18
REVIEWS AND COMMENTARY n STATE OF THE ART Radiology: Volume 280: Number 3—September 2016 n radiology.rsna.org 675 1 From the Department of Radiology, Mount Sinai Beth Israel, New York, NY (J.E.S.); West Cancer Center, University of Tennessee Health Science Center, 7945 Wolf River Boulevard, Germantown, TN 38138 (D.K.P.); Department of Radiology and Medical Imaging, University of Virginia Health System, Charlottesville, Va (A.H.S.); and Department of Radiology, MedStar Georgetown University Hospital, Washington, DC (J.B.S.). Received August 5, 2014; revision requested September 18; revision received December 1; accepted December 16; final version accepted December 23. Address correspondence to D.K.P. (e-mail: dpowell@ westclinic.com). q RSNA, 2016 Uterine fibroids occur in approximately 50% of women over the age of 40 years, and an estimated 50% of those are symptomatic. Menorrhagia is the most common symptom and the primary indication for treatment, al- though bulk symptoms often occur and can be treated. Pharmacotherapy is typically inadequate unless it can be expected to successfully bridge to menopause or al- low for a less-invasive intervention. However, hormonal therapies have risks. Hysterectomy is still the most com- monly performed procedure for symptomatic fibroids and has the lowest rate of reintervention (compared with myomectomy or uterine artery embolization [UAE]), but rates of more serious complications are higher and pa- tient satisfaction and ability to return to normal activities may also be less favorable. Myomectomy is not necessarily less morbid than hysterectomy and may have a greater failure rate than UAE. Techniques and devices vary with little standardization, and operator experience is crucial to success. The largest studies of UAE show very low rates of serious complications and rapid recovery. UAE signif- icantly improves symptoms related to uterine fibroids in 85%–90% of patients. Herein, this article will discuss the nature of fibroids and their diagnosis, pharmacotherapy, surgical treatment, and nonsurgical interventional treat- ment, including UAE and magnetic resonance–guided fo- cused ultrasound. q RSNA, 2016 James E. Silberzweig, MD Daniel K. Powell, MD Alan H. Matsumoto, MD James B. Spies, MD, MPH Management of Uterine Fibroids: A Focus on Uterine-sparing Interventional Techniques 1 Learning Objectives: After reading the article and taking the test, the reader will be able to: n Describe the various treatment options for the management of uterine fibroids n Describe the treatment complications and outcomes for management of uterine fibroids n Identify the imaging and anatomic features relevant to management of uterine fibroids Accreditation and Designation Statement The RSNA is accredited by the Accreditation Council for Continuing Medical Education (ACCME) to provide continuing medical education for physicians. The RSNA designates this journal-based SA-CME activity for a maximum of 1.0 AMA PRA Category 1 Credit . Physicians should claim only the credit commensurate with the extent of their participation in the activity. Disclosure Statement The ACCME requires that the RSNA, as an accredited provider of CME, obtain signed disclosure statements from the authors, editors, and reviewers for this activity. For this journal-based CME activity, author disclosures are listed at the end of this article. Online SA-CME See www.rsna.org/education/search/ry This copy is for personal use only. To order printed copies, contact [email protected]

Management of Uterine Fibroids - CIRwebcir.org/.../2016/4_noviembre/rsna/management_uterine_fibroids.pdf · icantly improves symptoms related to uterine fibroids in 85%–90% of patients

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Management of Uterine Fibroids - CIRwebcir.org/.../2016/4_noviembre/rsna/management_uterine_fibroids.pdf · icantly improves symptoms related to uterine fibroids in 85%–90% of patients

REVIEWS AND COM

MENTARY

n State of the art

Radiology: Volume 280: Number 3—September 2016 n radiology.rsna.org 675

1 From the Department of Radiology, Mount Sinai Beth Israel, New York, NY (J.E.S.); West Cancer Center, University of Tennessee Health Science Center, 7945 Wolf River Boulevard, Germantown, TN 38138 (D.K.P.); Department of Radiology and Medical Imaging, University of Virginia Health System, Charlottesville, Va (A.H.S.); and Department of Radiology, MedStar Georgetown University Hospital, Washington, DC (J.B.S.). Received August 5, 2014; revision requested September 18; revision received December 1; accepted December 16; final version accepted December 23. Address correspondence to D.K.P. (e-mail: [email protected]).

q RSNA, 2016

Uterine fibroids occur in approximately 50% of women over the age of 40 years, and an estimated 50% of those are symptomatic. Menorrhagia is the most common symptom and the primary indication for treatment, al-though bulk symptoms often occur and can be treated. Pharmacotherapy is typically inadequate unless it can be expected to successfully bridge to menopause or al-low for a less-invasive intervention. However, hormonal therapies have risks. Hysterectomy is still the most com-monly performed procedure for symptomatic fibroids and has the lowest rate of reintervention (compared with myomectomy or uterine artery embolization [UAE]), but rates of more serious complications are higher and pa-tient satisfaction and ability to return to normal activities may also be less favorable. Myomectomy is not necessarily less morbid than hysterectomy and may have a greater failure rate than UAE. Techniques and devices vary with little standardization, and operator experience is crucial to success. The largest studies of UAE show very low rates of serious complications and rapid recovery. UAE signif-icantly improves symptoms related to uterine fibroids in 85%–90% of patients. Herein, this article will discuss the nature of fibroids and their diagnosis, pharmacotherapy, surgical treatment, and nonsurgical interventional treat-ment, including UAE and magnetic resonance–guided fo-cused ultrasound.

q RSNA, 2016

James E. Silberzweig, MDDaniel K. Powell, MDAlan H. Matsumoto, MDJames B. Spies, MD, MPH

Management of Uterine Fibroids: A Focus on Uterine-sparing Interventional Techniques1

Learning Objectives:

After reading the article and taking the test, the reader will be able to:

n Describe the various treatment options for the management of uterine fibroids

n Describe the treatment complications and outcomes for management of uterine fibroids

n Identify the imaging and anatomic features relevant to management of uterine fibroids

Accreditation and Designation StatementThe RSNA is accredited by the Accreditation Council for Continuing Medical Education (ACCME) to provide continuing medical education for physicians. The RSNA designates this journal-based SA-CME activity for a maximum of 1.0 AMA PRA Category 1 Credit™. Physicians should claim only the credit commensurate with the extent of their participation in the activity.

Disclosure Statement

The ACCME requires that the RSNA, as an accredited provider of CME, obtain signed disclosure statements from the authors, editors, and reviewers for this activity. For this journal-based CME activity, author disclosures are listed at the end of this article.

Online SA-CMESee www.rsna.org/education/search/ry

This copy is for personal use only. To order printed copies, contact [email protected]

Silvisol
Rectángulo
Page 2: Management of Uterine Fibroids - CIRwebcir.org/.../2016/4_noviembre/rsna/management_uterine_fibroids.pdf · icantly improves symptoms related to uterine fibroids in 85%–90% of patients

676 radiology.rsna.org n Radiology: Volume 280: Number 3—September 2016

STATE OF THE ART: Management of Uterine Fibroids Silberzweig et al

which when it occurs, results in hyaline or myxoid degeneration (6).

There is controversy over the effect of fibroids on fertility. However, a meta-analysis showed lower pregnancy rates and a greater incidence of miscarriage in patients with submucosal fibroids (12). Similarly, removal of submucosal fibroids has resulted in an increased rate of pregnancy (13). Fibroids have been shown to increase pregnancy risks, including intrauterine growth re-striction, placental abruption, preterm delivery, premature rupture of mem-branes, abnormal fetal lie, cesarean section, and blood loss (14).

Imaging Classification

Fibroids can be classified by their lo-cation: submucosal, intramural, and subserosal (9) (Fig 1). Pedunculated fibroids can be intracavitary or exo-phytic (9) and can undergo torsion or derive new blood supply from adjacent structures (parasitize) (6). Intramural fibroids are the most common type of fibroid, while submucosal fibroids are the least common type. However, a sub-mucosal fibroid may be more symptom-atic than a subserosal or small intra-mural fibroid, owing to its relationship with the endometrium. Bulk symptoms are more often seen with large intramu-ral or subserosal fibroids (6).

