Downloaded from https://journals.lww.com/dcrjournal by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWnYQp/IlQrHD3IJrtBKuSsQVRz8fA4yAY0a8W1YLRn6mHykFpaZ5LFvI= on 06/18/2018
Downloaded from https://journals.lww.com/dcrjournal by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWnYQp/IlQrHD3IJrtBKuSsQVRz8fA4yAY0a8W1YLRn6mHykFpaZ5LFvI= on 06/18/2018
Copyright © The American Society of Colon & Rectal Surgeons, Inc. Unauthorized reproduction of this article is prohibited.
755
DISEASES OF THE COLON & RECTUM VOLUME 61: 7 (2018)
T
he American Society of Colon and Rectal Surgeons
is dedicated to ensuring high-quality patient care
by advancing the science, prevention, and manage-
ment of disorders and diseases of the colon, rectum, and
anus. The Clinical Practice Guidelines Committee is com-
posed of society members who are chosen because they
have demonstrated expertise in the specialty of colon and
rectal surgery. This committee was created to lead interna-
tional efforts in defining quality care for conditions related
to the colon, rectum, and anus. This is accompanied by
developing clinical practice guidelines based on the best
available evidence. These guidelines are inclusive and not
prescriptive. The purpose of this guideline is to provide
information on which decisions can be made rather than
to dictate a specific form of treatment. These guidelines
are intended for use by all practitioners and healthcare
workers, as well as by patients who desire information
about the management of the conditions addressed by the
topics covered in these guidelines. It should be recognized
that these guidelines should not be deemed inclusive of all
proper methods of care or exclusive of methods of care
reasonably directed toward obtaining the same results.
The ultimate judgment regarding the appropriateness of
any specific management decision must be made by the
treating physician in light of all of the circumstances pres-
ent in the care of the patient in question.
STATEMENT OF THE PROBLEM
Squamous cell cancers of the anal canal and perianal re-
gion remain one of the least common malignancies aris-
ing from the alimentary tract. As of 2016, it is estimated
that 8200 new cases of squamous cell cancers of the anus
were diagnosed in the United States, with 1.7 times as
many women as men affected.
1
Within this same time pe-
riod, 1100 patients were estimated to have died of anal
cancer, with cancer deaths among women being 1.4 times
the number observed among men. Although squamous
cancers of the anus remain relatively rare GI malignan-
cies, 2 factors have nonetheless focused greater attention
toward this disease. The first is the observation that the
frequency of squamous cancers of the anus has increased
in the United States from the 1970s through the 2000s,
2
with a notable increase in incidence among men, and, in
particular, black men.
3
In addition, given the inverse re-
lationship between stage of disease and survival,
4
studies
using population-level data suggest that earlier detection
may improve survival from anal cancer, which makes anal
cancer an important and treatable public health concern.
Supplemental digital content is available for this article. Links to the
digital files are provided in the HTML and PDF versions of this article
on the journal’s Web site (www.dcrjournal.com).
Earn Continuing Education (CME) credit online at cme.lww.com.
Links to search strategies: See search strategies for anal dysplasia, HPV
vaccination, chemoradiation for squamous cell cancer of the anus, sal-
vage APR, and metastatic disease in supplemental digital content at
http://links.lww.com/DCR/A639.
Funding/Support: None reported.
Financial Disclosure: None reported.
Correspondence: Scott R. Steele, M.D., Department of Colorectal
Surgery, Cleveland Clinic, 9500 Euclid Ave, A30, Cleveland, OH 44915.
The American Society of Colon and Rectal Surgeons
Clinical Practice Guidelines for Anal Squamous Cell
Cancers (Revised 2018)
David B. Stewart, M.D.
1
Wolfgang B. Gaertner, M.D., M.Sc.
2
• Sean C. Glasgow, M.D.
3
Daniel O. Herzig, M.D.
4
• Daniel Feingold, M.D.
5
• Scott R. Steele, M.D.
5
Prepared on Behalf of the Clinical Practice Guidelines Committee of the American Society of Colon and Rectal Surgeons
1 Department of Surgery, University of Arizona, Tucson, Arizona
2 Division of Colorectal Surgery, University of Minnesota, Minneapolis, Minnesota
3 Division of Colorectal Surgery, Washington University School of Medicine, St. Louis, Missouri
4 Division of Colorectal Surgery, Columbia University, New York, New York
5 Division of Colorectal Surgery, Cleveland Clinic, Cleveland, Ohio
Dis Colon Rectum 2018; 61: 755–774
DOI: 10.1097/DCR.0000000000001114
© The ASCRS 2018
CLINICAL PRACTICE GUIDELINES
Copyright © The American Society of Colon & Rectal Surgeons, Inc. Unauthorized reproduction of this article is prohibited.
STEWART ET AL: ANAL CANCER CLINICAL PRACTICE GUIDELINES
756
The second factor that has resulted in a paradigm shift in
understanding the etiology of anal cancer is the discovery
that the human papilloma virus (HPV), especially HPV
serotypes 16 and 18,
5
is the primary cause of squamous
cancers of the anus,
6,7
making anal cancer a sequela of a
sexually transmitted disease. This aspect of anal carcino-
genesis reinforces the concept that it is a potentially pre-
ventable disease and that if evidence-based screening and
preventative measures were developed and consistently
applied, decreases in cancer-related deaths would follow.
The modifiable risk of death from anal cancer is further
underscored by large studies that have documented that
50% of patients with anal cancer present with localized,
node-negative disease, which is associated with high cure
rates; one third of patients will present with node-positive
disease, whereas only 10% to 15% will present with dis-
tant metastases.
8
Thus, even without effective preventative
measures, the majority of patients with anal cancer are po-
tentially curable at the time of diagnosis and treatment.
The multiple risk factors associated with developing
squamous cancers of the anus are well documented and
can be grouped into the 2 broad categories of HPV and
immunosuppression, although there is also an association
between these categories as well. Among the HPV-related
risk factors include lifetime number of sexual partners,
9
a history of previous sexually transmitted diseases of any
kind,
10
a history of anogenital warts,
11
anoreceptive in-
tercourse,
12
and a history of cervical, vaginal, or vulvar
cancer.
13
Risk factors related to immunity include a di-
agnosis of HIV,
14
autoimmune disorders such as lupus
and sarcoidosis,
15
and being the recipient of a solid organ
transplant.
16
Female sex
3
and cigarette smoking
17
are also
associated with developing anal malignancies.
ANATOMIC CONSIDERATIONS AND TERMINOLOGY
The management of anal cancers requires a multidisci-
plinary approach, and the unfamiliarity of nonsurgical
disciplines with anorectal anatomy can create ambiguity
in describing the location and the clinical stage of anal
cancers across disciplines. The anal canal, as viewed by
colorectal surgeons, is 4 to 5 cm in length beginning at
the distal rectum, where the mucosa blends into the anal
transitional zone (ATZ) epithelium, which then transi-
tions to nonkeratinized squamous epithelium as it further
transitions into keratinized perianal skin at the anal verge.
An ATZ, located several millimeters proximal to the den-
tate line and extending for 0.5 to 1.5 cm in length, repre-
sents a region of naturally occurring intestinal metaplasia,
representing a transition from the columnar epithelium of
the distal rectum to the modified squamous epithelium of
the anal canal, referred to as anoderm. Because of the pres-
ence of metaplasia in the ATZ, this region is particularly
susceptible to HPV infection.
18
In addition, the variety of
tissue types in the ATZ have been associated with a num-
ber of subtypes of squamous cancers of the anus of both
keratinizing and nonkeratinizing histologies. Although
there were previous efforts to distinguish between histo-
logical subtypes of anal cancer, all of these subtypes are
now grouped together, because multiple histological vari-
ants can exist within the same malignancy,
19
and because
the natural history and survival of these subtypes are simi-
lar when stratified by treatment and cancer stage.
20
Because the anatomic landmarks of the anus will not
be easily identifiable by nonsurgical providers who are also
untrained in techniques such as anoscopy and proctosco-
py, a simplified taxonomy
21
has been suggested. An anal
canal cancer would be any lesion that cannot be complete-
ly visualized with distraction of the gluteal cheeks, whereas
a perianal (which replaces the term anal margin) lesion
can be completely visualized with distraction of the gluteal
cheeks, and that is still within 5 cm of the anal orifice. Any
lesion >5 cm from the anal orifice would be classified as a
skin lesion and would not be considered related to the GI
tract.
Confusion often arises over the various pathology
terms commonly used to describe lesions involving the
anus and perianal skin. The Lower Anogenital Squa-
mous Terminology project unified terminology for all
HPV-related squamous precursor lesions with a 2-tiered
nomenclature system.
22,23
This system simply designates
noninvasive pathology as either low-grade or high-grade
squamous intraepithelial lesions (LSILs and HSILs) based
on histological findings such as mitotic activity, depth of
dermal involvement, and abnormalities in squamous cell
differentiation. LSILs include anal intraepithelial neopla-
sia (AIN) 1, whereas HSILs encompass AIN-2 and AIN-
3 designations. The distinction between condylomas and
LSILs is somewhat arbitrary; condylomas generally appear
as bland exophytic, papillary proliferations with viral cy-
topathic changes, whereas LSILs tend to be flat lesions.
23
Older terms such as Bowens disease should no longer be
used. Throughout this Clinical Practice Guideline, the
terms LSIL and HSIL will be used, although reference to
AIN may appear when directly quoting published research
findings.
This guideline only discusses the management of
premalignant and malignant squamous neoplasms of the
anus and perianal region, excluding other, rarer malignan-
cies. The abbreviation SCC will be used to refer to squa-
mous cell cancers.
METHODS
These guidelines were built on the most recent Ameri-
can Society of Colon and Rectal Surgeons Practice Pa-
rameters for Anal Squamous Neoplasms, published in
2012.
24
An organized search of MEDLINE, PubMed,
Copyright © The American Society of Colon & Rectal Surgeons, Inc. Unauthorized reproduction of this article is prohibited.
DISEASES OF THE COLON & RECTUM VOLUME 61: 7 (2018)
757
Embase, and the Cochrane Database of Collected Re-
views was queried June 2015 through January 2018,
searching for relevant publications with no limitations
regarding date of publication. Retrieved publications
were limited to the English language, but no limits on
year of publication were applied. The search strate-
gies were based on the key words anal cancer and anal
squamous cancer as primary search terms, with addi-
tional, key-word searches including AIN, anal intraep-
ithelial neoplasia, Nigro protocol, anal HPV, LSIL, and
HSIL. Searches were also performed based on various
treatments for anal cancers, including “anal cancer
AND radiation, “anal cancer AND chemoradiother-
apy, anal cancer AND surgery, anal cancer AND
abdominoperineal resection, anal cancer AND anal
dysplasia, and “anal cancer and lymphadenectomy.
Directed searches of the embedded references from
the primary articles were also performed in certain
circumstances. Prospective randomized controlled tri-
als (RCTs) and meta-analyses were given preference in
developing these guidelines. The final grade of recom-
mendation was performed using the Grades of Recom-
mendation, Assessment, Development, and Evaluation
system (Table 1).
25
Premalignant Neoplasms of the Anal Canal and
Perianal Region
Patients at increased risk for anal squamous neoplasms
should be identified by history, physical examination,
and laboratory testing, noting that the risk is higher in
HIV-positive individuals, men who have sex with men
(MSM), and women with a history of cervical dysplasia.
Grade of Recommendation: Strong recommendation
based on moderate-quality evidence, 1B.
Multiple large (300- to 1200-patient) cohort studies have
identified risk factors for anal dysplasia and cancer.
26,27
A systematic review and meta-analysis noted that the
pooled prevalence of HPV-16 in HIV-positive MSM was
35.4%.
28
The prevalence of AIN was 29.1%, and the inci-
dence of anal cancer was 45.9 per 100,000 men. For HIV-
negative MSM, the prevalence of HPV-16 was 11.8%, the
prevalence of AIN was 21.5%, and the incidence of anal
cancer was 5.1 per 100,000 men. A cohort of 171 HIV-
positive women noted that 12.9% had HSILs.
29
Cervi-
cal dysplasia can also guide risk assessment in women; a
population-based study of 89,018 women with cervical
HSILs matched with control subjects without cervical
dysplasia demonstrated an increased rate of anal cancer
TABLE 1. Grade scoring system
Description Benet vs risk and burdens
Methodologic quality of
supporting evidence Implications
1A Strong recommendation,
High-quality evidence
Benets clearly outweigh risk
and burdens or vice versa
RCTs without important
limitations or overwhelming
evidence from observational
studies
Strong recommendation, can
apply to most patients in
most circumstances without
reservation
1B Strong recommendation,
Moderate-quality
evidence
Benets clearly outweigh risk
and burdens or vice versa
RCTs with important limitations
(inconsistent results,
methodologic aws, indirect
or imprecise) or exceptionally
strong evidence from
observational studies
Strong recommendation, can
apply to most patients in
most circumstances without
reservation
1C Strong recommendation,
Low- or very-low–quality
evidence
Benets clearly outweigh risk
and burdens or vice versa
Observational studies or case
series
Strong recommendation but
may change when higher-
quality evidence becomes
available
2A Weak recommendation,
High-quality evidence
Benets closely balanced with
risks and burdens
RCTs without important
limitations or overwhelming
evidence from observational
studies
Weak recommendation, best
action may dier depending
on circumstances or patient
or societal values
2B Weak recommendations,
Moderate-quality
evidence
Benets closely balanced with
risks and burdens
RCTs with important limitations
(inconsistent results,
methodologic aws, indirect
or imprecise) or exceptionally
strong evidence from
observational studies
Weak recommendation, best
action may dier depending
on circumstances or patient
or societal values
2C Weak recommendation,
Low- or very-low–quality
evidence
Uncertainty in the estimates of
benets, risks, and burdens;
benets, risks, and burdens
may be closely balanced
Observational studies or case
series
Very weak recommendations;
other alternatives may be
equally reasonable
Adapted with permission from Chest 2006;129:174–181.
25
RCT = randomized controlled trial.
Copyright © The American Society of Colon & Rectal Surgeons, Inc. Unauthorized reproduction of this article is prohibited.
STEWART ET AL: ANAL CANCER CLINICAL PRACTICE GUIDELINES
758
(relative risk = 6.68 (95% CI, 3.64–12.25)) and HSILs
(relative risk = 4.97 (95% CI, 3.26–7.57)).
30
Standardized nomenclature with a 2-tiered system
should be used. Biomarkers, including p16, should be
used selectively to clarify equivocal high-grade lesions.
Grade of Recommendation: Strong recommendation
based on low- or very-lowquality evidence, 1C.
