hysterectomy Women of any age who have had a total hysterectomy and have no history of Same as normal-risk womend
cervical cancer or precancer should not be screened. If there is a history of
CIN 2 or worse, screen for 20 years even if screening extends to beyond age 65
HPV vaccinated No change from above No change from above
aProviders should consider screening within 1 year of onset of sexual activity (101).
bSome experts would recommend continued cytology screening every 6 months in women with CD4 counts less than 200/mm3 or a history of HPV infection.
cAnnual cervical cytology screening is recommended for immunosuppressed women without HIV (90).

dData insufficient in this population.

ASMscience.org/MicrobiolSpectrum 9
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Sun, 27 Nov 2016 18:41:47
Burd and Dean

FIGURE 3 Algorithm for screening and diagnosis of cervical cancer in immunosuppressed


women. For HIV-infected women (90), screen at 6 months, 12 months, then annually;
consider screening within 1 year of onset of sexual activity (92); some experts would
recommend continued cytology screening every 6 months in women with CD4 counts
less than 200/mm3 or a history of HPV infection. For immunosuppressed women without
HIV, annual cervical cytology screening is recommended (90).

infection with multiple HPV types is higher for HIV- could particularly be of benefit in screening of immuno-
infected women than for the general population (56). suppressed women.
This higher rate of multiple HPV infections is found in HPV16 is associated with the majority (48 to 72%) of
both the presence and absence of abnormal cytology (56). high-grade cervical lesions in normal-risk women (105,
The utility of detection of high-risk HPV or HPV 106). A large meta-analysis as well as subsequent studies
genotyping in conjunction with cytology as is endorsed have found that HPV16 is less prevalent (32 to 51%)
for the general population has not been determined for among HIV-infected women with high-grade cervical
immunosuppressed women. The concern is that HPV lesions, and that other subtypes (11, 18, 33, 51, 52, 53,
DNA tests may be less specific in immunocompromised 58, and 61) and infection with multiple HPV types are
women and might not be as effective for screening be- significantly more common (56, 105, 107). These find-
cause of the high prevalence and persistence of HPV ings suggest that genotype testing may have less of a role
among women with HIV (56). Specificity and positive in this population.
predictive value may be improved by detecting E6/E7
mRNA, rather than HPV DNA as has been found in the Diagnosis
general population, but this screening strategy has not Cytology
been studied in immunosuppressed populations. Studies Cervical cytology using Papanicolaou-stained smears has
that compare detection of high-risk HPV with cytology been the standard screening test for detection of prema-
and development of precancerous and cancerous lesions lignant lesions and cervical cancer since 1941. Liquid-
are needed. Results of pilot studies have raised concerns based, thin-layer systems are a more recent technology
about normal cytology and unrecognized high-risk HPV for preparation of smears that requires placing cervical
infection, especially in HIV-infected women 21 to 30 specimens into a vial of liquid preservative. The speci-
years of age (98, 103, 104). Follow-up observations in mens are processed to remove debris and reduce exces-
one of the studies showed that HIV-infected women sive white and red blood cells. The remaining specimen is
with normal cervical cytology and high-risk HPV were then sedimented if the BD SurePath (BD Diagnostics–
4-fold more likely than women without high-risk HPV TriPath, Burlington, NC) system is used, or filtered if the
to have an abnormal finding on colposcopic examina- ThinPrep (Hologic Inc., Marlborough, MA) system is
tion (103). From these results, it appears that detection used, to produce a monolayer of cells on a glass micro-
of high-risk HPV and early colposcopic examination scope slide, which is then stained. The liquid-based pro-

10 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Sun, 27 Nov 2016 18:41:47
Human Papillomavirus

cess improves disease detection by making diagnostically assessed as positive after a single examination should be
relevant cells more clearly visible and results in fewer reviewed a second time for verification.
unsatisfactory slides when compared to conventionally
prepared smears (108). Stained slides are examined man- Molecular HPV tests
ually using a standard light microscope or by an imaging HPV DNA testing in HIV-infected women or adoles-
system (e.g., ThinPrep Imaging System, Hologic Inc.) cents is not recommended at this time, although it has
that uses a computer to automatically scan the slide and now been incorporated into cervical cancer screening for
mark potentially abnormal cells with larger or darker normal-risk women because of the limitations of cytol-
nuclei for review by a cytotechnologist and interpreta- ogy (114) (Table 3). The most common commercially
tion by a pathologist for final diagnosis. The consistent available molecular tests are designed to detect HPV
and objective evaluation of cytology smears afforded by DNA from high-risk types and generate a pooled result
imaging systems is thought to improve diagnostic accu- without identification of the specific type present. There
racy (109). are important differences among the various tests. The
The Bethesda System has been the standard termi- tests include the Digene Hybrid Capture 2 High-Risk
nology used for reporting cervical cytology since 1988. HPV DNA Test (Qiagen, Redwood City, CA) using hy-
The system has been modified several times, and the brid capture technology, the Cervista HPV HR test
nomenclature currently in use was developed in 2001 (Hologic Gen-Probe Inc., San Diego, CA) using fluoro-
(110) (Table 1). Precancerous cytologic abnormalities in metric invader technology, and the cobas HPV real-time
squamous epithelial cells are stratified in several cate- PCR test (Roche Molecular Systems Inc., Pleasanton,
gories. Atypical squamous cells (ASC) are the most CA). The cobas HPV test generates an aggregate result
common abnormal finding and are divided into ASC-US and indicates the presence of HPV16 and/or HPV18.
and ASC-H. LSIL is mild dysplasia caused by HPV in- Other tests are designed specifically to detect HPV16
fection and characterized by squamous cells with nu- and HPV18 (or HPV18/45). Commercially available
clear atypia, perinuclear halo, and dense cytoplasm. genotyping assays include the Cervista HPV16/18 test
HSIL is a more severe abnormality that includes mod- and the Aptima HPV16 18/45. Many studies have de-
erate to severe dysplasia and carcinoma in situ. If LAST termined that detection of HPV E6/E7 mRNA is more
terminology is accepted, squamous histology and cy- specific than DNA-based assays when measured against
tology descriptions will be consistent (Table 1). The clinical disease with a histological diagnosis of moderate
Bethesda system also allows for description of glandular cervical intraepithelial neoplasia (CIN 2) or worse (115,
cell abnormalities which are caused by HPV but are far 116). Currently, the only available test that detects HPV
less common and are categorized as atypical glandular mRNA is the Aptima HPV Assay (Hologic Gen-Probe
cells (AGC) with identification of whether they are en- Inc.). The HPV molecular tests are approved by the FDA
dometrial or endocervical if possible (111). “Adeno- for cervical cancer screening in combination with a Pap
carcinoma in situ” and “AGC favor neoplastic” are test among women over age 30 and for follow-up testing
included as subcategories of AGC and can be difficult to of women with abnormal cytology results. The cobas HPV
detect because the ability to completely sample the test was recently approved as a primary screening test.
endocervical canal for cytologic analysis is somewhat It is important to note that HPV tests, as approved by
limited and glandular lesions may be inaccessible. the FDA, are performed on a fixed volume of sample
Cervical cytology has been very effective in cervical
cancer screening programs but carries a false-negative TABLE 3 Role for HPV testing in HIV-infected womena
(119)
rate of about 5% to 20% (112). Inadequate sampling of
the transformation zone and small lesion size may ac- Normal-risk women HIV-infected women
count for much of the reduced sensitivity, but there is Triage ASC-US cytology result Triage ASC-US cytology result?a
also some interobserver variability as well as screening Cotest with cytology for Follow-up after colposcopy or
errors by cytotechnologists and interpretive errors by women ≥age 30 years treatment (loop electrosurgical
excision procedure/cone biopsy
pathologists that contribute. Steps to reduce laboratory
Postmenopausal women
reading errors were mandated under the Clinical Labo- with LSIL
ratory Improvement Act of 1988 (CLIA), which requires Follow-up after colposcopy or
that a random sample of at least 10% of cytology smears treatment (loop electrosurgical
are rescreened by a pathologist or senior cytotechnol- excision procedure/cone biopsy