Although a high-quality ultrasonog-raphy (US) examination may be suffi-cient for evaluation in patients with straightforward cases of fibroids (for instance to estimate the size of a dom-inant fibroid), imaging evaluation is most reliably performed with magnetic resonance (MR) imaging to determine

of women 40 years of age, peaking in the 5th decade of life (1,2). Although an estimated 50% of women with fi-broids experience symptoms (3), epide-miology studies demonstrate a two to nine times increased prevalence among African-Americans and a peak inci-dence occurring 5–10 years earlier as compared with Caucasians (2,4). Rates among Asians and Hispanics appear to be comparable to Caucasians (2). Fibroids are thought to arise from the myometrium following neoplastic trans-formation of a single smooth muscle cell (5), with subsequent development of a connective tissue component and a pseudocapsule (6). Fibroid pathogen-esis remains to be elucidated and there is controversy over the contribution of proposed causes, including cytogenetic abnormalities, abnormal extracellular matrix expression, growth factors, cy-tokines, and chemokines (7). There is increasing evidence that progesterone receptors have a major impact on the growth of fibroids (8).

Factors associated with a reduced risk for fibroids include a later age of menarche, menopause, and child bear-ing. Increased risk for fibroids has been associated with use of estrogens, obe-sity, red meat consumption, a family history of fibroids, tubal ligation, prior pelvic inflammatory disease, prior in-trauterine device expulsion, hyperten-sion, and diabetes (2).

Clinical Features (Signs and Symptoms)

Symptomatic fibroids are most com-monly associated with menorrhagia (9) and are the most common cause of abnormal menstrual bleeding (10). Uterine fibroids may also cause meno-metrorrhagia, anemia, dysmenorrhea, dyspareunia, chronic (pelvic and back) pain, bloating, pelvic fullness, consti-pation, tenesmus, infertility, hydrone-phrosis, and urinary frequency (9). The growth of fibroids appears to be hor-monally driven (9), accounting for their peak occurrence during reproductive age, enlargement during pregnancy, and regression after menopause (6). Fi-broids are susceptible to ischemia (11),

Published online10.1148/radiol.2016141693 Content code:

Radiology 2016; 280:675–692

Abbreviations:FDA = Food and Drug AdministrationFUS = focused ultrasoundGnRHa = gonadatropin-releasing hormone agonistHIFU = high-intensity focused ultrasoundUAE = uterine artery embolization

Conflicts of interest are listed at the end of this article.

Essentials

n Preprocedural MR imaging is a useful tool for the classification of fibroids, to determine the best method of treatment, and to detect other potential causes of symptoms.

n Pharmacotherapy can bridge the perimenopausal period to meno-pause or result in a reduction in fibroid volume and blood loss to possibly allow a less-invasive in-tervention; however, hormonal therapies have risks.

n Hysterectomy remains the most commonly performed procedure for symptomatic fibroids and has the lowest rate of reintervention (compared with myomectomy or uterine artery embolization [UAE]), but rates of more se-rious complications are higher and patient satisfaction and ability to return to normal activ-ities may also be less favorable.

n Myomectomy is not necessarily less morbid than hysterectomy and may have a greater failure rate than UAE; techniques and devices vary with little standardi-zation and operator experience is crucial to success.

n The largest studies of UAE show rapid recovery and very low rates of serious complications; UAE significantly improves symptoms related to uterine fibroids in 85%–90% of patients.

n MR-guided focused ultrasound may have an application for the treatment of patients with symp-tomatic fibroids that are limited in number and volume, demon-strate no calcifications, show ho-mogeneously low signal intensity on T2-weighted MR images, and can be safely targeted with the MR-guided focused ultrasound technology.

U terine fibroids (leiomyomas) are the most common benign neo-plasm of the female genitourinary

tract and are present in more than 50%

Page 3: Management of Uterine Fibroids - CIRwebcir.org/.../2016/4_noviembre/rsna/management_uterine_fibroids.pdf · icantly improves symptoms related to uterine fibroids in 85%–90% of patients

Radiology: Volume 280: Number 3—September 2016 n radiology.rsna.org 677

STATE OF THE ART: Management of Uterine Fibroids Silberzweig et al

with any hormone regimen compared with management with hysterectomy (311 patients), showed that 25% of medically treated patients went on to undergo hysterectomy, while 25% of patients with abnormal bleeding and 50% with chronic pain still complained

are often associated with alleviation of symptoms, large fibroids may not shrink for some time after menopause and treatment may be indicated (9).

PharmacotherapyMedical therapy is limited in the man-agement of symptomatic fibroids. Oral contraceptive pills are a common first-line therapy (23) but are mainly used to regulate the menstrual cycle and may not reduce the volume of menstrual flow. Oral contraceptive pills carry side effects, including a 20%–40% increased risk of stroke in older smokers with hypertension (23), venous thrombo-embolism, liver adenomas, gallstones, weight gain, acne, breast tenderness, fluid retention, mood disturbances, and sexual dysfunction (9). Levonorgestrel-releasing intrauterine contraceptive de-vices have also been shown to decrease fibroid-associated menorrhagia (9,24). However, progestin-only treatments have not shown any benefit in reduc-ing fibroid size (9). The Maine Wom-en’s Health Study, which examined management of fibroids (380 patients)

the characteristics, number, size, and location of fibroids and to assess for other pathologic conditions such as adenomyosis (15–17). The recognition of adenomyosis is important as it may alter the treatment approach (18), pa-tient counseling, and expectations (19). Adenomyosis is a diffuse or focal endo-metrial basal layer invasion of the myo-metrium that can cause symptoms simi-lar to fibroids, and it is best seen on MR images as a thickened junctional zone (.12 mm), with scattered T1- and T2-hyperintense foci of endometrial glands (6) (Fig 2). Studies reveal that pretreat-ment MR imaging evaluation may alter therapy in about 20% of patients with fibroids (16,20).

Noninterventional and Surgical Treatment Options

Observational placebo studies have largely shown no improvement in fi-broid-related symptoms with watchful waiting prior to menopause (9,21,22). Moreover, while decreasing estrogen levels in the perimenopausal period

Figure 1

Figure 1: Illustration of fibroid location classification. A type I utero-ovarian anastomosis is depicted on the left of the figure (straight arrow) with flow from the ovarian artery toward the uterus in the tubo-ovarian segment feeding into the intramural portion of the uterine artery (curved arrowheads indicate direction of blood flow). A type II utero-ovarian anastomosis is depicted on the right of the figure (open arrowhead) with at least partial direct supply of the fibroid by the ovarian artery without prior connection to the uterine artery. Note that less common extrauterine locations, such as cervical or broad ligament fibroids, are not included (the incidences of which have not been well studied).

Figure 2

Figure 2: Sagittal T2-weighted MR image of a uterus with adenomyosis, indicated by junctional zone thickening greater than 12 mm (T2- and T1-hyperintense foci of myometrial glandular tissue support the diagnosis, not depicted here).

Page 4: Management of Uterine Fibroids - CIRwebcir.org/.../2016/4_noviembre/rsna/management_uterine_fibroids.pdf · icantly improves symptoms related to uterine fibroids in 85%–90% of patients

678 radiology.rsna.org n Radiology: Volume 280: Number 3—September 2016

STATE OF THE ART: Management of Uterine Fibroids Silberzweig et al

menorrhagia in patients with fibroids and minimal bulk symptoms who are finished with child-bearing since the endometrial cavity develops synechiae (9). However, results are inconsistent and not always enduring (44). Uterine artery ligation, performed either lapa-roscopically or transvaginally, has not been well studied (9). A small random-ized trial of UAE versus laparoscopic uterine artery occlusion showed su-periority of UAE: 17% symptom re-currence and 100% rate of complete fibroid infarction at MR imaging with UAE, compared with 48% symptom recurrence and 23% fibroid infarc-tion with ligation (45). Another study comparing 225 laparoscopic uterine artery occlusion and 112 UAE patients reported equivalent fibroid volume re-ductions of 41.2% and 49.2%, respec-tively, without reporting on symptom improvements (46).

Uterine Artery (Fibroid) Embolization

Procedure HistoryUAE was first performed in France in 1994 by Merland, a neuroradiologist, who subsequently collaborated with Ravina, a gynecologist, to reduce blood loss prior to myomectomy (47). Sub-sequently, in 1997, Goodwin et al re-ported on a series of UAE patients in the United States with fibroid-related menorrhagia, demonstrating success-ful reduction of fibroid-related symp-toms (48). The procedure has since been validated as a safe, efficacious, and cost-effective treatment option for symptomatic fibroids. About 14 000 UAEs are estimated to be performed per year in the United States (49).

Pelvic Arterial Anatomy and PitfallsThe internal iliac (hypogastric) artery is classically described as having ante-rior and posterior divisions, with the uterine artery arising from the anterior division, but this appears to occur only 51% of the time (50). Alternatively, four angiographic patterns of uterine artery branching have been described from the superior gluteal, inferior glu-teal, and the internal iliac (hypogastric)

years in 2005 were for fibroids (ap-proximately 200 000 per year) (32). Open hysterectomy still accounts for close to two-thirds of all hysterec-tomies performed (32).