There are advantages in standardizing terminology in de-
fining the disease and treatments. The 2-tiered system
22,23
defined in 2012 provides the most appropriate system for
standardizing definitions. Two retrospective studies have
shown that there can be inconsistent interrater reliability
when examining histology specimens.
31
Evidence support-
ing this 2-tiered system is based in part on 1 prospective
blinded study that reported that interrater reliability for
cytology using a 2-tiered system was 85% and that p16
staining and HPV oncogene messenger RNA analysis im-
proved the ability to come to a diagnostic consensus.
32
Individuals with anal dysplasia should be followed
at regular intervals with a history, physical examina-
tion, and a discussion of screening options. Grade of
Recommendation: Weak recommendation based on
moderate-quality evidence, 2B.
Prevention of cervical cancer with screening for precan-
cerous lesions has been proven effective.
31
Whether a simi-
lar program of screening and destruction of precancerous
anal lesions will lead to reductions in anal cancer is a mat-
ter of considerable debate. A 2012 systematic review and
meta-analysis suggested that, as part of their natural his-
tory, rates of progression to anal cancer are substantially
lower than those observed for cervical cancer.
28
Regardless
of which, if any, intensive screening program is selected,
individuals with anal dysplasia should have a periodic of-
fice visit to assess for any new or modifiable risk factors,
including a digital anorectal examination. Even in an in-
tensive screening program, nearly all the cancers that de-
veloped were detectable on digital examination.
33
It is not clear that screening will prevent a cancer from
occurring, but there is evidence that cancers detected dur-
ing a screening program are identified at an early stage.
33
Multiple cohort studies have shown progression from low-
grade to high-grade dysplasia and from dysplasia to cancer
even under regimented surveillance. A cohort of 91 HIV-
positive patients treated for anal dysplasia followed for >1
year showed that 75.8% had recurrent dysplasia, 46.0%
progressed to high-grade dysplasia, and 2.3% developed
anal cancer.
34
Studies of 4 cohorts of patients in intensive
treatment programs have separately estimated the devel-
opment of cancer despite treatment, although all recorded
the risk differently. One estimated the rate of progression
to cancer at 6.9 cases per 100 person-years of follow up,
35
another estimated the Kaplan–Meier probability of SCC
at 3 years at 1.97%,
36
a third estimated the duration to de-
velopment of SCC at 57 to 62 months,
33
and a fourth esti-
mated the 5-year cumulative incidence of SCC at 1.70%.
37
There is consensus among experts that an HSIL is the
precursor to invasive cancer.
33,38
What remains unclear
is whether screening to identify and ablate premalignant
lesions will decrease the incidence of SCC. Nevertheless,
even if the progression to cancer cannot be halted, early
diagnosis of a cancer justifies follow-up, and a history and
examination to identify and treat visible and palpable le-
sions and/or a discussion of screening options is justified.
Screening with anal cytology (or anal Papanicolaou (Pap)
tests) may be considered in high-risk populations as part
of a comprehensive screening program, but the sensitivity
and specificity of the test do not support its use for uni-
versal screening. Grade of Recommendation: Weak rec-
ommendations based on moderate-quality evidence, 2B.
Similar to cervical Pap smear cytology, a swab or brush
sample from the anal canal to include the ATZ can be
evaluated for cytological evidence of dysplasia. Although
the data demonstrating effectiveness for cervical cancer
screening and prevention are well founded, substantial
efforts to prove its role in anal cancer screening have not
been conclusive. Because screening tests perform better as
the prevalence of a condition is higher, screening a higher-
risk population will be more effective. Most studies screen
high-risk individuals, including MSM, HIV-positive per-
sons, and/or women with a history of cervical dysplasia.
Results from studies that have performed both anal cytol-
ogy and histologic evaluation are shown in Table 2.
12,29,39–52
The sensitivity and specificity are limited, because the gold
standard is the finding of HSILs on biopsies. Currently
the ability of anal cytology to identify patients at risk for
dysplasia is inconclusive, and an association between anal
cytology and reduced rates of anal cancer has not been dem-
onstrated. The decision to perform anal Pap tests should be
a shared decision with the patient, including a discussion
about how abnormal tests will be further evaluated.
HPV testing may be used as an adjunct to screening for
anal cancer. Grade of Recommendation: Weak recom-
mendations based on moderate-quality evidence, 2B.
The presence of HPV, especially subtypes 16 and 18, is
associated with the majority of anal cancers. Biomarkers
screen for the presence of high-risk HPV to estimate the
risk of dysplasia. The main limitation to this strategy is
the high prevalence of HPV in the high-risk population.
Currently available options include HPV DNA testing,
HPV DNA genotyping for HPV-16 and HPV-18, HPV-E6/
E7 mRNA testing, and p16/Ki-67 immunostaining based
on either anal cytology or biopsy. P16 is a tumor suppres-
sor gene product that indicates HPV integration into the
host genome. The Lower Anogenital Squamous Terminol-
ogy guidelines recommend the use of p16 in borderline
HSIL/LSIL cases, with strong positive staining leading to
Copyright © The American Society of Colon & Rectal Surgeons, Inc. Unauthorized reproduction of this article is prohibited.
DISEASES OF THE COLON & RECTUM VOLUME 61: 7 (2018)
759
Table 2. Studies of anal cytology and histologic evaluation for squamous intraepithelial lesions
Study Location
No./type of
patient
Anal cytology
ndings
Anal biopsy
ndings
Sensitivity/specicity for
cytology detecting HSIL
Jin et al,
39
2017 Sydney, NSW, Australia 617 MSM Negative 41.0%
ASC-US 17.5
LSIL 8.0%
ASC-H 15.0%
HSIL 18.5%
Negative 29.6%
LSIL 31.0%
HSIL 39.0%
SCC 0.2%
Sensitivity 83.2%
Specicity 52.4%
PPV 45.8%
NPV 86.6%
Schoeld et al,
12
2016
Manchester,
United Kingdom
284 MSM Negative 56%
Inadequate 2.1%
LSIL 31.4%
HSIL 10.5%
Normal 4.5%
AIN-1 54.9%
AIN-2 31.7%
AIN-3 7.6%
SCC 1.3%
Sensitivity 76.5%
Specicity 54.6%
PPV 45.8%
NPV 86.6%
Heard et al,
29
2015 Multicenter 171 HIV-positive women Negative 70.7%
ASC-US or LSIL 18.7%
ASC-H or HSIL 10.0%
SCC 0.6%
Normal 59.2%
Benign 20.7%
LG-AIN 10.7%
HG-AIN 5.9%
SCC 0.6%
Cheng et al,
40
2015 Taiwan 196 HIV-positive men Negative 63.8%
ASC-US 16.8%
LSIL 14.8%
HSIL 4.6%
HG-AIN 7.1% Sensitivity 64.0%
Specicity 66.0%
PPV 12.7%
NPV 96.0%
Sendagorta
et al,
41
2014
Spain 298 HIV-positive MSM Negative 60.2%
ASC-US 5.7%
LSIL 24.1%
HSIL 10%
Normal 19.3%
Benign 10.9%
AIN-1 14.3%
AIN-2 35.3%
AIN-3 20.2%
Incomplete data
Botes et al,
42
2013 Australia 262 HIV-positive MSM Negative 23.0%
ASC-US 15.8%
ASC-H 9.6%
LSIL 39.9%
HSIL 4.5
No biopsy 11.9%
Benign 19.7%
LG-AIN 13.9%
HG-AIN 54.5%
Incomplete data
Wentzensen
et al,
43
2012
San Francisco, California 363 HIV-positive patients Negative 30.8%
ASC-US 20.1%%
ASC-H 7.4%
LSIL 18.5%
HSIL 16.5%
No biopsy 19.3%
Benign 23.4%
AIN-1 34.7%
AIN-2 15.2%
AIN-3 6.9%
Williams et al,
44
2010 Australia 154 patients Negative 5.4%
ASC-US 18.7%
ASC-H 1.2%
LSIL 47.3%
HSIL 27.4%
Benign 11.6%
LSIL 50.0%
HSIL 38.6
Sensitivity 96.0%
Specicity 14.0%
PPV 89.0%
NPV 31.0%
Salit et al,
45
2010 Toronto, Ontario,
Canada
401 HIV-positive MSM Negative 33.0%
ASC-US 12.0%
LSIL 43.0%
HSIL 12.0%
Normal 59.2%
Benign 32.0%
LG-AIN 43.0%
HG-AIN 25.0%
Sensitivity 84.0%
Specicity 39.0%
NPV 88.0%
PPV 31.0%
Nathan et al,
46
2010 London,
United Kingdom
395 patients Negative 32.6%
ASC-US 26.7%
LSIL 32.6%
HSIL 8.0%
Normal 24.2%
LG-AIN 50.2%
HG-AIN 25.6%
Sensitivity 81.0%
Specicity 37.0%
NPV 85.0%
PPV 30.0%
Fox et al,
47
2005 London,
United Kingdom
99 MSM Negative 26.0%
LSIL 59.0%
HSIL 15.0%
Normal 16.0%
AIN-1 18.0%
AIN-2 37.0%
AIN-3 26.0%
Sensitivity 83.0%
Specicity 38.0%
NPV 86.0%
PPV 33.0%
Arain et al,
48
2005 Los Angeles, California 198 patients Negative 31.7%
ASC-US or LSIL 55.2%
ASC-H or HSIL 13.1%
Normal 18.3%
AIN-1 21.1%
AIN-2 35.2%
AIN-3 25.4%
Sensitivity 98.0%
Specicity 50.0%
(Continued)
Copyright © The American Society of Colon & Rectal Surgeons, Inc. Unauthorized reproduction of this article is prohibited.
STEWART ET AL: ANAL CANCER CLINICAL PRACTICE GUIDELINES
760
an HSIL diagnosis and weak or absent staining supporting
an LSIL diagnosis. The use of p16 staining, or any of the
biomarkers, in a screening setting is less clear.
Two prospective studies have compared anal cytology
with HPV testing, as have multiple retrospective cohorts.
The ANALOGY trial prospectively evaluated screen-
ing with cytology, high-risk HPV typing, and high-reso-
lution anoscopy (HRA). With HPV testing only 59% of
HSILs would have been detected.
12
The Australian study
used high-risk HPV viral load and high-risk HPV-E6/E7
mRNA, as well as cytology and HRA.
39
Compared with
cytology for the detection of HSIL, HPV testing showed
similar sensitivity and improved specificity for the detec-
tion of HSILs, especially in the HIV-negative group.
HRA may be considered as a screening option for pa-
tients at high risk for cancer when performed by cli-
nicians with appropriate training in the procedure.
Recommendation: Weak recommendation based on
moderate-quality evidence, 2B.
HRA is a procedure performed in the office or in the op-
erating room using magnification and topical agents such
as acetic acid and Lugol’s solution to identify, biopsy, and
ablate lesions not visible by conventional examination or
anoscopy. The procedure can be more cost-effective if per-
formed in the office. In addition, because it is superior to
cytology or HPV testing in identifying HSILs, HRA may
be more cost-effective than other strategies; the cost per
HSIL found has been estimated to be $809.39. A prospec-
tive screening study of high-risk MSM evaluated all 3
modalities of HPV testing, cytology, and HRA.
12
In a co-
hort of 284 MSM, AIN-3 was detected in 17 patients, with
screening HRA finding 16, HPV-16/18 testing finding 10,
any HPV testing finding 16, high-grade cytology detecting
3, and any abnormal cytology detecting 12 subjects. How-
ever, only 15% of the cohort tested negative for HPV, rep-
resenting a methodologic weakness in this study. Cytology
missed nearly one third of high-risk lesions, suggesting
that HRA would have the most clinical use for screening.
The effectiveness of HRA to prevent the progression
of dysplasia or development of cancer has been evaluated
in retrospective cohort studies. A retrospective review of
246 patients treated with HRA-targeted destruction of
HSIL/LSIL over a 10-year period was published in 2008.
53
A recurrent HSIL was seen in 57% of patients at an aver-
age of 19 months. Despite treatment, 1.2% progressed to
invasive cancer.
A cohort of 727 MSM followed for a median of 2.2 years
was published in 2014. With ablation of all HRA-identified
lesions and with regular follow-up, the rate of recurrence at
1 year was 53% in HIV-positive patients and 49% in HIV-
negative patients. Over the follow-up period, 5 patients devel-
oped cancer, with the probability of cancer 1.97% at 3 years.
36
There are few comparisons of HRA with other treatment
strategies. A retrospective review of 424 patients compared
HRA with expectant management in 2 cohorts, one treated
by 3 clinicians who followed patients with expectant man-
agement and the other treated by 2 clinicians who followed
patients with HRA.
54
Anal cancer occurred in 1 of the HRA
patients and 2 of the expectant management patients. The
5-year progression rate was similar in the 2 cohorts. Selection
bias and the possibility for type II error limit these findings.
Papaconstantinou
et al,
49
2005
Dallas, Texas 47 patients Negative 23.4%
ASC-US or LSIL 57.4%
ASC-H or HSIL 14.9%
SCC 4.3%
Normal 42.6%
AIN-1 17.0%
AIN-2 12.8%
AIN-3 21.3%
SCC 6.4%
Sensitivity 42.0%
Specicity 96.0%
Mathews et al,
50
2004 San Diego, California 1732 patients Negative 43.2%
ASC-US 34.1
LSIL 15.3%
HSIL 7.5%
Incomplete data Sensitivity 85.0%
Specicity 56.0%
Panther et al,
51
2004 Boston, Massachusetts 153 MSM Negative 12.4%
ASC-US 19.6%
LSIL 47.1%
HSIL 20.9%
Normal 21.6%
AIN-1 37.3%
AIN-2 14.4%
AIN-3 25.5%
SCC 1.3%
Sensitivity 47.0%
Specicity 90.0%
Palefsky et al,
52
1997 San Francisco, California 407 patients Negative 72.4%
ASC-US 15.1%
LSIL 12.1%
HSIL 0.4%
Normal 11.9%
LG-AIN 79.3%
HG-AIN 8.9%
Sensitivity 69.0%
Specicity 59.0%
MSM = men who have sex with men; ASC-US = atypical squamous cells of undetermined signicance; ASC-H = atypical squamous cells, cannot exclude high-grade squa-
mous intraepithelial lesion; LSIL = low-grade squamous intraepithelial lesion; HSIL = high-grade squamous intraepithelial lesion; SCC = squamous cell cancers; AIN = anal
intraepithelial neoplasia; LG = low-grade; HG = high-grade; PPV = positive predictive value; NPV = negative predictive value.
