SlideShare una empresa de Scribd logo
1 de 14
Descargar para leer sin conexión
Clinical Expert Series 
Chronic Pelvic Pain 
John F. Steege, MD, and Matthew T. Siedhoff, MD, MSCR 
As opposed to the satisfying solutions found in the management of acute pain, chronic pelvic 
pain can be a vexing problem for the patient and physician. Seldom is a single source or cause 
found, and nearly always the condition is influenced by the broader social and psychological 
context of the patient. In this article, we discuss the evaluation of chronic pelvic pain, often 
considering pain as the disease itself, and identify peripheral generators, which gynecologists 
can address to help reduce their contributions to symptoms. 
(Obstet Gynecol 2014;124:616–29) 
DOI: 10.1097/AOG.0000000000000417 
The American College of Obstetricians and Gyne-cologists 
proposed the following definition of 
chronic pelvic pain: noncyclic pain of 6 or more 
months’ duration that localizes to the anatomic pelvis, 
anterior abdominal wall at or below the umbilicus, the 
lumbosacral back or the buttocks, and is of sufficient 
severity to cause functional disability or lead to med-ical 
care.1 The problem costs the economy of the 
United States well in excess of $3 billion2 and costs 
women greatly in the areas of work, family responsi-bilities, 
and relationships. The practicing gynecologist 
finds the problem time-consuming, challenging, and 
less rewarding than the usually successful obstetric 
and gynecologic endeavors. 
Traditional thinking about chronic pelvic pain has 
emphasized observable organic pathology (eg, endo-metriosis, 
adhesions), but the connection between these 
problems and pain symptoms is actually more tenuous 
than previously thought. For example, even the most 
optimistic clinical reports note pain relief in only 60– 
70% of women undergoing laparoscopic surgery for 
endometriosis,3,4 suggesting that in many women, there 
is more to the story than just the implants. More and 
more data confirming the coexistence of one or more 
other chronic pain disorders in patients—conditions such 
as interstitial cystitis (also known as painful bladder syn-drome), 
irritable bowel syndrome, temporomandibular 
joint disorder, migraine headaches, vulvodynia, and fi-bromyalgia— 
suggest that perhaps we should be treating 
pelvic pain itself as the disease rather than as just a man-ifestation 
of a specific pathologic change.5 In this review, 
we describe chronic pelvic pain and point out common 
peripheral pain generators—nociceptive stimuli—that can 
be modulated by gynecologic interventions. 
HISTORY OF PELVIC PAIN CONCEPTS 
Over the past 60 years, the study of chronic pelvic 
pain has gone through significant changes in 
approach. Before the advent of laparoscopy and based 
in a Cartesian framework in which pain perception 
should be proportional to the degree of tissue damage, 
gynecologists were understandably reluctant to oper-ate 
on any pathology not large enough to palpate. In 
addition, much of clinical medicine was practiced 
from the perspective of the mind–body split: causa-tion 
of symptoms was distinctly divided between 
physical and psychological sources. Although this 
model was sufficient to address most causes of acute 
pain, it fails to interpret the majority of chronic pain 
disorders in gynecology as well as other areas of med-icine. 
The gate-control theory, an alternative pro-posed 
by Melzack and Wall in 1965, suggests that 
pain information flows in two directions: 1) nocicep-tive 
signals from peripheral tissue ascend through the 
spinal cord to higher centers; and 2) central centers 
can modulate, using descending signals altering spinal 
From the Department of Obstetrics and Gynecology, University of North 
Carolina at Chapel Hill, Chapel Hill, North Carolina. 
Continuing medical education for this article is available at http://links.lww. 
com/AOG/A541. 
Corresponding author: John F. Steege, MD, Department of Obstetrics and 
Gynecology, School of Medicine, University of North Carolina at Chapel Hill, 
Chapel Hill, NC; e-mail: jfsteege@med.unc.edu. 
Financial Disclosure 
The authors did not report any potential conflicts of interest. 
© 2014 by The American College of Obstetricians and Gynecologists. Published 
by Lippincott Williams & Wilkins. 
ISSN: 0029-7844/14 
616 VOL. 124, NO. 3, SEPTEMBER 2014 OBSTETRICS & GYNECOLOGY
cord neurotransmitter and interneuron activity, the 
transmission of these nociceptive signals from the 
periphery. Deterioration of these regulatory processes 
were thought to potentially account for development 
of chronic pain states by allowing too many peripheral 
signals to pass through the spinal cord “gates.” Vari-ation 
in patients’ relative degree of gate opening could 
thus explain why similar amounts of physical tissue 
damage result in different degrees of pain perception.6 
While these theory changes were stimulating the 
field of pain research, gynecologists were busy devel-oping 
laparoscopy and with it the hope that treating 
visible pathology could fix chronic pelvic pain. A 
focus on “laparoscopy-negative” patients in chronic 
pelvic pain clinics emerged, implying that if some 
pathology were found, it must be a “real” cause for 
pain. Subsequent experience has shown that although 
treatment of laparoscopically diagnosed pathology 
can be helpful, the clinical reality is more complex: 
1) in many instances, visible pathology found at lapa-roscopy 
may be incidental and not related to the pain; 
2) in those with pathology that does contribute to 
nociception, the pain experienced by the patient 
may differ from another patient with anatomically 
similar pathology; and 3) pain from a laparoscopic 
finding is often best understood in the larger context 
of a centralized pain disorder. 
Research of the 1980s added the observation of 
distressingly high rates of physical and sexual abuse, 
especially in the chronic pelvic pain population. These 
observations led to the speculation that the experience 
of abuse may make a person more vulnerable to the 
development of chronic pelvic pain or perhaps be 
a specific cause for pain. In relation to pain, abuse, 
particularly that which occurs in formative years, may 
serve to alter the response to nociception and central 
pain processing. That said, not all abused patients go 
on to have chronic pain nor do all patients with pain 
have a history of abuse, so it might be the response to 
trauma that plays a key role in development of chronic 
pain. Health care providers need to take into account 
the presence of abuse in a patient’s history when de-tected 
but be careful to avoid necessarily concluding 
a causal relationship in that patient’s pain. 
Melzack’s neuromatrix theory7 added the pivotal 
notion of neuroplasticity, which suggests that experi-ence 
can change the neurophysiologic behavior of the 
central nervous system in a manner that influences the 
subsequent processing of nociceptive stimuli. It may 
explain the apparent development of pain responses 
to stimuli usually thought of as nonpainful (allodynia) 
as well as exaggerated responses to painful stimuli 
(hyperalgesia). Every practicing gynecologist has seen 
patients whose pain responses seem out of proportion 
to the pathology found. This may reflect the emo-tional 
meaning of the problem for the patient as well 
as past or present trauma, but it may also be the result 
of sensitization of spinal cord interneurons that have 
become pain amplifiers as a result of being on the 
receiving end of peripheral nociceptive stimuli for pro-longed 
periods. On the positive side, neuroplasticity 
suggests that given enough time and the right treat-ment, 
even seemingly intractable chronic pain prob-lems 
can improve substantially. 
The concept of central sensitization adds the 
most recent layer to theoretical understanding of 
chronic pain: the notion that multiple repetitions of 
lower-level stimuli may over time result in a more 
severe central perception of pain. This centralized 
pain hypersensitivity helps us understand how mul-tiple 
organ systems can be recruited into the syn-drome, 
incorporating genetic and social factors in 
pain amplification. Whatever the nociceptive stimulus 
(bowel, bladder, muscle, uterus), it can register as pain 
at a lower threshold in the patient with a centralized 
pain disorder (Box 1). 
Box 1. Definitions of Pain Terms 
Allodynia—pain resulting from a nonnoxious stimulus 
Hyperalgesia—painful sensation of abnormal severity 
after noxious stimulation 
Neuropathic pain—pain persisting after healing of 
disease or trauma-induced tissue damage 
Neuroplasticity—the malleability of central pain perception 
mechanisms in response to chronic pain states 
Nociceptor—a nerve receptor for pain 
This understanding contributes to the framework 
a clinician needs when taking a clinic history of pelvic 
pain and performing the physical examination. The 
inquiry should set out in pursuit of all the factors 
relevant to a person’s pain, not simply trying to identify 
one factor with an acceptably plausible relationship to 
her complaints. Once the patient is aware that the cli-nician 
is keeping an open mind about multiple contrib-uting 
factors, she may become less defensive about any 
inquiries in emotional areas. 
EVALUATION OF PATIENTS WITH CHRONIC 
PELVIC PAIN 
History-Taking 
The site, duration, pattern during activities, relation to 
position changes, and association with bodily func-tions 
are all important elements of pain. For example, 
pain that is absent in the morning but worsens 
VOL. 124, NO. 3, SEPTEMBER 2014 Steege and Siedhoff Chronic Pelvic Pain 617
progressively during the day may be associated with 
pelvic floor muscle dysfunction, whereas a tender 
“spot” of dyspareunia might be related to nodular cul 
de sac endometriosis. A previous review supplies a very 
detailed description of both history-taking and physical 
examination techniques that are useful in the evalua-tion 
of chronic pelvic pain.8 We focus here on the most 
frequently useful elements of this process. 
The chronology of a patient’s pain is critical. As a 
pain syndrome develops, pain can be present over a 
progressively larger area despite stable detectable 
pathology. Interpreting this as the breakdown or wear-ing 
out of physiologic systems that deal with pain sig-nals 
has some biologic validity and may make sense to 
the patient. The clinician may need to counter the idea 
patients sometimes have that endometriosis “flares” 
like rheumatoid disease or spreads like a malignancy. 
From a cognitive perspective, it is invaluable to 
discern the patient’s and her family’s ideas about the 
causes of and future for her pain. Fears of cancer can 
be discovered even if this diagnosis was never even 
remotely considered by the clinician. Less dramatic 
but equally powerful attributions of cause can emerge 
such as pelvic infection that is the result of sexual acts 
remote in time, arguments with a spouse, divine ret-ribution, 
and so forth. 
When a patient has been under that care of 
previous health care providers, it is important to 
review the effects of previous treatments and the 
evolution of how the patient has thought about her 
own disease. For example, when a prior treatment 
for mild endometriosis was only briefly successful, 
did she (and her health care provider?) see this as 
evidence for increasing severity of disease or did it 
make them question the diagnosis in the first place? 
In a referral-based practice, it is a frequent event to 
deal with a patient with mild disease who has not 
responded to any of four or five medical or surgical 
measures that would ordinarily improve symptoms. 
She then sees herself as having increasing and 
invasive disease, prompting even more aggressive 
treatment, rather than wondering if something else is 
contributing to her pain. 
Physical Examination 
The physical examination begins with observing the 
patient’s gait, comfort with sitting, ease of going from 
sitting to standing, and from sitting on the table to 
lying down. Dysfunctions of the pelvic floor and hip 
muscles may be manifested by discomfort with these 
motions. For example, if the patient sits up on one 
buttock during the interview, one might suspect pelvic 
floor muscle pain. 
With the patient sitting on the examination table, 
it can be helpful to begin the examination with the 
back, evaluating for tenderness of the spine, para-spinous 
muscles, and sacroiliac joints. This may 
identify pain generators and sites for therapeutic 
intervention, but it also allows for touch to begin in 
a very nonthreatening way. Gynecologic assessment is 
uncomfortable to some degree for almost all women, 
but the patient with chronic pelvic pain is particularly 
vulnerable. Moving from back to abdomen to pelvis 
can establish trust and reduce fear. 
The patient is then asked to recline on the table, 
observing how comfortably she can do this. The 
abdominal wall is examined with and without flexed 
rectus muscles. A positive Carnett sign (increased 
tenderness when palpation is done in the presence of 
abdominal wall flexion) implies at least a contribution 
to the pain from abdominal wall myofascial sources. 
Decreased pain during this maneuver implies a higher 
contribution from visceral sources. On occasion, gentle 
fingertip palpation of the abdominal wall can detect 
such trigger points in the musculature. Rarely, a sub-cutaneous 
abdominal wall endometrioma is discov-ered, 
a diagnosis supported by a history of predictable, 
cyclic, focal tenderness. An abdominal wall endome-trioma, 
or a hernia, is often more readily appreciated 
by examining the patient in the standing position. 
Pelvic examination then begins with external 
review of the vulva and vestibule. Gentle palpation 
with a cotton swab can detect areas of sensitivity 
consistent with vulvar vestibular syndrome in the 
introitus or trigger points higher in the vagina. 
Intrinsic cervical allodynia (from cervical conization 
or after obstetric cervical laceration) may be detected 
in the same manner with gentle cotton-tipped appli-cator 
palpation. 
Guiding a patient through contraction–relaxation 
sequences of the abdominal, thigh, and vaginal introi-tal 
muscles can reduce the discomfort of the examina-tion 
and can indicate the patient’s degree of control 
over muscle tension. Single-digit palpation of the leva-tor 
plate, piriformis, and obturator muscles can elicit 
the tenderness of pelvic floor tension myalgia. This 
condition can present as a sequel to some other pelvic 
pain or a problem in itself. Discomfort is usually felt as 
pelvic pressure and radiation pain to the sacrum, near 
the insertions of the levator plate muscles. The exact 
sequence of the examination should be adjusted in 
view of the information obtained during the history: 
examine the areas less likely to be tender first, saving 
the more tender areas for the end. Palpating the most 
tender areas first can elevate anxiety and distort pain 
responses from subsequent areas. 
618 Steege and Siedhoff Chronic Pelvic Pain OBSTETRICS & GYNECOLOGY
Single-digit palpation should also be used to 
discover areas of tenderness in the cervix, uterus, 
adnexa, bladder, and urethra. Premature addition of 
the abdominal hand to the examination adds in 
nociceptive signals from abdominal wall myofascial 
components that may lead the examiner to over-attribute 
pain to the viscera. Finally, the abdominal 
hand is added to assess size, shape, and mobility of 
pelvic structures. Adnexal thickening and mobility, 
pelvic relaxation, coccygeal tenderness, and foci of 
pain that reproduce dyspareunia should be noted. A 
rectovaginal examination is important when deep 
infiltrating endometriosis is suspected. 
During all components of the physical examina-tion, 
it is important to not only ask the question “Does 
this hurt?,” but also “Do you feel pain where I am 
pressing or somewhere else?” and “Is this the pain 
you were describing earlier—is this your pain?” If 
a patient answers affirmatively to the final question, 
it can be helpful to point out what structure you are 
palpating (eg, pelvic floor muscle compared with 
ovary). As you are going along through the examina-tion, 
it can be useful to mention the items you are 
adding to the list of possible contributing factors to 
her pain, thus reinforcing what you discussed during 
the history in terms of pain being multifactorial. 
Having concluded the most important part of the 
evaluation, the history and physical, you are now 
prepared to discuss diagnostic possibilities and a treat-ment 
plan. What follows is a review of the commonly 
described contributors to pelvic pain, which might be 
part of that discussion. 
CONTRIBUTIONS OF PERIPHERAL 
PAIN GENERATORS 
This section deserves an important caveat. Although we 
believe that types of tissue damage or other nociceptive 
input can generate pain, they cannot be viewed in 
isolation of the patient’s individual central pain process-ing. 
Management strategies are discussed in more 
detail, but, in general, the goal of the treating health 
care provider involves trying to dampen overall pain 
signaling sensitivity—“turning down the master vol-ume”— 
and looking for areas in the periphery that can 
be “tuned up” toward better functioning. This effort is 
more likely to be successful in the patient who takes an 
active role in her own improvement and avoids seeing 
herself as helpless in the face of her discomforts.9 
The following peripheral generators represent areas 
where we can intervene but should not be described to 
patients with chronic pelvic pain as the only cause of 
their pain. Because these conditions can be completely 
asymptomatic in many patients, their importance needs 
interpretation in the context of her overall health, taking 
into account her other pain disorders and general 
capacity for dealing with life’s challenges. 
Endometriosis 