In general, myomectomy is best suited for the treatment of one to three dominant fibroids in accessible loca-tions (9). About one-fifth of patients who undergo myomectomy will require a second surgery at 5 years (33). A me-ta-analysis suggests that laparoscopic myomectomy results in fewer compli-cations, less pain, and shorter recov-ery compared with open myomectomy (14,34).

A variety of techniques and de-vices may be used in the laparoscopic removal of fibroids, including dissec-tion, enucleation, morcellation, and thermal myolysis (1,35–37). A recent controversy has arisen over the use of power morcellation with laparoscopic hysterectomy and myomectomy due to the potential risk of disseminating unsuspected uterine sarcoma. A ret-rospective report of 1091 uterine mor-cellations found a 0.09% rate of unex-pected sarcoma and 1.2% incidence of unsuspected neoplasms, with dissemi-nation occurring in two-thirds of these cases (38). A smaller review found one case (1% risk) of unsuspected atypical tumor after uterine morcellation in a low-risk population (39). Retrospec-tive reviews of surgical specimens in 1332 patients and 923 patients found a 0.23% rate of uterine sarcoma (with a 0.27% rate in the subset with rapidly growing fibroids) (40) and a 0.4% rate of unsuspected malignancy in hysterec-tomy specimens, respectively (41).

As such, a 2014 U.S. Food and Drug Administration (FDA) safety communi-cation discourages the use of laparo-scopic power morcellation, estimating a one in 350 risk for an unsuspected sarcoma and potential pelvic dissemi-nation (42). The American Congress of Obstetricians and Gynecologists issued a special report on the use of power morcellation in May 2014, suggesting an incidence of two in 1000 for unsus-pected sarcoma (43).

Endometrial thermal ablation is an option for the treatment of

of substantial symptoms after medical treatment (21).

Gonadatropin-releasing hormone agonists (GnRHas), such as leupro-lide (eg, Eligard; Sanofi-Aventis, Que-bec, Canada), create a hypoestrogenic menopause-like state by means of competitive inhibition on the pituitary-hypothalamic axis (9,25). GnRHa ther-apy shrinks fibroids by 35% within 8 weeks on average, with induction of amenorrhea (9,25). Maximum effects are achieved after 24 weeks of therapy, with volume reductions up to 70% (1), but return of symptoms and original fi-broid size recur in a similar timeframe after drug cessation (9,26). Long-term treatment is limited in 95% by side ef-fects. As such, GnRHa therapy may be considered in perimenopausal women as a bridge to menopause (1). GnRHa therapy has also been recommended for short-term use (,6 months) to fa-cilitate myomectomy, hysterectomy, or MR-guided focused ultrasound (FUS) therapy and allow for a preprocedure rise in hematocrit and shrinkage of the fibroid volume (1). However, these medications may be associated with more difficulty in surgical excision dur-ing myomectomy and higher recurrence rates. Therefore, presurgical GnRHa use is not universally accepted (14).

Surgical TreatmentHysterectomy is a definitive therapy for uterine fibroids with the lowest re-intervention rates and the highest rate of symptom relief in a nonrandomized controlled study compared with uter-ine artery embolization (UAE), myo-mectomy, and control subjects (27). However, only 72% of women report-ed improvement of symptoms after hysterectomy in the Maine Women’s Health Study (9,21). Moreover, while patient satisfaction (28) and major complication rates have been compa-rable in some studies (29), a number of large trials have shown greater rates of major complications with surgery (30,31) and patient dissatisfaction (31) compared with UAE. While hysterec-tomy rates for symptomatic fibroids are decreasing, 25% of hysterectomies performed in women younger than 45

Page 5: Management of Uterine Fibroids - CIRwebcir.org/.../2016/4_noviembre/rsna/management_uterine_fibroids.pdf · icantly improves symptoms related to uterine fibroids in 85%–90% of patients

Radiology: Volume 280: Number 3—September 2016 n radiology.rsna.org 679

STATE OF THE ART: Management of Uterine Fibroids Silberzweig et al

arteries, 90% of the time either trifur-cating with the superior gluteal and inferior gluteal arteries or arising as a first branch from the inferior gluteal artery (with near equal frequency) (51) (Fig 3).

The uterine artery has descending, transverse, ascending, and multiple in-tramural segments. The cervicovaginal branch typically arises from the mid- to distal portion of the transverse seg-ment, and the vaginal artery can have a common trunk with the uterine artery (10). These vessels should be delineated and avoided if possible during UAE to minimize the risk for vaginal mucosal ulceration, loss of sexual function (52), and dyspareunia (53,54) (Fig 4).

Utero-ovarian anastomoses may also be present, which can increase the risk of nontarget ovarian emboliza-tion, such as with type I anastomoses

Figure 3

Figure 3: Illustration of an angiographic classification of uterine artery branching patterns (IGA = inferior gluteal artery, SGA = superior gluteal artery, UA = uterine artery). Type I: Uterine artery as first branch of inferior gluteal artery. Type II: Uterine artery as second or third branch of inferior gluteal artery. Type III: Trifurcation. Type IIIa: Uterine artery and superior gluteal artery bifurcation with inferior gluteal artery as a branch of the latter. Type IV: Uterine artery as a first branch of the internal iliac artery. Type I and III comprise about 90% of cases with equal frequency.

Figure 4

Figure 4: Angiographic images before and after UAE. (a) Digital subtraction angiogram with a reverse curve catheter in the contralateral internal iliac artery opacifying type I or type III branching anatomy and a hypertrophied uterine artery (UA) and perifibroidal plexus. Note the cervicovaginal branch arising from the proximal uterine artery (arrows). (b) Postembolization image with evidence of successful embolization through a catheter tip (arrow) distal to the level of the cervicovaginal branch to avoid embolizing that artery.

Page 6: Management of Uterine Fibroids - CIRwebcir.org/.../2016/4_noviembre/rsna/management_uterine_fibroids.pdf · icantly improves symptoms related to uterine fibroids in 85%–90% of patients

680 radiology.rsna.org n Radiology: Volume 280: Number 3—September 2016

STATE OF THE ART: Management of Uterine Fibroids Silberzweig et al

Jordan, Utah) and polyvinyl alcohol particles (various manufacturers). Spherical polyvinyl alcohol particles have shown comparatively unaccept-able rates of failure in three random-ized controlled trials (59,65,66). Gel-atin sponge embolization has been successfully used, although considered less precise, and has recently been studied in Korea with superior results compared with embolization using poly-vinyl alcohol (61). Polyzene F-coated hydrogel microspheres (67,68) and a pingyangmycin lipiodol emulsion and silk-segment embolic (69) have shown good results.

The arteriographic end point of embolization was initially described as a static column of contrast material in the uterine artery based on the use of polyvinyl alcohol particles. However, with calibrated microspheres, it is suffi-cient to observe sluggish antegrade flow in the uterine artery (53) (visualization of contrast material for at least five cardiac cycles) (59) and a pruned-tree

Technical Details and CostMost UAE procedures are performed with intravenous sedation (29) by means of a right common femoral ap-proach with a reverse curved 4- or 5-F catheter to access the contralateral in-ternal iliac artery followed by formation of a Waltman loop to access the ipsi-lateral internal iliac artery (48,59–61) (Fig 5). Some investigators suggest the routine use of a microcatheter, for ex-ample, a 2.7-F Progreat (Terumo, Som-erset, NJ) to preserve flow in the uter-ine artery and facilitate access to distal branches (48,59,61–64).

Perifibroidal plexus vessels are gen-erally 500–800 microns in caliber (56), which is the basis for the selection of either 500–700 or 700–900-micron–sized particles (59). In the United States and Europe, particulate material is by far the most common embolic agent used for UAE. Several embolic materials are FDA-approved for UAE of uterine fibroids, including tris-acryl gelatin spheres (Embospheres, Merit, South

(21.7%, ovarian to intramural uterine flow) (Fig 5) or type III (6.6%, tubo-ovarian segment to ovarian flow) (55) (Figs 1, 6). The use of microspheres greater than 500 microns theoretically helps avoid nontarget passage of the embolic agent through these anas-tomoses (56). Additionally, a type II utero-ovarian anastomosis may be a cause of UAE failure due to partial or complete arterial supply of the fibroid directly from the ovarian artery (3.9% incidence) (55) (Fig 1). Ovarian artery catheterization and embolization may be required (53) (Fig 7).

Many operators forgo an initial aortography examination and perform a completion aortography examination (10). However, aortography increases the estimated radiation dose by 20% and has an 18% sensitivity for detecting collateral ovarian artery supply (57). The effects on dose of cone-beam com-puted tomography to identify regions of persistent perfusion within fibroids remains unclear (58).