Table 2. Continued
Study Location
No./type of
patient
Anal cytology
ndings
Anal biopsy
ndings
Sensitivity/specicity for
cytology detecting HSIL
Copyright © The American Society of Colon & Rectal Surgeons, Inc. Unauthorized reproduction of this article is prohibited.
DISEASES OF THE COLON & RECTUM VOLUME 61: 7 (2018)
761
Performing HRA requires specialized training and on-
going practice to perform good-quality examinations.
55,56
In addition, patient compliance, patient tolerance, and the
risk of over treatment have led some clinicians to wait to
start performing HRA until additional data are available.
Although HRA effectively identifies HSIL, whether HRA
with ablation of HSIL can reduce the incidence of cancer
or whether it can do so more consistently than conven-
tional anoscopy remains unclear.
Topical imiquimod, fluorouracil, trichloroacetic acid
and cidofovir with close long-term follow-up are each
options for the treatment of LSIL or HSIL. Grade of
Recommendation: Weak recommendation based on
moderate-quality evidence, 2B.
Two randomized trials and 1 prospective cohort study have
evaluated imiquimod. A placebo-controlled RCT of 64 HIV-
positive patients with HSILs who were followed with HRA
suggested superior clearance of HSILs with imiquimod
(42.9% vs 4.0%).
57
With a median follow-up of 33 months,
61% showed a sustained absence of HSILs. An RCT compar-
ing imiquimod with topical fluorouracil and electrocautery
found that 24% of HSILs had resolved and 11% had a partial
response with imiquimod.
58
Although electrocautery had a
statistically significant improvement in response when the
p value was calculated for the 3 treatment options, the dif-
ference was not significant if imiquimod and electrocautery
were compared head to head. A third prospective cohort
study evaluated imiquimod 5 days per week and found that
the overall response rate for HSILs was 66%.
59
Two retrospective studies have evaluated topical fluoro-
uracil. A retrospective review of 46 patients with HSILs or
LSILs showed that 57% responded to topical fluorouracil,
with 39% having a complete response.
60
A second review
of 11 patients with dysplasia (although only 5 had HSILs)
showed a decrease in dysplasia in 6 (55%) of 11 patients.
61
Two retrospective cohort studies have examined the use
of trichloroacetic acid. A review of 98 HSILs from 72 patients
demonstrated that 28.7% of lesions had resolution or down-
grading to LSILs on follow-up, although recurrence occurred
in 20.8% of the lesions.
62
A review of 54 men showed that,
on a per-lesion basis, 72% of HSILs cleared to LSIL or less.
63
Cidofovir has been evaluated in 1 prospective pilot
study and 1 retrospective cohort study. The pilot study in-
cluded 16 HIV-positive patients with HSILs and revealed
a complete response rate of 10 (62.5%) of 16, although
2 (20.0%) of 10 had recurrent HSILs at the 24-week end
point of the study.
64
A small cohort of 24 patients with
HSILs demonstrated that 51% had responsive disease,
with a complete response observed in 15% of patients.
65
Ablative treatments with conventional anoscopy or
HRA are appropriate therapies for HSILs. Grade of
Recommendation: Weak recommendation based on
moderate-quality evidence, 2B.
If HRA is used as the primary screening modality, ablative
therapies can be used as first-line treatment for dysplasia.
A prospective cohort of 98 patients with HSILs treated
with infrared coagulation (IRC) showed that 74% had no
additional evidence of HSILs on short-term follow-up.
66
There was a comparison to expectant management in that
study, but the control group was derived from patients who
delayed or declined treatment, and patients who missed
follow-up were excluded. In light of these limitations, the
findings in this study are best viewed as a cohort treated
with IRC. Three reviews have been published from a single
center, and it is unclear how much overlap exists.
67–69
Find-
ings from the most recent review, a retrospective review
of 96 MSM with HSILs treated with 1 IRC showed that
82% of HIV-positive and 90% of HIV-negative individu-
als were free from HSILs, but recurrence rates were high
in the study, and nearly one third of patients were lost to
follow-up.
69
A retrospective review of 78 HSILs in 68 HIV-
positive MSM showed 64% efficacy per treated lesion.
70
A
retrospective review of 66 patients with HSILs treated with
IRC reported that only 13% had recurrent HSILs at 1 year
of follow-up.
71
Electrocautery ablation was reviewed in a cohort of
232 MSM with HSIL.
72
Initial clearance rates were high
(75%–80%), but recurrences were common (53%–61%),
and 1 patient in the cohort developed cancer despite treat-
ment. A cohort of 83 patients treated for HSIL with elec-
trocautery found a complete or partial response in 66.2%.
73
With a mean follow-up of 12.1 months, 25.4% of patients
had a high-grade recurrence. A single-center review of
3 ablative techniques, electrocautery, IRC, or laser treat-
ments, showed no differences in the rate of recurrence.
36
One prospective RCT compared imiquimod, topical
fluorouracil, and electrocautery in HIV-positive MSM
with confirmed AIN.
58
Resolution of AIN was achieved
in 24% of patients in the imiquimod group, 17% of the
topical fluorouracil group, and 39% of the electrocautery
group (p = 0.008 for electrocautery vs fluorouracil; p =
0.10 for electrocautery vs imiquimod).
Vaccination against HPV in men and women under
age 26 years for primary prevention is typically recom-
mended. Vaccination of individuals with anal dysplasia
for secondary prevention of dysplasia and cancer is not
recommended. Grade of Recommendation: Weak rec-
ommendation based on high-quality evidence, 2A.
The availability of bivalent, quadrivalent, and now non-
avalent vaccines has created considerable promise that the
next generation of individuals will be largely exempt from
HPV-related neoplasms. Although there are convincing
data for use of the vaccine in pre-exposure young individ-
uals, the off-label use of the vaccine in those with anal dys-
plasia is of considerable interest. A systematic review and
meta-analysis of the efficacy of the vaccine in cervical dys-
plasia showed that it had no effect.
74
A systematic review
Copyright © The American Society of Colon & Rectal Surgeons, Inc. Unauthorized reproduction of this article is prohibited.
STEWART ET AL: ANAL CANCER CLINICAL PRACTICE GUIDELINES
762
including all of the sites reported that 9 of 12 studies per-
formed in patients with active disease showed decreased
disease recurrence, but no study reported improved out-
comes without clinical disease.
75
However, enthusiasm for
the vaccine for secondary prevention or regression of dys-
plasia gained attention after a study of 602 MSM who were
randomly assigned to receive either the quadrivalent HPV
vaccine or placebo. Those who received the vaccine had
a lower rate of intraepithelial neoplasia, and the rate of
HSILs was reduced by 54%.
5
A comparative cohort study
of 202 patients with previous HSILs treated with or with-
out the quadrivalent vaccine demonstrated that vaccine
treatment was associated with decreased risk of recurrent
HSILs.
76
A prospective, randomized, National Institutes of
Health–funded trial including 574 patients has been com-
pleted and presented in abstract form, and the results are
available at clinicaltrials.gov (NCT01461096). The num-
ber of participants with biopsy-proven HSIL occurrences
and recurrences at 1 year or abnormal cytology at 1, 2, or
3 years was the same.
Patients who have been treated for anal dysplasia may
be observed without regular cytology, HPV testing, or
HRA; however, treatment of visible or palpable disease
should be offered. Grade of Recommendation: Weak
recommendation based on low or very low-quality
evidence, 2C.
Although it is generally accepted that HSIL is a precursor to
invasive cancer, there remain no studies that compare more
intensive screening and/or HRA protocols with office exami-
nations, conventional anoscopy, and treatment of visible or
palpable disease. A review of 574 patients in an HRA pro-
gram showed that 24% of patients with HSIL had spontane-
ous regression to LSIL.
77
The progression to cancer is often
multifactorial, including contributions from medication
compliance, immunosuppression, HIV viral load, ongoing
exposure to HPV, smoking, and a variety of other factors.
In the largest prospective randomized US trial, which is
currently enrolling, the control arm of the study includes no
treatment of HRA-identified HSILs. The Topical or Ablative
Treatment in Preventing Anal Cancer in Patients With HIV
and Anal High-Grade Squamous Intraepithelial Lesions
trial (NCT02135419) will screen HIV-positive patients with
HRA and targeted biopsies. Previously untreated individu-
als who have HSILs identified will be randomly assigned to
treatment or monitoring. For patients who are treated, the
clinician decides whether the patient should have topical
treatment or ablative treatment. Patients with topical treat-
ment may receive imiquimod, fluorouracil, or trichloro-
acetic acid. Patients with ablative treatment may have IRC,
electrocautery, or laser ablation. In the study, number and
frequency of the treatments is left to the discretion of the
treating physician. For patients who are observed, examina-
tions and cytology are performed every 6 months, with bi-
opsies of any visible lesions. Therefore, although screening
and preventative treatment remain controversial, expectant
management with treatment of visible or palpable disease
remains an option unless or until emerging evidence sug-
gests that screening and ablation of subclinical lesions are
beneficial to reduce the incidence of anal cancer.
MALIGNANT NEOPLASMS OF THE ANAL CANAL
AND PERIANAL REGION
Pretreatment Evaluation
A disease-specific history and physical examina-
tion should be performed, emphasizing symptoms,
risk factors, and signs of advanced disease. Grade of
Recommendation: Strong recommendation based on
low-quality evidence, 1C.
Most patients present with a slow-growing mass involv-
ing either the anal canal or the perianal skin.
78
Pain and
bleeding are common, occurring in approximately half
of patients, although <20% of patients may be asymp-
tomatic.
10,79
Diagnosis of anal SCC may often be delayed,
mainly because of nonspecific anorectal symptoms, which
are frequently attributed to benign anorectal pathology,
such as hemorrhoids, in <70% to 80% of patients.
78,80
Pa-
tients with locally advanced anal cancers may also pres-
ent with foreign body sensation, symptoms related to
anal stenosis, and inguinal pain (commonly representing
inguinal lymph node metastases). Risk factors associated
with anal SCC include HPV infection; HIV seropositiv-
ity; a history of other HPV-related genital neoplasias,
such as cervical cancer; cervical intraepithelial neoplasia;
vulvar cancer; vulvar intraepithelial neoplasia; previous
sexually acquired diseases; cigarette smoking; anoreceptive
intercourse; multiple sexual partners; a history of solid-
organ transplant; and other forms of immunosuppres-
sion.
9,11,81–86
Because the incidence of anal cancer is higher
among men practicing anoreceptive intercourse, as well as
among patients positive for HIV, a high index of suspicion
should be maintained in these patients who present with
anorectal complaints.
87
Additional historical factors such
as previous radiation and inadequately controlled HIV
may limit chemoradiotherapy (CRT) and radical surgery
and are important variables to investigate at the time of
diagnosis.
Physical examination should focus primarily on ano-
rectal examination and evaluation of the inguinofemoral
nodes.
88
Digital anorectal examination should be performed
to identify the lesion location and to evaluate for fixation
and/or the presence of invasion of local structures, such as
the vagina or the anal sphincter mechanism. Anoscopy or
proctoscopy with biopsy is essential to establish the size of
the lesion, to determine its location within the anal canal,
and to confirm diagnosis. The presence of palpable inguinal
lymphadenopathy can suggest the need for fine-needle aspi-
ration or core biopsy to confirm malignant involvement and
Copyright © The American Society of Colon & Rectal Surgeons, Inc. Unauthorized reproduction of this article is prohibited.
DISEASES OF THE COLON & RECTUM VOLUME 61: 7 (2018)
763
to help guide radiation planning. In general, metastatic dis-
ease is difficult to detect on physical examination, although a
complete physical examination should be performed to help
identify any potential sites of distant spread that may war-
rant additional evaluation. All patients with a new diagnosis
of anal SCC should undergo basic laboratory studies, in-
cluding a complete blood count, renal and hepatic function
tests, and an assessment of their HIV status if not already
known. Women should undergo a cervical Pap test, and men
should undergo penile examination to exclude premalignant
or malignant lesions. Although the immunohistochemical
expression of p16 and Ki-67 has been shown to correlate
with the degree of anal intraepithelial neoplasia,
89
their role
in the evaluation of anal SCC is still being defined.
Endoscopic and radiologic evaluation should be per-
formed to help determine tumor extension and assess for
metastatic disease. Grade of Recommendation: Strong
recommendation based on low-quality evidence, 1C.
Biopsy should be performed under direct vision or with
anoscopy. Although anal cancer is not a risk factor for the de-
velopment of colon cancer, colorectal neoplasms have been
demonstrated in <15% of patients with anal cancer; there-
fore, colonoscopy should be performed to rule out synchro-
nous colorectal neoplasms.
88,89
A CT of the chest, abdomen,
and pelvis with intravenous contrast enhancement should
be performed to evaluate for distant metastatic disease and
lymphadenopathy, including evaluating the inguinal lymph
nodes, which may warrant biopsy in the setting of clinical or
radiographic abnormalities.
90
Because SCC can metastasize
to the brain, a CT of the head may be performed if the patient
has symptoms or signs of central nervous system involve-
ment. With CRT being the mainstay of treatment for anal
SCC, accurate anatomic imaging of the primary tumor is
highly recommended, because it enables optimal radiother-
apy planning and allows for posttreatment comparisons. En-
doanal ultrasound (EAUS) and MRI are at present the 2 most
accepted modalities for determining primary tumor depth,
anal sphincter involvement, and perirectal lymph node in-
volvement.
91,92
There is only 1 study to date
93
that directly
compares EAUS (using 2-dimensional imaging) with MRI in
the primary staging of anal SCC, with comparable results in
assessing primary tumor size and perirectal lymph node sta-
tus. Although EAUS is traditionally considered to be superior
to MRI for small superficial tumors, this has not been report-
ed in the current literature. Additional considerations with
EAUS include that it is operator dependent and may cause
significant discomfort in patients with anal stenosis.
2-[
18
F] Fluoro-2-deoxy-D-glucose positron emission to-
mography (PET)/CT may be considered as an adjunct
radiologic study in the staging of anal SCC, although it
does not replace CT scanning for clinical staging. Grade
of Recommendation: Strong recommendation based on
low-quality evidence, 1C.
Staging for SCC of the anal canal focuses on size of the pri-
mary lesion and locoregional lymph node involvement. As
such, clinical evaluation including size is critically impor-
tant to determine proper staging. The most widely used
clinical staging system is the American Joint Committee
on Cancer and International Union Against Cancer TNM
classification (Table 3), which defines T stage by maxi-
mum tumor diameter. This staging system does not take
into account sphincter muscle and perianal skin involve-
ment or the presence of a perianal or anovaginal fistula,
which are important prognostic factors that have not been
well studied in the era of modern CRT.