Evidence that endometriosis causes pain results from 
observations that the disease is more commonly 
encountered in women undergoing laparoscopy for 
pain than for other reasons10 and that laparoscopic 
treatment results in better pain reduction than diagnos-tic 
surgery alone.11–13 However, amount of disease does 
not correlate with symptom severity14,15; endometriosis 
is a frequently encountered incidental finding, and peri-toneal 
implants do not localize to symptom location.16 
A variety of proposed mechanisms explaining pain 
associated with endometriosis exist, including inflam-matory, 
nociceptive, and neuropathic.17 There is no 
pathognomonic symptom associated with endometri-osis, 
however. Many of the symptoms often attributed 
to endometriosis (eg, dyspareunia, dysmenorrhea, 
abnormal bowel or bladder function) are commonly 
found in functional disorders such as irritable bowel 
syndrome or interstitial cystitis, making it difficult to 
understand the relevant contribution of endometriosis 
to chronic pelvic pain. Deeply infiltrating endometri-osis— 
fibrotic, vascular, desmoplastic tissue destruction— 
is a biologically different disease, in which detectable 
physical examination or imaging findings relate to spe-cific 
symptoms such as tender rectovaginal nodularity 
or ovarian endometrioma with dyspareunia or dysche-zia 
with intrinsic rectal involvement.18–20 The relation-ship 
of a related condition, adenomyosis, to pelvic pain 
is less established, but the disease should be considered, 
especially when symptoms include dysmenorrhea or 
heavy bleeding.21 
Pelvic Adhesions 
Early reviews supported the role of adhesions as 
a significant contributor in chronic pelvic pain.10,22 
More recent investigations23 demonstrate a relatively 
weak correlation between adhesions and chronic 
pelvic pain, much less than other factors such as psy-chosomatic 
symptoms and substance abuse. Few, if any, 
well-designed studies demonstrate effective treatment of 
chronic pelvic pain with adhesiolysis. Unfortunately, in 
an effort to provide some explanation for complex pain 
disorders, health care providers often still posit adhe-sions 
as an etiology, even when a patient’s surgical his-tory 
includes only laparoscopy with findings of minimal 
or no endometriosis, pelvic inflammatory disease, or 
other conditions associated with meaningful adhesions. 
This explanation can happen even when the patient’s 
pain escalation is remote from her last surgery. 
VOL. 124, NO. 3, SEPTEMBER 2014 Steege and Siedhoff Chronic Pelvic Pain 619
Adhesions may play some role in pain conditions in 
some women, but the relative contribution is probably 
small. Also, the putative treatment—repeat surgical 
intervention—can add new contributions to pain syn-dromes 
such as the effect of surgical trauma, disappoint-ment 
from lack of pain relief, feeding the psychological 
need of being “ill” with more surgery, and, in the worst 
case, generating a complication such as enterotomy. 
Pelvic Support 
Most women in pain clinics are in their third or fourth 
decade of life, whereas pelvic organ prolapse affects 
significantly older women, suggesting a very minimal 
role for support problems in chronic pelvic pain. 
Pelvic relaxation usually leads to reports of heaviness, 
pressure, dropping sensations, or aching. In attempt-ing 
to hold in prolapsing organs, the patient may tense 
the levator plate, leading to tenderness during daily 
activities and intercourse. Fear of (or actual) loss of 
urinary control during coitus can add to the discom-fort 
by impairing physiologic sexual response. 
The retroverted uterus is another potential contrib-utor 
to chronic pelvic pain, particularly in the form of 
deep dyspareunia. Clearly for many women, retrover-sion 
is an innocent anatomic variant, but for those with 
pain, uncontrolled clinical series of uterine suspension 
procedures suggest changing the position of the uterus 
to an axial or anteverted position can improve dyspar-eunia 
by elevating the fundus out of the posterior cul de 
sac24–26 and allowing for better vaginal expansion as 
a natural part of the sexual response cycle.27 
Pelvic Congestion 
Overfilling (congestion) of the pelvic venous system 
has been implicated as a cause of dull chronic aching 
pain that usually is bilateral, worse at the end of the 
day after prolonged standing, premenstrually, and 
postcoitally. Some studies suggest the condition is 
present in nearly one-third of women with chronic 
pelvic pain,28 but there is no agreed-on reference stan-dard 
for diagnosis despite individual technical regi-mens 
involving venography, magnetic resonance, 
and ultrasonography.29 Hormonal suppression,30,31 
percutaneous embolotherapy, and surgery (vein liga-tion, 
hysterectomy and salpingo-oophorectomy) rep-resent 
available treatments,32 but study protocols 
involving these interventions are diverse and few have 
been investigated in controlled trials.33 
Residual Ovary 
When the uterus has been removed, with or without 
removal of one ovary, the remaining ovary or ovaries 
become symptomatic in a small percentage of women.34 
Pain from the ovary can be increased by confinement 
within postoperative adhesions, rupture or leakage of 
a cyst prompting additional adhesion formation, or 
attachment of the ovary to the sigmoid colon or vaginal 
apex by postoperative adhesions. In the case of attach-ment 
to the vaginal apex, deep dyspareunia can result 
when the area is struck. 
Ovarian Remnant 
A more difficult situation can develop if a small 
fragment of ovarian tissue is left behind during 
attempted oophorectomy.35 In most instances, this 
happens when challenging dissection is required, such 
as cases of extensive pelvic adhesions or deeply infil-trating 
endometriosis. Within 1–3 years of the attemp-ted 
oophorectomy, continued follicle-stimulating 
hormone stimulation will result in growth of the ovar-ian 
fragment, often producing an intermittently symp-tomatic 
pelvic mass located along the course of the 
ovarian vascular supply. A postulated mechanism for 
pain generation includes the cystic enlargement of the 
mass confined within fibrotic adhesions. If the rem-nant 
developed because endometriosis made for diffi-cult 
oophorectomy, that disease is often found in the 
remnant and probably also serves as a pain generator. 
Classic remnant symptoms include absence of vaso-motor 
symptoms after ostensible bilateral oophorec-tomy 
and the presence of cyclic unilateral pain. Like 
in the case of the residual ovary, the remnant can 
produce dyspareunia if it is located close to the vaginal 
apex. When performing oophorectomy, it is best to 
open the pararectal space and completely skeletonize 
the infundibulopelvic ligament not only to avoid com-plications 
such as ureteral injury, but also to prevent 
ovarian remnant syndrome. In difficult cases, dividing 
the pedicle at or above the pelvic brim, like in risk-reduction 
prophylactic oophorectomy, is prudent. 
Vaginal Apex Pain 
After hysterectomy, pain may persist or recur because 
of intrinsic sensitivity of the vaginal apex. Although the 
cuff may appear to have healed perfectly well, gentle 
examination with a cotton-tipped applicator may reveal 
focal sensitivity of a moderate to severe degree, many 
times located in one lateral fornix or the other and often 
replicating the reported pain of dyspareunia. When the 
cotton applicator examination is not done, the unaware 
examiner may then, noting pain on traditional biman-ual 
examination, mistakenly conclude that the source of 
nociception lies cephalad, for example, in a remaining 
ovary, pelvic scarring, or bowel adhesions. 
The diagnosis may be confirmed by noting 
elimination of the pain after injection of a local 
620 Steege and Siedhoff Chronic Pelvic Pain OBSTETRICS & GYNECOLOGY
anesthetic. The condition is generally considered 
neuropathic by virtue of the character of the pain 
(burning, stinging, sharp) and that neuropathic treat-ments 
(overnight application of lidocaine, oral medi-cations 
such as nortriptyline, amitriptyline, gabapentin, 
etc) seem to benefit some patients. Laparoscopic 
revision of the vaginal cuff may give good initial relief 
in approximately two-thirds of patients, but pain tends 
to recur to a degree over the subsequent 2–3 years, 
although perhaps at a less intense level.36,37 
Musculoskeletal Problems 
Musculoskeletal changes can become involved with 
chronic pelvic pain, either as the primary problem or 
as a secondary reaction to the pelvic pain. The 
muscular problem that most often produces pelvic 
pain is pelvic floor tension myalgia (also called levator 
spasm or levator ani syndrome).38 The clinical symp-tom 
profile commonly includes pain with sitting flat 
on a chair, worsening pelvic pressure over the course of 
the day, and midvaginal dyspareunia. Intermittent or 
constant painful contraction of the levator plate can be 
present as a primary problem or a reaction to some 
other source of pain. The patient with levator pain will 
often sit up on one buttock during the interview and will 
report that pelvic floor soreness persisted for 1–2 days 
after palpation of the muscles during pelvic examination. 
The piriformis and obturator muscles warrant 
further emphasis because they are seldom appreciated 
as possible sources of pain. These muscles are external 
rotators of the leg, and rotation against resistance can 
allow detection of tender spasm of the muscles during 
the pelvic examination. The sciatic nerve can traverse 
the belly of the piriformis as a normal anatomic variant, 
producing symptoms similar to sciatica when the muscle 
is in spasm. Pain caused by the iliopsoas complex can 
manifest as anterior hip or groin pain and is diagnosed 
with passive or active extension at the hip. 
The mainstay of treatment of muscular compo-nents 
of pelvic pain is physical therapy, most com-monly 
performed by a therapist with specialized 
training in the treatment of female pelvic pain. This 
subspecialty of the field has grown enormously over 
the past 15 years in obvious response to growing 
recognition of muscular elements to pelvic pain and in 
recognition that it works.39 Most physical therapists 
specializing in women’s care will address pelvic floor 
and hip muscles issues with transvaginal muscle treat-ments 
as well as other treatment measures. Other 
measures that are sometimes helpful are self-directed 
pelvic floor contraction and relaxation exercises, mus-cle 
relaxants, vaginal valium, and Botox injections. 
When the problem is severe enough to require these 
measures, it is generally advised that they be used 
along with simultaneous physical therapy. 
Medical Comorbidity 
Peripheral pain generation in chronic pelvic pain 
often involves nongynecologic systems.40,41 A careful 
history and close physical examination of gastrointes-tinal, 
urologic, musculoskeletal, and neurologic sys-tems 
are needed to evaluate these additional 
contributions to chronic pelvic pain. Most of the avail-able 
literature examines these problems indepen-dently 
of each other and without reference to their 
relevance to chronic pelvic pain or to the overall prev-alence 
of these disorders in chronic pelvic pain. The 
gastrointestinal system is the most common nongyne-cologic 
contributor to chronic pelvic pain, typically 
manifesting as irritable bowel syndrome. Perhaps sec-ond 
in terms of prevalence, functional urinary prob-lems 
(ie, interstitial cystitis) are another common 
comorbid contributor in chronic pelvic pain. These 
as well as others such as migraine, temporomandibu-lar 
joint disorder, and fibromyalgia should be viewed 
in the context of a larger centralized pain problem and 
treatment should be symptom-specific. 
Psychological comorbidity also travels with 
chronic pelvic pain. Depression, anxiety, anger–hos-tility, 
somatization, and catastrophization9 (the belief 
that things are as bad as they can be and are not likely 
to improve) are all more common among women with 
chronic pain than in women in a control group.42,43 
Whether mood disorder is a predisposing factor to, or 
a result of, chronic pain is not clear and may differ 
from patient to patient, but the situation is symbiotic 
at a minimum. Many women with chronic pelvic pain 
fear physicians’ recognition of mood disorder leads to 
the conclusion that their pain problem is not “real” or 
“in her head.” Thus, from a treatment perspective, it 
makes sense to attend to each disorder to the degree it 
manifests, but not necessarily attempt to discern cau-sality 
in a gynecologic visit. 
Studies vary in describing the prevalence of sexual 
abuse,44,45 but regardless, it is difficult to judge whether 
these events are directly relevant to present pain and 
hence demand attention or whether they contribute to 
a psychologically vulnerable substrate influenced by 
subsequent physical and emotional events. In these 
circumstances, it may be worthwhile to suggest further 
mental health evaluation as an exploratory measure, 
being careful not to imply that the patient is being 
referred because the physician is certain that the abuse 
is related to the development of the pain. 
Lastly, marital distress and sexual dysfunction, 
particularly dyspareunia, are common further 
VOL. 124, NO. 3, SEPTEMBER 2014 Steege and Siedhoff Chronic Pelvic Pain 621
burdens for patients with pain. Although some 
women report satisfactory sexual functioning before 
the onset of pain symptoms, others appear to have 
long-standing impairments in sexual response. In our 
experience, sexual difficulties are often the problem 
that makes a person seek (or is encouraged by her 
partner to seek) help for her pain. 
DIAGNOSTIC STRATEGIES 
Recognizing a Chronic Pain Syndrome 
Many women can experience pain for longer than 6 
months without becoming debilitated. Although their 
pain is chronic, such women are not described as 
having a chronic pain syndrome. The following are the 
common clinical hallmarks of this syndrome46: 1) dura-tion 
of 6 months or longer, 2) incomplete relief by most 
previous treatments, 3) significantly impaired physical 
function at home or at work, 4) signs of depression 
(sleep disturbance, weight loss, loss of appetite), 5) 
hypersensitive response to nociceptive stimuli, and 6) 
altered family roles. 
Of the signs of depression, sleep disturbance is 
usually the first to appear. Careful questioning is 
needed to distinguish awakening caused by pain from 
awakening that just happens. In the true vegetative 
sign, the person usually cannot get back to sleep even 
if pain is relieved (by medication or other means). 
The alteration of family roles is perhaps the most 
important of those mentioned. This includes changed 
responsibilities for household, children, finances, and 
so forth. Initially intended as helpful, such changes 
may eventually diminish the patient’s self-esteem and 
progressively reduce her family’s interactions with her 
to little more than checking on her pain. Over time, 
this covertly reinforces the symptom of pain and im-parts 
to it unintended value as a major means of main-taining 
communication within the family. 
Imaging Studies 
If the physical examination is relatively benign and is 
not severely limited by body habitus, extensive 
imaging usually adds little to the database needed 
before laparoscopy is performed. This is especially 
true in the case of organ-specific studies (intravenous 
pyelography, barium enema, colonoscopy) in the 
absence of symptoms or signs pointing to an explicit 
organ problem (eg, blood in the stools). Intervening 
for chronic pelvic pain on the basis of an imaging study 
finding alone is unlikely to be fruitful. On the other 
hand, with a specific question in mind—for example, 
pelvic ultrasonography to confirm ovarian endometrio-ma, 
magnetic resonance imaging when adenomyosis is 
suspected, or lower endoscopic ultrasonography to rule 
out invasive rectal endometriosis—imaging can be quite 
helpful. 
Laboratory Studies 
Relatively few hematologic or chemical measures are 
useful in diagnosing chronic pelvic pain. An elevated 
leukocyte count and erythrocyte sedimentation rate 
may make the clinician suspect chronic pelvic inflam-matory 
disease even when cervical probes are nega-tive 
for the most common sexually transmitted 
infections. Serum CA-125 can confirm suspicions of 
deeply infiltrating endometriosis in those without 
prior surgical evaluation but is not sufficiently sensi-tive 
to detect early-stage disease. In those with 
advanced endometriosis, the anti-Müllerian hormone 
level, a measure of ovarian reserve, can help fertility 
counseling in a woman considering extirpative surgical 
treatment for her disease. In those status postbilateral 
oophorectomy with remnant ovarian tissue, follicle-stimulating 
hormone and estradiol levels remain in 
premenopausal ranges. Women using replacement 
estrogen therapy should stop 3 weeks before these 
levels are measured. 
Anesthetic Blocks 
Injection of small volumes of a local anesthetic, 1–5 
mL of 1% lidocaine or 0.25–0.5% bupivacaine, blocks 
pain from either an entrapped segmental nerve (eg, 
ilioinguinal) or an abdominal wall trigger point. Such 
blocks can be therapeutic as well as diagnostic. Many 
anesthesia pain clinics administer epidural or spinal 
anesthetics to distinguish pain arising from peripheral 
organs from pain that has become completely central 
in origin. 
In some instances, it is useful to attempt diagnos-tic 
and therapeutic transvaginal blocks with the same 
local anesthetics for vaginal apex pain, as discussed 
previously. A series of three or four blocks adminis-tered 
1–2 weeks apart may provide durable relief in 
some instances. 
Psychologic Tests and Interviews 
In the attempt to distinguish physical from psycho-logical 
contributions to pain, many studies of chronic 
pelvic pain have used traditional psychological instru-ments 