Figure 5

Figure 5: Illustration of a reversed curve catheter in the iliac arteries and aorta from a common femoral approach, forming a Waltman loop to access the ipsilateral internal iliac artery with a microcatheter accessing the uterine artery. The inset depicts ideal position of the microcatheter tip for embolization, bypassing the cervicovaginal branches and a type III utero-ovarian anas-tomosis (straight arrow), where there is at least partial supply of the ovary from the tubo-ovarian segment of the uterine artery (curved arrows indicate direction of blood flow). While embolic material should not be injected into this anastomosis, studies have suggested that these measure less than 500 microns and standard 500–700 and 700–900 micron particles should bypass these anastomoses.

Page 7: Management of Uterine Fibroids - CIRwebcir.org/.../2016/4_noviembre/rsna/management_uterine_fibroids.pdf · icantly improves symptoms related to uterine fibroids in 85%–90% of patients

Radiology: Volume 280: Number 3—September 2016 n radiology.rsna.org 681

STATE OF THE ART: Management of Uterine Fibroids Silberzweig et al

menorrhagia, anemia, and bulk-relat-ed symptoms (77). Contraindications to UAE include pelvic inflammatory disease, uterine malignancy, or preg-nancy. Relative contraindications include severe renal insufficiency not managed by dialysis or a refractory coagulopathy (10). Pedunculated subserosal fibroids

increases in radiation-induced cancer” and that it is “high unlikely that the radiation dose from UAE alone would directly cause infertility” (75).

Patient SelectionIndications for UAE include uterine fi-broids causing symptoms, primarily

appearance of the perifibroidal plexus (54) (Fig 8).

Preprocedure administration of 1 g cefazolin (eg, Ancef; GlaxoSmithKline, Philadelphia, Pa) intravenously is a widely accepted prophylactic antibiotic of choice. However, this has not been vali-dated and the risks of a side effect from the antibiotic may be greater than those of an infection (10). The presence of an intrauterine device, or IUD, does not ap-pear to increase the risk of endometritis (70). Abstention from use of tampons, swimming, bathing, or sexual intercourse is likely only necessary for 7–10 days.

Aggressive postprocedure pain management allows same-day discharge with a medication regimen consisting of a narcotic analgesic, a nonsteroidal anti-inflammatory drug, and an anti-emetic, tapering these medications over the course of 7 days. Patients should be educated about postembolization syndrome (pyrexia, pain, and nausea) and how to manage it. This syndrome occurs in at least half of patients (28,29) and lasts approximately 72 hours (10).

Multiple studies have demonstrated significantly shorter mean hospital stays with UAE when compared with surgical treatment (from one-half to one-fifth of the time), with an associated cost savings (28–30,71) and a greater net hospital income with UAE (72). How-ever, there may be no long-term savings given the higher rates of reintervention with UAE (73).

Radiation dose for UAE is directly in the middle range for interventional radiology procedures, including elective and emergency procedures (74). Mean effective dose associated with UAE has been reported to be 77.8 mSv, which can be decreased to 11.3 mSv after in-tense dose reduction (including avoid-ing digital subtraction angiography), with doses as low as 3.3 mSv in one study (the equivalent of three abdom-inal radiographs) (75). Calculated ab-sorbed skin doses have been reported as representing only 2.2% of the risk from medical radiation (75), rarely ap-proaching thresholds for transient ery-thema (76). A review of radiation dose studies concludes that standard UAE “is very unlikely to cause any significant

Figure 6

Figure 6: Digital subtraction flush aortogram demonstrates hypertrophied uterine arteries (arrows) and ovarian arteries with type I utero-ovarian anastomoses, where the ovarian arteries continue as the tubo-ovarian segment of the uterine arteries connecting to the intramural segment of the uterine arteries. OA = ovarian artery, IMA = inferior mesenteric artery, Int. Iliac = internal iliac artery, Ext. Iliac = external iliac artery, SGA = superior gluteal artery, IGA = inferior gluteal artery, UA = uterine artery, bottom OA = obturator artery (off internal iliac), PA = pudendal artery.

Page 8: Management of Uterine Fibroids - CIRwebcir.org/.../2016/4_noviembre/rsna/management_uterine_fibroids.pdf · icantly improves symptoms related to uterine fibroids in 85%–90% of patients

682 radiology.rsna.org n Radiology: Volume 280: Number 3—September 2016

STATE OF THE ART: Management of Uterine Fibroids Silberzweig et al

Recent studies have demonstrated equivalent safety and efficacy of UAE even in patients with large (.10 cm) fibroids or uterine volumes (.700 mL) compared with those with smaller vol-umes (81,82). Efficacy has even been shown in 22 patients with “megauter-us” (.1600 mL), 80% of whom avoided hysterectomy (83).

Adenomyosis (with or without fi-broids) can be treated with UAE, al-though the patient should be counseled that the results might not be as durable as with symptoms due to fibroids alone. Small studies have shown substan-tial short-term symptom improvement in 84% (84) and 92.3% (85). How-ever, 2-year results from another small study showed higher recurrence rates compared with UAE for fibroids (19). Better outcomes may be seen in adeno-myosis with lower T2 signal intensity on MR images, focal areas of adenomyosis (18,86), and use of smaller particles (18) and hydrogel-coated microspheres

The role of endometrial biopsy and Papanicolau test to exclude cancer prior to UAE has not been defined. Un-fortunately, malignancy is not reliably distinguished from leiomyomata, either by means of symptoms or imaging. Re-ports that diffusion-weighted imaging and apparent diffusion coefficient char-acterization of presumed fibroids may be used to predict risk of underlying leiomyosarcoma have yet to be sub-stantiated (80). Moreover, two retro-spective studies of patients with rapid fibroid growth suggest no increased risk for malignancy and that premenopausal women with symptoms and imaging findings consistent with fibroids can be treated without undue consideration for sarcoma (38,40). However, a dis-cussion should occur with the patient about the small risk (approximately one in 350–1000 or less) (38,40,43) that a malignancy could be present, under-standing that this risk is probably less in younger patients.

had once been considered a relative contraindication due to the theoretical risk of sloughing the fibroid into the peritoneal cavity or bowel and peri-toneal inflammation (78). However, a prospective study of UAE in 12 pa-tients with 15 pedunculated fibroids showed no serious complications and symptom improvement in all patients at 2 years of follow-up (79). The risk of intracavitary dislodgement and ex-pulsion of pedunculated submuco-sal fibroids, requiring extraction, has not been well studied. A review of 39 UAE patients with submucosal fibroids suggests that an interface-dimension ratio may be used in predicting the likelihood of fibroid sloughing into the uterine cavity. In this small study, the mean ratio was 0.65 in those fibroids that became at least partially intracavi-tary compared with 0.32 in those that did not and 0.81 in those fibroids that became completely intracavitary (64) (Fig 9).

Figure 7

Figure 7: Angiographic images before and after UAE. (a) Opacification of the left perifibroidal plexus from a catheter tip in the distal left uterine artery. Note the rel-atively small arteries feeding the cranial portion of the fibroid (arrow). (b) Digital subtraction flush aortogram demonstrates a hypertrophied left ovarian artery (arrow). (c) Digital subtraction aortogram of a subselected left ovarian artery demonstrates a type II utero-ovarian anastomosis with partial supply of the fibroid (arrow) directly from the ovarian artery, which can be a cause of treatment failure unless a separate ovarian embolization is performed. Note the peri-ovarian plexus (arrowhead) also supplied by the ovarian artery, which should be avoided with embolization.

Page 9: Management of Uterine Fibroids - CIRwebcir.org/.../2016/4_noviembre/rsna/management_uterine_fibroids.pdf · icantly improves symptoms related to uterine fibroids in 85%–90% of patients

Radiology: Volume 280: Number 3—September 2016 n radiology.rsna.org 683

STATE OF THE ART: Management of Uterine Fibroids Silberzweig et al

(68). Diffuse adenomosis with general-ized thickening of the junctional zone seems to be a useful predictor for re-currence after UAE (68).

OutcomesA fibroid-specific self-administered uterine fibroid symptom and quality of life (UFS-QoL) questionnaire has been developed to assess symptom resolu-tion and quality of life, including a men-orrhagia questionnaire and sexual func-tion scale (87). Its initial development and validation showed the ability of this questionnaire to discriminate between UAE patients and normal volunteers as well as between UAE patients with dif-ferent outcomes and has proved to very important in the assessment of quality of life after UAE and for the FIBROID registry (87–89).