ogist for quality control (113). In addition, all slides a Data insufficient in this population.

ASMscience.org/MicrobiolSpectrum 11
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Sun, 27 Nov 2016 18:41:47
Burd and Dean

randomly obtained from the collection vial used for liq- ablative (e.g., cryotherapy, laser vaporization, electro-
uid-based cytology or a cervical brush in preservative cautery, diathermy, cold coagulation) or excisional (e.g.,
fluid. Because cytology analyzes only cells and HPV tests loop electrosurgical excision procedure, laser coniza-
analyze cells plus extracellular material, they are not ex- tion, cold knife conization) to remove or destroy the
actly comparable (117). The extracellular material ex- abnormal cells. An excisional procedure is indicated in
amined in HPV tests may contain a large amount of patients with recurrent or persistent high-grade CIN or
nucleic acids from free virions as are found in produc- if colposcopy is unsatisfactory (119). For HIV-infected
tive transient infections. When HPV tests are performed adolescents, standard treatment guidelines for adoles-
using an enriched cellular fraction obtained after centri- cents and young adults are considered to be safe and
fugation rather than a random whole sample from the effective (119). Recurrence is more common in these pa-
collection vial, the specificity of the HPV test for detection tients, and close observation as defined in the guidelines
of corresponding abnormal cytology is improved (117). for management of CIN 1 and CIN 2 is important. For
Another aspect is that even molecular HPV tests with a patients with CIN 1 who may not be compliant, fol-
high analytical sensitivity of about 20 to 400 HPV DNA lowing the more stringent management guidelines for
copies per reaction are not sensitive enough to detect all CIN 2 may be preferable (119).
cancers (118). The lower limit can easily be reached when The success of treatment has been monitored using a
few clonal (abnormal) cells are present in the sample. wide variety of follow-up approaches including repeat
cytology, HPV testing, cytology and HPV cotesting, or
Management colposcopy alone or in combination with cytology or
There is controversy about management of abnormal HPV tests. There are no established guidelines for test-
cytology in HIV-infected women. ASCCP guidelines in ing, and follow-up intervals vary among reports from
2001 presented separate recommendations for the man- 3 to 12 months (119, 121). Although posttreatment
agement of ASC-US in women with HIV infection and data from randomized trials are not available, HPV
other immunosuppressive conditions. These separate testing is generally considered to be more sensitive than
guidelines were eliminated in 2006 in favor of managing cytology and may allow for earlier diagnosis of persis-
immunosuppressed women in the same manner as the tent or recurrent disease, while a negative HPV test may
general population. ASC-US is managed in the general allow for less intense management (119, 121). Further
population either by repeat cytology (at 1 year is ac- work to refine follow-up strategies is needed for the gen-
ceptable) or by high-risk HPV testing, which is preferred eral population as well as specifically for immunocom-
(119). Other guidelines suggest that there are not enough promised patients.
data to recommend HPV testing alone in the manage-
ment of ASC-US and suggest that either immediate re- Prevention
ferral to colposcopy or repeat cytology in 6 to 12 months Prophylactic vaccination
should be used (120). Some authorities recommend that The FDA has approved two LI (major capsid protein)
all HIV-infected women with atypical squamous cell of virus-like particle vaccines that also contain adjuvants to
undetermined significance be referred for only immedi- enhance both major histocompatibility complex class I
ate colposcopy and directed biopsy, with further treat- (to activate cytotoxic [CD8+] T lymphocytes) and class II
ment based on those results (102). (to activate T-helper [CD4+] lymphocytes) presentation
For any lesion greater than ASC-US (i.e., ASC-H, pathways. Gardasil (Merck & Co., Whitehouse Station,
LSIL, HSIL or AGC), referral for colposcopy is recom- NJ) is a quadrivalent vaccine that protects against disease
mended (102, 119). It is also recommended that HIV- caused by HPV types 6, 11, 16, and 18, and Cervarix
infected women with cervical HSIL should be referred (GlaxoSmithKline Biologicals, Rixensart, Belgium) is a
for anal cytologic screening as they are at increased risk bivalent vaccine that protects against HPV types 16 and
for anal dysplasia and cancer (102). 18. The quadrivalent vaccine is approved for both fe-
males and males 9 to 26 years of age while the bivalent
Treatment monitoring vaccine is approved only for females 9 to 26 years of age.
Although HPV is more difficult to treat and more likely The vaccine is administered in three doses, with the first
to recur with increased immunosuppression, cervical dose of the quadrivalent or bivalent vaccine recom-
cancer precursors in HIV-infected and other immunosup- mended for girls 11 to 12 years old and the quadrivalent
pressed women should generally be managed accord- vaccine for boys 11 or 12 years old, but either vaccine
ing to ASCCP guidelines (119). Treatment may include can be administered as early as age 9 years. The second