that were developed to measure general 
psychopathology or personality factors. These psy-chometric 
instruments generally have uncertain face 
value for patients with chronic pain, and their use can 
support the patient’s fears that the health care pro-vider 
thinks her pain is imagined. Psychometric tests 
are most useful when they are interpreted by a psy-chologist 
who has interviewed the patient, and they 
622 Steege and Siedhoff Chronic Pelvic Pain OBSTETRICS & GYNECOLOGY
serve best as a means to better understand the pa-tient’s 
strengths and weaknesses rather than as a means 
to decide who has “organic” compared with “psycho-logical” 
pathology or who needs surgery. The newer 
specialty of pain psychology holds promise in assist-ing 
women dealing with chronic pelvic pain to better 
focus on the positives in their lives and encourages 
them to take an active role in dealing with their pain. 
Laparoscopy 
Great strides have been made in operative laparoscopy 
in the past three decades. Laparoscopy can be useful 
diagnostically and therapeutically (even in the face of 
negative findings), but when a chronic pain syndrome 
is clinically evident, results of laparoscopic treatment 
alone, despite comparable pathology, are much less 
impressive. For a patient with the clinical markers of 
chronic pain syndrome listed earlier, a complete 
workup should be performed before laparoscopy. 
In some puzzling cases, laparoscopy under local 
anesthesia can be used to “pain map” the pelvis.47 A 2- 
mm laparoscope and a small suprapubic probe are 
placed with the use of short-acting, reversible intrave-nous 
analgesia (eg, remifentanil) and local anesthetic. 
Having been oriented to the procedure beforehand, as 
each organ is touched, the patient is asked if the site is 
painful, to rate it on an ordinal scale from 1 to 10, and 
if the discomfort represents her pain. It is possible in 
some cases to block the superior hypogastric plexus 
during pain mapping to better predict benefit from 
presacral neurectomy.48 In this approach, mapping 
is done before and after injecting 10 mL of 1% lido-caine 
just underneath the peritoneum over the sacrum 
using a 7-inch, 22-gauge spinal needle. 
Limiting patient characteristics for using pain 
mapping include high states of anxiety and obesity, 
in which the torque required to move an instrument 
against a thick abdominal wall can provide a distract-ing 
nociceptive stimulus. Once thought to be the 
“holy grail” of chronic pelvic pain diagnostics, better 
understanding of the central mechanisms of chronic 
pain and the relationship among various named pain 
disorders has led to diminished use of laparoscopic 
pain mapping in routine practice. 
The enthusiasm for laparoscopy has its negative 
aspects as well. When pain recurs after the first 
laparoscopic treatment of endometriosis, for example, 
many patients and their gynecologists will automati-cally 
assume that this is the result of a return of their 
disease. The surgeon must also consider the possibil-ity 
that the improvement seen after the first laparos-copy 
was a nonspecific effect of the surgery and that, 
in fact, the “disease” seen and treated was not actually 
relevant to the pain. The second laparoscopy, in 
perhaps the majority of circumstances, reveals less 
disease than was present at the first surgery.49 This 
would suggest that the pain is multifactorial and that 
over time, in many cases, the relative importance of 
the endometriosis may diminish. Ironically, in most 
practices, medical and surgical treatment is neverthe-less 
escalated in such situations. We would recom-mend 
instead starting all over again, making no 
assumption ahead of time that the endometriosis is 
playing a role at all. 
MANAGEMENT 
Relatively straightforward pain problems are not 
challenging to manage such as treating isolated 
dysmenorrhea with hormonal suppression or 
a chronic tubo-ovarian abscess with adnexectomy. 
More often, however, chronic pelvic pain represents 
a complex and nuanced syndrome in which treatment 
may vary considerably depending on the patient. When 
pain itself is the disease, the goal of treatment is not 
necessarily complete eradication of pain, but rather 
finding strategies that afford more functional living. 
Neuromodulatory medications (eg, tricyclic antidepres-sants, 
neurotransmitter reuptake inhibitors, neuroleptics), 
psychological adjuncts (eg, cognitive–behavioral ther-apy, 
pain psychotherapy, sexual counseling), and com-plementary 
strategies (eg, mindfulness-based medication, 
yoga, acupuncture) can be useful to dampen central 
hypersensitivity. For the peripheral elements, physical 
therapy, diet modification, peripheral nerve blocks, 
and surgery can be helpful depending on the target pain 
generator. In all cases, good sleep hygiene, exercise, 
smoking cessation, healthy eating, and social support 
are important foundational elements that improve the 
effectiveness of chronic pelvic pain treatment. 
Medication Use 
Analgesics 
Analgesics such as nonsteroidal anti-inflammatory 
drugs and opioid narcotics can be quite effective for 
acute conditions, but their use in chronic pain is 
marred by a host of adverse outcomes and limited 
efficacy associated with long-term use. Dose-related 
effects of medications such as acetaminophen (liver 
toxicity) and cyclooxygenase inhibitors (gastric and 
renal damage) are well known, but they are not major 
offenders in the realm of tolerance and withdrawal. 
Opioid narcotics, on the other hand, are notoriously 
dangerous in regard to these consequences in addition 
to problems such as narcotic bowel syndrome and 
opioid-induced hyperalgesia. Some patients benefit 
from structured narcotic therapy, but, if they are to be 
VOL. 124, NO. 3, SEPTEMBER 2014 Steege and Siedhoff Chronic Pelvic Pain 623
used for chronic pelvic pain, clinicians should screen 
carefully for factors associated with high risk for 
misuse50,51 and set nonnegotiable ground rules such 
as limiting prescriptions to one health care provider, 
not entertaining requests for early refills, mandating 
scheduled urine drug screens, and refraining from 
uncontrolled dose escalation. Newer opioid medica-tions 
such as oxymorphone and tapentadol are con-sidered 
less euphoria-generating than the more 
commonly used narcotics such as hydrocodone, oxy-codone, 
and hydromorphone (Table 1). 
Antidepressants and Neuroleptics 
These classes of drugs are commonly used in the 
treatment of chronic pain, including chronic pelvic 
pain, although few controlled trials have been 
carried out specifically in this population. Although 
no longer first line for mood disorders, tricyclic 
Table 1. Narcotics Used in Pain Management 
Drug Dosing Side Effects 
Hydrocodone bitartrate with 
acetaminophen 
5–10 mg hydrocodone either every 
6 or every 8 h 
Lightheadedness, dizziness, sedation, nausea 
and vomiting, constipation (these are common 
side effects of all narcotics) 
Lortab 2.5/500, 5/500, or 
7.5/500 
Can use additional acetaminophen 
between doses 
Vicodin 5/750 
Lorcet 10/650 
Lorcet Plus 7.5/650 
Oxycodone hydrochloride 1 tablet every 6 or every 8 h Common effects 
Percocet 5/325 Additional acetaminophen 
between doses 
Oxycodone controlled release 10–40 mg every 12 h Common effects 
Oxycontin 
Methadone hydrochloride 2.5 mg every 8 h to 10 mg every 6 h Common effects, lower extremity edema or joint 
swelling may occur and require discontinuation, 
concurrent use of desipramine may increase 
methadone blood level, cautious use in patients 
on monoamine oxidase inhibitors 
Dolphine 5 or 10 mg scored 
tablets 
Commonly 15–20 mg per day total 
Acetaminophen with codeine 1–2 tablets every 6–8 h Common effects, constipation very likely, nausea 
and vomiting more common than with other 
narcotics, more common allergy—rash 
Tylenol #3: 300 mg 
acetaminophen with 30 mg 
codeine 
Morphine sulfate 15–60 mg every 12 h (controlled-release 
tablets) 
Common effects, higher doses increase risk 
of respiratory depression 
MS Contin 
Oramorph 
Avinza 
Fentanyl transdermal system 25-microgram patch, 1 every 72 h Common effects, patch must be kept from heat 
sources or dose may be increased, extreme 
caution in patients on other central nervous 
system medications, respiratory depression 
can result 
Duragesic Also available in 50 or 75 
micrograms 
Actiq Always start with lowest dose 
Fentora 
Tramadol 50 mg every 6–8 h Common adverse effects, less gastrointestinal, 
less euphoric effects 
Ultram 
Tapentadol 50 mg every 6–8 h; 50–100 mg daily 
for extended release 
Nucynta, Nucynta ER 
624 Steege and Siedhoff Chronic Pelvic Pain OBSTETRICS & GYNECOLOGY
antidepressants such as amitriptyline, nortriptyline, 
and desipramine have a long record of being effective 
in treating chronic pain.52 Newer-generation neuro-transmitter 
reuptake inhibitors such as duloxetine and 
desvenlafaxine can also be useful. Neuroleptics such as 
gabapentin, pregabalin, and lamotrigine are generally 
used when symptoms are more specifically neuropathic 
in nature. It is important to discuss with patients that, 
although they tend to diminish with continued use, all 
of these medications have central side effects, some of 
which are predictable and others quite idiosyncratic. 
When higher doses or multiple agents are used, it 
can be helpful to consult with a psychiatrist or psy-chopharmacologist 
to avoid complications such as 
severe mood dysregulation or serotonin syndrome 
(Table 2). 
Anxiolytics 
Anxiolytic drugs are certainly widely prescribed by 
gynecologists, although it is uncertain how often they 
are given for pain. In one study, alprazolam had 
a surprising degree of analgesic effect in moderate-to-high 
doses in patients with chronic pain of malignant 
origin and concomitant mood changes or anxiety.53 
These patients were already receiving narcotics, 
which may suggest that alprazolam potentiates the 
analgesic effect of narcotics. Their role in conjunction 
with nonnarcotic analgesics is uncertain, and the 
addiction potential is obvious. 
Hormonal Medications 
Combined oral contraceptives are effective in reduc-ing 
dysmenorrhea and cyclic symptoms associated 
with endometriosis. It is not uncommon, however, to 
meet resistance to using these medications in women 
with chronic pelvic pain—either because they were pre-viously 
used and did not cure the entirety of the pain 
syndrome and were thus deemed ineffective or because 
of sensitivity to side effects such as nausea, an under-standable 
consequence in a viscerally hypersensitive 
group. Avoiding ultralow-dose 20-microgram ethinyl 
estradiol formulations can help reduce unscheduled 
bleeding, important for women who may closely asso-ciate 
bleeding with pain. Outside of deeply infiltrating 
endometriosis, progestin-only formulations, by enteral 
or parenteral route, run the risk of exacerbating depres-sive 
symptoms in a vulnerable population. A notable 
exception includes the levonorgestrel intrauterine sys-tem, 
which has little systemic absorption and can con-trol 
dysmenorrhea and pain from endometriosis in 
a low-risk, reversible, long-acting manner. 
The use of gonadotropin-releasing hormone 
agonists deserves special mention. They have been 
recommended to distinguish gynecologic from 
Table 2. Antidepressants and Neuroleptics Used in Pain Management 
Medication Class Dosing Consideration, Side Effects 
Tricyclic antidepressants Tricyclics commonly cause sedation, dry mouth, 
weight gain, constipation; this can be exploited 
for urinary frequency problems; nortriptyline or 
desipramine used more frequently when patients 
have constipation, generally administer at night 
Amitriptyline Start 10–25 mg nightly, titrate 
to 75–150 mg 
Nortriptyline Start 25 mg nightly, titrate to 
50–150 mg 
Desipramine Start 25 mg nightly, titrate to 
50–150 mg 
Serotonin–norepinephrine 
reuptake inhibitors 
Most common side effects are nausea and central 
nervous effects such as lethargy and dizziness, 
provide warning signs of serotonin syndrome for 
patients on other serotonergic drugs and do not 
stop abruptly 
Duloxetine Start 30 mg daily, titrate to 60, 
occasionally 90 mg 
Desvenlafaxine 50–100 mg daily 
Gamma-aminobutyric 
acid analogues 
No major drug interactions, reduce central nervous 
effects by slow titration (for example, over 4 wk); 
do not stop abruptly 
Gabapentin 100–300 mg divided 3 times daily 
Pregabalin 75 mg twice daily 
VOL. 124, NO. 3, SEPTEMBER 2014 Steege and Siedhoff Chronic Pelvic Pain 625
nongynecologic sources of pain based on interpreta-tion 
of some data,54 but pain relief after gonadotropin-releasing 
hormone treatment does not make a gyneco-logic 
(including endometriosis) diagnosis. In that 
study, patients without endometriosis improved with 
the same the frequency as those with the disease, and 
these agents also relieve symptoms of other functional 
conditions such as irritable bowel syndrome. Further-more, 
pain thresholds have been shown to be lower 
premenstrually, even in women without chronic pelvic 
pain. The effect of the menstrual cycle itself in patients 
with chronic pain has not been well-explored, but it 
seems likely that it may impart some cyclicity even to 
conditions unrelated to the reproductive tract. Cyclic-ity 
of symptoms must therefore be interpreted with 
caution, and the disappearance of symptoms or of 
their cyclicity by pharmacologically obliterating the 
menstrual cycle does not demonstrate a gynecologic 
cause. Gonadotropin-releasing hormone agonists can 
be useful when differential diagnosis includes ovarian 
remnant syndrome or residual ovary syndrome or in 
the treatment of deeply infiltrating endometriosis, but 
its use in treating general chronic pelvic pain is limited 
by cost and morbidity. Contrary to popular belief, 
gonadotropin-releasing hormone agonists are not 
more effective than other more benign hormonal ma-nipulations 
directed at pelvic pain.55–57 For deeply 
infiltrating endometriosis, aromatase inhibitors with 
norethindrone may work similarly without as signifi-cant 
hypoestrogenic effects.58,59 
Surgery 
Two basic surgical approaches have been used to treat 
chronic pelvic pain: removing pelvic organs and 
treating visible disease while leaving the pelvic organs 
in place. The use of both approaches is guided by 
clinical experience, because high-quality scientific 
data regarding the role of surgery in pelvic pain are 
sparse. Expansion of literature on the topic, including 
stratification for characteristics that lead to strong or 
poor response, would be most welcome. 
In the United States, approximately 12% of 
hysterectomies are performed with pelvic pain as the 
primary indication.60,61 Although hysterectomy should 
not be viewed as a final curative step in an algorithm 
for treating chronic pelvic pain in general, it can be 
quite helpful for central uterine pain, dyspareunia asso-ciated 
with fundal tenderness, dysmenorrhea, abnor-mal 
bleeding, and cyclic symptoms. Quality of life, 
with particular attention to the effect of pain and 
depression on outcomes, was studied using data from 
the Maryland Women’s Health Study, in which 1,249 
women were evaluated at several-month intervals up to 
2 years after hysterectomy. Not surprisingly, women 
without pain or depression had the highest functional 
levels, but even among women with pain and depres-sion, 
pelvic pain decreased from 97% to 19%, and there 
was a reduction by half in limited physical and social 
function. Women with depression only had improve-ment 
in impaired mental health (85% down to 33%) 
and dyspareunia decreased in all groups.62 Another 
study suggests hysterectomy done for various condi-tions, 
including pelvic pain, outperforms medical or 
uterus-preserving surgical treatment on quality-of-life 
measures.63 Furthermore, the majority of hysterecto-mies 
in these studies were performed using laparot-omy. 
Newer randomized controlled trial data suggest 
improved quality of life laparoscopic over abdominal 
hysterectomy durable to 4 years out from surgery.64 
When considering hysterectomy in the treatment of 
chronic pelvic pain, it is essential to discuss with pa-tients 
which pain symptoms are likely to improve after 
surgery and that some may remain. Just because hys-terectomy 
does not fix every problem does not exclude 
it from being a helpful aspect of treatment provided 
attention is still provided to centralized pain disorders 
and symptoms linked to other organ systems. 
Although many patients and health care providers 
still consider endometriosis nearly synonymous with 
pelvic pain, as discussed previously, the relationship 
is probably more tenuous than previously thought. 
Laparoscopic treatment, even of mild disease, however, 
does provide pain relief.10–13 Some studies suggest 
superiority of excision to ablation of endometriosis im-plants, 
and this approach probably makes good clinical 
sense, but there is insufficient evidence to conclude one 
is definitively superior to the other in pain reduc-tion. 
65,66 Specific symptom improvement after deeply 
infiltrating endometriosis (gastrointestinal, urologic, cul 
de sac) resection is best documented in the litera-ture. 
67,68 Given the complexity of these operations 
and the potential for harm to adjacent vital structures, 
advanced endometriosis resection should be performed 
by experienced surgeons. The benefits of surgical exci-sion 
may be prolonged by subsequent medical therapy. 
Numerous studies have been done of postsurgical treat-ment 
with hormonal medications. Oral contraceptive 
pills, danazol, progestins, aromatase inhibitors, and 
gonadotropin-releasing hormone agonists (with and 
without add-back estrogen and progestin) have all been 
shown to be effective.69–74 Although gonadotropin-releasing 
hormone agonists have become perhaps the 
most widely used of these, definitive evidence for their 
superiority is lacking. The more economical and less 