A number of landmark studies have shown greater safety and shorter hos-pital stays (on average by 4 days) with UAE compared with hysterectomy (9,28,30,90). A 2012 Cochrane review of five randomized controlled trials of UAE compared with hysterectomy and

Figure 8

Figure 8: Angiographic images before and after UAE. (a) Digital subtraction angiogram with a reverse curve catheter forming a Waltman loop with the tip in the right internal iliac artery opacifying a probable trifurcation into the superior gluteal artery (SGA), inferior gluteal artery (IGA), and uterine artery (UA). Note the hypertrophied uterine artery (arrowhead) and perifibroidal plexus (arrows). (b) Magnified oblique fluoroscopic image with a reverse curve catheter in the internal iliac artery and a microcatheter in the uterine artery shows evidence of successful embolization, including pruning of the perifibroidal plexus, sluggish antegrade flow in the uterine artery (arrowhead) and reflux of contrast material into the superior and inferior gluteal arteries (arrows).

Figure 9

Figure 9: Sagittal T2-weighted MR image of a uterus with a submucosal fibroid and superimposed interface-diameter measurement lines, which can be used to predict the likelihood of fibroid expulsion after UAE (those with ratios greater than 0.65 and 0.81 are more likely to become at least partially intracavitary or entirely intracavitary, respectively) (64). Note the interface line was measured in two strokes to approximate the arc of the subtended endometrial component.

myomectomy concluded that UAE has a similar patient satisfaction rate com-pared with surgical treatments, while being associated with shorter length of hospital stay and disability (91). How-ever, UAE had more minor complica-tions and a higher likelihood of sur-gical reintervention within 2–5 years (91). While overall complication rates are higher with UAE in some studies (25%–50%), at least half of the report-ed complications are due to postembo-lization syndrome and are rarely seri-ous (28,30). In contrast, hysterectomy complications are more often severe (28). Reported rates of clinical suc-cess after UAE are high, ranging from 82% to 90% (28,30,31,73,88,92–94) for menorrhagia after a year and from 77% to 86% with dysmenorrhea and bulk symptoms (93,94).

The 2008 FIBROID registry, which involved 3160 UAE patients from 72 sites, found relatively low rates of re-intervention. At 3 years of follow-up in 1278 patients, further treatment with hysterectomy, myomectomy, or re-peat UAE was estimated to be 9.79%,

2.82%, and 1.83%, respectively (88). Similarly, further treatment was re-quired in 10.5% of 200 patients (92) and 6% of 400 patients (93) in other studies. In a study of 200 UAE patients, two of three patients with partial in-farction showed regrowth of the resid-ual fibroid at imaging follow-up. Both of these patients had recurrent symp-toms, suggesting that percent of fibroid infarction rather than volume reduction is important for predicting outcomes (95).

Pelvic pain is by far the most com-mon symptom after UAE and is expect-ed to last 48–72 hours, but it can last for up to a month (10). Vaginal dis-charge associated with the procedure typically resolves after 1 month (10). Major complications have been re-ported in 4.5% of UAE patients in the HOPEFUL trial compared with 14.8% in the surgical arm (31) and in 1.3% in the EMMY trial compared with 14.5% of surgery patients (29). A 400-patient

Page 10: Management of Uterine Fibroids - CIRwebcir.org/.../2016/4_noviembre/rsna/management_uterine_fibroids.pdf · icantly improves symptoms related to uterine fibroids in 85%–90% of patients

684 radiology.rsna.org n Radiology: Volume 280: Number 3—September 2016

STATE OF THE ART: Management of Uterine Fibroids Silberzweig et al

myomectomy in the United Kingdom has yet to report results.

Focused Ultrasound Therapy

FUS or high-intensity focused ultra-sound (HIFU) technology has existed since the 1940s. It was first used clini-cally in the liver in 1942 (104), for neu-rologic disease in the mid-1950s (105), and for tumor treatment in 1995 (106). FUS has been used in the treatment of breast, brain, eye, prostate, bladder, re-nal, pancreatic, metastatic bone, liver, and uterine tumors (107). However, the need for image guidance to facilitate precise targeting and to provide tem-perature feedback initially limited its clinical use (108). Early US-guided FUS therapy has been slow to affect clinical practice due to the limited ability to vi-sualize the target lesion (109), assess outcome during a session, and perform thermometry (110). A six-patient series from China in 2002 demonstrated the feasibility of MR-guided FUS fibroid treatment, with symptomatic improve-ment in five patients (111). Since then, small phase I, II, and III trials have been performed, demonstrating the safety, feasibility, and efficacy of MR-guided FUS therapy for uterine fibroids (105–109).

The ExAblate 2000 device (InSightec, Haifa, Israel) is an MR-guided system that received U.S. FDA approval in 2004 for uterine fibroid ablation. Var-ious other commercial systems are available but not FDA approved, includ-ing the Sonalleve (Philips Healthcare, the Netherlands) volumetric ablation system (112,113), as well as US-guided systems (eg, Ablatherm, FEP-BY02, and Sonoblate) (114). The MR technol-ogies can be used with 1.5- and 3-T MR imaging units and require a special MR imaging–compatible table with the em-bedded US transducer in a sealed de-gassed water bath (to improve acoustic coupling) (107) (Fig 10).

FUS/HIFU therapy causes destruc-tion of tissue by thermal injury from absorption of sound wave energy and vibratory effects and secondarily by cav-itation through the generation of micro-bubbles. The high-intensity ultrasound

patients but a greater rate of reinter-vention (36.7% compared with 6.1%) without a significant difference in total symptomatic relief (98).

The need for hysterectomy due to a complication of UAE occurs in less than 1% of patients (14). The Soci-ety of Interventional Radiology qual-ity improvement guidelines from 2010 suggested complication thresholds of 20% for prolonged vaginal discharge, 3% for permanent amenorrhea in pa-tients younger than 45 years and 15% in those older than 45 years, 15% for transcervical expulsion of fibroids, 3% for septicemia, less than 1% for thromboembolic disease, and less than 1% for nontarget embolization (99). Three fatalities have been reported after UAE (14), including fatal pul-monary embolism (100), lethal sepsis from pyometrium (10), and death from ovarian cancer (101).

Fertility and PregnancyFertility after UAE has not been conclu-sively established, although most stud-ies suggest ovarian failure is rare (77), and data suggest that fertility rates are likely similar after myomectomy (53). However, while successful pregnancies after UAE have been reported, UAE may have a negative impact on sub-sequent pregnancy, including greater rates of spontaneous abortion (46), failed pregnancy attempts (102), abnor-mal placental position (103), and Ce-sarian section (31,100).

A small study showed no differ-ence in rates of conception after UAE and laparoscopic uterine artery occlu-sion but a significantly greater rate of spontaneous abortion in the UAE group (56% compared with 10.5%), without a significant difference in preterm de-liveries, cesarean sections, or malpre-sentation (46). A prospective random-ized trial of UAE versus myomectomy in 121 women showed better reproduc-tive outcomes for myomectomy after 2 years (102), with greater rates of preg-nancy and labor and fewer spontaneous abortions, but similar perinatal and ob-stetrical results (102). A dedicated on-going trial (FEMME) measuring quality of life and pregnancy after UAE versus

British study reported a 5% morbid-ity rate after UAE, based on American Congress of Obstetricians and Gyne-cologists definitions, compared with 38.6% for myomectomy and 40.1% for hysterectomy (62). Late complications in patients in that study were mostly related to fibroid sloughing and pas-sage (none of whom had pedunculated submucosal fibroids), which should always be considered when patients present with pain, fever, or recurrent bleeding or discharge. Overall, the reported rates of expulsion or trans-vaginal sloughing of a necrotic fibroid are around 5% but may be as high as 20%–25% in patients with dominant submucosal fibroids (53).

Complications after UAE include endometritis requiring antibiotics, delayed contrast material reactions, tubo-ovarian or uterine abscess, amen-orrhea, urinary tract infection, urinary retention, fibroid sloughing requiring hysteroscopic extraction or hysterec-tomy, and uterine infarction. Nontarget embolization of the ovaries is a risk and around 3%–5% of patients will become amenorrheic, although the risk for this complication appears to be higher in older patients (96). There are also re-ports of nontarget embolization lead-ing to buttock claudication, and sciatic nerve, bladder, and vaginal mucosal in-jury (10).