94–96
Although not typically a part of the routine evalua-
tion, 2-[
18
F] fluoro-2-deoxy-D-glucose PET/CT has been
shown to identify distant metastases that are not detected
by physical examination or other imaging modalities in
17% to 25% of patients,
97,98
resulting in a reported change
in treatment (ie, radiotherapy) in <19% of cases.
99–101
In
addition, retrospective studies evaluating the role and im-
pact of PET/CT in anal SCC have shown that metabolic
tumor volume at the primary tumor site, as well as hy-
permetabolic inguinal lymphadenopathy, correlates with
overall survival.
102–104
The measurement of metabolic
tumor volume at the primary cancer site, as well as with
potential pelvic and inguinal lymph node metastases, also
influences preradiation planning by radiation oncologists.
The primary treatment for all squamous cell cancers of
the anal canal, and for most perianal squamous cell can-
cers, is CRT. Grade of Recommendation: Strong recom-
mendation based on high-quality evidence, IA.
Before the initial case series by Nigro et al
105
describing
patients with squamous anal cancers treated with mul-
timodal neoadjuvant therapy, patients with these malig-
nancies were treated with abdominoperineal resection
(APR). The outcomes associated with primary APR were
dismal,
106,107
with locoregional recurrence rates as high
as 50% and 5-year survival rates ranging from 40% to
70%.
108,109
In addition, the morbidity of an APR during
this time period was significant. By contrast, even with
the limitations of delivering radiotherapy in his day, Ni-
gro and colleagues provided follow-up data to his original
series,
110,111
culminating in an evaluation of 104 patients
with squamous cancers of the anus who were treated with
30 Gy of radiotherapy combined with 5-fluorouracil (5-
FU) and mitomycin-C (MMC). In 97 patients, no gross
tumor remained. In this same series there were 31 patients
who underwent an APR regardless of whether there was
clinical evidence of tumor persistence, and 22 of these pa-
tients were completely free of any histological evidence of
cancer.
Currently, CRT is the standard of care for the treat-
ment of all anal canal squamous cell cancers and for all
perianal squamous cell cancers that are not well-differen-
tiated, node-negative, T1 lesions amenable to wide local
Copyright © The American Society of Colon & Rectal Surgeons, Inc. Unauthorized reproduction of this article is prohibited.
STEWART ET AL: ANAL CANCER CLINICAL PRACTICE GUIDELINES
764
excision. Although there are slight variations in how ra-
diation is administered, there are certain principles that
should be followed for every patient.
112,113
A multifield
technique should be used to deliver a minimum radiation
dose of 45.0 Gy, which is typically delivered to the primary
cancer in 25 fractions, each of 1.8 Gy over an 5-week time
interval. The radiation field should initially extend from
the border of L5 to S1, spreading distally to incorporate
the entire pelvis, including the anus and the inguinofemo-
ral nodes, terminating onto the perianal skin 2.5 cm distal
to the anus. Radiation oncologists should also make ef-
forts to reduce radiation exposure to the femoral heads be-
cause of the risk of avascular necrosis.
114
After the first 30.6
Gy is administered, the additional 14.4 Gy of scheduled
radiation should be delivered with the cephalad aspect of
the radiation field lowered to the distal aspect of the sac-
roiliac joints while also sparing the inguinal nodes from
additional treatment for those patients with no inguinal
nodal disease. In addition, for any lesions larger than a T1
cancer or for those that are node positive, a boost of 9 to
14 Gy to the primary tumor and to the involved nodes is
recommended. In this setting, the total dose of radiation
delivered would be 54 to 59 Gy.
The selection and doses of chemotherapeutic agents
for CRT can vary from among several options. Classically,
5-FU is infused at 1000 mg/m
2
on days 1 to 4 and days 29
to 32, with MMC administered in bolus form at 10 mg/
m
2
on days 1 and 29.
112
Alternatively, capecitabine, an oral
fluoropyrimidine prodrug, can be substituted for 5-FU at
a dose of 825 mg/m
2
twice daily.
115,116
When capecitabine is
selected, the dose of MMC is often increased to 12 mg/m
2
at the discretion of the treating medical oncologist.
CRT is associated with significant rates of acute toxic-
ity. In the Radiation Therapy Oncology Group (RTOG)
9811 study,
112
a total of 90% of patients treated with
MMC-based CRT experienced some form of GI toxicity,
ranging from nausea to vomiting and diarrhea. The inci-
dence of grade III or IV nonhematologic toxicity was 74%
in this group, whereas the incidence of severe long-term
toxic effects was 11%. This high incidence of significant
toxicities has prompted investigations into alternative
CRT regimens, as discussed below.
Patients preparing for CRT need to be counseled re-
garding sexual and reproductive health choices. Both men
and women of child-producing ages should be counseled
regarding sperm and ova banking before the onset of
therapy. Women should be asked if they have undergone
a recent gynecologic examination with screening for cer-
vical HPV, given the frequent association between ano-
rectal HPV and HPV of the cervix. Female patients who
are planning on sexual activity at any point after CRT
should be counseled regarding the use of vaginal dilators
to prevent vaginal stenosis resulting in the inability for
coitus. These issues are easily and frequently overlooked
and should typically be included in pretherapy counsel-
ing to preserve quality of life for long-term survivors of
anal cancer.
Multimodal therapy involving chemotherapy combined
with radiotherapy provides superior locoregional con-
trol compared with treatment with radiotherapy alone.
Grade of Recommendation: Strong recommendation
based on high-quality evidence, IA.
Cancers with squamous histology are typically radiosensi-
tive, and with the initial implementation of radiation as
monotherapy for squamous cancers of the anus, a high rate
of locoregional disease control and avoidance of surgery
was documented with smaller, earlier-stage cancers.
117,118
With larger, bulkier cancers, however, persistence and
recurrence rates were frequently encountered, and this
raised questions regarding a possible role for chemothera-
py in combination with radiotherapy. The first large study
to evaluate the role of multimodality therapy in anal can-
TABLE 3. American Joint Committee on Cancer and International
Union Against Cancer TNM classication of anal cancer
Primary tumor (T)
TX Primary tumor cannot be assessed
T0 No evidence of primary tumor
Tis High-grade squamous intraepithelial lesion (previously
termed carcinoma in situ, Bowen disease, anal
intraepithelial neoplasia II–III, high-grade anal
intraepithelial neoplasia)
T1
Tumor 2 cm
T2
Tumor >2 cm but 5 cm
T3 Tumor >5 cm
T4 Tumor of any size invading adjacent organ(s), such as
the vagina, urethra, or bladder
Regional lymph nodes (N)
NX Regional lymph nodes cannot be assessed
N0 No regional lymph node metastasis
N1 Metastasis in inguinal, mesorectal, internal iliac, or
external iliac nodes
N1a Metastasis in inguinal, mesorectal, or internal iliac lymph
nodes
N1b Metastasis in external iliac lymph nodes
N1c Metastasis in external iliac with any N1a nodes
Distant metastasis (M)
MX Distant metastasis cannot be assessed
cM0 No distant metastasis
cM1 Distant metastasis
pM1 Distant metastasis, microscopically conrmed
When T is... And N is... And M is...
Then the stage
group is...
Tis N0 M0 0
T1 N0 M0 I
T1 N1 M0 IIIA
T2 N0 M0 IIA
T2 N1 M0 IIIA
T3 N0 M0 IIB
T3 N1 M0 IIIC
T4 N0 M0 IIIB
T4 N1 M0 IIIC
Any T Any N M1 IV
Copyright © The American Society of Colon & Rectal Surgeons, Inc. Unauthorized reproduction of this article is prohibited.
DISEASES OF THE COLON & RECTUM VOLUME 61: 7 (2018)
765
cer was the United Kingdom Coordinating Committee on
Cancer Research Anal Cancer Trial (ACT I) Working Par-
ty.
119
In this trial, 585 patients were randomly assigned to
receive either 45 Gy of radiation in 20 to 25 fractions over
4 to 5 weeks or to receive the same form of radiotherapy
but with the addition of 5-FU and MMC. With a median
follow-up time of 42 months, 59% of the radiotherapy pa-
tients had experienced a local failure compared with 36%
of the CRT group, amounting to a 46% reduction in local
failure associated with the use of CRT. Similar rates of tu-
mor response were observed in both treatment groups at
6 weeks. Although the risk of death in the CRT arm was
lower, there was no difference in overall survival between
the 2 arms at 3 years after treatment. Not unexpectedly,
the CRT cohort was associated with a higher incidence
of early morbidity, whereas late adverse events related to
treatment occurred with similar frequency. This study was
instrumental in establishing CRT as the first-line therapy
for squamous cancers of the anal canal. A recent analysis
of this original study population
120
has provided 13-year
follow-up, demonstrating that CRT is associated with 25.3
fewer locoregional recurrences and 12.5 fewer anal can-
cer–related deaths for every 100 patients treated with CRT
compared with every 100 patients treated with radiation
alone. In addition, although there was a 9.1% increase in
deaths unrelated to anal cancer during the first 5 years af-
ter CRT, this increased risk of noncancer-related mortality
disappeared after 10 years. Local failure rates favored CRT
(57% vs 32%) at 5 years, as did colostomy-free survival at
5 years (37% vs 47%).
The year following ACT I, the European Organization
for Research and Treatment of Cancer Radiotherapy and
Gastrointestinal Cooperative Groups published a study
(22861)
121
evaluating the role of multimodality therapy
as compared with radiation alone for anal cancer. In this
randomized study, 110 patients were assigned to either 45
Gy followed by a boost of variable dosing depending on
tumor response versus the same radiation regimen com-
bined with 5-FU and MMC, the latter of which was pro-
vided as a bolus only on the first treatment day. Patients in
the CRT arm experienced a complete response rate of 80%
compared with 54% in the radiation arm, with improved
locoregional control and with superior progression-free
and colostomy-free survival rates. As with the United
Kingdom Coordinating Committee on Cancer Research
study, there were no differences in overall survival rates at
3 years after treatment.
The combination of 5-FU and MMC in conjunction
with radiotherapy remains as first-line multimodal
therapy for the treatment of squamous cancers of the
anus. Grade of Recommendation: Strong recommenda-
tion based on high-quality evidence, IA.
MMC has a significant toxicity profile, including he-
matologic toxicity in the form of bone marrow sup-
pression with neutropenia and thrombocytopenia,
pulmonary toxicity, and acute kidney injuries.
122
Be-
cause of earlier, albeit small, studies suggesting com-
parable oncologic outcomes and lower rates of toxicity
associated with omission of MMC, the RTOG 8704 tri-
al
123
was performed by the RTOG and the Eastern Co-
operative Oncology Group to evaluate whether MMC
could be removed from CRT while maintaining the
same disease control and cure rates. During this trial,
310 patients were randomly assigned to receive either
radiotherapy (ranging from 45.0 to 50.4 Gy) with 5-FU
infused for 4 days versus the same radiotherapy regi-
men combined with 5-FU and MMC at 10 mg/m
2
as
a bolus over 2 days. After treatment, biopsies yielding
histology positive for cancer did not differ to a statisti-
cally significant degree between either treatment arm
(15.0% vs 7.7%; p = 0.135). At 4 years after treatment,
colostomy rates (9% vs 22%; p = 0.002), colostomy
free-survival (71% vs 59%; p = 0.14), and disease-free
survival (73% vs 51%; p = 0.0003) favored the cohort
that received MMC. There was no difference in overall
survival, whereas toxicity was significantly higher in
the MMC cohort (23% vs 7% for grade 4 and 5 toxic-
ity; p < 0.001). Although toxicity with MMC was high-
er, the results of the study nonetheless supported the
inclusion of MMC in CRT compared with the use of
5-FU and radiation alone.
Although the net benefit of including MMC with
5-FU and radiation was established early in the evolution
of multimodality anal cancer therapy, lingering concerns
regarding MMC-related toxicity lead to attempts to sub-
stitute MMC with other agents. Several important clinical
trials have evaluated whether cisplatin might offer im-
proved toxicity rates while providing comparable, or su-
perior, disease control. The ACT II trial,
124
which remains
the largest clinical trial on anal cancer to date, enrolled
patients with squamous cancers of the anus from a total
of 59 centers across the United Kingdom. Patients were
randomly assigned to 1 of 4 groups, receiving either MMC
or cisplatin, with 5-FU and radiotherapy (50.4 Gy) with or
without 2 infusions of maintenance chemotherapy con-
sisting of 5-FU and cisplatin provided at weeks 11 and 14.
A total of 940 patients were enrolled, with a median fol-
low-up of 5.1 years. Complete response rates were similar
in both MMC and cisplatin treatment groups at 26 weeks
after therapy, with similar incidences of treatment-related
toxicities. There was no significant difference in 3-year
progression-free survival between the MMC and cisplatin
cohorts, and colostomy rates were similar between MMC-
and cisplatin-treated patients. Because of similar rates of
grade 3 and 4 adverse events, with no significant difference
in complete response rates or progression-free survival,
this study concluded that first-line therapy for squamous
cancers of the anus should continue as 5-FU and MMC
combined with radiotherapy.
Copyright © The American Society of Colon & Rectal Surgeons, Inc. Unauthorized reproduction of this article is prohibited.
STEWART ET AL: ANAL CANCER CLINICAL PRACTICE GUIDELINES
766
A second major study investigating the benefit of
MMC versus cisplatin was published by Ajani et al
112
in the RTOG 9811 study, a multicenter RCT. Study
patients were randomly assigned to receive 5-FU and
MMC with 45 to 59 Gy of radiotherapy or to receive
induction 5-FU with cisplatin followed by CRT with
5-FU and cisplatin. A total of 644 patients were evaluat-
ed with a median follow-up time of 2.5 years. There was
no significant difference between the treatment arms
in terms of 5-year disease-free survival or 5-year over-
all survival; 5-year locoregional recurrence rates and
distant metastasis rates were also similar between the
groups. A significantly higher colostomy rate was asso-
ciated with the cisplatin treatment arm (10% vs 19%;
p = 0.02), whereas the MMC arm experienced a higher
incidence of hematologic toxicity. This study conclud-
ed that the routine use of cisplatin in place of MMC is
not recommended. A subsequent analysis of this same
study population was undertaken by Gunderson et al,
125
published in 2012, to determine the long-term effects
of MMC versus cisplatin on recurrence and survival. In
this analysis, both disease-free and overall survival were
actually improved in the MMC-treated group compared
with the cisplatin group, with a trend toward improved
colostomy-free survival and locoregional control. These
results strengthened the recommendations to include
MMC in first-line multimodality therapy for anal squa-
mous cancers.