12 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Sun, 27 Nov 2016 18:41:47
Human Papillomavirus

dose should be administered 1 to 2 months after the first Therapeutic vaccination


dose, and the third dose 6 months after the first dose, There is limited literature regarding therapeutic HPV
with a minimum interval of 12 weeks between the second vaccination. Clearance of a naturally acquired HPV in-
and third dose (122). Catch-up vaccination using the fection is mediated by a specific cell-mediated immune
standard dosing intervals is also recommended for response in which dendritic cells stimulate T-helper type
females and males aged 13 through 26 years old if they 1 lymphocytes, which then elicit the production of
were not previously vaccinated or if they have not cytotoxic T lymphocytes that attack infected cells. The
completed the three-dose series (122). Vaccination in the ability of an HPV vaccine to stimulate a specific cellular
general population induces a cell-mediated immune re- immune response could potentially have a therapeutic
sponse and a strong humoral immune response with effect (124). The current FDA-approved L1 VLP vac-
antibody titers several times higher than typically seen cines were designed to prevent HPV infection primarily
after natural infections (123, 124). Vaccination is by stimulating a neutralizing antibody response and
recommended regardless of the presence of abnormal were not intended for therapeutic purposes. Several
cervical cytology, as it may protect against one or more short-term studies suggest that the current vaccines may
HPV types to which the individual has not been exposed. be somewhat effective in patients with existing HPV
In addition, the vaccine has been shown to be effective in infection, while other studies have not found a signifi-
older individuals, and vaccination up to age 45 has been cant effect (125, 127–129).
recommended by some groups (125). Vaccines that are designed to boost specific T-cell
Several published studies have shown that HPV responses could potentially be therapeutically effective.
vaccines are well tolerated in immunocompromised Since the L1 and L2 structural capsid late proteins ap-
individuals and that a specific humoral immune response pear only in fully differentiated epithelial cells later in
was produced with high rates of seroconversion in the course of infection, therapeutic vaccines have been
HIV-infected individuals after three doses and 100% directed toward stimulating T-cell responses to the HPV
seroconversion after four doses in one study (25, 90). E6 and E7 viral oncoproteins, which are continuously
Seroconversion rates, however, have been less than op- expressed throughout the course of infection (130). Ap-
timal in vaccinated organ transplant recipients (126). proaches have included administration of peptide anti-
Although immunosuppression does not prevent suc- gens or recombinant proteins, live viral vector vaccines,
cessful immunization, most studies have found that whole-cell vaccines, and plasmid DNA vaccines as well
antibody titers are lower in immunocompromised indi- as administration of E7-pulsed dendritic cells (130).
viduals than in immunocompetent individuals (25, 90, DNA vaccines have been favored since they are stable,
126). Among HIV-infected individuals, antibody titers safe for multiple administrations, can provide sustained
are higher in those who are on treatment with antiretro- release of antigenic proteins, and can be delivered by a
viral agents. While safety has been established, optimal variety of methods (130). Modifications that lead to
dosing schedules and the efficacy of vaccination in im- enhanced immunogenicity are being investigated (130).
munocompromised individuals will require additional Effective therapeutic vaccination in the face of CD4+
study. T-cell compromise or depletion is of concern in some
The Advisory Committee on Immunization Practices primary immune deficiencies and in HIV-infected indi-
(ACIP) and IDSA recommend HPV vaccination for im- viduals. In a murine immunodeficiency model, a DNA
munocompromised men and women, including those vaccine against HPV E6 and E7 was shown to be ca-
with HIV infection, as well as for men who have sex with pable of generating a specific CD8+ T-cell-mediated
men. Vaccination is recommended regardless of the se- immune response in the absence of CD4+ T cells and was
verity of immunosuppression and regardless of history protective when challenged with subcutaneous admin-
of abnormal cervical cytology. The dosing schedule for istration of E6- and E7-expressing tumor cells, although
immunocompromised individuals is the same as for no antibody response was detected (131). Further in-
immunocompetent individuals, with the three-dose se- vestigations are needed to determine the long-term effect
ries given at age 11 or 12 years and at age 13 through 26 of therapeutic vaccination in management strategies
years if not previously vaccinated (102, 122). Immuno- for both immune-competent and immunocompromised
compromised individuals should continue to have rou- individuals. A chimeric vaccine that contains preventive
tine cervical and anal cancer screening tests and visual and therapeutic components may ultimately be ideal
and digital pelvic and anorectal examinations even if and could be used for both treatment and prophylaxis
they have been vaccinated. (123).

ASMscience.org/MicrobiolSpectrum 13
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Sun, 27 Nov 2016 18:41:47
Burd and Dean