physiologically intrusive approach would seem to favor 
sex steroids over gonadotropin-releasing hormone 
626 Steege and Siedhoff Chronic Pelvic Pain OBSTETRICS & GYNECOLOGY
agonists. Most troublesome in reviewing all of these 
studies is the observation that dyspareunia is the symp-tom 
that is most refractory to treatment, testifying to its 
multifactorial nature. 
Presacral neurectomy through surgical interrup-tion 
of the superior hypogastric plexus has been 
described in clinical series as effective at treating central 
uterine pain, dysmenorrhea, and endometriosis.75,76 In 
the most rigorous study, Zullo et al4 investigated the 
question with a double-masked randomized trial and 
demonstrated a 20% difference in pain improvement 
when presacral neurectomy was added to endometri-osis 
excision in women with a midline component to 
their pain. When considering presacral neurectomy, 
patients must be counseled about risks of postoperative 
bowel and bladder dysfunction and should also be 
offered more conservative (eg, levonorgestrel intrauter-ine 
systemic) as well as more definitive (ie, hysterec-tomy) 
treatments. 
An ovarian remnant should be removed if it is 
persistently symptomatic despite attempts at medical 
suppression and if menopause cannot be expected in 
the patient’s near future. The dissection should be 
detailed and should include all the peritoneum sur-rounding 
the mass. The pararectal space, and paraves-ical 
space if needed, should be opened systematically 
and the ureter and pelvic sidewall vessels exposed and 
carefully freed fromthe specimen.Usually there is a vas-cular 
supply along the tract of the infundibulopelvic 
ligament, and it is prudent to divide the pedicle well 
above the pelvic brim. When a gonadotropin-releasing 
hormone agonist has been used preoperatively for 
symptom control or to distinguish the relative contribu-tions 
made by the remnant and other pelvic pathology, 
the remnant tissue may become so small as to make it 
difficult to identify. Hence, if a palpable (or visible by 
imaging) mass disappears after gonadotropin-releasing 
hormone agonist treatment, it may be wise to allow 
time for it to regrow before pursuing surgical excision. 
When the remnant is small, some surgeons have stim-ulated 
the remnant with clomiphene citrate to make it 
easier to find.77 
Alternative Treatments 
Psychological disorders should be treated in chronic 
pelvic pain, whether independently present or the result 
of a long-standing pain disorder. Some practitioners 
may find cognitive–behavioral or biofeedback therapy 
useful in reducing automatic responses to painful stim-uli. 
Sexual counseling, couples counseling, and psycho-therapy 
can be helpful adjuncts. Alternative strategies 
such as mindfulness-based meditation, yoga, and acu-puncture 
may have roles in individual cases, but none is 
so clearly applicable or effective that its automatic use is 
supported in cases of chronic pelvic pain. 
Management Overview 
The most effective clinical approach requires simul-taneous 
treatment of as many factors as possible: 
anatomic, musculoskeletal, functional bowel and 
bladder, psychological, and so forth. The patient and 
physician must contract for the long term and work 
from a rehabilitation perspective rather than hope that 
the latest single addition to the treatment will prove to 
be the answer. The physician, to prevent frustration 
and feelings of defeat, must often play the role of 
helping to manage and relieve the pain while helping 
to maximize function, even when pain persists. To the 
surgically trained gynecologist who prefers a clear-cut 
single answer to a clinical problem, this can be the 
most difficult part of dealing with the problem of 
chronic pelvic pain. It is important to free oneself 
from the responsibility of needing to “fix” a patient’s 
chronic pelvic pain. Although the compassionate 
health care provider can be an invaluable aide, much 
of the work in improving from pelvic pain is the bur-den 
of the patient herself. 
REFERENCES 
1. Chronic pelvic pain. ACOG Practice Bulletin No. 51. American 
College of Obstetricians and Gynecologists. Obstet Gynecol 
2004;103:589–605. 
2. Mathias SD, Kuppermann M, Liberman RF, Lipschutz RC, 
Steege JF. Chronic pelvic pain: prevalence, health-related quality 
of life, and economic correlates. Obstet Gynecol 1996;87:321–7. 
3. Deguara CS, Pepas L, Davis C. Does minimally invasive sur-gery 
for endometriosis improve pelvic symptoms and quality of 
life? Curr Opin Obstet Gynecol 2012;24:241–4. 
4. Zullo F, Palomba S, Zupi E, Russo T, Morelli M, Cappiello F, 
et al. Effectiveness of presacral neurectomy in women with 
severe dysmenorrhea caused by endometriosis who were trea-ted 
with laparoscopic conservative surgery: a 1-year prospective 
randomized double-blind controlled trial. Am J Obstet Gynecol 
2003;189:5–10. 
5. Warren JW, Morozov V, Howard FM. Could chronic pelvic 
pain be a functional somatic syndrome? Am J Obstet Gynecol 
2011;205:199.e1–5. 
6. Melzack R, Wall PD. Pain mechanisms: a new theory. Science 
1965;150:971–9. 
7. Melzack R. Pain and the neuromatrix in the brain. J Dent Educ 
2001;65:1378–82. 
8. Howard FM. Chronic pelvic pain. Obstet Gynecol 2003;101: 
594–611. 
9. Martin CE, Johnson E, Wechter ME, Leserman J, Zolnoun DA. 
Catastrophizing: a predictor of persistent pain among women 
with endometriosis at 1 year. Hum Reprod 2011;26:3078–84. 
10. Howard FM. The role of laparoscopy in chronic pelvic pain: 
promise and pitfalls. Obstet Gynecol Surv 1993;48:357–87. 
11. Sutton CJ, Ewen SP, Whitelaw N, Haines P. Prospective, ran-domized, 
double-blind, controlled trial of laser laparoscopy in 
VOL. 124, NO. 3, SEPTEMBER 2014 Steege and Siedhoff Chronic Pelvic Pain 627
the treatment of pelvic pain associated with minimal, mild, and 
moderate endometriosis. Fertil Steril 1994;62:696–700. 
12. Sutton CJ, Pooley AS, Ewen SP, Haines P. Follow-up report on 
a randomized controlled trial of laser laparoscopy in the treat-ment 
of pelvic pain associated with minimal to moderate endo-metriosis. 
Fertil Steril 1997;68:1070–4. 
13. Abbott J, Hawe J, Hunter D, Holmes M, Finn P, Garry R. 
Laparoscopic excision of endometriosis: a randomized, 
placebo-controlled trial. Fertil Steril 2004;82:878–84. 
14. Porpora MG, Koninckx PR, Piazze J, Natili M, Colagrande S, 
Cosmi EV. Correlation between endometriosis and pelvic pain. 
J Am Assoc Gynecol Laparosc 1999;6:429–34. 
15. Milingos S, Protopapas A, Kallipolitis G, Drakakis P, 
Loutradis D, Liapi A, et al. Endometriosis in patients with 
chronic pelvic pain: is staging predictive of the efficacy of lap-aroscopic 
surgery in pain relief? Gynecol Obstet Invest 2006; 
62:48–54. 
16. Hsu AL, Sinaii N, Segars J, Nieman LK, Stratton P. Relating 
pelvic pain location to surgical findings of endometriosis. 
Obstet Gynecol 2011;118:223–30. 
17. Howard FM. Endometriosis and mechanisms of pelvic pain. 
J Minim Invasive Gynecol 2009;16:540–50. 
18. Vercellini P, Fedele L, Aimi G, Pietropaolo G, Consonni D, 
Crosignani PG. Association between endometriosis stage, 
lesion type, patient characteristics and severity of pelvic pain 
symptoms: a multivariate analysis of over 1000 patients. Hum 
Reprod 2007;22:266–71. 
19. Fauconnier A, Chapron C, Dubuisson JB, Vieira M, Dousset B, 
Bréart G. Relation between pain symptoms and the anatomic loca-tion 
of deep infiltrating endometriosis. Fertil Steril 2002;78:719–26. 
20. Gruppo Italiano per lo Studio dell’Endometriosi. Relationship 
between stage, site and morphological characteristics of pelvic 
endometriosis and pain. Hum Reprod 2001;16:2668–71. 
21. Taran FA, Weaver AL, Coddington CC, Stewart EA. Under-standing 
adenomyosis: a case control study. Fertil Steril 2010; 
94:1223–8. 
22. Hammoud A, Gago LA, Diamond MP. Adhesions in patients 
with chronic pelvic pain: a role for adhesiolysis? Fertil Steril 
2004;82:1483–91. 
23. Latthe P, Mignini L, Gray R, Hills R, Khan K. Factors predis-posing 
women to chronic pelvic pain: systematic review. BMJ 
2006;332:749–55. 
24. Halperin R, Padoa A, Schneider D, Bukovsky I, Pansky M. 
Long-term follow-up (5-20 years) after uterine ventrosuspension 
for chronic pelvic pain and deep dyspareunia. Gynecol Obstet 
Invest 2003;55:216–9. 
25. Batioglu S, Zeyneloglu HB. Laparoscopic plication and suspen-sion 
of the round ligament for chronic pelvic pain and dyspar-eunia. 
J Am Assoc Gynecol Laparosc 2000;7:547–51. 
26. Carter JE. Carter-Thomason uterine suspension and positioning 
by ligament investment, fixation and truncation. J Reprod Med 
1999;44:417–22. 
27. Masters WH, Johnson VE; Reproductive Biology Research 
Foundation. Human sexual response. 1st ed. Boston (MA): Little, 
Brown; 1966. 
28. Duncan CH, Taylor HC Jr. A psychosomatic study of pelvic 
congestion. Am J Obstet Gynecol 1952;64:1–12. 
29. Freedman J, Ganeshan A, Crowe PM. Pelvic congestion syn-drome: 
the role of interventional radiology in the treatment of 
chronic pelvic pain. Postgrad Med J 2010;86:704–10. 
30. Farquhar CM, Rogers V, Franks S, Pearce S, Wadsworth J, 
Beard RW. A randomized controlled trial of medroxyproges-terone 
acetate and psychotherapy for the treatment of pelvic 
congestion. Br J Obstet Gynaecol 1989;96:1153–62. 
31. Soysal ME, Soysal S, Vicdan K, Ozer S. A randomized con-trolled 
trial of goserelin and medroxyprogesterone acetate in 
the treatment of pelvic congestion. Hum Reprod 2001;16:931–9. 
32. Perry CP. Current concepts of pelvic congestion and chronic 
pelvic pain. JSLS 2001;5:105–10. 
33. Tu FF, Hahn D, Steege JF. Pelvic congestion syndrome-associated 
pelvic pain: a systematic review of diagnosis and 
management. Obstet Gynecol Surv 2010;65:332–40. 
34. Bukovsky I, Liftshitz Y, Langer R, Weinraub Z, Sadovsky G, 
Caspi E. Ovarian residual syndrome. Surg Gynecol Obstet 
1988;167:132–4. 
35. Steege JF. Ovarian remnant syndrome. Obstet Gynecol 1987; 
70:64–7. 
36. Lamvu G, Robinson B, Zolnoun D, Steege JF. Vaginal apex resec-tion: 
a treatment option for vaginal apex pain. Obstet Gynecol 
2004;104:1340–6. 
37. Sharp HT, Dodson MK, Langer KM, Doucette RC, Norton PA. 
The role of vaginal apex excision in the management of persis-tent 
posthysterectomy dyspareunia. Am J Obstet Gynecol 
2000;183:1385–8. 
38. Sinaki M, Merritt JL, Stillwell GK. Tension myalgia of the 
pelvic floor. Mayo Clin Proc 1977;52:717–22. 
39. FitzGerald MP, Payne CK, Lukacz ES, Yang CC, Peters KM, 
Chai TC, et al. Randomized multicenter clinical trial of myofascial 
physical therapy in women with interstitial cystitis/painful bladder 
syndrome and pelvic floor tenderness. J Urol 2012;187:2113–8. 
40. Williams RE, Hartmann KE, Sandler RS, Miller WC, 
Savitz LA, Steege JF. Recognition and treatment of irritable 
bowel syndrome among women with chronic pelvic pain. Am 
J Obstet Gynecol 2005;192:761–7. 
41. Tu FF, As-Sanie S, Steege JF. Prevalence of pelvic musculoskel-etal 
disorders in a female chronic pelvic pain clinic. J Reprod 
Med 2006;51:185–9. 
42. Walker E, Katon W, Harrop-Griffiths J, Holm L, Russo J, 
Hickok LR. Relationship of chronic pelvic pain to psychiatric 
diagnoses and childhood sexual abuse. Am J Psychiatry 1988; 
145:75–80. 
43. Slocumb JC, Kellner R, Rosenfeld RC, Pathak D. Anxiety and 
depression in patients with the abdominal pelvic pain syn-drome. 
Gen Hosp Psychiatry 1989;11:48–53. 
44. Reiter RC, Gambone JC. Demographic and historic variables 
in women with idiopathic chronic pelvic pain. Obstet Gynecol 
1990;75:428–32. 
45. Rapkin AJ, Kames LD, Darke LL, Stampler FM, Naliboff BD. 
History of physical and sexual abuse in women with chronic 
pelvic pain. Obstet Gynecol 1990;76:92–6. 
46. Steege JF, Metzger DA, Levy BS. Chronic pelvic pain: an inte-grated 
approach. 1st ed. Philadelphia (PA): Saunders; 1998. 
47. Yunker A, Steege J. Practical guide to laparoscopic pain map-ping. 
J Minim Invasive Gynecol 2010;17:8–11. 
48. Zupi E, Sbracia M, Marconi D, Zullo F, Santi K, Solima E, et al. 
Pain mapping during minilaparoscopy in infertile patients with-out 
pathology. J Am Assoc Gynecol Laparosc 1999;6:51–4. 
49. Yeung P Jr, Sinervo K, Winer W, Albee RB Jr. Complete lap-aroscopic 
excision of endometriosis in teenagers: is postopera-tive 
hormonal suppression necessary? Fertil Steril 2011;95: 
1909–12, 1912.e1. 
50. Warner EA. Opioids for the treatment of chronic noncancer 
pain. Am J Med 2012;125:1155–61. 
628 Steege and Siedhoff Chronic Pelvic Pain OBSTETRICS & GYNECOLOGY
51. Manchikanti L, Abdi S, Atluri S, Balog CC, Benyamin RM, 
Boswell MV, et al. American Society of Interventional Pain 
Physicians (ASIPP) guidelines for responsible opioid prescrib-ing 
in chronic non-cancer pain: Part 2—guidance. Pain Physician 
2012;15:S67–116. 
52. An update on the drug treatment of neuropathic pain. Part 1: 
antidepressants [published erratum appears in Drug Ther Bull 
2012;50:132]. Drug Ther Bull 2012;50:114–7. 
53. Fernandez F, Adams F, Holmes VF. Analgesic effect of alpra-zolam 
in patients with chronic, organic pain of malignant ori-gin. 
J Clin Psychopharmacol 1987;7:167–9. 
54. Ling FW. Randomized controlled trial of depot leuprolide in 
patients with chronic pelvic pain and clinically suspected endo-metriosis. 
Pelvic Pain Study Group. Obstet Gynecol 1999;93: 
51–8. 
55. Brown J, Kives S, Akhtar M. Progestagens and anti-progestagens 
for pain associated with endometriosis. The Co-chrane 
Database of Systematic Reviews 2012, Issue 3. Art No.: 
CD002122. DOI: 10.1002/14651858.CD002122.pub2. 
56. Petta CA, Ferriani RA, Abrao MS, Hassan D, Rosa E Silva JC, 
Podgaec S, et al. Randomized clinical trial of a levonorgestrel-releasing 
intrauterine system and a depot GnRH analogue for 
the treatment of chronic pelvic pain in women with endometri-osis. 
Hum Reprod 2005;20:1993–8. 
57. Howard FM. An evidence-based medicine approach to the 
treatment of endometriosis-associated chronic pelvic pain: 
placebo-controlled studies. J Am Assoc Gynecol Laparosc 
2000;7:477–88. 
58. Pavone ME, Bulun SE. Aromatase inhibitors for the treatment 
of endometriosis. Fertil Steril 2012;98:1370–9. 
59. Ferrero S, Gillott DJ, Venturini PL, Remorgida V. Use of 
aromatase inhibitors to treat endometriosis-related pain symp-toms: 
a systematic review. Reprod Biol Endocrinol 2011;9:89. 
60. Lee NC, Dicker RC, Rubin GL, Ory HW. Confirmation of the 
preoperative diagnoses for hysterectomy. Am J Obstet Gynecol 
1984;150:283–7. 
61. Wu JM, Wechter ME, Geller EJ, Nguyen TV, Visco AG. Hys-terectomy 
rates in the United States, 2003. Obstet Gynecol 
2007;110:1091–5. 
62. Hartmann KE, Ma C, Lamvu GM, Langenberg PW, Steege JF, 
Kjerulff KH. Quality of life and sexual function after hysterec-tomy 
in women with preoperative pain and depression. Obstet 
Gynecol 2004;104:701–9. 
63. Kuppermann M, Learman LA, Schembri M, Gregorich SE, 
Jackson RA, Jacoby A, et al. Contributions of hysterectomy 
and uterus-preserving surgery to health-related quality of life. 
Obstet Gynecol 2013;122:15–25. 
64. Nieboer TE, Hendriks JC, Bongers MY, Vierhout ME, 
Kluivers KB. Quality of life after laparoscopic and abdominal 
hysterectomy: a randomized controlled trial. Obstet Gynecol 
2012;119:85–91. 
65. Healey M, Ang WC, Cheng C. Surgical treatment of endome-triosis: 
a prospective randomized double-blinded trial compar-ing 
excision and ablation. Fertil Steril 2010;94:2536–40. 
66. Wright J, Lotfallah H, Jones K, Lovell D. A randomized trial of 
excision versus ablation for mild endometriosis. Fertil Steril 
2005;83:1830–6. 
67. Bassi MA, Podgaec S, Dias JA Jr, D’Amico Filho N, Petta CA, 
Abrao MS. Quality of life after segmental resection of the rec-tosigmoid 
by laparoscopy in patients with deep infiltrating 
endometriosis with bowel involvement. J Minim Invasive 
Gynecol 2011;18:730–3. 
68. Nezhat C, Nezhat F, Nezhat CH, Nasserbakht F, Rosati M, 
Seidman DS. Urinary tract endometriosis treated by laparos-copy. 
Fertil Steril 1996;66:920–4. 
69. Bianchi S, Busacca M, Agnoli B, Candiani M, Calia C, 
Vignali M. Effects of 3 month therapy with danazol after lapa-roscopic 
surgery for stage III/IV endometriosis: a randomized 
study. Hum Reprod 1999;14:1335–7. 
70. Morgante G, Ditto A, La Marca A, De Leo V. Low-dose dana-zol 
after combined surgical and medical therapy reduces the 
incidence of pelvic pain in women with moderate and severe 
endometriosis. Hum Reprod 1999;14:2371–4. 
71. Telimaa S, Rönnberg L, Kauppila A. Placebo-controlled com-parison 
of danazol and high-dose medroxyprogesterone acetate 
in the treatment of endometriosis after conservative surgery. 
Gynecol Endocrinol 1987;1:363–71. 
72. Hornstein MD, Hemmings R, Yuzpe AA, Heinrichs WL. Use 
of nafarelin versus placebo after reductive laparoscopic surgery 
for endometriosis. Fertil Steril 1997;68:860–4. 
73. Vercellini P, Frontino G, De Giorgi O, Aimi G, Zaina B, 
Crosignani PG. Comparison of a levonorgestrel-releasing intra-uterine 
device versus expectant management after conservative 
surgery for symptomatic endometriosis: a pilot study. Fertil 
Steril 2003;80:305–9. 
74. Ferrero S, Camerini G, Seracchioli R, Ragni N, Venturini PL, 
Remorgida V. Letrozole combined with norethisterone acetate 
compared with norethisterone acetate alone in the treatment of 
pain symptoms caused by endometriosis. Hum Reprod 2009; 
24:3033–41. 
75. Latthe PM, Proctor ML, Farquhar CM, Johnson N, Khan KS. 
Surgical interruption of pelvic nerve pathways in dysmenor-rhea: 
a systematic review of effectiveness. Acta Obstet Gynecol 
Scand 2007;86:4–15. 
76. Zullo F, Palomba S, Zupi E, Russo T, Morelli M, Sena T, et al. 
Long-term effectiveness of presacral neurectomy for the treat-ment 
of severe dysmenorrhea due to endometriosis. J Am Assoc 
Gynecol Laparosc 2004;11:23–8. 
77. Kaminski PF, Sorosky JI, Mandell MJ, Broadstreet RP, 
Zaino RJ. Clomiphene citrate stimulation as an adjunct in locat-ing 
ovarian tissue in ovarian remnant syndrome. Obstet Gynecol 
1990;76:924–6. 
VOL. 124, NO. 3, SEPTEMBER 2014 Steege and Siedhoff Chronic Pelvic Pain 629