UAE also compares well to myo-mectomy, with better symptom re-duction and a reduced rate of rein-tervention. A 2006 study comparing 70 patients treated with UAE and 69 patients with myomectomy showed symptom reduction in 88.3% of UAE patients compared with 75.4% after myomectomy. UAE patients reported significantly higher median quality of life scores at 6 months, persisting through 2 years of follow-up. Adverse events occurred in 42% of patients in the myomectomy group compared with 26% of patients in the UAE group (97). A smaller comparison study with an average of 17 months of patient follow-up showed shorter procedure times, hospital stays, and disability periods and higher hemoglobin levels in UAE patients versus myomectomy

Page 11: Management of Uterine Fibroids - CIRwebcir.org/.../2016/4_noviembre/rsna/management_uterine_fibroids.pdf · icantly improves symptoms related to uterine fibroids in 85%–90% of patients

Radiology: Volume 280: Number 3—September 2016 n radiology.rsna.org 685

STATE OF THE ART: Management of Uterine Fibroids Silberzweig et al

skin burn or intestinal perforation, re-spectively. Placing vitamin E capsules on an abdominal scar during the MR imag-ing is helpful in determining if the scar is in the acoustic path, although scars are often readily recognized on MR im-ages. If the targeted fibroid tissue is too deep, the acoustic energy is unlikely to be effective (as the transducer is unable to focus the energy at that depth); if the patient has a large amount of sub-cutaneous fat, the need to use higher ultrasound energies increases the risk of a skin burn (107,118). Obtaining a pretreatement MR imaging study with the patient in the prone position to de-fine the anatomy prior to FUS therapy is helpful in treatment planning (108).

Currently, patients with more than four fibroids or fibroid volumes greater than 500 mL are not considered to be good candidates for MR-guided FUS therapy (107). Maximal fibroid size that can be treated with MR-guided FUS has not been established, but a retrospective study showed the best results in fibroids less than 50 mL in volume (115). Many studies have used a maximum diameter

the number of sonications performed for a typical ablation ranged from six to 31, with MR occupancy times ranging from 3 hours 19 minutes to 4 hours 55 minutes per case (117). In contrast, a 2013 retrospective study of 115 patients had a mean of 74 sonications during a treatment session (115). The Sonal-leve system’s larger sonication size al-lows for greater treatment efficiency as compared with the InSightec unit. Aver-age treatment time with the Sonalleve system has been reported as 3.67 hours with 99 sonications performed (requir-ing an average of 54.6 minutes of son-ication) (112) and 4.07 hours with 432 sonications (with an average of 2.33 hours of sonication time) (113).

Patient SelectionIndications for treatment are similar to the indications for UAE and myomec-tomy. The number, size, location, and characteristics of the fibroids will be used to determine if a patient is a can-didate. In addition, if the patient has an abdominal scar or intestines in the acoustic path, the patient is at risk for a

waves bypass nontarget structures and are focused on a target to cause ther-mally induced protein denaturation and coagulative necrosis, leading to cell death (107,114). The ablation volume of a sonication varies from a few mil-limeters up to a centimeter with the ExAblate and 16 mm 3 40 mm with the Sonalleve system (113), depending on the energy applied, the type of tis-sue being treated, and the attenuation of the sound waves by intervening tis-sue (107). During and after each son-ication, using the shift in proton reso-nance frequency that occurs with rises in temperature, a dynamic thermal map is created (107). The temperature-de-pendent proton frequency shift is linear over a wide temperature range (includ-ing above the coagulation threshold); therefore slight elevations in adjacent tissues can safely be used to monitor for nontarget damage (108).

At the time of FDA approval of MR-guided FUS in 2004, the manufacturer’s indications for use for the ExAblate 2000 system allowed for a maximum of 33% of the fibroid volume to be treat-ed, due to concerns for injury to the normal uterine tissue. In 2006, the FDA allowed for up to 50% of the fibroid volume to be treated (108). In 2009, the FDA allowed for the treatment of 100% of the fibroid volume, with the stipulation that the treatment focal spot should not be within 10 mm of the se-rosal surface of the uterus (115,116). Therefore, studies employing MR-guid-ed FUS using the FDA standards prior to 2009 were biased toward less than optimal results.

Use of MR-guided FUS therapy is limited by the time required to perform quality assurance, synchronize the ul-trasound transducer and MR imaging scanner coordinates, create a treatment plan, acquire the MR images, perform each acoustic ablation (termed sonica-tion), allow for cooling of the technol-ogy, and reposition the treatment focal spot. While cooling time has the great-est impact on procedure time, advances have been made in the efficiency of treatment by increasing sonication sizes and decreasing sonication times. In a 2008 study with the ExAblate system,

Figure 10

Figure 10: Drawing of an MR-guided FUS table. The ultrasound transducer is built into the table with a degassed water bath and a gel pad to enhance acoustic coupling. The bottom half of a two-part pelvic coil is also built into the table and the top half is placed over the patient.

Page 12: Management of Uterine Fibroids - CIRwebcir.org/.../2016/4_noviembre/rsna/management_uterine_fibroids.pdf · icantly improves symptoms related to uterine fibroids in 85%–90% of patients

686 radiology.rsna.org n Radiology: Volume 280: Number 3—September 2016

STATE OF THE ART: Management of Uterine Fibroids Silberzweig et al

trapped air and contaminants from in-terfering or absorbing the FUS beam that could predispose to a skin burn. Prophylaxis to prevent deep venous thrombosis is recommended (107,120).

Successful positioning of the fibroid over the FUS transducer is confirmed with rapid gradient-echo localizer se-quences, followed by a baseline tripla-nar T2-weighted planning study. The radiologist manually outlines the treat-ment target area in a single plane, and software then interpolates the three-dimensional fibroid treatment volume (107). Electronic barriers to sound are manually placed in orthogonal planes to prevent any portion of the individual acoustic waves from traversing key an-atomic structures (bowel, pubic bone, and skin scars). Additional electronic barriers are placed adjacent to nerve roots emanating from the sacral foram-ina to minimize the heating of nerves. A focal spot distance of at least 4 cm from the sacrum should be maintained to avoid sacral heating and nerve damage or pain. Electronic fiducial markers are also placed as a reference to monitor movement during the treatment session. If too much movement occurs, treat-ment replanning is required. Subthera-peutic test sonications are performed in the coronal plane (perpendicular to the ultrasound beam) and sagittal (parallel) plane to assess targeting accuracy, and adjustments are made to synchronize the FUS transducer coordinates with the MR imaging unit. MR images can also be used to scrutinize the abdomen sur-face for air bubbles overlying the skin. Skin heating can be assessed on thermal mapping images (131).

Treatment proceeds with gradually in-creasing ultrasound energy (107) (Fig 11). Acoustic power, spot size, and sonica-tion frequency and duration can be ad-justed with a desire to generate targeted tissue heating to 65°C for the ExAblate system (107) and 60°–62°C for the Sonalleve system (112). Each treatment sonication lasts about 20–30 seconds, with an 80–100-second cooling period interspersed for both the ExAblate and Sonalleve systems (107,112). Patients are instructed to use a “stop sonication” button device if they experience severe

and 103 clinically eligible patients, only 25% were eligible anatomically (based on MR imaging findings) and only 14% were ultimately eligible due to feasibil-ity and safety concerns (125). A 2013 retrospective study of 123 patients had a 6.5% technical failure rate due to the presence of bowel (115). Vari-ous maneuvers have been used to in-crease the number of patients who can be treated with FUS. A study of 21 patients showed the feasibility of using a degassed water-filled balloon placed beneath a prone patient to successfully displace bowel from the acoustic path (117). Similarly, a 230-patient study showed significant decrease in screen-ing failures after use of a bowel dis-placement technique: sequential nor-mal saline bladder filling, ultrasound coupling gel rectal filling, followed by bladder emptying (126).

Currently, the U.S. FDA advises precaution with use of MR-guided FUS after UAE because it has not been stud-ied. Interactions with embolic agents may limit results or lead to complica-tions with beam scattering. Addition-ally, the effects of MR-guided FUS on fertility have not been well studied (107). Currently, the recommendation from InSightec is that an intrauterine device should be removed if it is in the direct treatment pathway but may be otherwise left in place, although this has not been well studied. Procedural timing with respect to phases of the menstrual cycle has not shown signifi-cant differences in outcomes (117).

Treatment of adenomyosis with FUS therapy has been studied at multiple in-stitutions in Asia, with reported benefit in 89.9% at 18 months (127), 89% at 6 months (128), and 77% at 12 months (129). There is also a case report of term vaginal delivery after MR-guided FUS treatment of focal adenomyosis in a 33-year-old woman with an inability to conceive (130).