REFERENCES 22. Hoy T, Singhal PK, Willey VJ, Insinga RP. 2009. Assessing incidence
1. Groves IJ, Coleman N. 2015. Pathogenesis of human papillomavirus- and economic burden of genital warts with data from a US commercially
associated mucosal disease. J Pathol 235:527–538. insured population. Curr Med Res Opin 25:2343–2351.
2. Doorbar J, Quint W, Banks L, Bravo IG, Stoler M, Broker TR, Stanley 23. Jemal A, Simard EP, Dorell C, Noone AM, Markowitz LE, Kohler B,
MA. 2012. The biology and life-cycle of human papillomaviruses. Vaccine Eheman C, Saraiya M, Bandi P, Saslow D, Cronin KA, Watson M,
30(Suppl 5):F55–F70. Schiffman M, Henley SJ, Schymura MJ, Anderson RN, Yankey D,
3. zur Hausen H. 2002. Papillomaviruses and cancer: from basic studies to Edwards BK. 2013. Annual Report to the Nation on the Status of Cancer,
clinical application. Nat Rev Cancer 2:342–350. 1975-2009, featuring the burden and trends in human papillomavirus
(HPV)-associated cancers and HPV vaccination coverage levels. J Natl
4. zur Hausen H. 2009. Papillomaviruses in the causation of human
Cancer Inst 105:175–201.
cancers – a brief historical account. Virology 384:260–265.
24. Palefsky J. 2007. Human papillomavirus infection in HIV-infected
5. Forman D, de Martel C, Lacey CJ, Soerjomataram I, Lortet-Tieulent J,
persons. Top HIV Med 15:130–133.
Bruni L, Vignat J, Ferlay J, Bray F, Plummer M, Franceschi S. 2012.
Global burden of human papillomavirus and related diseases. Vaccine 30 25. Denny LA, Franceschi S, de Sanjosé S, Heard I, Moscicki AB, Palefsky
(Suppl 5):F12–F23. J. 2012. Human papillomavirus, human immunodeficiency virus and
immunosuppression. Vaccine 30(Suppl 5):F168–F174.
6. Dunne EF, Park IU. 2013. HPV and HPV-associated diseases. Infect Dis
Clin North Am 27:765–778. 26. Freiberger D, Lewis L, Helfand L. 2015. Human papillomavirus-
related high-grade squamous intraepithelial lesions of the esophagus, skin,
7. Egawa N, Egawa K, Griffin H, Doorbar J. 2015. Human papilloma-
and cervix in an adolescent lung transplant recipient: a case report and
viruses; epithelial tropisms, and the development of neoplasia. Viruses
literature review. Transpl Infect Dis 17:98–102.
7:3863–3890.
27. Palefsky JM. 1997. Cutaneous and genital HPV-associated lesions in
8. Stanley M. 2008. Immunobiology of HPV and HPV vaccines. Gynecol
HIV-infected patients. Clin Dermatol 15:439–447.
Oncol 109(Suppl):S15–S21.
28. Gormley RH, Kovarik CL. 2009. Dermatologic manifestations of
9. Arbyn M, Tommasino M, Depuydt C, Dillner J. 2014. Are 20 human
HPV in HIV-infected individuals. Curr HIV/AIDS Rep 6:130–138.
papillomavirus types causing cervical cancer? J Pathol 234:431–435.
29. Partridge JM, Koutsky LA. 2006. Genital human papillomavirus in-
10. Halec G, Alemany L, Lloveras B, Schmitt M, Alejo M, Bosch FX, Tous
fection in men. Lancet Infect Dis 6:21–31.
S, Klaustermeier JE, Guimerà N, Grabe N, Lahrmann B, Gissmann L,
Quint W, Bosch FX, de Sanjose S, Pawlita M, Retrospective International 30. Wheless L, Jacks S, Mooneyham Potter KA, Leach BC, Cook J. 2014.
Survey and HPV Time Trends Study Group, Retrospective International Skin cancer in organ transplant recipients: more than the immune system.
Survey and HPV Time Trends Study Group. 2014. Pathogenic role of the J Am Acad Dermatol 71:359–365.
eight probably/possibly carcinogenic HPV types 26, 53, 66, 67, 68, 70, 73 31. Gaiser MR, Textor S, Senger T, Schädlich L, Waterboer T, Kaufmann
and 82 in cervical cancer. J Pathol 234:441–451. AM, Süsal C, Pawlita M, Enk AH, Gissmann L, Lonsdorf AS. 2015.
11. Finnen RL, Erickson KD, Chen XS, Garcea RL. 2003. Interac- Evaluation of specific humoral and cellular immune responses against the
tions between papillomavirus L1 and L2 capsid proteins. J Virol 77:4818– major capsid L1 protein of cutaneous wart-associated alpha-papilloma-
4826. viruses in solid organ transplant recipients. J Dermatol Sci 77:37–45.
12. Doorbar J. 2006. Molecular biology of human papillomavirus infec- 32. Bouwes Bavinck JN, Feltkamp M, Struijk L, ter Schegget J. 2001.
tion and cervical cancer. Clin Sci (Lond) 110:525–541. Human papillomavirus infection and skin cancer risk in organ transplant
recipients. J Investig Dermatol Symp Proc 6:207–211.
13. Van Tine BA, Dao LD, Wu SY, Sonbuchner TM, Lin BY, Zou N,
Chiang CM, Broker TR, Chow LT. 2004. Human papillomavirus (HPV) 33. Bouwes Bavinck JN, Berkhout RJ. 1997. HPV infections and immu-
origin-binding protein associates with mitotic spindles to enable viral nosuppression. Clin Dermatol 15:427–437.
DNA partitioning. Proc Natl Acad Sci USA 101:4030–4035. 34. Patel T, Morrison LK, Rady P, Tyring S. 2010. Epidermodysplasia
14. Middleton K, Peh W, Southern S, Griffin H, Sotlar K, Nakahara T, verruciformis and susceptibility to HPV. Dis Markers 29:199–206.
El-Sherif A, Morris L, Seth R, Hibma M, Jenkins D, Lambert P, Coleman 35. Rizzo JD, Curtis RE, Socié G, Sobocinski KA, Gilbert E, Landgren O,
N, Doorbar J. 2003. Organization of human papillomavirus productive Travis LB, Travis WD, Flowers ME, Friedman DL, Horowitz MM,
cycle during neoplastic progression provides a basis for selection of di- Wingard JR, Deeg HJ. 2009. Solid cancers after allogeneic hematopoietic
agnostic markers. J Virol 77:10186–10201. cell transplantation. Blood 113:1175–1183.
15. Stanley M. 2006. Immune responses to human papillomavirus. 36. Gewirtzman A, Bartlett B, Tyring S. 2008. Epidermodysplasia ver-
Vaccine 24(Suppl 1):S16–S22. ruciformis and human papilloma virus. Curr Opin Infect Dis 21:141–146.
16. Schiffman M, Wentzensen N. 2013. Human papillomavirus infection 37. Rogers HD, Macgregor JL, Nord KM, Tyring S, Rady P, Engler DE,
and the multistage carcinogenesis of cervical cancer. Cancer Epidemiol Grossman ME. 2009. Acquired epidermodysplasia verruciformis. J Am
Biomarkers Prev 22:553–560. Acad Dermatol 60:315–320.
17. Klingelhutz AJ, Roman A. 2012. Cellular transformation by human 38. Parkin DM, Bray F, Ferlay J, Pisani P. 2005. Global cancer statistics,
papillomaviruses: lessons learned by comparing high- and low-risk 2002. CA Cancer J Clin 55:74–108.
viruses. Virology 424:77–98. 39. Beachler DC, D'Souza G. 2013. Oral human papillomavirus infection
18. DiMaio D, Petti LM. 2013. The E5 proteins. Virology 445:99–114. and head and neck cancers in HIV-infected individuals. Curr Opin Oncol
19. Kadaja M, Silla T, Ustav E, Ustav M. 2009. Papillomavirus DNA 25:503–510.
replication – from initiation to genomic instability. Virology 384:360– 40. Beachler DC, Weber KM, Margolick JB, Strickler HD, Cranston RD,
368. Burk RD, Wiley DJ, Minkoff H, Reddy S, Stammer EE, Gillison ML,
20. Maglennon GA, McIntosh PB, Doorbar J. 2014. Immunosuppression D’Souza G. 2012. Risk factors for oral HPV infection among a high
facilitates the reactivation of latent papillomavirus infections. J Virol prevalence population of HIV-positive and at-risk HIV-negative adults.
88:710–716. Cancer Epidemiol Biomarkers Prev 21:122–133.
21. Satterwhite CL, Torrone E, Meites E, Dunne EF, Mahajan R, Ocfemia 41. Gillison ML, Shah KV. 2001. Human papillomavirus-associated head
MC, Su J, Xu F, Weinstock H. 2013. Sexually transmitted infections and neck squamous cell carcinoma: mounting evidence for an etiologic
among US women and men: prevalence and incidence estimates, 2008. role for human papillomavirus in a subset of head and neck cancers. Curr
Sex Transm Dis 40:187–193. Opin Oncol 13:183–188.