Más contenido relacionado

La actualidad más candente

CaitlinB_SummerPoster_final___
CaitlinB_SummerPoster_final___CaitlinB_SummerPoster_final___
CaitlinB_SummerPoster_final___
Caitlin Barrett
 
Physical activity in the treatment of fibromyalgia (1)
Physical activity in the treatment of fibromyalgia (1)Physical activity in the treatment of fibromyalgia (1)
Physical activity in the treatment of fibromyalgia (1)
DanielaClarosV
 

La actualidad más candente (19)

Cancer Pain Management
Cancer Pain Management Cancer Pain Management
Cancer Pain Management
 
Multidisciplinary pain management rsuh dr. takdri
Multidisciplinary pain management rsuh dr. takdriMultidisciplinary pain management rsuh dr. takdri
Multidisciplinary pain management rsuh dr. takdri
 
Chronic Pain After Surgery
Chronic Pain After SurgeryChronic Pain After Surgery
Chronic Pain After Surgery
 
March 2017 vol3 issue1
March 2017 vol3 issue1March 2017 vol3 issue1
March 2017 vol3 issue1
 
CaitlinB_SummerPoster_final___
CaitlinB_SummerPoster_final___CaitlinB_SummerPoster_final___
CaitlinB_SummerPoster_final___
 
Maladaptive movement and motor control impairments as underlying mechanism
Maladaptive movement and motor control impairments as underlying mechanismMaladaptive movement and motor control impairments as underlying mechanism
Maladaptive movement and motor control impairments as underlying mechanism
 
Opioid Induced Hyperalgesia
Opioid Induced HyperalgesiaOpioid Induced Hyperalgesia
Opioid Induced Hyperalgesia
 
Pain management
Pain managementPain management
Pain management
 
Physical activity in the treatment of fibromyalgia (1)
Physical activity in the treatment of fibromyalgia (1)Physical activity in the treatment of fibromyalgia (1)
Physical activity in the treatment of fibromyalgia (1)
 
Cancer Pain
Cancer PainCancer Pain
Cancer Pain
 
Facet joint cervical
Facet joint cervicalFacet joint cervical
Facet joint cervical
 
Edwards et al-2006 Catastrophizing
Edwards et al-2006 CatastrophizingEdwards et al-2006 Catastrophizing
Edwards et al-2006 Catastrophizing
 
De kaak in al zijn facetten
De kaak in al zijn facettenDe kaak in al zijn facetten
De kaak in al zijn facetten
 
Common Brain Mechanisms Between Pain & Addiction
Common Brain Mechanisms Between Pain & AddictionCommon Brain Mechanisms Between Pain & Addiction
Common Brain Mechanisms Between Pain & Addiction
 
Neuroticism & PCS
Neuroticism & PCSNeuroticism & PCS
Neuroticism & PCS
 
Pain management peter
Pain management peterPain management peter
Pain management peter
 
Pain management and prn medications
Pain management and prn medicationsPain management and prn medications
Pain management and prn medications
 
Updates in pain management
Updates in pain managementUpdates in pain management
Updates in pain management
 
Efficacy of classification-based_cft_in_nsclbp
Efficacy of classification-based_cft_in_nsclbpEfficacy of classification-based_cft_in_nsclbp
Efficacy of classification-based_cft_in_nsclbp
 

Similar a 00006250 201409000-00021

The association between a history of lifetime traumatic events and pain sever...
The association between a history of lifetime traumatic events and pain sever...The association between a history of lifetime traumatic events and pain sever...
The association between a history of lifetime traumatic events and pain sever...
Paul Coelho, MD
 
The conundrum of opioid tapering in long term opioid therapy for chronic pain...
The conundrum of opioid tapering in long term opioid therapy for chronic pain...The conundrum of opioid tapering in long term opioid therapy for chronic pain...
The conundrum of opioid tapering in long term opioid therapy for chronic pain...
Paul Coelho, MD
 
Chronic Overlapping Pain Conditions
Chronic Overlapping Pain ConditionsChronic Overlapping Pain Conditions
Chronic Overlapping Pain Conditions
Paul Coelho, MD
 
Supplement maarten-loos
Supplement maarten-loosSupplement maarten-loos
Supplement maarten-loos
R_Roumen
 

Similar a 00006250 201409000-00021 (20)

gtg_41 CPP.pdf
gtg_41 CPP.pdfgtg_41 CPP.pdf
gtg_41 CPP.pdf
 
Predicting transition to chronic pain.
Predicting transition to chronic pain.Predicting transition to chronic pain.
Predicting transition to chronic pain.
 
Gt41 initialmanagementchronicpelvicpain2005
Gt41 initialmanagementchronicpelvicpain2005Gt41 initialmanagementchronicpelvicpain2005
Gt41 initialmanagementchronicpelvicpain2005
 
Medical Management of Chronic Pelvic Pain: The Evidence.
Medical Management of Chronic Pelvic Pain: The Evidence.Medical Management of Chronic Pelvic Pain: The Evidence.
Medical Management of Chronic Pelvic Pain: The Evidence.
 
Austin Pain & Relief
Austin Pain & ReliefAustin Pain & Relief
Austin Pain & Relief
 
The association between a history of lifetime traumatic events and pain sever...
The association between a history of lifetime traumatic events and pain sever...The association between a history of lifetime traumatic events and pain sever...
The association between a history of lifetime traumatic events and pain sever...
 
MP Journal Of Anesthesiology Vol 1
MP Journal Of Anesthesiology Vol 1MP Journal Of Anesthesiology Vol 1
MP Journal Of Anesthesiology Vol 1
 
Jospt.2014 abd aneurysm
Jospt.2014 abd aneurysmJospt.2014 abd aneurysm
Jospt.2014 abd aneurysm
 
P 517
P 517P 517
P 517
 
Pain, Heat, & Emotion. NEJM
Pain, Heat, & Emotion. NEJMPain, Heat, & Emotion. NEJM
Pain, Heat, & Emotion. NEJM
 
PCP AC 2021 - Back Pain Basics
PCP AC 2021 - Back Pain BasicsPCP AC 2021 - Back Pain Basics
PCP AC 2021 - Back Pain Basics
 
The conundrum of opioid tapering in long term opioid therapy for chronic pain...
The conundrum of opioid tapering in long term opioid therapy for chronic pain...The conundrum of opioid tapering in long term opioid therapy for chronic pain...
The conundrum of opioid tapering in long term opioid therapy for chronic pain...
 
2014 acute pain management in infants
2014 acute pain management in infants 2014 acute pain management in infants
2014 acute pain management in infants
 
Intensity of chronic pain — the wrong metric
Intensity of chronic pain — the wrong metric Intensity of chronic pain — the wrong metric
Intensity of chronic pain — the wrong metric
 
Overlapping chronic pain conditions
Overlapping chronic pain conditionsOverlapping chronic pain conditions
Overlapping chronic pain conditions
 
Chronic Overlapping Pain Conditions
Chronic Overlapping Pain ConditionsChronic Overlapping Pain Conditions
Chronic Overlapping Pain Conditions
 
Supplement maarten-loos
Supplement maarten-loosSupplement maarten-loos
Supplement maarten-loos
 
ANTIDEPRESSANTS IN CHRONIC PAIN RELIEF- A REVIEW
ANTIDEPRESSANTS IN CHRONIC PAIN RELIEF- A REVIEWANTIDEPRESSANTS IN CHRONIC PAIN RELIEF- A REVIEW
ANTIDEPRESSANTS IN CHRONIC PAIN RELIEF- A REVIEW
 
The dark side of opioids
The dark side of opioidsThe dark side of opioids
The dark side of opioids
 
The dark side_of_opioids_in_pain_management__.3
The dark side_of_opioids_in_pain_management__.3The dark side_of_opioids_in_pain_management__.3
The dark side_of_opioids_in_pain_management__.3
 

Más de Jonny Luna (12)

C059553
C059553C059553
C059553
 
Mecanismo
MecanismoMecanismo
Mecanismo
 
Guias15
Guias15Guias15
Guias15
 
A03v56n1
A03v56n1A03v56n1
A03v56n1
 
9789584476180.04
9789584476180.049789584476180.04
9789584476180.04
 
Optimal evaluation of_the_infertile_female(1)
Optimal evaluation of_the_infertile_female(1)Optimal evaluation of_the_infertile_female(1)
Optimal evaluation of_the_infertile_female(1)
 