Patient Preparation and ProcedureEfforts to minimize bowel activity during a treatment session include a low resi-due diet, bowel cathartics, 6-hour fast-ing, and glucagon. The lower abdominal wall is shaved and cleansed to prevent

of 10 cm (approximately 520 mL) for a fibroid as the limit for inclusion, al-though an ongoing comparison study of UAE and MR-guided FUS has a limit of 3 cm (119). Some studies have used no limitations on proximity to the endome-trium (108), while others maintained a 1.5 cm distance from the endometrium and 1 cm distance from the junctional zone (120). GnRHa pretreatment has been attempted in one small study to reduce fibroid volume prior to ablation of fibroids 10 cm or larger with good results (121). Increased fibroid vascu-larity has been suggested as a predictor of treatment failure with greater per-fusion on dynamic contrast-enhanced MR images correlating with worse treatment efficacy (122,123). In addi-tion, homogeneously T2-hypointense fibroids without infarction or areas of cystic degeneration on MR images have shown better response rates with FUS/HIFU therapy than heterogeneous and T2-hyperintense fibroids (118,124). However, a retrospective study of 282 patients undergoing MR-guided FUS found smaller, but adequate, nonperfu-sion volume ratios (used as a surrogate for the percentage of ablated tissue) in patients with heterogeneous fibroids (120). Similarly, a retrospective study from the United States of 115 patients showed no significant difference in non-perfusion volume based on T2 signal in-tensity or homogeneity (115). However, higher rates of skin burns (12.5%) and technical failures have been suggested with T2-hyperintense fibroids (as well as subcutaneous fat thickness . 20 mm), likely due to the need for higher acoustic energies, which also resulted in more terminations of procedures (33%) owing to pain during treatment (118).

Contraindications to MR-guided FUS include pedunculated fibroids, lower abdominal wall scarring in the acoustic path, potential malignancy, pelvic infection, pregnancy, calcified fi-broids, fibroids too close to the sacral nerve roots, and loops of bowel within the acoustic path. Unfortunately, ana-tomic limitations may not be discovered until the patient is on the procedure table. In a study of 333 patients seeking to undergo MR-guided FUS treatment

Page 13: Management of Uterine Fibroids - CIRwebcir.org/.../2016/4_noviembre/rsna/management_uterine_fibroids.pdf · icantly improves symptoms related to uterine fibroids in 85%–90% of patients

Radiology: Volume 280: Number 3—September 2016 n radiology.rsna.org 687

STATE OF THE ART: Management of Uterine Fibroids Silberzweig et al

but not until the gadolinium-based con-trast agent has effectively been elimi-nated, as the FDA has cautioned that the high-energy beam may release toxic free gadolinium. However, a study of MR-guided FUS treatment of breast

T1-weighted unenhanced and gado-linium-enhanced MR imaging study is performed to assess and calculate the degree of ablation and nonperfused volume (Fig 11). A second session may be performed for inadequate treatment

pain, internal heating, skin burning, or pain radiating down their legs (107).

A cumulative electronically calcu-lated thermal map of the treatment foci provides a prediction of the ab-lated volume. A triplanar fat-saturated

Figure 11

Figure 11: Images of MR-guided FUS treatment with the ExAblate system. (a) Sagittal T2-weighted MR image shows planned sonication path (blue) and treatment zones for each sonication (green rectangles). The radiologist manually outlines the fibroid and the volume is interpolated by the computer (red). The patient’s bladder has been filled to displace anterior bowel cranially out of the ultrasound beam path. (b) Coronal T2-weighted MR image shows planned treatment sonication zones en face (green circles) and electronic fiducials to track motion (red). (c) Intraprocedural thermometry (measuring water proton resonance frequency shift) obtained every 2–4 seconds throughout sonication (blue path), with the portion of the focal spot reaching the desired temperature threshold (depicted in red). The last image shows the accumulated thermal dosimetry that reached the desired threshold during sonication (green), with the prior accumulated foci of sonication (dark blue) that reached the threshold temperature. (d) Real-time sonication thermometry is plotted with temperature measurements over time. (e) Coronal T2-weighted MR image and accumulated dosimetry after sonication. Additional sonications could be planned at this point before administration of gadolinium-based contrast agent if the predicted ablation zone is inadequate. (f) Coronal fat-saturated T1-weighted postcontrast MR image shows the nonenhancing, nonperfused ablated tissue. Note how the nonperfused volume corresponds to the dosimetry map.

Page 14: Management of Uterine Fibroids - CIRwebcir.org/.../2016/4_noviembre/rsna/management_uterine_fibroids.pdf · icantly improves symptoms related to uterine fibroids in 85%–90% of patients

688 radiology.rsna.org n Radiology: Volume 280: Number 3—September 2016

STATE OF THE ART: Management of Uterine Fibroids Silberzweig et al

are reports of successful child bear-ing after the procedure as well as use of the treatment to address infertility (130,142–145). A registry maintained by the ExAblate manufacturer report-ed 54 pregnancies in 51 women with a mean time to conception of 8 months, 41% resulting in live births, 64% de-livering vaginally, 28% experiencing spontaneous abortion, 11% electing abortion, and 20% with ongoing preg-nancies at the time of the report (146).

United States and British studies in 2009 modeled cost-effectiveness of MR-guided FUS compared with other treatments, predicting total costs and quality-adjusted life-years (147,148). Quality-adjusted life-years were simi-lar for UAE, MR-guided FUS, myomec-tomy, hysterectomy, and pharmaco-therapy (in decreasing order of added quality of life) in the U.S. study (147), while MR-guided FUS had only a small gain in quality-adjusted life-years and cost savings in the British study (148). Myomectomy was more costly and less effective than both MR-guided FUS and UAE (147).

Future Directions

Another percutaneous image-guided fi-broid treatment technique being studied is radiofrequency ablation. A 2012 Dan-ish study of 43 patients who underwent US-guided radiofrequency ablation for symptomatic fibroids showed significant mean symptom severity score reduction of 48.6%, mean fibroid reduction vol-ume of 69.7%, no adverse events, and a 4.7% rate of subsequent hysterectomy after 9 months. Success rates with larger fibroids have been shown in some stud-ies, while incomplete ablation was seen in others. Further study is still required to determine the safety precautions needed to protect adjacent structures, such as intraperitoneal saline instillation and normal tissue margins (149).

Conclusion

Various techniques are available and are being investigated for the treatment of symptomatic uterine fibroids. Treat-ment approach in many cases depends

damage includes focal abdominal wall edema and deep vein thrombosis (re-ported in one patient in the FDA Man-ufacturer and User Facility Device Ex-perience adverse event database) (107). Thermal damage to the bowel is a seri-ous complication and several cases have been reported on the FDA database.

OutcomesPelvic pain and pressure improve soon after the procedure, with a decrease in menorrhagia and uterine volume taking approximately three menstrual cycles to occur. There is a correlation between the nonperfused volume and symptom resolution, with enduring symptom re-lief and a lower likelihood of needing additional treatments with nonperfused volumes greater than 50%–60% (118). Patients with greater than 30% non-perfused volume, compared with those with less, are three times more likely to have a 10-point improvement on the uterine fibroid symptom and quality of life questionnaire (UFS-QoL) symptom severity scale (108). Moreover, with nonperfused volume of 80% or more, the reduction in fibroid volume is signif-icantly greater than with less than 80% nonperfused volume (135).

In a 2011 study of 130 women, 85.7% reported symptom improvement at 3 months, 92.9% at 6 months, and 87.6% at 12 months (136). However, in an update of this report, patients with lower nonperfused volume (on average, 45.5%), younger age, and heteroge-neously bright fibroids on T2-weighted images were more likely to require ad-ditional treatment: 19% at 36 months and 23% at 48 months (137). Phase III trials have shown significant reductions in symptoms in 78%–80% of women after 6–12 months (14,108). Short-term studies have also corroborated significant decreases in symptom se-verity scores among nearly all patients (115,138–140). A multicenter random-ized UAE-controlled trial, the FIRSTT trial, is currently recruiting patients (119), and a 3-year, 1000-patient inter-national registry began to collect data on MR-guided FUS in 2013 (141).

Pregnancy after MR-guided FUS has not been well studied, although there

cancer is using gadolinium-enhanced images to guide therapy and no ad-verse events have been reported (107). In addition, a study of HIFU treatment of gadolinium-enhanced rat tumor and muscle showed no dissociation of gado-pentetate dimeglumine (132).

Following the procedure, patients are monitored for 30–60 minutes, although early study patients were monitored for up to 3 hours (120,133,134). The major-ity of patients require only nonsteroidal anti-inflammatory analgesics (117). In one study, only 10% of patients required any analgesics in the first 72 hours (acet-aminophen or nonsteroidal anti-inflam-matory drugs), with the most common symptom being mild to moderate gen-eralized discomfort (in 25% of patients) (117). Women can often return to work the following day (107).

ComplicationsSevere skin burns requiring skin grafting can occur at the site of a scar without pa-tients experiencing pain during the pro-cedure (presumably due to scar dener-vation). Most studies have not excluded patients simply because of lower abdom-inal scarring and have made exclusion decisions based on the extent of scarring and expected ultrasound beam path. It has been suggested that evaluation for persistence of skin sensation is an im-portant determinant for patient inclusion (107). First-degree burns in two patients occurred in a retrospective of 115 pa-tients, both which resolved after 1 week (115). No burns were reported in a 2007 multicenter prospective trial of 160 pa-tients (108). A 2007 study of 35 patients reported one first-degree burn (2.9%) that resolved within 2 weeks (134).