14 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Sun, 27 Nov 2016 18:41:47
Human Papillomavirus

42. Madeleine MM, Finch JL, Lynch CF, Goodman MT, Engels EA. 59. Savani BN, Stratton P, Shenoy A, Kozanas E, Goodman S, Barrett AJ.
2013. HPV-related cancers after solid organ transplantation in the United 2008. Increased risk of cervical dysplasia in long-term survivors of allo-
States. Am J Transplant 13:3202–3209. geneic stem cell transplantation—implications for screening and HPV
43. Katz J, Islam MN, Bhattacharyya I, Sandow P, Moreb JS. 2014. Oral vaccination. Biol Blood Marrow Tr 14:1072–1075.
squamous cell carcinoma positive for p16/human papilloma virus in post 60. Wang CY, Brodland DG, Su WP. 1995. Skin cancers associated with
allogeneic stem cell transplantation: 2 cases and review of the literature. acquired immunodeficiency syndrome. Mayo Clin Proc 70:766–772.
Oral Surg Oral Med Oral Pathol Oral Radiol 118:e74–e78. 61. Reusser NM, Downing C, Guidry J, Tyring SK. 2015. HPV carcino-
44. Johnson LG, Madeleine MM, Newcomer LM, Schwartz SM, Daling mas in immunocompromised patients. J Clin Med 4:260–281.
JR. 2004. Anal cancer incidence and survival: the surveillance, epidemi- 62. Paradisi A, Waterboer T, Sampogna F, Tabolli S, Simoni S, Pawlita M,
ology, and end results experience, 1973-2000. Cancer 101:281–288. Abeni D. 2011. Seropositivity for human papillomavirus and incidence
45. Tong WW, Hillman RJ, Kelleher AD, Grulich AE, Carr A. 2014. Anal of subsequent squamous cell and basal cell carcinomas of the skin in
intraepithelial neoplasia and squamous cell carcinoma in HIV-infected patients with a previous nonmelanoma skin cancer. Br J Dermatol 165:
adults. HIV Med 15:65–76. 782–791.
46. Darwich L, Videla S, Cañadas MP, Piñol M, García-Cuyàs F, Vela S, 63. Blessing K, McLaren KM, Benton EC, Barr BB, Bunney MH, Smith
Molina-López RA, Coll J, Sirera G, Clotet B, Can Ruti H, Can Ruti HIV- IW, Beveridge GW. 1989. Histopathology of skin lesions in renal allograft
HPV Team. 2013. Distribution of human papillomavirus genotypes in recipients—an assessment of viral features and dysplasia. Histopathology
anal cytological and histological specimens from HIV-infected men who 14:129–139.
have sex with men and men who have sex with women. Dis Colon Rectum 64. Gutiérrez-Dalmau A, Revuelta I, Ferrer B, Mascaró JM Jr,
56:1043–1052. Oppenheimer F, Albanell J, Campistol JM. 2010. Distinct immunohisto-
47. Silverberg MJ, Lau B, Justice AC, Engels E, Gill MJ, Goedert JJ, chemical phenotype of nonmelanoma skin cancers between renal trans-
Kirk GD, D’Souza G, Bosch RJ, Brooks JT, Napravnik S, Hessol NA, plant and immunocompetent populations. Transplantation 90:986–992.
Jacobson LP, Kitahata MM, Klein MB, Moore RD, Rodriguez B, Rourke 65. Pfefferle R, Marcuzzi GP, Akgül B, Kasper HU, Schulze F, Haase I,
SB, Saag MS, Sterling TR, Gebo KA, Press N, Martin JN, Dubrow R, Wickenhauser C, Pfister H. 2008. The human papillomavirus type 8 E2
North American AIDS Cohort Collaboration on Research and Design protein induces skin tumors in transgenic mice. J Invest Dermatol 128:
(NA-ACCORD) of IeDEA. 2012. Risk of anal cancer in HIV-infected and 2310–2315.
HIV-uninfected individuals in North America. Clin Infect Dis 54:1026–
66. U.S. Preventive Services Task Force. 2013. Screening for Oral Cancer:
1034. Final Recommendation Statement. AHRQ Publication No. 13-05186-
48. Roka S, Rasoul-Rockenschaub S, Roka J, Kirnbauer R, Mühlbacher F, EF-2. Agency for Healthcare Research & Quality, Rockville MD.
Salat A. 2004. Prevalence of anal HPV infection in solid-organ transplant
67. Rethman MP, Carpenter W, Cohen EE, Epstein J, Evans CA, Flaitz
patients prior to immunosuppression. Transpl Int 17:366–369. CM, Graham FJ, Hujoel PP, Kalmar JR, Koch WM, Lambert PM, Lingen
49. Patel HS, Silver AR, Northover JM. 2007. Anal cancer in renal MW, Oettmeier BW Jr, Patton LL, Perkins D, Reid BC, Sciubba JJ, Tomar