7 1
7 17 1
7 1
 
Nihms204875
Nihms204875Nihms204875
Nihms204875
 
628362
628362628362
628362
 
guidelines summary
guidelines summaryguidelines summary
guidelines summary
 
Anatomía de tórax
Anatomía de tóraxAnatomía de tórax
Anatomía de tórax
 
Anatomia del dorso
Anatomia del dorsoAnatomia del dorso
Anatomia del dorso
 

00006250 201409000-00021

  • 1. Clinical Expert Series Chronic Pelvic Pain John F. Steege, MD, and Matthew T. Siedhoff, MD, MSCR As opposed to the satisfying solutions found in the management of acute pain, chronic pelvic pain can be a vexing problem for the patient and physician. Seldom is a single source or cause found, and nearly always the condition is influenced by the broader social and psychological context of the patient. In this article, we discuss the evaluation of chronic pelvic pain, often considering pain as the disease itself, and identify peripheral generators, which gynecologists can address to help reduce their contributions to symptoms. (Obstet Gynecol 2014;124:616–29) DOI: 10.1097/AOG.0000000000000417 The American College of Obstetricians and Gyne-cologists proposed the following definition of chronic pelvic pain: noncyclic pain of 6 or more months’ duration that localizes to the anatomic pelvis, anterior abdominal wall at or below the umbilicus, the lumbosacral back or the buttocks, and is of sufficient severity to cause functional disability or lead to med-ical care.1 The problem costs the economy of the United States well in excess of $3 billion2 and costs women greatly in the areas of work, family responsi-bilities, and relationships. The practicing gynecologist finds the problem time-consuming, challenging, and less rewarding than the usually successful obstetric and gynecologic endeavors. Traditional thinking about chronic pelvic pain has emphasized observable organic pathology (eg, endo-metriosis, adhesions), but the connection between these problems and pain symptoms is actually more tenuous than previously thought. For example, even the most optimistic clinical reports note pain relief in only 60– 70% of women undergoing laparoscopic surgery for endometriosis,3,4 suggesting that in many women, there is more to the story than just the implants. More and more data confirming the coexistence of one or more other chronic pain disorders in patients—conditions such as interstitial cystitis (also known as painful bladder syn-drome), irritable bowel syndrome, temporomandibular joint disorder, migraine headaches, vulvodynia, and fi-bromyalgia— suggest that perhaps we should be treating pelvic pain itself as the disease rather than as just a man-ifestation of a specific pathologic change.5 In this review, we describe chronic pelvic pain and point out common peripheral pain generators—nociceptive stimuli—that can be modulated by gynecologic interventions. HISTORY OF PELVIC PAIN CONCEPTS Over the past 60 years, the study of chronic pelvic pain has gone through significant changes in approach. Before the advent of laparoscopy and based in a Cartesian framework in which pain perception should be proportional to the degree of tissue damage, gynecologists were understandably reluctant to oper-ate on any pathology not large enough to palpate. In addition, much of clinical medicine was practiced from the perspective of the mind–body split: causa-tion of symptoms was distinctly divided between physical and psychological sources. Although this model was sufficient to address most causes of acute pain, it fails to interpret the majority of chronic pain disorders in gynecology as well as other areas of med-icine. The gate-control theory, an alternative pro-posed by Melzack and Wall in 1965, suggests that pain information flows in two directions: 1) nocicep-tive signals from peripheral tissue ascend through the spinal cord to higher centers; and 2) central centers can modulate, using descending signals altering spinal From the Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. Continuing medical education for this article is available at http://links.lww. com/AOG/A541. Corresponding author: John F. Steege, MD, Department of Obstetrics and Gynecology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC; e-mail: jfsteege@med.unc.edu. Financial Disclosure The authors did not report any potential conflicts of interest. © 2014 by The American College of Obstetricians and Gynecologists. Published by Lippincott Williams & Wilkins. ISSN: 0029-7844/14 616 VOL. 124, NO. 3, SEPTEMBER 2014 OBSTETRICS & GYNECOLOGY
  • 2. cord neurotransmitter and interneuron activity, the transmission of these nociceptive signals from the periphery. Deterioration of these regulatory processes were thought to potentially account for development of chronic pain states by allowing too many peripheral signals to pass through the spinal cord “gates.” Vari-ation in patients’ relative degree of gate opening could thus explain why similar amounts of physical tissue damage result in different degrees of pain perception.6 While these theory changes were stimulating the field of pain research, gynecologists were busy devel-oping laparoscopy and with it the hope that treating visible pathology could fix chronic pelvic pain. A focus on “laparoscopy-negative” patients in chronic pelvic pain clinics emerged, implying that if some pathology were found, it must be a “real” cause for pain. Subsequent experience has shown that although treatment of laparoscopically diagnosed pathology can be helpful, the clinical reality is more complex: 1) in many instances, visible pathology found at lapa-roscopy may be incidental and not related to the pain; 2) in those with pathology that does contribute to nociception, the pain experienced by the patient may differ from another patient with anatomically similar pathology; and 3) pain from a laparoscopic finding is often best understood in the larger context of a centralized pain disorder. Research of the 1980s added the observation of distressingly high rates of physical and sexual abuse, especially in the chronic pelvic pain population. These observations led to the speculation that the experience of abuse may make a person more vulnerable to the development of chronic pelvic pain or perhaps be a specific cause for pain. In relation to pain, abuse, particularly that which occurs in formative years, may serve to alter the response to nociception and central pain processing. That said, not all abused patients go on to have chronic pain nor do all patients with pain have a history of abuse, so it might be the response to trauma that plays a key role in development of chronic pain. Health care providers need to take into account the presence of abuse in a patient’s history when de-tected but be careful to avoid necessarily concluding a causal relationship in that patient’s pain. Melzack’s neuromatrix theory7 added the pivotal notion of neuroplasticity, which suggests that experi-ence can change the neurophysiologic behavior of the central nervous system in a manner that influences the subsequent processing of nociceptive stimuli. It may explain the apparent development of pain responses to stimuli usually thought of as nonpainful (allodynia) as well as exaggerated responses to painful stimuli (hyperalgesia). Every practicing gynecologist has seen patients whose pain responses seem out of proportion to the pathology found. This may reflect the emo-tional meaning of the problem for the patient as well as past or present trauma, but it may also be the result of sensitization of spinal cord interneurons that have become pain amplifiers as a result of being on the receiving end of peripheral nociceptive stimuli for pro-longed periods. On the positive side, neuroplasticity suggests that given enough time and the right treat-ment, even seemingly intractable chronic pain prob-lems can improve substantially. The concept of central sensitization adds the most recent layer to theoretical understanding of chronic pain: the notion that multiple repetitions of lower-level stimuli may over time result in a more severe central perception of pain. This centralized pain hypersensitivity helps us understand how mul-tiple organ systems can be recruited into the syn-drome, incorporating genetic and social factors in pain amplification. Whatever the nociceptive stimulus (bowel, bladder, muscle, uterus), it can register as pain at a lower threshold in the patient with a centralized pain disorder (Box 1). Box 1. Definitions of Pain Terms Allodynia—pain resulting from a nonnoxious stimulus Hyperalgesia—painful sensation of abnormal severity after noxious stimulation Neuropathic pain—pain persisting after healing of disease or trauma-induced tissue damage Neuroplasticity—the malleability of central pain perception mechanisms in response to chronic pain states Nociceptor—a nerve receptor for pain This understanding contributes to the framework a clinician needs when taking a clinic history of pelvic pain and performing the physical examination. The inquiry should set out in pursuit of all the factors relevant to a person’s pain, not simply trying to identify one factor with an acceptably plausible relationship to her complaints. Once the patient is aware that the cli-nician is keeping an open mind about multiple contrib-uting factors, she may become less defensive about any inquiries in emotional areas. EVALUATION OF PATIENTS WITH CHRONIC PELVIC PAIN History-Taking The site, duration, pattern during activities, relation to position changes, and association with bodily func-tions are all important elements of pain. For example, pain that is absent in the morning but worsens VOL. 124, NO. 3, SEPTEMBER 2014 Steege and Siedhoff Chronic Pelvic Pain 617
  • 3. progressively during the day may be associated with pelvic floor muscle dysfunction, whereas a tender “spot” of dyspareunia might be related to nodular cul de sac endometriosis. A previous review supplies a very detailed description of both history-taking and physical examination techniques that are useful in the evalua-tion of chronic pelvic pain.8 We focus here on the most frequently useful elements of this process. The chronology of a patient’s pain is critical. As a pain syndrome develops, pain can be present over a progressively larger area despite stable detectable pathology. Interpreting this as the breakdown or wear-ing out of physiologic systems that deal with pain sig-nals has some biologic validity and may make sense to the patient. The clinician may need to counter the idea patients sometimes have that endometriosis “flares” like rheumatoid disease or spreads like a malignancy. From a cognitive perspective, it is invaluable to discern the patient’s and her family’s ideas about the causes of and future for her pain. Fears of cancer can be discovered even if this diagnosis was never even remotely considered by the clinician. Less dramatic but equally powerful attributions of cause can emerge such as pelvic infection that is the result of sexual acts remote in time, arguments with a spouse, divine ret-ribution, and so forth. When a patient has been under that care of previous health care providers, it is important to review the effects of previous treatments and the evolution of how the patient has thought about her own disease. For example, when a prior treatment for mild endometriosis was only briefly successful, did she (and her health care provider?) see this as evidence for increasing severity of disease or did it make them question the diagnosis in the first place? In a referral-based practice, it is a frequent event to deal with a patient with mild disease who has not responded to any of four or five medical or surgical measures that would ordinarily improve symptoms. She then sees herself as having increasing and invasive disease, prompting even more aggressive treatment, rather than wondering if something else is contributing to her pain. Physical Examination The physical examination begins with observing the patient’s gait, comfort with sitting, ease of going from sitting to standing, and from sitting on the table to lying down. Dysfunctions of the pelvic floor and hip muscles may be manifested by discomfort with these motions. For example, if the patient sits up on one buttock during the interview, one might suspect pelvic floor muscle pain. With the patient sitting on the examination table, it can be helpful to begin the examination with the back, evaluating for tenderness of the spine, para-spinous muscles, and sacroiliac joints. This may identify pain generators and sites for therapeutic intervention, but it also allows for touch to begin in a very nonthreatening way. Gynecologic assessment is uncomfortable to some degree for almost all women, but the patient with chronic pelvic pain is particularly vulnerable. Moving from back to abdomen to pelvis can establish trust and reduce fear. The patient is then asked to recline on the table, observing how comfortably she can do this. The abdominal wall is examined with and without flexed rectus muscles. A positive Carnett sign (increased tenderness when palpation is done in the presence of abdominal wall flexion) implies at least a contribution to the pain from abdominal wall myofascial sources. Decreased pain during this maneuver implies a higher contribution from visceral sources. On occasion, gentle fingertip palpation of the abdominal wall can detect such trigger points in the musculature. Rarely, a sub-cutaneous abdominal wall endometrioma is discov-ered, a diagnosis supported by a history of predictable, cyclic, focal tenderness. An abdominal wall endome-trioma, or a hernia, is often more readily appreciated by examining the patient in the standing position. Pelvic examination then begins with external review of the vulva and vestibule. Gentle palpation with a cotton swab can detect areas of sensitivity consistent with vulvar vestibular syndrome in the introitus or trigger points higher in the vagina. Intrinsic cervical allodynia (from cervical conization or after obstetric cervical laceration) may be detected in the same manner with gentle cotton-tipped appli-cator palpation. Guiding a patient through contraction–relaxation sequences of the abdominal, thigh, and vaginal introi-tal muscles can reduce the discomfort of the examina-tion and can indicate the patient’s degree of control over muscle tension. Single-digit palpation of the leva-tor plate, piriformis, and obturator muscles can elicit the tenderness of pelvic floor tension myalgia. This condition can present as a sequel to some other pelvic pain or a problem in itself. Discomfort is usually felt as pelvic pressure and radiation pain to the sacrum, near the insertions of the levator plate muscles. The exact sequence of the examination should be adjusted in view of the information obtained during the history: examine the areas less likely to be tender first, saving the more tender areas for the end. Palpating the most tender areas first can elevate anxiety and distort pain responses from subsequent areas. 618 Steege and Siedhoff Chronic Pelvic Pain OBSTETRICS & GYNECOLOGY
  • 4. Single-digit palpation should also be used to discover areas of tenderness in the cervix, uterus, adnexa, bladder, and urethra. Premature addition of the abdominal hand to the examination adds in nociceptive signals from abdominal wall myofascial components that may lead the examiner to over-attribute pain to the viscera. Finally, the abdominal hand is added to assess size, shape, and mobility of pelvic structures. Adnexal thickening and mobility, pelvic relaxation, coccygeal tenderness, and foci of pain that reproduce dyspareunia should be noted. A rectovaginal examination is important when deep infiltrating endometriosis is suspected. During all components of the physical examina-tion, it is important to not only ask the question “Does this hurt?,” but also “Do you feel pain where I am pressing or somewhere else?” and “Is this the pain you were describing earlier—is this your pain?” If a patient answers affirmatively to the final question, it can be helpful to point out what structure you are palpating (eg, pelvic floor muscle compared with ovary). As you are going along through the examina-tion, it can be useful to mention the items you are adding to the list of possible contributing factors to her pain, thus reinforcing what you discussed during the history in terms of pain being multifactorial. Having concluded the most important part of the evaluation, the history and physical, you are now prepared to discuss diagnostic possibilities and a treat-ment plan. What follows is a review of the commonly described contributors to pelvic pain, which might be part of that discussion. CONTRIBUTIONS OF PERIPHERAL PAIN GENERATORS This section deserves an important caveat. Although we believe that types of tissue damage or other nociceptive input can generate pain, they cannot be viewed in isolation of the patient’s individual central pain process-ing. Management strategies are discussed in more detail, but, in general, the goal of the treating health care provider involves trying to dampen overall pain signaling sensitivity—“turning down the master vol-ume”— and looking for areas in the periphery that can be “tuned up” toward better functioning. This effort is more likely to be successful in the patient who takes an active role in her own improvement and avoids seeing herself as helpless in the face of her discomforts.9 The following peripheral generators represent areas where we can intervene but should not be described to patients with chronic pelvic pain as the only cause of their pain. Because these conditions can be completely asymptomatic in many patients, their importance needs interpretation in the context of her overall health, taking into account her other pain disorders and general capacity for dealing with life’s challenges. Endometriosis Evidence that endometriosis causes pain results from observations that the disease is more commonly encountered in women undergoing laparoscopy for pain than for other reasons10 and that laparoscopic treatment results in better pain reduction than diagnos-tic surgery alone.11–13 However, amount of disease does not correlate with symptom severity14,15; endometriosis is a frequently encountered incidental finding, and peri-toneal implants do not localize to symptom location.16 A variety of proposed mechanisms explaining pain associated with endometriosis exist, including inflam-matory, nociceptive, and neuropathic.17 There is no pathognomonic symptom associated with endometri-osis, however. Many of the symptoms often attributed to endometriosis (eg, dyspareunia, dysmenorrhea, abnormal bowel or bladder function) are commonly found in functional disorders such as irritable bowel syndrome or interstitial cystitis, making it difficult to understand the relevant contribution of endometriosis to chronic pelvic pain. Deeply infiltrating endometri-osis— fibrotic, vascular, desmoplastic tissue destruction— is a biologically different disease, in which detectable physical examination or imaging findings relate to spe-cific symptoms such as tender rectovaginal nodularity or ovarian endometrioma with dyspareunia or dysche-zia with intrinsic rectal involvement.18–20 The relation-ship of a related condition, adenomyosis, to pelvic pain is less established, but the disease should be considered, especially when symptoms include dysmenorrhea or heavy bleeding.21 Pelvic Adhesions Early reviews supported the role of adhesions as a significant contributor in chronic pelvic pain.10,22 More recent investigations23 demonstrate a relatively weak correlation between adhesions and chronic pelvic pain, much less than other factors such as psy-chosomatic symptoms and substance abuse. Few, if any, well-designed studies demonstrate effective treatment of chronic pelvic pain with adhesiolysis. Unfortunately, in an effort to provide some explanation for complex pain disorders, health care providers often still posit adhe-sions as an etiology, even when a patient’s surgical his-tory includes only laparoscopy with findings of minimal or no endometriosis, pelvic inflammatory disease, or other conditions associated with meaningful adhesions. This explanation can happen even when the patient’s pain escalation is remote from her last surgery. VOL. 124, NO. 3, SEPTEMBER 2014 Steege and Siedhoff Chronic Pelvic Pain 619
  • 5. Adhesions may play some role in pain conditions in some women, but the relative contribution is probably small. Also, the putative treatment—repeat surgical intervention—can add new contributions to pain syn-dromes such as the effect of surgical trauma, disappoint-ment from lack of pain relief, feeding the psychological need of being “ill” with more surgery, and, in the worst case, generating a complication such as enterotomy. Pelvic Support Most women in pain clinics are in their third or fourth decade of life, whereas pelvic organ prolapse affects significantly older women, suggesting a very minimal role for support problems in chronic pelvic pain. Pelvic relaxation usually leads to reports of heaviness, pressure, dropping sensations, or aching. In attempt-ing to hold in prolapsing organs, the patient may tense the levator plate, leading to tenderness during daily activities and intercourse. Fear of (or actual) loss of urinary control during coitus can add to the discom-fort by impairing physiologic sexual response. The retroverted uterus is another potential contrib-utor to chronic pelvic pain, particularly in the form of deep dyspareunia. Clearly for many women, retrover-sion is an innocent anatomic variant, but for those with pain, uncontrolled clinical series of uterine suspension procedures suggest changing the position of the uterus to an axial or anteverted position can improve dyspar-eunia by elevating the fundus out of the posterior cul de sac24–26 and allowing for better vaginal expansion as a natural part of the sexual response cycle.27 Pelvic Congestion Overfilling (congestion) of the pelvic venous system has been implicated as a cause of dull chronic aching pain that usually is bilateral, worse at the end of the day after prolonged standing, premenstrually, and postcoitally. Some studies suggest the condition is present in nearly one-third of women with chronic pelvic pain,28 but there is no agreed-on reference stan-dard for diagnosis despite individual technical regi-mens involving venography, magnetic resonance, and ultrasonography.29 Hormonal suppression,30,31 percutaneous embolotherapy, and surgery (vein liga-tion, hysterectomy and salpingo-oophorectomy) rep-resent available treatments,32 but study protocols involving these interventions are diverse and few have been investigated in controlled trials.33 Residual Ovary When the uterus has been removed, with or without removal of one ovary, the remaining ovary or ovaries become symptomatic in a small percentage of women.34 Pain from the ovary can be increased by confinement within postoperative adhesions, rupture or leakage of a cyst prompting additional adhesion formation, or attachment of the ovary to the sigmoid colon or vaginal apex by postoperative adhesions. In the case of attach-ment to the vaginal apex, deep dyspareunia can result when the area is struck. Ovarian Remnant A more difficult situation can develop if a small fragment of ovarian tissue is left behind during attempted oophorectomy.35 In most instances, this happens when challenging dissection is required, such as cases of extensive pelvic adhesions or deeply infil-trating endometriosis. Within 1–3 years of the attemp-ted oophorectomy, continued follicle-stimulating hormone stimulation will result in growth of the ovar-ian fragment, often producing an intermittently symp-tomatic pelvic mass located along the course of the ovarian vascular supply. A postulated mechanism for pain generation includes the cystic enlargement of the mass confined within fibrotic adhesions. If the rem-nant developed because endometriosis made for diffi-cult oophorectomy, that disease is often found in the remnant and probably also serves as a pain generator. Classic remnant symptoms include absence of vaso-motor symptoms after ostensible bilateral oophorec-tomy and the presence of cyclic unilateral pain. Like in the case of the residual ovary, the remnant can produce dyspareunia if it is located close to the vaginal apex. When performing oophorectomy, it is best to open the pararectal space and completely skeletonize the infundibulopelvic ligament not only to avoid com-plications such as ureteral injury, but also to prevent ovarian remnant syndrome. In difficult cases, dividing the pedicle at or above the pelvic brim, like in risk-reduction prophylactic oophorectomy, is prudent. Vaginal Apex Pain After hysterectomy, pain may persist or recur because of intrinsic sensitivity of the vaginal apex. Although the cuff may appear to have healed perfectly well, gentle examination with a cotton-tipped applicator may reveal focal sensitivity of a moderate to severe degree, many times located in one lateral fornix or the other and often replicating the reported pain of dyspareunia. When the cotton applicator examination is not done, the unaware examiner may then, noting pain on traditional biman-ual examination, mistakenly conclude that the source of nociception lies cephalad, for example, in a remaining ovary, pelvic scarring, or bowel adhesions. The diagnosis may be confirmed by noting elimination of the pain after injection of a local 620 Steege and Siedhoff Chronic Pelvic Pain OBSTETRICS & GYNECOLOGY