The European Organization for Research and Treat-
ment of Cancer 22011-40014 trial
126
evaluated the role
of cisplatin not as a replacement for MMC but as a
substitute for 5-FU. In this study, 88 patients with ei-
ther node-negative cancers >4 cm in diameter or those
with node-positive disease were randomly assigned to
radiotherapy totaling 59.4 Gy, with a 2-week gap in ra-
diotherapy deliberately introduced in the middle of the
therapy period, combined with either MMC/cisplatin or
MMC/5-FU. The cisplatin-treated patients demonstrat-
ed a trend toward lower compliance with receipt of CRT.
Toxicity rates were similar between the groups, with the
exception of a higher incidence of grade 3 hematologic
toxicity in the cisplatin group. Survival analyses with
this trial were made difficult because of median event-
free, overall, and progression-free survivals not being
reached. Despite this study concluding that the combi-
nation of MMC and cisplatin is promising, additional
data regarding this form of CRT are required before its
role can be determined.
The use of induction chemotherapy before CRT
has also been evaluated, with disappointing results.
The UNICANCER ACCORD 03 study
127
was a 4-arm,
prospective randomized trial designed to assess both
the role of induction chemotherapy consisting of 5-FU
on days 1 through 4 and days 29 through 32, and cis-
platin on days 1 and 29, before CRT, as well as to test
the effect of a higher dose applied to a radiation boost.
Patients were randomly assigned to the following: 1)
receive 2 cycles of induction chemotherapy, CRT, and
a standard-dose boost (15 Gy); 2) receive 2 cycles of
induction chemotherapy, CRT, and a high-dose boost
(20–25 Gy); 3) receive CRT and a standard-dose boost;
or 4) receive CRT and a high-dose boost. Of the 307
patients enrolled, 283 received a complete treatment
course, with a median follow-up of 50 months. No
benefit to either induction chemotherapy or high-dose
boost radiation was observed, with comparable partial
and complete tumor response rates, locoregional fail-
ure rates, and 3-year colostomy-free, event-free, and
overall survival. A recent small retrospective study (n =
38)
128
of patients with T4 anal cancers treated with 45
Gy of radiotherapy with concurrent 5-FU and cisplatin
(n = 27) versus patients treated with induction chemo-
therapy with 5-FU and cisplatin followed by CRT (n =
11) was conducted. There was no statistically signifi-
cant difference between the 2 groups based on 5-year
overall, disease-free, or relapse-free survival, although
5-year colostomy-free survival was much higher in the
group receiving induction chemotherapy (100% vs
38%; p = 0.0006), leading the authors to suggest that
induction chemotherapy has a role in the management
of T4 anal cancers. At this time, there is insufficient
high-quality data to support the use of induction che-
motherapy outside of a clinical trial.
No oncologic benefit exists for providing radiation dos-
es >59 Gy. Grade of Recommendation: Strong recom-
mendation based on moderate-quality evidence, IB.
Once doses of radiotherapy exceed 40 Gy, the incidence
of radiation-related toxicities increases. There is good
evidence that the minimum dose of radiation for anal
squamous cancers should be 45 Gy, although the op-
timal dose is a matter of debate, in large part because
of different radiation delivery techniques and differ-
ent tumor stages included in the studies on this sub-
ject, all of which influence toxicity and response rates.
The data on optimal radiation dosing have either been
the subject of smaller and nonrandomized studies or
have been evaluated in randomized studies looking at
such issues as adjusting the dose of boost radiation to
the total dosage. The number of studies dedicated to
the subject are few, although 1 such study
129
consisted
of a small number (n = 69) of early stage Tis and T1
cancers. The study population was extremely hetero-
geneous, with 26 subjects undergoing local excision
before radiotherapy and with varying doses of exter-
nal beam radiotherapy, with or without brachytherapy,
with 8 patients treated with brachytherapy alone. The
study concluded, on the basis of local control and tox-
icity rates, that for T1 cancers a radiation dosing range
of 50 to 60 Gy was optimal. One of the only other ret-
Copyright © The American Society of Colon & Rectal Surgeons, Inc. Unauthorized reproduction of this article is prohibited.
DISEASES OF THE COLON & RECTUM VOLUME 61: 7 (2018)
767
rospective studies on this subject
130
included patients
with stage I to III disease, although with a study popu-
lation of only 43 subjects and with a median follow-up
of only 42 months. Locoregional control was improved
for patients who received >50 Gy (86.5% vs 34.0%; p =
0.012), although no dosage ceiling was suggested from
these data.
An older study, the RTOG 9208 trial,
131
was a clini-
cal trial that enrolled only 47 patients with at least T1
squamous anal cancers who received CRT with 5-FU
and MMC with 59.6 Gy of radiation, which included a
2-week rest period during CRT. The study patients were
compared with patients enrolled in the previous RTOG
8704 trial who had received 40.0 to 50.4 Gy of radia-
tion. Despite a higher dose of radiation, patients in the
9208 trial actually had a higher colostomy rate at 1 and
2 years after treatment, without an improvement in lo-
coregional control. No benefit to higher doses exceeding
59 Gy was noted, although concerns for escalating inci-
dences in toxicity were raised. The lack of efficacy with
higher doses of radiation is also echoed in the results
of the previously mentioned ACCORD 03 trial, which
found no benefit to increasing radiation dosing by using
high-dose boost treatments.
Missed treatments should be avoided, because they are
strongly associated with inferior disease control. Grade
of Recommendation: Strong recommendation based on
moderate-quality evidence, IB.
The ACT II trial primarily evaluated the role of cispla-
tin with CRT, and the use of maintenance chemotherapy
with 5-FU and cisplatin in addition to CRT, finding no
advantage to either of these alternative approaches. This
study touted a high rate of complete response (90%),
with similar 3-year progression-free survival in the
MMC and cisplatin groups, and with an overall 3-year
colostomy-free survival of 74%, with 75% of patients
having locoregional control with organ preservation.
The authors of this study attributed these excellent re-
sults in part to high compliance rates with the 50.4 Gy
of planned radiation and in part because of the ability
to provide CRT in a shorter time interval by avoiding
a planned treatment gap, which was a common feature
of CRT before this study. A similar observation can be
gleaned from the Eastern Cooperative Oncology Group
E4292 trial,
132
a study of 33 patients designed to evalu-
ate whether cisplatin could replace MMC as part of CRT.
This study accrued a total of 13 patients who did not re-
ceive a planned 2-week treatment break, which the larger
number of study patients did receive. Complete response
rates of 78% were noted in patients without a treatment
break, which was higher than in subjects who did receive
a break in therapy. The RTOG 9208 trial
131
included a
2-week treatment break, which was associated with in-
creased locoregional failure rates and lower colostomy-
free survival rates. Treatment breaks and interruptions to
CRT should be avoided if possible, although they occur
as frequently as in 80% of patients with anal cancer,
133
making treatment compliance a major and potentially
modifiable factor associated with survival.
Surveillance
Disease surveillance should typically start 8 to 12 weeks
from the completion of CRT. Grade of Recommendation:
Strong recommendation based on moderate-quality
evidence, 1B.
Slow tumor regression after completion of CRT is based
on evidence from trials, such ACT II, in which randomly
assigned patients had clinical complete response docu-
mented at 11 and 26 weeks postcompletion of CRT. Data
from this trial indicate that 29% of patients who did
not demonstrate a complete remission at 11 weeks had
achieved a complete response by 26 weeks, with durable
response.
124
Most patients are reasonably comfortable with anal
examination by the eighth week after completion of CRT,
although earlier evaluation to ensure symptom control
should be tailored to the individual. The National Com-
prehensive Cancer Network guidelines
134
recommend
evaluation at 8 to 12 weeks after completion of CRT. Clini-
cal assessment should include digital rectal examination,
anoscopy, and palpation of the inguinal lymph nodes. Be-
cause of slow tumor regression, biopsies for persistent dis-
ease are typically avoided at 8 to 12 weeks and <6 months
post-CRT.
Posttreatment imaging is most frequently undertaken
3 months from the completion of CRT, when treatment-
related fibrosis and residual tumor could be distin-
guished.
135
Imaging recommendations from the National
Comprehensive Cancer Network guidelines include CT
of the chest, abdomen, and pelvis annually for 3 years (if
T3–T4 or inguinal node positive).
Surveillance involving digital rectal examination, anos-
copy, and imaging should be continued for 5 years after
completion of CRT. Grade of Recommendation: Strong
recommendation based on moderate-quality evidence, 1B.
After the first evaluation post-CRT, additional surveil-
lance including digital anorectal examination, anosco-
py, and palpation of inguinal lymph nodes every 3 to 6
months for those in complete remission or every 4 weeks
until remission <6 months is recommended in patients
with evidence of persistent disease.
134
Patients with re-
sidual changes require close follow-up and documenta-
tion of any changes identified. New tissue thickening,
growth, or ulceration should be evaluated. Examination
under anesthesia and potential targeted biopsy should
be considered in patients with persistent abnormalities
5 to 6 months after CRT, stenosis, pain, or scarring pre-
Copyright © The American Society of Colon & Rectal Surgeons, Inc. Unauthorized reproduction of this article is prohibited.
STEWART ET AL: ANAL CANCER CLINICAL PRACTICE GUIDELINES
768
venting adequate surveillance examinations. The major-
ity of relapses and thus the most efficient time for more
frequent surveillance are within the first 3 years of com-
pletion of CRT. The ACT II trial with long-term follow-
up demonstrated <1% of relapses occurring beyond 3
years.
124
Other trials suggest that this may be closer to
5%; thus, there seems to be some rationale to continue
surveillance out to 5 years.
125,136
CT scans of the chest, abdomen, and pelvis annually
for 3 years for patients with T3 to T4 tumors or positive
inguinal lymph nodes are also recommended. There are
no formal recommendations with regard to post-CRT
EAUS, MRI, or PET/CT. One retrospective study evaluat-
ing the impact of PET/CT on assessing for residual tumor
in 52 patients after CRT showed negative predictive and
positive predictive values of 100% and 71%.
137
Vercellino
et al
138
showed a sensitivity for the detection of persistent
or recurrent disease of 93% and specificity of 81% with
PET/CT, leading to changes in management in 20% of pa-
tients. The role of post-CRT imaging with MRI and PET/
CT to detect locoregional persistent or recurrent tumor
seems to be evolving; however, its true ability to impact
earlier salvage compared with clinical evaluation remains
inconclusive.
Treatment of Recurrent or Persistent Disease
APR is effective salvage therapy for persistent or recur-
rent disease. Grade of Recommendation: Strong recom-
mendation based on moderate-quality evidence, 1B.
Approximately 20% to 30% of patients will have persis-
tent or recurrent disease after primary CRT.
133
Predictors
of recurrent and persistent locoregional disease after de-
finitive CRT include higher T and N stages at original
presentation, HIV-positive status, and the inability to
complete CRT.
133,139
Although patients with persistent
disease who present within 6 months of completing CRT
have been reported to have worse prognosis,
140–142
recent
data suggest that persistent and recurrent disease does
not show any significant difference in survival, and pa-
tients with late recurrence (>24 mo) may have a better
prognosis.
143
APR is recommended for persistent or recurrent dis-
ease for salvage therapy, with reported 5-year locoregional
control in 30% to 77% of patients
144–148
and 5-year overall
survival rates of 24% to 69%.
141,142,145–149
Positive surgi-
cal margins (microscopic or macroscopic), male sex, and
higher Charlson comorbidity index portend a worse prog-
nosis, whereas younger age (<55 y), as well as T1 to T2 and
N0 to N1 disease have been associated with higher overall
survival after APR.
145,150
Major wound complications are common, reported in
36% to 80%.
145
Muscle flap reconstruction has been asso-
ciated with significantly lower rates of such complications
without major donor-site morbidity.
148,151,152
Pedicled
muscle flaps have shown a significantly lower rate of recip-
ient site complications than V–Y advancement flaps, with
the vertical rectus abdominis myocutaneous flap showing
superior results compared with the gracilis flap in terms of
the overall reduction of complications.
153
Patients with HIV or AIDS who present with anal cancer
as the first manifestation of their immunosuppression,
and who are not medically deconditioned, can be safely
treated according to the same regimens as immunocom-
petent patients. Grade of Recommendation: Strong rec-
ommendation based on medium-quality evidence, IC.
As the medical management of HIV has improved
and as the life expectancy of patients with HIV has
increased, older and smaller retrospective studies that
suggested poorer outcomes for HIV-positive patients
treated for anal cancer have been questioned in terms
of their generalizability to the management of current
HIV patients with anal cancer. One of these earlier ret-
rospective studies suggesting that well-compensated
HIV patients could be treated similarly to HIV-negative
patients was published in 1999 by Hoffman et al.
154
In
this small retrospective analysis of 17 HIV-positive pa-
tients with anal cancer treated with either CRT or ra-
diation alone, patients with CD4 counts 200 cells per
cubic millimeter had excellent disease control with an
incidence of treatment toxicities similar to what has
been described in the literature for immunocompetent
patients. Patients with a CD4 count <200 cells per cubic
millimeter (n = 8) had a significantly higher incidence
of treatment toxicities, although with 7 of these 8 pa-
tients achieving disease control. A more recent publi-
cation from 2010
155
evaluated 21 HIV-positive patients
receiving CRT. Completion of treatment was achieved
in all 21 patients, with a complete response of 81% and
6 patients (29%) dying of disease. The authors noted
that the 5-year local control, cancer-specific, and over-
all survival rates were 59%, 75%, and 67%. Because the
majority of the patients were able to complete CRT
without dose reductions in either radiation or che-
motherapy, this study also supported the concept that
HIV-positive patients could be treated according to
standard regimens with the expectation of good toler-
ance to therapy. Another recent retrospective study
156
of 36 HIV-positive patients treated with standard regi-
mens of CRT again demonstrated completion of treat-
ment in all of the patients, with complete response rates
of 86% and with 5-year local control, colostomy-free,
cancer-specific, and overall survival rates of 72%, 87%,
77%, and 74%. This study noted a CRT-related decline
in CD4 counts for <6 years after treatment, although
this decrease in CD4 counts was not associated with
increased HIV-related morbidity, with oncologic out-
comes comparable to descriptions of the HIV-negative
population in the literature.
Copyright © The American Society of Colon & Rectal Surgeons, Inc. Unauthorized reproduction of this article is prohibited.