transplant patients. Int J Colorectal Dis 22:1–5. SL, Wyatt AD Jr, Aravamudhan K, Frantsve-Hawley J, Cleveland JL,
50. Hernandez BY, Barnholtz-Sloan J, German RR, Giuliano A, Goodman Meyer DM, American Dental Association Council on Scientific Affairs
MT, King JB, Negoita S, Villalon-Gomez JM. 2008. Burden of invasive Expert Panel on Screening for Oral Squamous Cell Carcinomas. 2012.
squamous cell carcinoma of the penis in the United States, 1998-2003. Evidence-based clinical recommendations regarding screening for oral
Cancer 113(Suppl):2883–2891. squamous cell carcinomas. Tex Dent J 129:491–507.
51. Hernandez BY, Goodman MT, Unger ER, Steinau M, Powers A, 68. Mirghani H, Amen F, Moreau F, Guigay J, Ferchiou M, Melkane AE,
Lynch CF, Cozen W, Saber MS, Peters ES, Wilkinson EJ, Copeland G, Hartl DM, Lacau St Guily J. 2014. Human papilloma virus testing in
Hopenhayn C, Huang Y, Watson M, Altekruse SF, Lyu C, Saraiya M, oropharyngeal squamous cell carcinoma: what the clinician should know.
HPV Typing of Cancer Workgroup. 2014. Human papillomavirus ge- Oral Oncol 50:1–9.
notype prevalence in invasive penile cancers from a registry-based United 69. Weinberger PM, Yu Z, Haffty BG, Kowalski D, Harigopal M,
States population. Front Oncol 4:9. Brandsma J, Sasaki C, Joe J, Camp RL, Rimm DL, Psyrri A. 2006.
52. Backes DM, Kurman RJ, Pimenta JM, Smith JS. 2009. Systematic Molecular classification identifies a subset of human papillomavirus–
review of human papillomavirus prevalence in invasive penile cancer. associated oropharyngeal cancers with favorable prognosis. J Clin Oncol
Cancer Cause Control 20:449–457. 24:736–747.
53. IARC Working Group on the Evaluation of Carcinogenic Risks to 70. Begum S, Gillison ML, Ansari-Lari MA, Shah K, Westra WH.
Humans. 2012. Biological agents. Volume 100 B. A review of human 2003. Detection of human papillomavirus in cervical lymph nodes: a
carcinogens. IARC Monogr Eval Carcinog Risks Hum 100(Pt B):1–441. highly effective strategy for localizing site of tumor origin. Clin Cancer Res
54. International Collaboration of Epidemiological Studies of Cervical 9:6469–6475.
Cancer. 2007. Comparison of risk factors for invasive squamous cell 71. Syrjänen S. 2005. Human papillomavirus (HPV) in head and neck
carcinoma and adenocarcinoma of the cervix: collaborative reanalysis of cancer. J Clin Virol 32(Suppl 1):S59–S66.
individual data on 8,097 women with squamous cell carcinoma and 1,374 72. Kerr DA, Pitman MB, Sweeney B, Arpin RN III, Wilbur DC, Faquin
women with adenocarcinoma from 12 epidemiological studies. Int J WC. 2014. Performance of the Roche cobas 4800 high-risk human pap-
Cancer 120:885–891. illomavirus test in cytologic preparations of squamous cell carcinoma of
55. Dugué PA, Rebolj M, Garred P, Lynge E. 2013. Immunosuppression the head and neck. Cancer Cytopathol 122:167–174.
and risk of cervical cancer. Expert Rev Anticancer Ther 13:29–42. 73. Shi W, Kato H, Perez-Ordonez B, Pintilie M, Huang S, Hui A,
56. Nguyen ML, Flowers L. 2013. Cervical cancer screening in immu- O’Sullivan B, Waldron J, Cummings B, Kim J, Ringash J, Dawson LA,
nocompromised women. Obstet Gynecol Clin North Am 40:339–357. Gullane P, Siu L, Gillison M, Liu FF. 2009. Comparative prognostic value
57. Brickman C, Palefsky JM. 2015. Human papillomavirus in the HIV- of HPV16 E6 mRNA compared with in situ hybridization for human
infected host: epidemiology and pathogenesis in the antiretroviral era. oropharyngeal squamous carcinoma. J Clin Oncol 27:6213–6221.
Curr HIV/AIDS Rep 12:6–15. 74. Lewis JS Jr, Thorstad WL, Chernock RD, Haughey BH, Yip JH,
58. Wang Y, Brinch L, Jebsen P, Tanbo T, Kirschner R. 2012. A clinical Zhang Q, El-Mofty SK. 2010. p16 positive oropharyngeal squamous cell
study of cervical dysplasia in long-term survivors of allogeneic stem cell carcinoma: an entity with a favorable prognosis regardless of tumor HPV
transplantation. Biol Blood Marrow Tr 18:747–753. status. Am J Surg Pathol 34:1088–1096.