  • 6. anesthetic. The condition is generally considered neuropathic by virtue of the character of the pain (burning, stinging, sharp) and that neuropathic treat-ments (overnight application of lidocaine, oral medi-cations such as nortriptyline, amitriptyline, gabapentin, etc) seem to benefit some patients. Laparoscopic revision of the vaginal cuff may give good initial relief in approximately two-thirds of patients, but pain tends to recur to a degree over the subsequent 2–3 years, although perhaps at a less intense level.36,37 Musculoskeletal Problems Musculoskeletal changes can become involved with chronic pelvic pain, either as the primary problem or as a secondary reaction to the pelvic pain. The muscular problem that most often produces pelvic pain is pelvic floor tension myalgia (also called levator spasm or levator ani syndrome).38 The clinical symp-tom profile commonly includes pain with sitting flat on a chair, worsening pelvic pressure over the course of the day, and midvaginal dyspareunia. Intermittent or constant painful contraction of the levator plate can be present as a primary problem or a reaction to some other source of pain. The patient with levator pain will often sit up on one buttock during the interview and will report that pelvic floor soreness persisted for 1–2 days after palpation of the muscles during pelvic examination. The piriformis and obturator muscles warrant further emphasis because they are seldom appreciated as possible sources of pain. These muscles are external rotators of the leg, and rotation against resistance can allow detection of tender spasm of the muscles during the pelvic examination. The sciatic nerve can traverse the belly of the piriformis as a normal anatomic variant, producing symptoms similar to sciatica when the muscle is in spasm. Pain caused by the iliopsoas complex can manifest as anterior hip or groin pain and is diagnosed with passive or active extension at the hip. The mainstay of treatment of muscular compo-nents of pelvic pain is physical therapy, most com-monly performed by a therapist with specialized training in the treatment of female pelvic pain. This subspecialty of the field has grown enormously over the past 15 years in obvious response to growing recognition of muscular elements to pelvic pain and in recognition that it works.39 Most physical therapists specializing in women’s care will address pelvic floor and hip muscles issues with transvaginal muscle treat-ments as well as other treatment measures. Other measures that are sometimes helpful are self-directed pelvic floor contraction and relaxation exercises, mus-cle relaxants, vaginal valium, and Botox injections. When the problem is severe enough to require these measures, it is generally advised that they be used along with simultaneous physical therapy. Medical Comorbidity Peripheral pain generation in chronic pelvic pain often involves nongynecologic systems.40,41 A careful history and close physical examination of gastrointes-tinal, urologic, musculoskeletal, and neurologic sys-tems are needed to evaluate these additional contributions to chronic pelvic pain. Most of the avail-able literature examines these problems indepen-dently of each other and without reference to their relevance to chronic pelvic pain or to the overall prev-alence of these disorders in chronic pelvic pain. The gastrointestinal system is the most common nongyne-cologic contributor to chronic pelvic pain, typically manifesting as irritable bowel syndrome. Perhaps sec-ond in terms of prevalence, functional urinary prob-lems (ie, interstitial cystitis) are another common comorbid contributor in chronic pelvic pain. These as well as others such as migraine, temporomandibu-lar joint disorder, and fibromyalgia should be viewed in the context of a larger centralized pain problem and treatment should be symptom-specific. Psychological comorbidity also travels with chronic pelvic pain. Depression, anxiety, anger–hos-tility, somatization, and catastrophization9 (the belief that things are as bad as they can be and are not likely to improve) are all more common among women with chronic pain than in women in a control group.42,43 Whether mood disorder is a predisposing factor to, or a result of, chronic pain is not clear and may differ from patient to patient, but the situation is symbiotic at a minimum. Many women with chronic pelvic pain fear physicians’ recognition of mood disorder leads to the conclusion that their pain problem is not “real” or “in her head.” Thus, from a treatment perspective, it makes sense to attend to each disorder to the degree it manifests, but not necessarily attempt to discern cau-sality in a gynecologic visit. Studies vary in describing the prevalence of sexual abuse,44,45 but regardless, it is difficult to judge whether these events are directly relevant to present pain and hence demand attention or whether they contribute to a psychologically vulnerable substrate influenced by subsequent physical and emotional events. In these circumstances, it may be worthwhile to suggest further mental health evaluation as an exploratory measure, being careful not to imply that the patient is being referred because the physician is certain that the abuse is related to the development of the pain. Lastly, marital distress and sexual dysfunction, particularly dyspareunia, are common further VOL. 124, NO. 3, SEPTEMBER 2014 Steege and Siedhoff Chronic Pelvic Pain 621
  • 7. burdens for patients with pain. Although some women report satisfactory sexual functioning before the onset of pain symptoms, others appear to have long-standing impairments in sexual response. In our experience, sexual difficulties are often the problem that makes a person seek (or is encouraged by her partner to seek) help for her pain. DIAGNOSTIC STRATEGIES Recognizing a Chronic Pain Syndrome Many women can experience pain for longer than 6 months without becoming debilitated. Although their pain is chronic, such women are not described as having a chronic pain syndrome. The following are the common clinical hallmarks of this syndrome46: 1) dura-tion of 6 months or longer, 2) incomplete relief by most previous treatments, 3) significantly impaired physical function at home or at work, 4) signs of depression (sleep disturbance, weight loss, loss of appetite), 5) hypersensitive response to nociceptive stimuli, and 6) altered family roles. Of the signs of depression, sleep disturbance is usually the first to appear. Careful questioning is needed to distinguish awakening caused by pain from awakening that just happens. In the true vegetative sign, the person usually cannot get back to sleep even if pain is relieved (by medication or other means). The alteration of family roles is perhaps the most important of those mentioned. This includes changed responsibilities for household, children, finances, and so forth. Initially intended as helpful, such changes may eventually diminish the patient’s self-esteem and progressively reduce her family’s interactions with her to little more than checking on her pain. Over time, this covertly reinforces the symptom of pain and im-parts to it unintended value as a major means of main-taining communication within the family. Imaging Studies If the physical examination is relatively benign and is not severely limited by body habitus, extensive imaging usually adds little to the database needed before laparoscopy is performed. This is especially true in the case of organ-specific studies (intravenous pyelography, barium enema, colonoscopy) in the absence of symptoms or signs pointing to an explicit organ problem (eg, blood in the stools). Intervening for chronic pelvic pain on the basis of an imaging study finding alone is unlikely to be fruitful. On the other hand, with a specific question in mind—for example, pelvic ultrasonography to confirm ovarian endometrio-ma, magnetic resonance imaging when adenomyosis is suspected, or lower endoscopic ultrasonography to rule out invasive rectal endometriosis—imaging can be quite helpful. Laboratory Studies Relatively few hematologic or chemical measures are useful in diagnosing chronic pelvic pain. An elevated leukocyte count and erythrocyte sedimentation rate may make the clinician suspect chronic pelvic inflam-matory disease even when cervical probes are nega-tive for the most common sexually transmitted infections. Serum CA-125 can confirm suspicions of deeply infiltrating endometriosis in those without prior surgical evaluation but is not sufficiently sensi-tive to detect early-stage disease. In those with advanced endometriosis, the anti-Müllerian hormone level, a measure of ovarian reserve, can help fertility counseling in a woman considering extirpative surgical treatment for her disease. In those status postbilateral oophorectomy with remnant ovarian tissue, follicle-stimulating hormone and estradiol levels remain in premenopausal ranges. Women using replacement estrogen therapy should stop 3 weeks before these levels are measured. Anesthetic Blocks Injection of small volumes of a local anesthetic, 1–5 mL of 1% lidocaine or 0.25–0.5% bupivacaine, blocks pain from either an entrapped segmental nerve (eg, ilioinguinal) or an abdominal wall trigger point. Such blocks can be therapeutic as well as diagnostic. Many anesthesia pain clinics administer epidural or spinal anesthetics to distinguish pain arising from peripheral organs from pain that has become completely central in origin. In some instances, it is useful to attempt diagnos-tic and therapeutic transvaginal blocks with the same local anesthetics for vaginal apex pain, as discussed previously. A series of three or four blocks adminis-tered 1–2 weeks apart may provide durable relief in some instances. Psychologic Tests and Interviews In the attempt to distinguish physical from psycho-logical contributions to pain, many studies of chronic pelvic pain have used traditional psychological instru-ments that were developed to measure general psychopathology or personality factors. These psy-chometric instruments generally have uncertain face value for patients with chronic pain, and their use can support the patient’s fears that the health care pro-vider thinks her pain is imagined. Psychometric tests are most useful when they are interpreted by a psy-chologist who has interviewed the patient, and they 622 Steege and Siedhoff Chronic Pelvic Pain OBSTETRICS & GYNECOLOGY
  • 8. serve best as a means to better understand the pa-tient’s strengths and weaknesses rather than as a means to decide who has “organic” compared with “psycho-logical” pathology or who needs surgery. The newer specialty of pain psychology holds promise in assist-ing women dealing with chronic pelvic pain to better focus on the positives in their lives and encourages them to take an active role in dealing with their pain. Laparoscopy Great strides have been made in operative laparoscopy in the past three decades. Laparoscopy can be useful diagnostically and therapeutically (even in the face of negative findings), but when a chronic pain syndrome is clinically evident, results of laparoscopic treatment alone, despite comparable pathology, are much less impressive. For a patient with the clinical markers of chronic pain syndrome listed earlier, a complete workup should be performed before laparoscopy. In some puzzling cases, laparoscopy under local anesthesia can be used to “pain map” the pelvis.47 A 2- mm laparoscope and a small suprapubic probe are placed with the use of short-acting, reversible intrave-nous analgesia (eg, remifentanil) and local anesthetic. Having been oriented to the procedure beforehand, as each organ is touched, the patient is asked if the site is painful, to rate it on an ordinal scale from 1 to 10, and if the discomfort represents her pain. It is possible in some cases to block the superior hypogastric plexus during pain mapping to better predict benefit from presacral neurectomy.48 In this approach, mapping is done before and after injecting 10 mL of 1% lido-caine just underneath the peritoneum over the sacrum using a 7-inch, 22-gauge spinal needle. Limiting patient characteristics for using pain mapping include high states of anxiety and obesity, in which the torque required to move an instrument against a thick abdominal wall can provide a distract-ing nociceptive stimulus. Once thought to be the “holy grail” of chronic pelvic pain diagnostics, better understanding of the central mechanisms of chronic pain and the relationship among various named pain disorders has led to diminished use of laparoscopic pain mapping in routine practice. The enthusiasm for laparoscopy has its negative aspects as well. When pain recurs after the first laparoscopic treatment of endometriosis, for example, many patients and their gynecologists will automati-cally assume that this is the result of a return of their disease. The surgeon must also consider the possibil-ity that the improvement seen after the first laparos-copy was a nonspecific effect of the surgery and that, in fact, the “disease” seen and treated was not actually relevant to the pain. The second laparoscopy, in perhaps the majority of circumstances, reveals less disease than was present at the first surgery.49 This would suggest that the pain is multifactorial and that over time, in many cases, the relative importance of the endometriosis may diminish. Ironically, in most practices, medical and surgical treatment is neverthe-less escalated in such situations. We would recom-mend instead starting all over again, making no assumption ahead of time that the endometriosis is playing a role at all. MANAGEMENT Relatively straightforward pain problems are not challenging to manage such as treating isolated dysmenorrhea with hormonal suppression or a chronic tubo-ovarian abscess with adnexectomy. More often, however, chronic pelvic pain represents a complex and nuanced syndrome in which treatment may vary considerably depending on the patient. When pain itself is the disease, the goal of treatment is not necessarily complete eradication of pain, but rather finding strategies that afford more functional living. Neuromodulatory medications (eg, tricyclic antidepres-sants, neurotransmitter reuptake inhibitors, neuroleptics), psychological adjuncts (eg, cognitive–behavioral ther-apy, pain psychotherapy, sexual counseling), and com-plementary strategies (eg, mindfulness-based medication, yoga, acupuncture) can be useful to dampen central hypersensitivity. For the peripheral elements, physical therapy, diet modification, peripheral nerve blocks, and surgery can be helpful depending on the target pain generator. In all cases, good sleep hygiene, exercise, smoking cessation, healthy eating, and social support are important foundational elements that improve the effectiveness of chronic pelvic pain treatment. Medication Use Analgesics Analgesics such as nonsteroidal anti-inflammatory drugs and opioid narcotics can be quite effective for acute conditions, but their use in chronic pain is marred by a host of adverse outcomes and limited efficacy associated with long-term use. Dose-related effects of medications such as acetaminophen (liver toxicity) and cyclooxygenase inhibitors (gastric and renal damage) are well known, but they are not major offenders in the realm of tolerance and withdrawal. Opioid narcotics, on the other hand, are notoriously dangerous in regard to these consequences in addition to problems such as narcotic bowel syndrome and opioid-induced hyperalgesia. Some patients benefit from structured narcotic therapy, but, if they are to be VOL. 124, NO. 3, SEPTEMBER 2014 Steege and Siedhoff Chronic Pelvic Pain 623
  • 9. used for chronic pelvic pain, clinicians should screen carefully for factors associated with high risk for misuse50,51 and set nonnegotiable ground rules such as limiting prescriptions to one health care provider, not entertaining requests for early refills, mandating scheduled urine drug screens, and refraining from uncontrolled dose escalation. Newer opioid medica-tions such as oxymorphone and tapentadol are con-sidered less euphoria-generating than the more commonly used narcotics such as hydrocodone, oxy-codone, and hydromorphone (Table 1). Antidepressants and Neuroleptics These classes of drugs are commonly used in the treatment of chronic pain, including chronic pelvic pain, although few controlled trials have been carried out specifically in this population. Although no longer first line for mood disorders, tricyclic Table 1. Narcotics Used in Pain Management Drug Dosing Side Effects Hydrocodone bitartrate with acetaminophen 5–10 mg hydrocodone either every 6 or every 8 h Lightheadedness, dizziness, sedation, nausea and vomiting, constipation (these are common side effects of all narcotics) Lortab 2.5/500, 5/500, or 7.5/500 Can use additional acetaminophen between doses Vicodin 5/750 Lorcet 10/650 Lorcet Plus 7.5/650 Oxycodone hydrochloride 1 tablet every 6 or every 8 h Common effects Percocet 5/325 Additional acetaminophen between doses Oxycodone controlled release 10–40 mg every 12 h Common effects Oxycontin Methadone hydrochloride 2.5 mg every 8 h to 10 mg every 6 h Common effects, lower extremity edema or joint swelling may occur and require discontinuation, concurrent use of desipramine may increase methadone blood level, cautious use in patients on monoamine oxidase inhibitors Dolphine 5 or 10 mg scored tablets Commonly 15–20 mg per day total Acetaminophen with codeine 1–2 tablets every 6–8 h Common effects, constipation very likely, nausea and vomiting more common than with other narcotics, more common allergy—rash Tylenol #3: 300 mg acetaminophen with 30 mg codeine Morphine sulfate 15–60 mg every 12 h (controlled-release tablets) Common effects, higher doses increase risk of respiratory depression MS Contin Oramorph Avinza Fentanyl transdermal system 25-microgram patch, 1 every 72 h Common effects, patch must be kept from heat sources or dose may be increased, extreme caution in patients on other central nervous system medications, respiratory depression can result Duragesic Also available in 50 or 75 micrograms Actiq Always start with lowest dose Fentora Tramadol 50 mg every 6–8 h Common adverse effects, less gastrointestinal, less euphoric effects Ultram Tapentadol 50 mg every 6–8 h; 50–100 mg daily for extended release Nucynta, Nucynta ER 624 Steege and Siedhoff Chronic Pelvic Pain OBSTETRICS & GYNECOLOGY
  • 10. antidepressants such as amitriptyline, nortriptyline, and desipramine have a long record of being effective in treating chronic pain.52 Newer-generation neuro-transmitter reuptake inhibitors such as duloxetine and desvenlafaxine can also be useful. Neuroleptics such as gabapentin, pregabalin, and lamotrigine are generally used when symptoms are more specifically neuropathic in nature. It is important to discuss with patients that, although they tend to diminish with continued use, all of these medications have central side effects, some of which are predictable and others quite idiosyncratic. When higher doses or multiple agents are used, it can be helpful to consult with a psychiatrist or psy-chopharmacologist to avoid complications such as severe mood dysregulation or serotonin syndrome (Table 2). Anxiolytics Anxiolytic drugs are certainly widely prescribed by gynecologists, although it is uncertain how often they are given for pain. In one study, alprazolam had a surprising degree of analgesic effect in moderate-to-high doses in patients with chronic pain of malignant origin and concomitant mood changes or anxiety.53 These patients were already receiving narcotics, which may suggest that alprazolam potentiates the analgesic effect of narcotics. Their role in conjunction with nonnarcotic analgesics is uncertain, and the addiction potential is obvious. Hormonal Medications Combined oral contraceptives are effective in reduc-ing dysmenorrhea and cyclic symptoms associated with endometriosis. It is not uncommon, however, to meet resistance to using these medications in women with chronic pelvic pain—either because they were pre-viously used and did not cure the entirety of the pain syndrome and were thus deemed ineffective or because of sensitivity to side effects such as nausea, an under-standable consequence in a viscerally hypersensitive group. Avoiding ultralow-dose 20-microgram ethinyl estradiol formulations can help reduce unscheduled bleeding, important for women who may closely asso-ciate bleeding with pain. Outside of deeply infiltrating endometriosis, progestin-only formulations, by enteral or parenteral route, run the risk of exacerbating depres-sive symptoms in a vulnerable population. A notable exception includes the levonorgestrel intrauterine sys-tem, which has little systemic absorption and can con-trol dysmenorrhea and pain from endometriosis in a low-risk, reversible, long-acting manner. The use of gonadotropin-releasing hormone agonists deserves special mention. They have been recommended to distinguish gynecologic from Table 2. Antidepressants and Neuroleptics Used in Pain Management Medication Class Dosing Consideration, Side Effects Tricyclic antidepressants Tricyclics commonly cause sedation, dry mouth, weight gain, constipation; this can be exploited for urinary frequency problems; nortriptyline or desipramine used more frequently when patients have constipation, generally administer at night Amitriptyline Start 10–25 mg nightly, titrate to 75–150 mg Nortriptyline Start 25 mg nightly, titrate to 50–150 mg Desipramine Start 25 mg nightly, titrate to 50–150 mg Serotonin–norepinephrine reuptake inhibitors Most common side effects are nausea and central nervous effects such as lethargy and dizziness, provide warning signs of serotonin syndrome for patients on other serotonergic drugs and do not stop abruptly Duloxetine Start 30 mg daily, titrate to 60, occasionally 90 mg Desvenlafaxine 50–100 mg daily Gamma-aminobutyric acid analogues No major drug interactions, reduce central nervous effects by slow titration (for example, over 4 wk); do not stop abruptly Gabapentin 100–300 mg divided 3 times daily Pregabalin 75 mg twice daily VOL. 124, NO. 3, SEPTEMBER 2014 Steege and Siedhoff Chronic Pelvic Pain 625