DISEASES OF THE COLON & RECTUM VOLUME 61: 7 (2018)
769
One of the largest retrospective studies on this
subject to date was a review of patient data from the
Veterans Affairs system,
157
analyzing 1184 individuals
treated for squamous cancers of the anus, of whom 175
were documented to be HIV positive. As with previous
studies, this study reported no difference in the receipt
of treatment based on HIV status, with 2-year observed
survival rates of 77% and 75% among HIV-positive and
HIV-negative individuals. Based on multivariate analy-
sis, HIV status was not associated with differences in
survival.
There are insufficient data to comment on whether
highly active antiretroviral therapy is associated with im-
proved outcomes after CRT. Patients with ongoing HIV/
AIDS disease–related complications present before their
diagnosis of anal cancer may not tolerate standard CRT
and may require dose reductions; this subpopulation of
patients with HIV is therefore at risk for higher CRT toxic-
ity rates and worse oncologic outcomes.
Perianal squamous cancers, which are well-differentiat-
ed, node-negative, T1 lesions, can be adequately treated
with wide-local excision with 1-cm margins of resec-
tion. All other anal margin cancers are preferentially
treated with CRT. Grade of Recommendation: Strong
recommendation based on low-quality evidence, IC.
Patients with well-differentiated, node-negative T1 lesions
involving the perianal skin are preferentially treated with
wide-local excision provided that 1-cm margins of resec-
tion can be obtained without compromising the patients
sphincter mechanism, which would compromise the
functional outcome of a local excision. The data in sup-
port of wide-local excision are from small retrospective
studies, although the outcomes of these studies are con-
sistently excellent, with 5-year survival rates of 100%.
158
In addition, wide-local excision avoids the toxicity asso-
ciated with radiotherapy. Any perianal squamous cancers
that do not meet the above-mentioned favorable criteria
should be treated with CRT, because there is a well-docu-
mented history of locoregional recurrence and decreased
survival when such cancers are treated with wide-local
excision.
106,159
Treatment of Distant Disease
Systemic chemotherapy should be considered for patients
with distant metastatic disease. Metastasectomy, radiation,
and radiofrequency ablation can be considered in selected
cases. Grade of Recommendation: Weak recommendation
based on low- or very-lowquality evidence, 2C.
Because of high response rates with chemoradiation, re-
ports comparing treatment of patients with metastatic dis-
ease are limited. The most extensive published experience
is a retrospective review of a 14-year experience at a single
center in France.
160
A total of 50 patients, 10 with synchro-
nous disease at the time of diagnosis and 40 with distant
failure after chemoradiation, were treated with multimodal
therapy. Patients received 1 chemotherapy regimen, 13
had surgical metastasectomy, 11 had radiotherapy, and 6
had radiofrequency ablation. Median overall survival was
20 months. Overall response rate was 56%. There was no
clear advantage of any chemotherapy regimen, but patients
who could also have resection, radiation, or ablation had
an overall survival of 22 months compared with 13 months
with chemotherapy alone (p = 0.002). A similar-sized cohort
has been published in abstract form, including 53 patients
treated with chemotherapy, with a median overall survival
of 38 months.
161,162
A small single-center study reported in
1999 that included 19 patients with metastatic anal cancer
treated with 5-FU and cisplatin described a response rate of
66%, with 1 patient achieving a complete response.
163
A sin-
gle-center report described the combination of carboplatin
and paclitaxel in 13 patients with a response rate of 62%,
with 2 patients achieving a complete response.
164
Targeted therapy to the epidermal growth factor re-
ceptor (EGFR) has been considered because of the high ex-
pression of EGFR in anal cancers.
165
The use of cetuximab
in the setting of localized disease has been limited by toxic-
ity when combined with traditional chemoradiation.
166,167
However, response rate was high, and it is possible that, in
the metastatic setting, without concurrent radiation, there
is a role for the addition of an EGFR inhibitor to cytotoxic
chemotherapy regimens. Because of the lack of uniform
treatment, there are also active clinical trials in this area
available at clinicaltrials.gov, examining novel approaches
including pembrolizumab and nivolumab.
REFERENCES
1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Can-
cer J Clin. 2016;66:7–30.
2. Nelson RA, Levine AM, Bernstein L, Smith DD, Lai LL. Chang-
ing patterns of anal canal carcinoma in the United States. J Clin
Oncol. 2013;31:1569–1575.
3. Johnson LG, Madeleine MM, Newcomer LM, Schwartz SM,
Daling JR. Anal cancer incidence and survival: the surveillance,
epidemiology, and end results experience, 1973-2000. Cancer.
2004;101:281–288.
4. Nielsen A, Munk C, Kjaer SK. Trends in incidence of anal can-
cer and high-grade anal intraepithelial neoplasia in Denmark,
1978-2008. Int J Cancer. 2012;130:1168–1173.
5. Palefsky JM, Giuliano AR, Goldstone S, et al. HPV vaccine
against anal HPV infection and anal intraepithelial neoplasia.
N Engl J Med. 2011;365:1576–1585.
6. Palefsky JM, Holly EA, Gonzales J, Berline J, Ahn DK,
Greenspan JS. Detection of human papillomavirus DNA in
anal intraepithelial neoplasia and anal cancer. Cancer Res.
1991;51:1014–1019.
7. Holly EA, Ralston ML, Darragh TM, Greenblatt RM, Jay
N, Palefsky JM. Prevalence and risk factors for anal squa-
mous intraepithelial lesions in women. J Natl Cancer Inst.
2001;93:843–849.
Copyright © The American Society of Colon & Rectal Surgeons, Inc. Unauthorized reproduction of this article is prohibited.
STEWART ET AL: ANAL CANCER CLINICAL PRACTICE GUIDELINES
770
8. Abbas A, Yang G, Fakih M. Management of anal cancer in 2010:
part 1–overview, screening, and diagnosis. Oncology (Williston
Park). 2010;24:364–369.
9. Frisch M, Glimelius B, van den Brule AJ, et al. Sexually trans-
mitted infection as a cause of anal cancer. N Engl J Med.
1997;337:1350–1358.
10. Ryan DP, Compton CC, Mayer RJ. Carcinoma of the anal canal.
N Engl J Med. 2000;342:792–800.
11. Daling JR, Weiss NS, Hislop TG, et al. Sexual practices, sexually
transmitted diseases, and the incidence of anal cancer. N Engl J
Med. 1987;317:973–977.
12. Schofield AM, Sadler L, Nelson L, et al. A prospective study
of anal cancer screening in HIV-positive and negative MSM.
AIDS. 2016;30:1375–1383.
13. Radecki Breitkopf C, Finney Rutten LJ, Findley V, et al. Aware-
ness and knowledge of human papillomavirus (HPV), HPV-
related cancers, and HPV vaccines in an uninsured adult clinic
population. Cancer Med. 2016;5:3346–3352.
14. Abramowitz L, Benabderrahmane D, Walker F, et al. Deter-
minants of macroscopic anal cancer and precancerous le-
sions in 1206 HIV-infected screened patients. Colorectal Dis.
2016;18:997–1004.
15. Hemminki K, Liu X, Ji J, Sundquist J, Sundquist K. Autoim-
mune disease and subsequent digestive tract cancer by histol-
ogy. Ann Oncol. 2012;23:927–933.
16. Patel HS, Silver AR, Northover JM. Anal cancer in renal trans-
plant patients. Int J Colorectal Dis. 2007;22:1–5.
17. Brickman C, Palefsky JM. Cancer in the HIV-infected host: epi-
demiology and pathogenesis in the antiretroviral era. Curr HIV/
AIDS Rep. 2015;12:388–396.
18. Pirog EC, Quint KD, Yantiss RK. P16/CDKN2A and Ki-67
enhance the detection of anal intraepithelial neoplasia and
condyloma and correlate with human papillomavirus de-
tection by polymerase chain reaction. Am J Surg Pathol.
2010;34:1449–1455.
19. Fenger C. Prognostic factors in anal carcinoma. Pathology.
2002;34:573–578.
20. Das P, Crane CH, Eng C, Ajani JA. Prognostic factors for squa-
mous cell cancer of the anal canal. Gastrointest Cancer Res.
2008;2:10–14.
21. Pineda CE, Welton ML. Management of anal squamous in-
traepithelial lesions. Clin Colon Rectal Surg. 2009;22:94–101.
22. Darragh TM, Colgan TJ, Cox JT, et al.; Members of LAST
Project Work Groups. The Lower Anogenital Squamous Ter-
minology standardization project for HPV-associated lesions:
background and consensus recommendations from the Col-
lege of American Pathologists and the American Society for
Colposcopy and Cervical Pathology. Arch Pathol Lab Med.
2012;136:1266–1297.
23. Yang EJ, Kong CS, Longacre TA. Vulvar and anal intraepithelial
neoplasia: terminology, diagnosis, and ancillary studies. Adv
Anat Pathol. 2017;24:136–150.
24. Steele SR, Varma MG, Melton GB, Ross HM, Rafferty JF, Buie
WD; Standards Practice Task Force of the American Society of
Colon and Rectal Surgeons. Practice parameters for anal squa-
mous neoplasms. Dis Colon Rectum. 2012;55:735–749.
25. Guyatt G, Gutterman D, Baumann MH, et al. Grading strength
of recommendations and quality of evidence in clinical guide-
lines: report from an american college of chest physicians task
force. Chest. 2006;129:174–181.
26. Palefsky JM, Holly EA, Ralston ML, et al. Anal squamous in-
traepithelial lesions in HIV-positive and HIV-negative homo-
sexual and bisexual men: prevalence and risk factors. J Acquir
Immune Defic Syndr Hum Retrovirol. 1998;17:320–326.
27. Chin-Hong PV, Vittinghoff E, Cranston RD, et al. Age-related
prevalence of anal cancer precursors in homosexual men: the
EXPLORE study. J Natl Cancer Inst. 2005;97:896–905.
28. Machalek DA, Poynten M, Jin F, et al. Anal human papillomavi-
rus infection and associated neoplastic lesions in men who have
sex with men: a systematic review and meta-analysis. Lancet
Oncol. 2012;13:487–500.
29. Heard I, Etienney I, Potard V, et al.; ANRS-C017 VIHGY Study
Group. High prevalence of anal human papillomavirus-associ-
ated cancer precursors in a contemporary cohort of asymptom-
atic HIV-infected women. Clin Infect Dis. 2015;60:1559–1568.
30. Ebisch RMF, Rutten DWE, IntHout J, et al. Long-lasting in-
creased risk of human papillomavirus-related carcinomas
and premalignancies after cervical intraepithelial neopla-
sia grade 3: a population-based cohort study. J Clin Oncol.
2017;35:2542–2550.
31. Schiffman M, Wentzensen N, Wacholder S, Kinney W, Gage JC,
Castle PE. Human papillomavirus testing in the prevention of
cervical cancer. J Natl Cancer Inst. 2011;103:368–383.
32. Darragh TM, Tokugawa D, Castle PE, et al. Interrater agreement
of anal cytology. Cancer Cytopathol. 2013;121:72–78.
33. Berry JM, Jay N, Cranston RD, et al. Progression of anal high-
grade squamous intraepithelial lesions to invasive anal cancer
among HIV-infected men who have sex with men. Int J Cancer.
2014;134:1147–1155.
34. Fazendin EA, Crean AJ, Fazendin JM, et al. Condyloma acumi-
natum, anal intraepithelial neoplasia, and anal cancer in the set-
ting of HIV: do we really understand the risk? Dis Colon Rectum.
2017;60:1078–1082.
35. Tinmouth J, Peeva V, Amare H, et al. Progression from peri-
anal high-grade anal intraepithelial neoplasia to anal cancer in
HIV-positive men who have sex with men. Dis Colon Rectum.
2016;59:836–842.
36. Goldstone SE, Johnstone AA, Moshier EL. Long-term outcome
of ablation of anal high-grade squamous intraepithelial le-
sions: recurrence and incidence of cancer. Dis Colon Rectum.
2014;57:316–323.
37. Cachay E, Agmas W, Mathews C. Five-year cumulative inci-
dence of invasive anal cancer among HIV-infected patients ac-
cording to baseline anal cytology results: an inception cohort
analysis. HIV Med. 2015;16:191–195.
38. Goldstone SE. Introduction. Semin Colon Rectal Surg.
2017;28:55–56.
39. Jin F, Roberts JM, Grulich AE, et al.; SPANC Research Team.
The performance of human papillomavirus biomarkers in pre-
dicting anal high-grade squamous intraepithelial lesions in gay
and bisexual men. AIDS. 2017;31:1303–1311.
40. Cheng SH, Wang CC, Chang SL, Chu FY, Hsueh YM. Oncogenic
human papillomavirus is not helpful for cytology screening of
the precursor lesions of anal cancers in Taiwanese men who are
infected with human immunodeficiency virus. Int J Clin Oncol.
2015;20:943–951.
41. Sendagorta E, Herranz P, Guadalajara H, et al. Prevalence of ab-
normal anal cytology and high-grade squamous intraepithelial
lesions among a cohort of HIV-infected men who have sex with
men. Dis Colon Rectum. 2014;57:475–481.
Copyright © The American Society of Colon & Rectal Surgeons, Inc. Unauthorized reproduction of this article is prohibited.
DISEASES OF THE COLON & RECTUM VOLUME 61: 7 (2018)
771
42. Botes LP, Pett S, Carr A, et al. Anal cytological abnormalities
are poor predictors of high-grade intraepithelial neoplasia
amongst HIV-positive men who have sex with men. Sex Health.
2013;10:9–17.
43. Wentzensen N, Follansbee S, Borgonovo S, et al. Human papil-
lomavirus genotyping, human papillomavirus mRNA expres-
sion, and p16/Ki-67 cytology to detect anal cancer precursors in
HIV-infected MSM. AIDS. 2012;26:2185–2192.
44. Williams VM, Metcalf C, French MA, McCloskey JC. Audit
of paired anal cytology and histopathology outcomes in pa-
tients referred to a public sexual health clinic. Sex Health.
2010;7:346–351.
45. Salit IE, Lytwyn A, Raboud J, et al. The role of cytology (Pap
tests) and human papillomavirus testing in anal cancer screen-
ing. AIDS. 2010;24:1307–1313.
46. Nathan M, Singh N, Garrett N, Hickey N, Prevost T, Sheaff M.
Performance of anal cytology in a clinical setting when mea-
sured against histology and high-resolution anoscopy findings.
AIDS. 2010;24:373–379.
47. Fox PA, Seet JE, Stebbing J, et al. The value of anal cytology and
human papillomavirus typing in the detection of anal intraepi-
thelial neoplasia: a review of cases from an anoscopy clinic. Sex
Transm Infect. 2005;81:142–146.