ASMscience.org/MicrobiolSpectrum 15
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Sun, 27 Nov 2016 18:41:47
Burd and Dean

75. Schache AG, Liloglou T, Risk JM, Filia A, Jones TM, Sheard J, from the College of American Pathologists and the American Society for
Woolgar JA, Helliwell TR, Triantafyllou A, Robinson M, Sloan P, Colposcopy and Cervical Pathology. Arch Pathol Lab Med 136:1266–
Harvey-Woodworth C, Sisson D, Shaw RJ. 2011. Evaluation of human 1297.
papilloma virus diagnostic testing in oropharyngeal squamous cell carci- 90. Schim van der Loeff MF, Mooij SH, Richel O, de Vries HJ, Prins JM.
noma: sensitivity, specificity, and prognostic discrimination. Clin Cancer 2014. HPV and anal cancer in HIV-infected individuals: a review. Curr
Res 17:6262–6271. HIV/AIDS Rep 11:250–262.
76. Westra WH. 2012. Detection of human papillomavirus in clinical 91. Glynne-Jones R, Nilsson PJ, Aschele C, Goh V, Peiffert D, Cervantes
samples. Otolaryngol Clin North Am 45:765–777. A, Arnold D, European Society for Medical Oncology (ESMO), European
77. Lewis JS Jr, Khan RA, Masand RP, Chernock RD, Zhang Q, Al-Naief Society of Surgical Oncology (ESSO), European Society of Radiotherapy
NS, Muller S, McHugh JB, Prasad ML, Brandwein-Gensler M, Perez- and Oncology (ESTRO). 2014. Anal cancer: ESMO-ESSO-ESTRO clini-
Ordonez B, El-Mofty SK. 2012. Recognition of nonkeratinizing mor- cal practice guidelines for diagnosis, treatment and follow-up. Eur J Surg
phology in oropharyngeal squamous cell carcinoma - a prospective cohort Oncol 40:1165–1176.
and interobserver variability study. Histopathology 60:427–436 10.1111 92. Committee on Practice Bulletins—Gynecology. 2012. ACOG Practice
/j.1365-2559.2011.04092.x. Bulletin Number 131: screening for cervical cancer. Obstet Gynecol
78. Bleeker MC, Heideman DA, Snijders PJ, Horenblas S, Dillner J, Meijer 120:1222–1238.
CJ. 2009. Penile cancer: epidemiology, pathogenesis and prevention. 93. Saslow D, Solomon D, Lawson HW, Killackey M, Kulasingam SL,
World J Urol 27:141–150. Cain J, Garcia FA, Moriarty AT, Waxman AG, Wilbur DC, Wentzensen
79. Mentrikoski MJ, Stelow EB, Culp S, Frierson HF Jr, Cathro HP. 2014. N, Downs LS Jr, Spitzer M, Moscicki AB, Franco EL, Stoler MH,
Histologic and immunohistochemical assessment of penile carcinomas in a Schiffman M, Castle PE, Myers ER, ACS-ASCCP-ASCP Cervical Cancer
North American population. Am J Surg Pathol 38:1340–1348. Guideline Committee. 2012. American Cancer Society, American Society
80. Chaux A, Cubilla AL, Haffner MC, Lecksell KL, Sharma R, Burnett for Colposcopy and Cervical Pathology, and American Society for Clinical
AL, Netto GJ. 2014. Combining routine morphology, p16(INK4a) im- Pathology screening guidelines for the prevention and early detection of
munohistochemistry, and in situ hybridization for the detection of human cervical cancer. CA Cancer J Clin 62:147–172.
papillomavirus infection in penile carcinomas: a tissue microarray study 94. Moyer VA, U.S. Preventive Services Task Force. 2012. Screening for
using classifier performance analyses. Urol Oncol 32:171–177. cervical cancer: U.S. Preventive Services Task Force recommendation
81. Gunia S, Erbersdobler A, Hakenberg OW, Koch S, May M. 2012. statement. Ann Intern Med 156:880–891, W312.
p16(INK4a) is a marker of good prognosis for primary invasive penile 95. Huh WK, Ault KA, Chelmow D, Davey DD, Goulart RA, Garcia FAR,
squamous cell carcinoma: a multi-institutional study. J Urol 187:899– Kinney WK, Massad LS, Mayeaux EJ, Saslow D, Schiffman M,
907. Wentzensen N, Lawson HW, Einstein MH. 2015. Use of primary high-
82. Ferrándiz-Pulido C, Masferrer E, de Torres I, Lloveras B, Hernandez- risk human papillomavirus testing for cervical cancer screening: interim
Losa J, Mojal S, Salvador C, Morote J, Ramon y Cajal S, Pujol RM, clinical guidance. Gynecol Oncol 136:178–182.
Garcia-Patos V, Toll A. 2013. Identification and genotyping of human 96. Massad LS, Ahdieh L, Benning L, Minkoff H, Greenblatt RM, Watts
papillomavirus in a Spanish cohort of penile squamous cell carcinomas: H, Miotti P, Anastos K, Moxley M, Muderspach LI, Melnick S. 2001.
correlation with pathologic subtypes, p16(INK4a) expression, and prog- Evolution of cervical abnormalities among women with HIV-1: evidence
nosis. J Am Acad Dermatol 68:73–82. from surveillance cytology in the women’s interagency HIV study.
83. Djajadiningrat RS, Jordanova ES, Kroon BK, van Werkhoven E, J Acquir Immune Defic Syndr 27:432–442.
de Jong J, Pronk DTM, Snijders PJF, Horenblas S, Heideman DAM. 2015. 97. Fife KH, Wu JW, Squires KE, Watts DH, Andersen JW, Brown DR.
Human papillomavirus prevalence in invasive penile cancer and associa- 2009. Prevalence and persistence of cervical human papillomavirus in-
tion with clinical outcome. J Urol 193:526–531. fection in HIV-positive women initiating highly active antiretroviral
84. Bezerra SM, Chaux A, Ball MW, Faraj SF, Munari E, Gonzalez- therapy. J Acquir Immune Defic Syndr 51:274–282.
Roiban N, Sharma R, Bivalacqua TJ, Burnett AL, Netto GJ. 2015. 98. Konopnicki D, Manigart Y, Gilles C, Barlow P, de Marchin J, Feoli F,
Human papillomavirus infection and immunohistochemical p16INK4a Larsimont D, Delforge M, De Wit S, Clumeck N. 2013. Sustained viral
expression as predictors of outcome in penile squamous cell carcinomas. suppression and higher CD4+ T-cell count reduces the risk of persistent
Hum Pathol 46:532–540. cervical high-risk human papillomavirus infection in HIV-positive
85. Tang DH, Clark PE, Giannico G, Hameed O, Chang SS, Gellert LL. women. J Infect Dis 207:1723–1729.
2015. Lack of P16ink4a over expression in penile squamous cell carci- 99. Krajewski C, Sucato G. 2014. Reproductive health care after trans-
noma is associated with recurrence after lymph node dissection. J Urol plantation. Best Pract Res Clin Obstet Gynaecol 28:1222–1234.
193:519–525. 100. Adler DH. 2010. The impact of HAART on HPV-related cervical
86. Darragh TM, Winkler B. 2011. Anal cancer and cervical cancer disease. Curr HIV Res 8:493–497.
screening: key differences. Cancer Cytopathol 119:5–19. 101. Kaplan JE, Benson C, Holmes KK, Brooks JT, Pau A, Masur H,
87. Castle PE, Follansbee S, Borgonovo S, Tokugawa D, Schwartz LM, Centers for Disease Control and Prevention (CDC), National Institutes of
Lorey TS, LaMere B, Gage JC, Fetterman B, Darragh TM, Rodriguez AC, Health, HIV Medicine Association of the Infectious Diseases Society of
Wentzensen N. 2013. A comparison of human papillomavirus genotype- America. 2009. Guidelines for prevention and treatment of opportunistic
specific DNA and E6/E7 mRNA detection to identify anal precancer infections in HIV-infected adults and adolescents: recommendations from
among HIV-infected men who have sex with men. Cancer Epidemiol CDC, the National Institutes of Health, and the HIV Medicine Associa-
Biomarkers Prev 22:42–49. tion of the Infectious Diseases Society of America. MMWR Recomm Rep
88. Park IU, Palefsky JM. 2010. Evaluation and management of anal 58(RR-4):1–207, quiz CE1–CE4.
intraepithelial neoplasia in HIV-negative and HIV-positive men who have 102. Aberg JA, Gallant JE, Ghanem KG, Emmanuel P, Zingman BS,
sex with men. Curr Infect Dis Rep 12:126–133. Horberg MA, Infectious Diseases Society of America. 2014. Primary care
89. Darragh TM, Colgan TJ, Cox JT, Heller DS, Henry MR, Luff RD, guidelines for the management of persons infected with HIV: 2013 update
McCalmont T, Nayar R, Palefsky JM, Stoler MH, Wilkinson EJ, Zaino by the HIV Medicine Association of the Infectious Diseases Society of
RJ, Wilbur DC, Members of LAST Project Work Groups. 2012. The America. Clin Infect Dis 58:1–10.
Lower Anogenital Squamous Terminology Standardization Project for 103. Musa J, Achenbach C, Taiwo B, Berzins B, Silas O, Daru PH, Agbaji
HPV-Associated Lesions: background and consensus recommendations O, Imade G, Sagay AS, Idoko JA, Kanki PJ, Murphy RL. 2014. High-risk

16 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Sun, 27 Nov 2016 18:41:47
Human Papillomavirus