  • 11. nongynecologic sources of pain based on interpreta-tion of some data,54 but pain relief after gonadotropin-releasing hormone treatment does not make a gyneco-logic (including endometriosis) diagnosis. In that study, patients without endometriosis improved with the same the frequency as those with the disease, and these agents also relieve symptoms of other functional conditions such as irritable bowel syndrome. Further-more, pain thresholds have been shown to be lower premenstrually, even in women without chronic pelvic pain. The effect of the menstrual cycle itself in patients with chronic pain has not been well-explored, but it seems likely that it may impart some cyclicity even to conditions unrelated to the reproductive tract. Cyclic-ity of symptoms must therefore be interpreted with caution, and the disappearance of symptoms or of their cyclicity by pharmacologically obliterating the menstrual cycle does not demonstrate a gynecologic cause. Gonadotropin-releasing hormone agonists can be useful when differential diagnosis includes ovarian remnant syndrome or residual ovary syndrome or in the treatment of deeply infiltrating endometriosis, but its use in treating general chronic pelvic pain is limited by cost and morbidity. Contrary to popular belief, gonadotropin-releasing hormone agonists are not more effective than other more benign hormonal ma-nipulations directed at pelvic pain.55–57 For deeply infiltrating endometriosis, aromatase inhibitors with norethindrone may work similarly without as signifi-cant hypoestrogenic effects.58,59 Surgery Two basic surgical approaches have been used to treat chronic pelvic pain: removing pelvic organs and treating visible disease while leaving the pelvic organs in place. The use of both approaches is guided by clinical experience, because high-quality scientific data regarding the role of surgery in pelvic pain are sparse. Expansion of literature on the topic, including stratification for characteristics that lead to strong or poor response, would be most welcome. In the United States, approximately 12% of hysterectomies are performed with pelvic pain as the primary indication.60,61 Although hysterectomy should not be viewed as a final curative step in an algorithm for treating chronic pelvic pain in general, it can be quite helpful for central uterine pain, dyspareunia asso-ciated with fundal tenderness, dysmenorrhea, abnor-mal bleeding, and cyclic symptoms. Quality of life, with particular attention to the effect of pain and depression on outcomes, was studied using data from the Maryland Women’s Health Study, in which 1,249 women were evaluated at several-month intervals up to 2 years after hysterectomy. Not surprisingly, women without pain or depression had the highest functional levels, but even among women with pain and depres-sion, pelvic pain decreased from 97% to 19%, and there was a reduction by half in limited physical and social function. Women with depression only had improve-ment in impaired mental health (85% down to 33%) and dyspareunia decreased in all groups.62 Another study suggests hysterectomy done for various condi-tions, including pelvic pain, outperforms medical or uterus-preserving surgical treatment on quality-of-life measures.63 Furthermore, the majority of hysterecto-mies in these studies were performed using laparot-omy. Newer randomized controlled trial data suggest improved quality of life laparoscopic over abdominal hysterectomy durable to 4 years out from surgery.64 When considering hysterectomy in the treatment of chronic pelvic pain, it is essential to discuss with pa-tients which pain symptoms are likely to improve after surgery and that some may remain. Just because hys-terectomy does not fix every problem does not exclude it from being a helpful aspect of treatment provided attention is still provided to centralized pain disorders and symptoms linked to other organ systems. Although many patients and health care providers still consider endometriosis nearly synonymous with pelvic pain, as discussed previously, the relationship is probably more tenuous than previously thought. Laparoscopic treatment, even of mild disease, however, does provide pain relief.10–13 Some studies suggest superiority of excision to ablation of endometriosis im-plants, and this approach probably makes good clinical sense, but there is insufficient evidence to conclude one is definitively superior to the other in pain reduc-tion. 65,66 Specific symptom improvement after deeply infiltrating endometriosis (gastrointestinal, urologic, cul de sac) resection is best documented in the litera-ture. 67,68 Given the complexity of these operations and the potential for harm to adjacent vital structures, advanced endometriosis resection should be performed by experienced surgeons. The benefits of surgical exci-sion may be prolonged by subsequent medical therapy. Numerous studies have been done of postsurgical treat-ment with hormonal medications. Oral contraceptive pills, danazol, progestins, aromatase inhibitors, and gonadotropin-releasing hormone agonists (with and without add-back estrogen and progestin) have all been shown to be effective.69–74 Although gonadotropin-releasing hormone agonists have become perhaps the most widely used of these, definitive evidence for their superiority is lacking. The more economical and less physiologically intrusive approach would seem to favor sex steroids over gonadotropin-releasing hormone 626 Steege and Siedhoff Chronic Pelvic Pain OBSTETRICS & GYNECOLOGY
  • 12. agonists. Most troublesome in reviewing all of these studies is the observation that dyspareunia is the symp-tom that is most refractory to treatment, testifying to its multifactorial nature. Presacral neurectomy through surgical interrup-tion of the superior hypogastric plexus has been described in clinical series as effective at treating central uterine pain, dysmenorrhea, and endometriosis.75,76 In the most rigorous study, Zullo et al4 investigated the question with a double-masked randomized trial and demonstrated a 20% difference in pain improvement when presacral neurectomy was added to endometri-osis excision in women with a midline component to their pain. When considering presacral neurectomy, patients must be counseled about risks of postoperative bowel and bladder dysfunction and should also be offered more conservative (eg, levonorgestrel intrauter-ine systemic) as well as more definitive (ie, hysterec-tomy) treatments. An ovarian remnant should be removed if it is persistently symptomatic despite attempts at medical suppression and if menopause cannot be expected in the patient’s near future. The dissection should be detailed and should include all the peritoneum sur-rounding the mass. The pararectal space, and paraves-ical space if needed, should be opened systematically and the ureter and pelvic sidewall vessels exposed and carefully freed fromthe specimen.Usually there is a vas-cular supply along the tract of the infundibulopelvic ligament, and it is prudent to divide the pedicle well above the pelvic brim. When a gonadotropin-releasing hormone agonist has been used preoperatively for symptom control or to distinguish the relative contribu-tions made by the remnant and other pelvic pathology, the remnant tissue may become so small as to make it difficult to identify. Hence, if a palpable (or visible by imaging) mass disappears after gonadotropin-releasing hormone agonist treatment, it may be wise to allow time for it to regrow before pursuing surgical excision. When the remnant is small, some surgeons have stim-ulated the remnant with clomiphene citrate to make it easier to find.77 Alternative Treatments Psychological disorders should be treated in chronic pelvic pain, whether independently present or the result of a long-standing pain disorder. Some practitioners may find cognitive–behavioral or biofeedback therapy useful in reducing automatic responses to painful stim-uli. Sexual counseling, couples counseling, and psycho-therapy can be helpful adjuncts. Alternative strategies such as mindfulness-based meditation, yoga, and acu-puncture may have roles in individual cases, but none is so clearly applicable or effective that its automatic use is supported in cases of chronic pelvic pain. Management Overview The most effective clinical approach requires simul-taneous treatment of as many factors as possible: anatomic, musculoskeletal, functional bowel and bladder, psychological, and so forth. The patient and physician must contract for the long term and work from a rehabilitation perspective rather than hope that the latest single addition to the treatment will prove to be the answer. The physician, to prevent frustration and feelings of defeat, must often play the role of helping to manage and relieve the pain while helping to maximize function, even when pain persists. To the surgically trained gynecologist who prefers a clear-cut single answer to a clinical problem, this can be the most difficult part of dealing with the problem of chronic pelvic pain. It is important to free oneself from the responsibility of needing to “fix” a patient’s chronic pelvic pain. Although the compassionate health care provider can be an invaluable aide, much of the work in improving from pelvic pain is the bur-den of the patient herself. REFERENCES 1. Chronic pelvic pain. ACOG Practice Bulletin No. 51. American College of Obstetricians and Gynecologists. Obstet Gynecol 2004;103:589–605. 2. Mathias SD, Kuppermann M, Liberman RF, Lipschutz RC, Steege JF. Chronic pelvic pain: prevalence, health-related quality of life, and economic correlates. Obstet Gynecol 1996;87:321–7. 3. Deguara CS, Pepas L, Davis C. Does minimally invasive sur-gery for endometriosis improve pelvic symptoms and quality of life? Curr Opin Obstet Gynecol 2012;24:241–4. 4. Zullo F, Palomba S, Zupi E, Russo T, Morelli M, Cappiello F, et al. Effectiveness of presacral neurectomy in women with severe dysmenorrhea caused by endometriosis who were trea-ted with laparoscopic conservative surgery: a 1-year prospective randomized double-blind controlled trial. Am J Obstet Gynecol 2003;189:5–10. 5. Warren JW, Morozov V, Howard FM. Could chronic pelvic pain be a functional somatic syndrome? Am J Obstet Gynecol 2011;205:199.e1–5. 6. Melzack R, Wall PD. Pain mechanisms: a new theory. Science 1965;150:971–9. 7. Melzack R. Pain and the neuromatrix in the brain. J Dent Educ 2001;65:1378–82. 8. Howard FM. Chronic pelvic pain. Obstet Gynecol 2003;101: 594–611. 9. Martin CE, Johnson E, Wechter ME, Leserman J, Zolnoun DA. Catastrophizing: a predictor of persistent pain among women with endometriosis at 1 year. Hum Reprod 2011;26:3078–84. 10. Howard FM. The role of laparoscopy in chronic pelvic pain: promise and pitfalls. Obstet Gynecol Surv 1993;48:357–87. 11. Sutton CJ, Ewen SP, Whitelaw N, Haines P. Prospective, ran-domized, double-blind, controlled trial of laser laparoscopy in VOL. 124, NO. 3, SEPTEMBER 2014 Steege and Siedhoff Chronic Pelvic Pain 627
  • 13. the treatment of pelvic pain associated with minimal, mild, and moderate endometriosis. Fertil Steril 1994;62:696–700. 12. Sutton CJ, Pooley AS, Ewen SP, Haines P. Follow-up report on a randomized controlled trial of laser laparoscopy in the treat-ment of pelvic pain associated with minimal to moderate endo-metriosis. Fertil Steril 1997;68:1070–4. 13. Abbott J, Hawe J, Hunter D, Holmes M, Finn P, Garry R. Laparoscopic excision of endometriosis: a randomized, placebo-controlled trial. Fertil Steril 2004;82:878–84. 14. Porpora MG, Koninckx PR, Piazze J, Natili M, Colagrande S, Cosmi EV. Correlation between endometriosis and pelvic pain. J Am Assoc Gynecol Laparosc 1999;6:429–34. 15. Milingos S, Protopapas A, Kallipolitis G, Drakakis P, Loutradis D, Liapi A, et al. Endometriosis in patients with chronic pelvic pain: is staging predictive of the efficacy of lap-aroscopic surgery in pain relief? Gynecol Obstet Invest 2006; 62:48–54. 16. Hsu AL, Sinaii N, Segars J, Nieman LK, Stratton P. Relating pelvic pain location to surgical findings of endometriosis. Obstet Gynecol 2011;118:223–30. 17. Howard FM. Endometriosis and mechanisms of pelvic pain. J Minim Invasive Gynecol 2009;16:540–50. 18. Vercellini P, Fedele L, Aimi G, Pietropaolo G, Consonni D, Crosignani PG. Association between endometriosis stage, lesion type, patient characteristics and severity of pelvic pain symptoms: a multivariate analysis of over 1000 patients. Hum Reprod 2007;22:266–71. 19. Fauconnier A, Chapron C, Dubuisson JB, Vieira M, Dousset B, Bréart G. Relation between pain symptoms and the anatomic loca-tion of deep infiltrating endometriosis. Fertil Steril 2002;78:719–26. 20. Gruppo Italiano per lo Studio dell’Endometriosi. Relationship between stage, site and morphological characteristics of pelvic endometriosis and pain. Hum Reprod 2001;16:2668–71. 21. Taran FA, Weaver AL, Coddington CC, Stewart EA. Under-standing adenomyosis: a case control study. Fertil Steril 2010; 94:1223–8. 22. Hammoud A, Gago LA, Diamond MP. Adhesions in patients with chronic pelvic pain: a role for adhesiolysis? Fertil Steril 2004;82:1483–91. 23. Latthe P, Mignini L, Gray R, Hills R, Khan K. Factors predis-posing women to chronic pelvic pain: systematic review. BMJ 2006;332:749–55. 24. Halperin R, Padoa A, Schneider D, Bukovsky I, Pansky M. Long-term follow-up (5-20 years) after uterine ventrosuspension for chronic pelvic pain and deep dyspareunia. Gynecol Obstet Invest 2003;55:216–9. 25. Batioglu S, Zeyneloglu HB. Laparoscopic plication and suspen-sion of the round ligament for chronic pelvic pain and dyspar-eunia. J Am Assoc Gynecol Laparosc 2000;7:547–51. 26. Carter JE. Carter-Thomason uterine suspension and positioning by ligament investment, fixation and truncation. J Reprod Med 1999;44:417–22. 27. Masters WH, Johnson VE; Reproductive Biology Research Foundation. Human sexual response. 1st ed. Boston (MA): Little, Brown; 1966. 28. Duncan CH, Taylor HC Jr. A psychosomatic study of pelvic congestion. Am J Obstet Gynecol 1952;64:1–12. 29. Freedman J, Ganeshan A, Crowe PM. Pelvic congestion syn-drome: the role of interventional radiology in the treatment of chronic pelvic pain. Postgrad Med J 2010;86:704–10. 30. Farquhar CM, Rogers V, Franks S, Pearce S, Wadsworth J, Beard RW. A randomized controlled trial of medroxyproges-terone acetate and psychotherapy for the treatment of pelvic congestion. Br J Obstet Gynaecol 1989;96:1153–62. 31. Soysal ME, Soysal S, Vicdan K, Ozer S. A randomized con-trolled trial of goserelin and medroxyprogesterone acetate in the treatment of pelvic congestion. Hum Reprod 2001;16:931–9. 32. Perry CP. Current concepts of pelvic congestion and chronic pelvic pain. JSLS 2001;5:105–10. 33. Tu FF, Hahn D, Steege JF. Pelvic congestion syndrome-associated pelvic pain: a systematic review of diagnosis and management. Obstet Gynecol Surv 2010;65:332–40. 34. Bukovsky I, Liftshitz Y, Langer R, Weinraub Z, Sadovsky G, Caspi E. Ovarian residual syndrome. Surg Gynecol Obstet 1988;167:132–4. 35. Steege JF. Ovarian remnant syndrome. Obstet Gynecol 1987; 70:64–7. 36. Lamvu G, Robinson B, Zolnoun D, Steege JF. Vaginal apex resec-tion: a treatment option for vaginal apex pain. Obstet Gynecol 2004;104:1340–6. 37. Sharp HT, Dodson MK, Langer KM, Doucette RC, Norton PA. The role of vaginal apex excision in the management of persis-tent posthysterectomy dyspareunia. Am J Obstet Gynecol 2000;183:1385–8. 38. Sinaki M, Merritt JL, Stillwell GK. Tension myalgia of the pelvic floor. Mayo Clin Proc 1977;52:717–22. 39. FitzGerald MP, Payne CK, Lukacz ES, Yang CC, Peters KM, Chai TC, et al. Randomized multicenter clinical trial of myofascial physical therapy in women with interstitial cystitis/painful bladder syndrome and pelvic floor tenderness. J Urol 2012;187:2113–8. 40. Williams RE, Hartmann KE, Sandler RS, Miller WC, Savitz LA, Steege JF. Recognition and treatment of irritable bowel syndrome among women with chronic pelvic pain. Am J Obstet Gynecol 2005;192:761–7. 41. Tu FF, As-Sanie S, Steege JF. Prevalence of pelvic musculoskel-etal disorders in a female chronic pelvic pain clinic. J Reprod Med 2006;51:185–9. 42. Walker E, Katon W, Harrop-Griffiths J, Holm L, Russo J, Hickok LR. Relationship of chronic pelvic pain to psychiatric diagnoses and childhood sexual abuse. Am J Psychiatry 1988; 145:75–80. 43. Slocumb JC, Kellner R, Rosenfeld RC, Pathak D. Anxiety and depression in patients with the abdominal pelvic pain syn-drome. Gen Hosp Psychiatry 1989;11:48–53. 44. Reiter RC, Gambone JC. Demographic and historic variables in women with idiopathic chronic pelvic pain. Obstet Gynecol 1990;75:428–32. 45. Rapkin AJ, Kames LD, Darke LL, Stampler FM, Naliboff BD. History of physical and sexual abuse in women with chronic pelvic pain. Obstet Gynecol 1990;76:92–6. 46. Steege JF, Metzger DA, Levy BS. Chronic pelvic pain: an inte-grated approach. 1st ed. Philadelphia (PA): Saunders; 1998. 47. Yunker A, Steege J. Practical guide to laparoscopic pain map-ping. J Minim Invasive Gynecol 2010;17:8–11. 48. Zupi E, Sbracia M, Marconi D, Zullo F, Santi K, Solima E, et al. Pain mapping during minilaparoscopy in infertile patients with-out pathology. J Am Assoc Gynecol Laparosc 1999;6:51–4. 49. Yeung P Jr, Sinervo K, Winer W, Albee RB Jr. Complete lap-aroscopic excision of endometriosis in teenagers: is postopera-tive hormonal suppression necessary? Fertil Steril 2011;95: 1909–12, 1912.e1. 50. Warner EA. Opioids for the treatment of chronic noncancer pain. Am J Med 2012;125:1155–61. 628 Steege and Siedhoff Chronic Pelvic Pain OBSTETRICS & GYNECOLOGY
  • 14. 51. Manchikanti L, Abdi S, Atluri S, Balog CC, Benyamin RM, Boswell MV, et al. American Society of Interventional Pain Physicians (ASIPP) guidelines for responsible opioid prescrib-ing in chronic non-cancer pain: Part 2—guidance. Pain Physician 2012;15:S67–116. 52. An update on the drug treatment of neuropathic pain. Part 1: antidepressants [published erratum appears in Drug Ther Bull 2012;50:132]. Drug Ther Bull 2012;50:114–7. 53. Fernandez F, Adams F, Holmes VF. Analgesic effect of alpra-zolam in patients with chronic, organic pain of malignant ori-gin. J Clin Psychopharmacol 1987;7:167–9. 54. Ling FW. Randomized controlled trial of depot leuprolide in patients with chronic pelvic pain and clinically suspected endo-metriosis. Pelvic Pain Study Group. Obstet Gynecol 1999;93: 51–8. 55. Brown J, Kives S, Akhtar M. Progestagens and anti-progestagens for pain associated with endometriosis. The Co-chrane Database of Systematic Reviews 2012, Issue 3. Art No.: CD002122. DOI: 10.1002/14651858.CD002122.pub2. 56. Petta CA, Ferriani RA, Abrao MS, Hassan D, Rosa E Silva JC, Podgaec S, et al. Randomized clinical trial of a levonorgestrel-releasing intrauterine system and a depot GnRH analogue for the treatment of chronic pelvic pain in women with endometri-osis. Hum Reprod 2005;20:1993–8. 57. Howard FM. An evidence-based medicine approach to the treatment of endometriosis-associated chronic pelvic pain: placebo-controlled studies. J Am Assoc Gynecol Laparosc 2000;7:477–88. 58. Pavone ME, Bulun SE. Aromatase inhibitors for the treatment of endometriosis. Fertil Steril 2012;98:1370–9. 59. Ferrero S, Gillott DJ, Venturini PL, Remorgida V. Use of aromatase inhibitors to treat endometriosis-related pain symp-toms: a systematic review. Reprod Biol Endocrinol 2011;9:89. 60. Lee NC, Dicker RC, Rubin GL, Ory HW. Confirmation of the preoperative diagnoses for hysterectomy. Am J Obstet Gynecol 1984;150:283–7. 61. Wu JM, Wechter ME, Geller EJ, Nguyen TV, Visco AG. Hys-terectomy rates in the United States, 2003. Obstet Gynecol 2007;110:1091–5. 62. Hartmann KE, Ma C, Lamvu GM, Langenberg PW, Steege JF, Kjerulff KH. Quality of life and sexual function after hysterec-tomy in women with preoperative pain and depression. Obstet Gynecol 2004;104:701–9. 63. Kuppermann M, Learman LA, Schembri M, Gregorich SE, Jackson RA, Jacoby A, et al. Contributions of hysterectomy and uterus-preserving surgery to health-related quality of life. Obstet Gynecol 2013;122:15–25. 64. Nieboer TE, Hendriks JC, Bongers MY, Vierhout ME, Kluivers KB. Quality of life after laparoscopic and abdominal hysterectomy: a randomized controlled trial. Obstet Gynecol 2012;119:85–91. 65. Healey M, Ang WC, Cheng C. Surgical treatment of endome-triosis: a prospective randomized double-blinded trial compar-ing excision and ablation. Fertil Steril 2010;94:2536–40. 66. Wright J, Lotfallah H, Jones K, Lovell D. A randomized trial of excision versus ablation for mild endometriosis. Fertil Steril 2005;83:1830–6. 67. Bassi MA, Podgaec S, Dias JA Jr, D’Amico Filho N, Petta CA, Abrao MS. Quality of life after segmental resection of the rec-tosigmoid by laparoscopy in patients with deep infiltrating endometriosis with bowel involvement. J Minim Invasive Gynecol 2011;18:730–3. 68. Nezhat C, Nezhat F, Nezhat CH, Nasserbakht F, Rosati M, Seidman DS. Urinary tract endometriosis treated by laparos-copy. Fertil Steril 1996;66:920–4. 69. Bianchi S, Busacca M, Agnoli B, Candiani M, Calia C, Vignali M. Effects of 3 month therapy with danazol after lapa-roscopic surgery for stage III/IV endometriosis: a randomized study. Hum Reprod 1999;14:1335–7. 70. Morgante G, Ditto A, La Marca A, De Leo V. Low-dose dana-zol after combined surgical and medical therapy reduces the incidence of pelvic pain in women with moderate and severe endometriosis. Hum Reprod 1999;14:2371–4. 71. Telimaa S, Rönnberg L, Kauppila A. Placebo-controlled com-parison of danazol and high-dose medroxyprogesterone acetate in the treatment of endometriosis after conservative surgery. Gynecol Endocrinol 1987;1:363–71. 72. Hornstein MD, Hemmings R, Yuzpe AA, Heinrichs WL. Use of nafarelin versus placebo after reductive laparoscopic surgery for endometriosis. Fertil Steril 1997;68:860–4. 73. Vercellini P, Frontino G, De Giorgi O, Aimi G, Zaina B, Crosignani PG. Comparison of a levonorgestrel-releasing intra-uterine device versus expectant management after conservative surgery for symptomatic endometriosis: a pilot study. Fertil Steril 2003;80:305–9. 74. Ferrero S, Camerini G, Seracchioli R, Ragni N, Venturini PL, Remorgida V. Letrozole combined with norethisterone acetate compared with norethisterone acetate alone in the treatment of pain symptoms caused by endometriosis. Hum Reprod 2009; 24:3033–41. 75. Latthe PM, Proctor ML, Farquhar CM, Johnson N, Khan KS. Surgical interruption of pelvic nerve pathways in dysmenor-rhea: a systematic review of effectiveness. Acta Obstet Gynecol Scand 2007;86:4–15. 76. Zullo F, Palomba S, Zupi E, Russo T, Morelli M, Sena T, et al. Long-term effectiveness of presacral neurectomy for the treat-ment of severe dysmenorrhea due to endometriosis. J Am Assoc Gynecol Laparosc 2004;11:23–8. 77. Kaminski PF, Sorosky JI, Mandell MJ, Broadstreet RP, Zaino RJ. Clomiphene citrate stimulation as an adjunct in locat-ing ovarian tissue in ovarian remnant syndrome. Obstet Gynecol 1990;76:924–6. VOL. 124, NO. 3, SEPTEMBER 2014 Steege and Siedhoff Chronic Pelvic Pain 629