48. Arain S, Walts AE, Thomas P, Bose S. The anal pap smear: cy-
tomorphology of squamous intraepithelial lesions. Cytojournal.
2005;2:4.
49. Papaconstantinou HT, Lee AJ, Simmang CL, et al. Screening
methods for high-grade dysplasia in patients with anal condy-
loma. J Surg Res. 2005;127:8–13.
50. Mathews WC, Sitapati A, Caperna JC, Barber RE, Tugend A,
Go U. Measurement characteristics of anal cytology, histopa-
thology, and high-resolution anoscopic visual impression in an
anal dysplasia screening program. J Acquir Immune Defic Syndr.
2004;37:1610–1615.
51. Panther LA, Wagner K, Proper J, et al. High resolution anoscopy
findings for men who have sex with men: inaccuracy of anal
cytology as a predictor of histologic high-grade anal intraepi-
thelial neoplasia and the impact of HIV serostatus. Clin Infect
Dis. 2004;38:1490–1492.
52. Palefsky JM, Holly EA, Hogeboom CJ, Berry JM, Jay N, Dar-
ragh TM. Anal cytology as a screening tool for anal squamous
intraepithelial lesions. J Acquir Immune Defic Syndr Hum Retro-
virol. 1997;14:415–422.
53. Pineda CE, Berry JM, Jay N, Palefsky JM, Welton ML. High-
resolution anoscopy targeted surgical destruction of anal high-
grade squamous intraepithelial lesions: a ten-year experience.
Dis Colon Rectum. 2008;51:829–835.
54. Crawshaw BP, Russ AJ, Stein SL, et al. High-resolution anoscopy
or expectant management for anal intraepithelial neoplasia for
the prevention of anal cancer: is there really a difference? Dis
Colon Rectum. 2015;58:53–59.
55. Mathews WC, Agmas W, Cachay E. Comparative accuracy of
anal and cervical cytology in screening for moderate to severe
dysplasia by magnification guided punch biopsy: a meta-analy-
sis. PLoS One. 2011;6:e24946.
56. Palefsky JM. Practising high-resolution anoscopy. Sex Health.
2012;9:580–586.
57. Fox PA, Nathan M, Francis N, et al. A double-blind, random-
ized controlled trial of the use of imiquimod cream for the
treatment of anal canal high-grade anal intraepithelial neopla-
sia in HIV-positive MSM on HAART, with long-term follow-
up data including the use of open-label imiquimod. AIDS.
2010;24:2331–2335.
58. Richel O, de Vries HJ, van Noesel CJ, Dijkgraaf MG, Prins JM.
Comparison of imiquimod, topical fluorouracil, and electro-
cautery for the treatment of anal intraepithelial neoplasia in
HIV-positive men who have sex with men: an open-label, ran-
domised controlled trial. Lancet Oncol. 2013;14:346–353.
59. van der Snoek EM, den Hollander JC, van der Ende ME. Im-
iquimod 5% cream for five consecutive days a week in an HIV-
infected observational cohort up to 32 weeks in the treatment
of high-grade squamous intraepithelial lesions. Sex Transm In-
fect. 2015;91:245–247.
60. Richel O, Wieland U, de Vries HJ, et al. Topical 5-fluoro-
uracil treatment of anal intraepithelial neoplasia in hu-
man immunodeficiency virus-positive men. Br J Dermatol.
2010;163:1301–1307.
61. Snyder SM, Siekas L, Aboulafia DM. Initial experience with
topical fluorouracil for treatment of hiv-associated anal in-
traepithelial neoplasia. J Int Assoc Physicians AIDS Care (Chic).
2011;10:83–88.
62. Cranston RD, Baker JR, Liu Y, Wang L, Elishaev E, Ho KS.
Topical application of trichloroacetic acid is efficacious for the
treatment of internal anal high-grade squamous intraepithelial
lesions in HIV-positive men. Sex Transm Dis. 2014;41:420–426.
63. Singh JC, Kuohung V, Palefsky JM. Efficacy of trichloroacetic
acid in the treatment of anal intraepithelial neoplasia in HIV-
positive and HIV-negative men who have sex with men. J Acquir
Immune Defic Syndr. 2009;52:474–479.
64. Sendagorta E, Bernardino JI, Álvarez-Gallego M, et al.; CI-
DAN127412 GESIDA Study Group. Topical cidofovir to treat
high-grade anal intraepithelial neoplasia in HIV-infected pa-
tients: a pilot clinical trial. AIDS. 2016;30:75–82.
65. Stier EA, Goldstone SE, Einstein MH, et al. Safety and efficacy
of topical cidofovir to treat high-grade perianal and vulvar in-
traepithelial neoplasia in HIV-positive men and women. AIDS.
2013;27:545–551.
66. Weis SE, Vecino I, Pogoda JM, Susa JS. Treatment of high-grade
anal intraepithelial neoplasia with infrared coagulation in a
primary care population of HIV-infected men and women. Dis
Colon Rectum. 2012;55:1236–1243.
67. Goldstone SE, Kawalek AZ, Huyett JW. Infrared coagulator: a
useful tool for treating anal squamous intraepithelial lesions.
Dis Colon Rectum. 2005;48:1042–1054.
68. Goldstone SE, Hundert JS, Huyett JW. Infrared coagulator ab-
lation of high-grade anal squamous intraepithelial lesions in
HIV-negative males who have sex with males. Dis Colon Rec-
tum. 2007;50:565–575.
69. Goldstone RN, Goldstone AB, Russ J, Goldstone SE. Long-term
follow-up of infrared coagulator ablation of anal high-grade
dysplasia in men who have sex with men. Dis Colon Rectum.
2011;54:1284–1292.
70. Cranston RD, Hirschowitz SL, Cortina G, Moe AA. A retrospec-
tive clinical study of the treatment of high-grade anal dysplasia
by infrared coagulation in a population of HIV-positive men
who have sex with men. Int J STD AIDS. 2008;19:118–120.
71. Sirera G, Videla S, Piñol M, et al.; HIV-HPV Study Group. Long-
term effectiveness of infrared coagulation for the treatment of
anal intraepithelial neoplasia grades 2 and 3 in HIV-infected
men and women. AIDS. 2013;27:951–959.
Copyright © The American Society of Colon & Rectal Surgeons, Inc. Unauthorized reproduction of this article is prohibited.
STEWART ET AL: ANAL CANCER CLINICAL PRACTICE GUIDELINES
772
72. Marks DK, Goldstone SE. Electrocautery ablation of high-grade
anal squamous intraepithelial lesions in HIV-negative and
HIV-positive men who have sex with men. J Acquir Immune
Defic Syndr. 2012;59:259–265.
73. Burgos J, Curran A, Landolfi S, et al. The effectiveness of elec-
trocautery ablation for the treatment of high-grade anal in-
traepithelial neoplasia in HIV-infected men who have sex with
men. HIV Med. 2016;17:524–531.
74. Miltz A, Price H, Shahmanesh M, Copas A, Gilson R. Systematic
review and meta-analysis of L1-VLP-based human papilloma-
virus vaccine efficacy against anogenital pre-cancer in women
with evidence of prior HPV exposure. PLoS One. 2014;9:e90348.
75. Dion GR, Teng S, Boyd LR, et al. Adjuvant human papillomavi-
rus vaccination for secondary prevention: a systematic review.
JAMA Otolaryngol Head Neck Surg. 2017;143:614–622.
76. Swedish KA, Factor SH, Goldstone SE. Prevention of recurrent
high-grade anal neoplasia with quadrivalent human papilloma-
virus vaccination of men who have sex with men: a nonconcur-
rent cohort study. Clin Infect Dis. 2012;54:891–898.
77. Tong WW, Jin F, McHugh LC, et al. Progression to and spontane-
ous regression of high-grade anal squamous intraepithelial lesions
in HIV-infected and uninfected men. AIDS. 2013;27:2233–2243.
78. Klas JV, Rothenberger DA, Wong WD, Madoff RD. Malignant
tumors of the anal canal: the spectrum of disease, treatment,
and outcomes. Cancer. 1999;85:1686–1693.
79. Robb BW, Mutch MG. Epidermoid carcinoma of the anal canal.
Clin Colon Rectal Surg. 2006;19:54–60.
80. Tanum G, Tveit K, Karlsen KO. Diagnosis of anal carcinoma:
doctor’s finger still the best? Oncology. 1991;48:383–386.
81. Santoso JT, Long M, Crigger M, Wan JY, Haefner HK. Anal in-
traepithelial neoplasia in women with genital intraepithelial
neoplasia. Obstet Gynecol. 2010;116:578–582.
82. Park IU, Ogilvie JW Jr, Anderson KE, et al. Anal human papillo-
mavirus infection and abnormal anal cytology in women with
genital neoplasia. Gynecol Oncol. 2009;114:399–403.
83. Ogilvie JW Jr, Park IU, Downs LS, Anderson KE, Hansberger J,
Madoff RD. Anal dysplasia in kidney transplant recipients. J Am
Coll Surg. 2008;207:914–921.
84. Welton ML, Sharkey FE, Kahlenberg MS. The etiology
and epidemiology of anal cancer. Surg Oncol Clin N Am.
2004;13:263–275.
85. Daling JR, Sherman KJ, Hislop TG, et al. Cigarette smoking and
the risk of anogenital cancer. Am J Epidemiol. 1992;135:180–189.
86. Chiao EY, Krown SE, Stier EA, Schrag D. A population-based
analysis of temporal trends in the incidence of squamous anal
canal cancer in relation to the HIV epidemic. J Acquir Immune
Defic Syndr. 2005;40:451–455.
87. Palefsky JM, Holly EA, Ralston ML, Jay N, Berry JM, Darragh
TM. High incidence of anal high-grade squamous intra-epithe-
lial lesions among HIV-positive and HIV-negative homosexual
and bisexual men. AIDS. 1998;12:495–503.
88. Anal canal. In: Greene FL, Page DL, Fleming ID, eds. AJCC
Cancer Staging Manual. 6th ed. New York, NY: Springer;
2002:125–130.
89. Bean SM, Eltoum I, Horton DK, Whitlow L, Chhieng DC. Im-
munohistochemical expression of p16 and Ki-67 correlates
with degree of anal intraepithelial neoplasia. Am J Surg Pathol.
2007;31:555–561.
90. Gordon PH. Current status: perianal and anal canal neoplasms.
Dis Colon Rectum. 1990;33:799–808.
91. Giovannini M, Bardou VJ, Barclay R, et al. Anal carcinoma:
prognostic value of endorectal ultrasound (ERUS)–results of
a prospective multicenter study. Endoscopy. 2001;33:231–236.
92. Jacopo M. Endoanal ultrasound for anal cancer staging. Int J
Colorectal Dis. 2011;26:385–386.
93. Otto SD, Lee L, Buhr HJ, Frericks B, Höcht S, Kroesen AJ.
Staging anal cancer: prospective comparison of transanal en-
doscopic ultrasound and magnetic resonance imaging. J Gas-
trointest Surg. 2009;13:1292–1298.
94. Boman BM, Moertel CG, O’Connell MJ, et al. Carcinoma of
the anal canal: a clinical and pathologic study of 188 cases.
Cancer. 1984;54:114–125.
95. Gaertner WB, Hagerman GF, Finne CO, et al. Fistula-associat-
ed anal adenocarcinoma: good results with aggressive therapy.
Dis Colon Rectum. 2008;51:1061–1067.
96. Grabenbauer GG, Kessler H, Matzel KE, Sauer R, Hohen-
berger W, Schneider IH. Tumor site predicts outcome after
radiochemotherapy in squamous-cell carcinoma of the anal
region: long-term results of 101 patients. Dis Colon Rectum.
2005;48:1742–1751.
97. Trautmann TG, Zuger JH. Positron emission tomography for
pretreatment staging and posttreatment evaluation in cancer
of the anal canal. Mol Imaging Biol. 2005;7:309–313.
98. Cotter SE, Grigsby PW, Siegel BA, et al. FDG-PET/CT in the
evaluation of anal carcinoma. Int J Radiat Oncol Biol Phys.
2006;65:720–725.
99. Krengli M, Milia ME, Turri L, et al. FDG-PET/CT imaging for
staging and target volume delineation in conformal radiother-
apy of anal carcinoma. Radiat Oncol. 2010;5:10.
100. Nguyen BT, Joon DL, Khoo V, et al. Assessing the impact of
FDG-PET in the management of anal cancer. Radiother Oncol.
2008;87:376–382.
101. Kidd EA, Dehdashti F, Siegel BA, Grigsby PW. Anal cancer
maximum F-18 fluorodeoxyglucose uptake on positron emis-
sion tomography is correlated with prognosis. Radiother On-
col. 2010;95:288–291.
102. Gauthé M, Richard-Molard M, Fayard J, Alberini JL, Cacheux
W, Lièvre A. Prognostic impact of tumour burden assessed by
metabolic tumour volume on FDG PET/CT in anal canal can-
cer. Eur J Nucl Med Mol Imaging. 2017;44:63–70.
103. Bazan JG, Koong AC, Kapp DS, et al. Metabolic tumor vol-
ume predicts disease progression and survival in patients
with squamous cell carcinoma of the anal canal. J Nucl Med.
2013;54:27–32.
104. Day FL, Link E, Ngan S, et al. FDG-PET metabolic response
predicts outcomes in anal cancer managed with chemoradio-
therapy. Br J Cancer. 2011;105:498–504.
105. Nigro ND, Vaitkevicius VK, Considine B Jr. Combined therapy
for cancer of the anal canal: a preliminary report. Dis Colon
Rectum. 1974;17:354–356.
106. Pintor MP, Northover JM, Nicholls RJ. Squamous cell carci-
noma of the anus at one hospital from 1948 to 1984. Br J Surg.
1989;76:806–810.
107. Beahrs OH, Wilson SM. Carcinoma of the anus. Ann Surg.
1976;184:422–428.
108. Dougherty BG, Evans HL. Carcinoma of the anal canal: a
study of 79 cases. Am J Clin Pathol. 1985;83:159–164.
109. Fuchshuber PR, Rodriguez-Bigas M, Weber T, Petrelli NJ.
Anal canal and perianal epidermoid cancers. J Am Coll Surg.
1997;185:494–505.
Copyright © The American Society of Colon & Rectal Surgeons, Inc. Unauthorized reproduction of this article is prohibited.
DISEASES OF THE COLON & RECTUM VOLUME 61: 7 (2018)
773
110. Buroker TR, Nigro N, Bradley G, et al. Combined therapy for
cancer of the anal canal: a follow-up report. Dis Colon Rectum.
1977;20:677–678.