human papilloma virus and cervical abnormalities in HIV-infected women 118. Depuydt CE, Criel AM, Benoy IH, Arbyn M, Vereecken AJ, Bogers
with normal cervical cytology. Infect Agent Cancer 9:36. JJ. 2012. Changes in type-specific human papillomavirus load predict
104. Keller MJ, Burk RD, Xie X, Anastos K, Massad LS, Minkoff H, progression to cervical cancer. J Cell Mol Med 16:3096–3104.
Xue X, D’Souza G, Watts DH, Levine AM, Castle PE, Colie C, Palefsky 119. Massad LS, Einstein MH, Huh WK, Katki HA, Kinney WK,
JM, Strickler HD. 2012. Risk of cervical precancer and cancer among Schiffman M, Solomon D, Wentzensen N, Lawson HW, 2012 ASCCP
HIV-infected women with normal cervical cytology and no evidence of Consensus Guidelines Conference. 2013. 2012 updated consensus guide-
oncogenic HPV infection. JAMA 308:362–369. lines for the management of abnormal cervical cancer screening tests and
105. Clifford GM, Gonçalves MA, Franceschi S, HPV and HIV Study cancer precursors. J Low Genit Tract Dis 17(Suppl 1):S1–S27.
Group. 2006. Human papillomavirus types among women infected with 120. Panel on Opportunistic Infections in HIV-Infected Adults and
HIV: a meta-analysis. AIDS 20:2337–2344. Adolescents. Guidelines for the prevention and treatment of opportunistic
106. de Sanjose S, et al, Retrospective International Survey and HPV Time infections in HIV-infected adults and adolescents: recommendations from
Trends Study Group. 2010. Human papillomavirus genotype attribution the Centers for Disease Control and Prevention, the National Institutes
in invasive cervical cancer: a retrospective cross-sectional worldwide of Health, and the HIV Medicine Association of the Infectious Diseases
study. Lancet Oncol 11:1048–1056. Society of America. Human Papillomavirus Disease section last updated
May 7, 2013. Available at http://aidsinfo.nih.gov/contentfiles/lvguidelines
107. Dames DN, Blackman E, Butler R, Taioli E, Eckstein S, Devarajan K,
Griffith-Bowe A, Gomez P, Ragin C, African Caribbean Cancer Consor- /adult_oi.pdf. Accessed on 2/2/2015.
tium. 2014. High-risk cervical human papillomavirus infections among 121. Cuschieri K, Bhatia R, Cruickshank M, Hillemanns P, Arbyn M.
human immunodeficiency virus-positive women in the Bahamas. PLoS 2016. HPV testing in the context of post-treatment follow up (test of cure).
One 9:e85429. J Clin Virol 76(Suppl 1):S56–S61.
108. Burd EM. 2003. Human papillomavirus and cervical cancer. Clin 122. Markowitz LE, Dunne EF, Saraiya M, Chesson HW, Curtis CR, Gee
Microbiol Rev 16:1–17. J, Bocchini JA Jr, Unger ER, Centers for Disease Control and Prevention
109. Miller FS, Nagel LE, Kenny-Moynihan MB. 2007. Implementation (CDC). 2014. Human papillomavirus vaccination: recommendations of
the Advisory Committee on Immunization Practices (ACIP). MMWR
of the ThinPrep imaging system in a high-volume metropolitan laboratory.
Diagn Cytopathol 35:213–217. Recomm Rep 63(RR-05):1–30.
110. Solomon D, Davey D, Kurman R, Moriarty A, O’Connor D, Prey M, 123. Dillner J, Arbyn M, Unger E, Dillner L. 2011. Monitoring of human
papillomavirus vaccination. Clin Exp Immunol 163:17–25.
Raab S, Sherman M, Wilbur D, Wright T Jr, Young N, Forum Group
Members, Bethesda 2001 Workshop. 2002. The 2001 Bethesda System: 124. Meyer SI, Fuglsang K, Blaakaer J. 2014. Cell-mediated immune
terminology for reporting results of cervical cytology. JAMA 287:2114– response: a clinical review of the therapeutic potential of human papillo-
2119. mavirus vaccination. Acta Obstet Gynecol Scand 93:1209–1218.
111. Ulker V, Numanoglu C, Akyol A, Kuru O, Akbayir O, Erim A, 125. Joura EA, Garland SM, Paavonen J, Ferris DG, Perez G, Ault KA,
Ongut C. 2013. Analyses of atypical glandular cells re-defined by the 2006 Huh WK, Sings HL, James MK, Haupt RM, FUTURE I and II Study
Bethesda System: histologic outcomes and clinical implication of follow- Group. 2012. Effect of the human papillomavirus (HPV) quadrivalent
up management. Eur J Gynaecol Oncol 34:457–461. vaccine in a subgroup of women with cervical and vulvar disease: retro-
spective pooled analysis of trial data. BMJ 344:e1401.
112. Gay JD, Donaldson LD, Goellner JR. 1985. False-negative results in
cervical cytologic studies. Acta Cytol 29:1043–1046. 126. Kumar D, Unger ER, Panicker G, Medvedev P, Wilson L, Humar A.
113. Clinical Laboratory Improvement Amendments of 1988. 1988. P.L. 2013. Immunogenicity of quadrivalent human papillomavirus vaccine in
organ transplant recipients. Am J Transplant 13:2411–2417.
100–578. Congr Rec 134:3828–3863.
127. Gustafson LW, Gade M, Blaakær J. 2014. Vulval cancer and HPV
114. Moscicki AB, Cox JT. 2010. Practice improvement in cervical
screening and management (PICSM): symposium on management of vaccination in recurrent disease. Clin Case Rep 2:243–246.
cervical abnormalities in adolescents and young women. J Low Genit 128. Mészner Z, Jankovics I, Nagy A, Gerlinger I, Katona G. 2015. Re-
Tract Dis 14:73–80. current laryngeal papillomatosis with oesophageal involvement in a 2 year
115. Dockter J, Schroder A, Eaton B, Wang A, Sikhamsay N, Morales L, old boy: successful treatment with the quadrivalent human papillomatosis
Giachetti C. 2009. Analytical characterization of the APTIMA HPV assay. vaccine. Int J Pediatr Otorhinolaryngol 79:262–266.
J Clin Virol 45(Suppl 1):S39–S47. 129. Coskuner ER, Ozkan TA, Karakose A, Dillioglugil O, Cevik I.
116. Sauter JL, Mount SL, St John TL, Wojewoda CM, Bryant RJ, Leiman 2014. Impact of the quadrivalent HPV vaccine on disease recurrence in
G. 2014. Testing of integrated human papillomavirus mRNA decreases men exposed to HPV infection: a randomized study. J Sex Med 11:2785–
colposcopy referrals: could a change in human papillomavirus detection 2791.
methodology lead to more cost-effective patient care? Acta Cytol 58:162– 130. Lin K, Roosinovich E, Ma B, Hung C-F, Wu T-C. 2010. Therapeutic
166. HPV DNA vaccines. Immunol Res 47:86–112.
117. Depuydt CE, Benoy IH, Beert JFA, Criel AM, Bogers JJ, Arbyn M. 131. Peng S, Song L, Knoff J, Wang JW, Chang YN, Hannaman D, Wu
2012. Clinical validation of a type-specific real-time quantitative human TC, Alvarez RD, Roden RB, Hung CF. 2014. Control of HPV-associated
papillomavirus PCR against the performance of hybrid capture 2 for the tumors by innovative therapeutic HPV DNA vaccine in the absence of
purpose of cervical cancer screening. J Clin Microbiol 50:4073–4077. CD4+ T cells. Cell Biosci 4:11.

ASMscience.org/MicrobiolSpectrum 17
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Sun, 27 Nov 2016 18:41:47

Das könnte Ihnen auch gefallen