111. Nigro ND. An evaluation of combined therapy for squamous
cell cancer of the anal canal. Dis Colon Rectum. 1984;27:763–766.
112. Ajani JA, Winter KA, Gunderson LL, et al. Fluorouracil, mi-
tomycin, and radiotherapy vs fluorouracil, cisplatin, and ra-
diotherapy for carcinoma of the anal canal: a randomized
controlled trial. JAMA. 2008;299:1914–1921.
113. Martenson JA, Lipsitz SR, Wagner H Jr, et al. Initial results of
a phase II trial of high dose radiation therapy, 5-fluorouracil,
and cisplatin for patients with anal cancer (E4292): an Eastern
Cooperative Oncology Group study. Int J Radiat Oncol Biol
Phys. 1996;35:745–749.
114. Meyer JJ, Willett CG, Czito BG. Emerging role of intensity-
modulated radiation therapy in anorectal cancer. Expert Rev
Anticancer Ther. 2008;8:585–593.
115. Meulendijks D, Dewit L, Tomasoa NB, et al. Chemoradiother-
apy with capecitabine for locally advanced anal carcinoma: an
alternative treatment option. Br J Cancer. 2014;111:1726–1733.
116. Thind G, Johal B, Follwell M, Kennecke HF. Chemoradiation
with capecitabine and mitomycin-C for stage I-III anal squa-
mous cell carcinoma. Radiat Oncol. 2014;9:124.
117. Papillon J, Mayer M, Montbarbon JF, Gerard JP, Chassard JL,
Bailly C. A new approach to the management of epidermoid
carcinoma of the anal canal. Cancer. 1983;51:1830–1837.
118. Papillon J, Montbarbon JF. Epidermoid carcinoma of the anal
canal: a series of 276 cases. Dis Colon Rectum. 1987;30:324–333.
119. UKCCCR Anal Cancer Trial Working Party; UK Co-ordinat-
ing Committee on Cancer Research. Epidermoid anal cancer:
results from the UKCCCR randomised trial of radiotherapy
alone versus radiotherapy, 5-fluorouracil, and mitomycin.
Lancet. 1996;348:1049–1054.
120. Northover J, Glynne-Jones R, Sebag-Montefiore D, et al.
Chemoradiation for the treatment of epidermoid anal cancer:
13-year follow-up of the first randomised UKCCCR Anal Can-
cer Trial (ACT I). Br J Cancer. 2010;102:1123–1128.
121. Bartelink H, Roelofsen F, Eschwege F, et al. Concomitant
radiotherapy and chemotherapy is superior to radiother-
apy alone in the treatment of locally advanced anal cancer:
results of a phase III randomized trial of the European Or-
ganization for Research and Treatment of Cancer Radiother-
apy and Gastrointestinal Cooperative Groups. J Clin Oncol.
1997;15:2040–2049.
122. Vinayan A, Glynne-Jones R. Anal cancer: what is the opti-
mum chemoradiotherapy? Best Pract Res Clin Gastroenterol.
2016;30:641–653.
123. Flam M, John M, Pajak TF, et al. Role of mitomycin in com-
bination with fluorouracil and radiotherapy, and of salvage
chemoradiation in the definitive nonsurgical treatment of epi-
dermoid carcinoma of the anal canal: results of a phase III ran-
domized intergroup study. J Clin Oncol. 1996;14:2527–2539.
124. James RD, Glynne-Jones R, Meadows HM, et al. Mitomycin or
cisplatin chemoradiation with or without maintenance che-
motherapy for treatment of squamous-cell carcinoma of the
anus (ACT II): a randomised, phase 3, open-label, 2 × 2 facto-
rial trial. Lancet Oncol. 2013;14:516–524.
125. Gunderson LL, Winter KA, Ajani JA, et al. Long-term update
of US GI intergroup RTOG 98-11 phase III trial for anal car-
cinoma: survival, relapse, and colostomy failure with concur-
rent chemoradiation involving fluorouracil/mitomycin versus
fluorouracil/cisplatin. J Clin Oncol. 2012;30:4344–4351.
126. Matzinger O, Roelofsen F, Mineur L, et al.; EORTC Radiation
Oncology and Gastrointestinal Tract Cancer Groups. Mitomy-
cin C with continuous fluorouracil or with cisplatin in com-
bination with radiotherapy for locally advanced anal cancer
(European Organisation for Research and Treatment of Cancer
phase II study 22011-40014). Eur J Cancer. 2009;45:2782–2791.
127. Peiffert D, Tournier-Rangeard L, Gérard JP, et al. Induction
chemotherapy and dose intensification of the radiation boost
in locally advanced anal canal carcinoma: final analysis of the
randomized UNICANCER ACCORD 03 trial. J Clin Oncol.
2012;30:1941–1948.
128. Moureau-Zabotto L, Viret F, Giovaninni M, et al. Is neoadju-
vant chemotherapy prior to radio-chemotherapy beneficial in
T4 anal carcinoma? J Surg Oncol. 2011;104:66–71.
129. Ortholan C, Ramaioli A, Peiffert D, et al. Anal canal carcino-
ma: early-stage tumors < or =10 mm (T1 or Tis): therapeutic
options and original pattern of local failure after radiotherapy.
Int J Radiat Oncol Biol Phys. 2005;62:479–485.
130. Ferrigno R, Nakamura RA, Dos Santos Novaes PE, et al.
Radiochemotherapy in the conservative treatment of anal
canal carcinoma: retrospective analysis of results and ra-
diation dose effectiveness. Int J Radiat Oncol Biol Phys.
2005;61:1136–1142.
131. John M, Pajak T, Flam M, et al. Dose escalation in chemora-
diation for anal cancer: preliminary results of RTOG 92-08.
Cancer J Sci Am. 1996;2:205–211.
132. Chakravarthy AB, Catalano PJ, Martenson JA, et al. Long-
term follow-up of a Phase II trial of high-dose radiation
with concurrent 5-fluorouracil and cisplatin in patients with
anal cancer (ECOG E4292). Int J Radiat Oncol Biol Phys.
2011;81:e607–e613.
133. Roohipour R, Patil S, Goodman KA, et al. Squamous-cell car-
cinoma of the anal canal: predictors of treatment outcome. Dis
Colon Rectum. 2008;51:147–153.
134. National Comprehensive Cancer Network. NCCN clini-
cal practice guidelines in oncology anal carcinoma, version
1.2018. https://www.nccn.org/professionals/physician_gls/re-
cently_updated.aspx. Accessed January 10, 2018.
135. Ciombor KK, Ernst RD, Brown G. Diagnosis and diag-
nostic imaging of anal canal cancer. Surg Oncol Clin N Am.
2017;26:45–55.
136. Abunassar M, Reinders J, Jonker DJ, Asmis T. Review of anal
cancer patients at the Ottawa hospital. Eur J Surg Oncol.
2015;41:653–658.
137. Teagle AR, Gilbert DC, Jones JR, Burkill GJ, McKinna F, Diz-
darevic S. Negative 18F-FDG-PET-CT may exclude resid-
ual or recurrent disease in anal cancer. Nucl Med Commun.
2016;37:1038–1045.
138. Vercellino L, Montravers F, de Parades V, et al. Impact of FDG
PET/CT in the staging and the follow-up of anal carcinoma.
Int J Colorectal Dis. 2011;26:201–210.
139. Das P, Bhatia S, Eng C, et al. Predictors and patterns of re-
currence after definitive chemoradiation for anal cancer. Int J
Radiat Oncol Biol Phys. 2007;68:794–800.
140. Ferenschild FT, Vermaas M, Hofer SO, Verhoef C, Eggermont
AM, de Wilt JH. Salvage abdominoperineal resection and peri-
neal wound healing in local recurrent or persistent anal cancer.
World J Surg. 2005;29:1452–1457.
Copyright © The American Society of Colon & Rectal Surgeons, Inc. Unauthorized reproduction of this article is prohibited.
STEWART ET AL: ANAL CANCER CLINICAL PRACTICE GUIDELINES
774
141. Papaconstantinou HT, Bullard KM, Rothenberger DA, Madoff
RD. Salvage abdominoperineal resection after failed Nigro
protocol: modest success, major morbidity. Colorectal Dis.
2006;8:124–129.
142. Stewart D, Yan Y, Kodner IJ, et al. Salvage surgery after failed
chemoradiation for anal canal cancer: should the para-
digm be changed for high-risk tumors? J Gastrointest Surg.
2007;11:1744–1751.
143. Severino NP, Chadi SA, Rosen L, et al. Survival following sal-
vage abdominoperineal resection for persistent and recurrent
squamous cell carcinoma of the anus: do these disease catego-
ries affect survival? Colorectal Dis. 2016;18:959–966.
144. Lefèvre JH, Corte H, Tiret E, et al. Abdominoperineal resection
for squamous cell anal carcinoma: survival and risk factors for
recurrence. Ann Surg Oncol. 2012;19:4186–4192.
145. Schiller DE, Cummings BJ, Rai S, et al. Outcomes of salvage
surgery for squamous cell carcinoma of the anal canal. Ann
Surg Oncol. 2007;14:2780–2789.
146. Mullen JT, Rodriguez-Bigas MA, Chang GJ, et al. Results
of surgical salvage after failed chemoradiation therapy for
epidermoid carcinoma of the anal canal. Ann Surg Oncol.
2007;14:478–483.
147. Goéré D, Bonnet S, Pocard M, Deutsch E, Lasser P, Elias D. On-
cologic and functional results after abdominoperineal resec-
tion plus pseudocontinent perineal colostomy for epidermoid
carcinoma of the anus. Dis Colon Rectum. 2009;52:958–963.
148. Lefevre JH, Parc Y, Kernéis S, et al. Abdomino-perineal resec-
tion for anal cancer: impact of a vertical rectus abdominis
myocutaneus flap on survival, recurrence, morbidity, and
wound healing. Ann Surg. 2009;250:707–711.
149. Vorob’ev GI, Shelygin IuA, Nechushkin MI, Rybakov EG. Re-
sults of surgical treatment of residual and recurrent anal tu-
mors [in Russian]. Khirurgiia (Mosk). 2008;8:4–9.
150. Mariani P, Ghanneme A, De la Rochefordière A, Girodet J,
Falcou MC, Salmon RJ. Abdominoperineal resection for anal
cancer. Dis Colon Rectum. 2008;51:1495–1501.
151. Hardt J, Mai S, Weiß C, Kienle P, Magdeburg J. Abdominoperineal
resection and perineal wound healing in recurrent, persistent, or
primary anal carcinoma. Int J Colorectal Dis. 2016;31:1197–1203.
152. Sunesen KG, Buntzen S, Tei T, Lindegaard JC, Nørgaard M,
Laurberg S. Perineal healing and survival after anal cancer sal-
vage surgery: 10-year experience with primary perineal recon-
struction using the vertical rectus abdominis myocutaneous
(VRAM) flap. Ann Surg Oncol. 2009;16:68–77.
153. Sheckter CC, Shakir A, Vo H, Tsai J, Nazerali R, Lee GK. Re-
construction following abdominoperineal resection (APR):
indications and complications from a single institution expe-
rience. J Plast Reconstr Aesthet Surg. 2016;69:1506–1512.
154. Hoffman R, Welton ML, Klencke B, Weinberg V, Krieg R. The
significance of pretreatment CD4 count on the outcome and
treatment tolerance of HIV-positive patients with anal cancer.
Int J Radiat Oncol Biol Phys. 1999;44:127–131.
155. Fraunholz I, Weiss C, Eberlein K, Haberl A, Rödel C. Concur-
rent chemoradiotherapy with 5-fluorouracil and mitomycin
C for invasive anal carcinoma in human immunodeficiency
virus-positive patients receiving highly active antiretroviral
therapy. Int J Radiat Oncol Biol Phys. 2010;76:1425–1432.
156. Fraunholz IB, Haberl A, Klauke S, Gute P, Rödel CM. Long-
term effects of chemoradiotherapy for anal cancer in patients
with HIV infection: oncological outcomes, immunological
status, and the clinical course of the HIV disease. Dis Colon
Rectum. 2014;57:423–431.
157. Chiao EY, Giordano TP, Richardson P, El-Serag HB. Human
immunodeficiency virus-associated squamous cell cancer of
the anus: epidemiology and outcomes in the highly active an-
tiretroviral therapy era. J Clin Oncol. 2008;26:474–479.
158. Schraut WH, Wang CH, Dawson PJ, Block GE. Depth of inva-
sion, location, and size of cancer of the anus dictate operative
treatment. Cancer. 1983;51:1291–1296.
159. Jensen SL, Hagen K, Harling H, Shokouh-Amiri MH, Nielsen
OV. Long-term prognosis after radical treatment for squa-
mous-cell carcinoma of the anal canal and anal margin. Dis
Colon Rectum. 1988;31:273–278.
160. Evesque L, Benezery K, Follana P, et al. Multimodal therapy
of squamous cell carcinoma of the anus with distant me-
tastasis: a single-institution experience. Dis Colon Rectum.
2017;60:785–791.
161. Pathak P, King BT, Ohinata A, et al. The treatment of meta-
static squamous cell carcinoma (SCCA) of the anal canal: a
single institution experience [abstract]. Gastroenterol Hepatol
(N Y). 2008;4:.
162. Eng C, Pathak P. Treatment options in metastatic squamous
cell carcinoma of the anal canal. Curr Treat Options Oncol.
2008;9:400–407.
163. Faivre C, Rougier P, Ducreux M, et al. 5-fluorouracile
and cisplatinum combination chemotherapy for meta-
static squamous-cell anal cancer [in French]. Bull Cancer.
1999;86:861–865.
164. Kim R, Byer J, Fulp WJ, Mahipal A, Dinwoodie W, Shibata D.
Carboplatin and paclitaxel treatment is effective in advanced
anal cancer. Oncology. 2014;87:125–132.
165. Paliga A, Onerheim R, Gologan A, et al. EGFR and K-ras
gene mutation status in squamous cell anal carcinoma: a role
for concurrent radiation and EGFR inhibitors? Br J Cancer.
2012;107:1864–1868.
166. Deutsch E, Lemanski C, Pignon JP, et al. Unexpected toxic-
ity of cetuximab combined with conventional chemoradio-
therapy in patients with locally advanced anal cancer: results
of the UNICANCER ACCORD 16 phase II trial. Ann Oncol.
2013;24:2834–2838.
167. Olivatto LO, Vieira FM, Pereira BV, et al. Phase 1 study of ce-
tuximab in combination with 5-fluorouracil, cisplatin, and
radiotherapy in patients with locally advanced anal canal car-
cinoma. Cancer. 2013;119:2973–2980.