In a 2007 study, one of 35 patients (2.9%) reported sciatic nerve pain, which subsided after a week (134), and one of 160 patients (0.6%) in a phase III study reported sonication-re-lated leg pain that resolved after 2 days (108). However, sciatic nerve damage can take months to resolve. The same study reported pain or discomfort re-lated to procedure positioning or uter-ine cramping in 47%–54% of patients. Vaginal discharge was reported in 9% of patients (108). Additional soft-tissue

Page 15: Management of Uterine Fibroids - CIRwebcir.org/.../2016/4_noviembre/rsna/management_uterine_fibroids.pdf · icantly improves symptoms related to uterine fibroids in 85%–90% of patients

Radiology: Volume 280: Number 3—September 2016 n radiology.rsna.org 689

STATE OF THE ART: Management of Uterine Fibroids Silberzweig et al

22. Friedman AJ, Hoffman DI, Comite F, Browneller RW, Miller JD. Treatment of leiomyomata uteri with leuprolide acetate depot: a double-blind, placebo-controlled, multicenter study. The Leuprolide Study Group. Obstet Gynecol 1991;77(5):720–725.

23. Lobo RA. The risk of stroke in postmeno-pausal women receiving hormonal therapy. Climacteric 2009;12(Suppl 1):81–85.

24. Zapata LB, Whiteman MK, Tepper NK, Jamieson DJ, Marchbanks PA, Curtis KM. Intrauterine device use among women with uterine fibroids: a systematic review. Con-traception 2010;82(1):41–55.

25. Rutgers JL, Spong CY, Sinow R, Heiner J. Leuprolide acetate treatment and myoma ar-terial size. Obstet Gynecol 1995;86(3):386–388.

26. Lethaby A, Vollenhoven B, Sowter M. Pre-operative GnRH analogue therapy before hysterectomy or myomectomy for uterine fibroids. Cochrane Database Syst Rev 2000 (2):CD000547.

27. Spies JB, Bradley LD, Guido R, Maxwell GL, Levine BA, Coyne K. Outcomes from leio-myoma therapies: comparison with normal controls. Obstet Gynecol 2010;116(3):641–652.

28. Edwards RD, Moss JG, Lumsden MA, et al. Uterine-artery embolization versus surgery for symptomatic uterine fibroids. N Engl J Med 2007;356(4):360–370.

29. Hehenkamp WJ, Volkers NA, Donderwinkel PF, et al. Uterine artery embolization versus hysterectomy in the treatment of symptom-atic uterine fibroids (EMMY trial): peri- and postprocedural results from a randomized controlled trial. Am J Obstet Gynecol 2005; 193(5):1618–1629.

30. Pinto I, Chimeno P, Romo A, et al. Uterine fibroids: uterine artery embolization versus abdominal hysterectomy for treatment—a prospective, randomized, and controlled clini-cal trial. Radiology 2003;226(2):425–431.

31. Dutton S, Hirst A, McPherson K, Nicholson T, Maresh M. A UK multicentre retrospec-tive cohort study comparing hysterectomy and uterine artery embolisation for the treatment of symptomatic uterine fibroids (HOPEFUL study): main results on medi-um-term safety and efficacy. BJOG 2007; 114(11):1340–1351.

32. Merrill RM. Hysterectomy surveillance in the United States, 1997 through 2005. Med Sci Monit 2008;14(1):CR24–CR31.

33. Reed SD, Newton KM, Thompson LB, Mc-Crummen BA, Warolin AK. The incidence of repeat uterine surgery following myomecto-my. J Womens Health (Larchmt) 2006;15(9): 1046–1052.

34. Jin C, Hu Y, Chen XC, et al. Laparoscopic versus open myomectomy: a meta-analysis of randomized controlled trials. Eur J Obstet Gy-necol Reprod Biol 2009;145(1):14–21.

35. Goldfarb HA. Laparoscopic coagulation of myoma (myolysis). Obstet Gynecol Clin North Am 1995;22(4):807–819.

8. Kim JJ, Sefton EC. The role of progester-one signaling in the pathogenesis of uter-ine leiomyoma. Mol Cell Endocrinol 2012; 358(2):223–231.

9. Lee JS, Spies JB. Management options for uterine fibroids. In: Geschwind JFH, Dake MD, eds. Abrams’ angiography: intervention-al radiology. 3rd ed. Philadelphia, Pa: Lippin-cott Williams & Wilkins, 2014; 239–245.

10. Carrillo TC. Uterine artery embolization in the management of symptomatic uterine fi-broids: an overview of complications and fol-low-up. Semin Intervent Radiol 2008;25(4): 378–386.

11. Buttram VC Jr, Reiter RC. Uterine leiomyo-mata: etiology, symptomatology, and man-agement. Fertil Steril 1981;36(4):433–445.

12. Pritts EA, Parker WH, Olive DL. Fibroids and infertility: an updated systematic review of the evidence. Fertil Steril 2009;91(4): 1215–1223.

13. Bosteels J, Kasius J, Weyeres S, Broekmans FJ, Mol BW, D’Hooghe TM. Hysteroscopy for treating subfertility associated with sus-pected major uterine cavity abnormalities. Cochrane Database Syst Rev 2015;21(2): CD009461.

14. Hurst BS, Matthews ML, Marshburn PB. Laparoscopic myomectomy for symptomatic uterine myomas. Fertil Steril 2005;83(1):1–23.

15. Rajan DK, Margau R, Kroll RR, et al. Clini-cal utility of ultrasound versus magnetic res-onance imaging for deciding to proceed with uterine artery embolization for presumed symptomatic fibroids. Clin Radiol 2011; 66(1):57–62.

16. Spielmann AL, Keogh C, Forster BB, Martin ML, Machan LS. Comparison of MRI and sonography in the preliminary evaluation for fibroid embolization. AJR Am J Roentgenol 2006;187(6):1499–1504.

17. Dudiak CM, Turner DA, Patel SK, Archie JT, Silver B, Norusis M. Uterine leiomyomas in the infertile patient: preoperative locali-zation with MR imaging versus US and hys-terosalpingography. Radiology 1988;167(3): 627–630.

18. Kim MD, Kim YM, Kim HC, et al. Uterine artery embolization for symptomatic adeno-myosis: a new technical development of the 1-2-3 protocol and predictive factors of MR imaging affecting outcomes. J Vasc Interv Radiol 2011;22(4):497–502.

19. Pelage JP, Jacob D, Fazel A, et al. Midterm results of uterine artery embolization for symptomatic adenomyosis: initial experi-ence. Radiology 2005;234(3):948–953.

20. Omary RA, Vasireddy S, Chrisman HB, et al. The effect of pelvic MR imaging on the diagnosis and treatment of women with pre-sumed symptomatic uterine fibroids. J Vasc Interv Radiol 2002;13(11):1149–1153.

21. Carlson KJ, Miller BA, Fowler FJ Jr. The Maine Women’s Health Study: II. Outcomes of nonsurgical management of leiomyomas, abnormal bleeding, and chronic pelvic pain. Obstet Gynecol 1994;83(4):566–572.

on the likelihood of success given the nature, size, and distribution of the fibroids; the patient’s anatomy; and treatment goals. UAE is a highly effec-tive, safe, and cost-effective approach. While there is risk of failure requiring reintervention, patient satisfaction may be higher and major morbidity lower compared with surgical techniques. In select patients, MR-guided FUS is prov-ing to be an even safer approach, with an extremely short recovery period and low morbidity.

Disclosures of Conflicts of Interest: J.E.S. disclosed no relevant relationships. D.K.P. dis-closed no relevant relationships. A.H.M. Ac-tivities related to the present article: disclosed no relevant relationships. Activities not related to the present article: grants from W.L. Gore, Medtronic, and Cook; member of data safety monitoring board for Trivascular, Bolton Med-ical, and W.L. Gore; member of scientific ad-visory board for Boston Scientific; member of advisory board for Tenex Medical and BrightWa-ter; consultant for Medicines Co; Stockholder, Volcano Medical. Other relationships: disclosed no relevant relationships. J.B.S. Activities re-lated to the present article: disclosed no rele-vant relationships. Activities not related to the present article: consultant, Boston Scientific and Merit Medical. Other relationships: disclosed no relevant relationships.

References 1. Wallach EE, Vlahos NF. Uterine myomas: an

overview of development, clinical features, and management. Obstet Gynecol 2004; 104(2):393–406.

2. Schwartz SM. Epidemiology of uterine leio-myomata. Clin Obstet Gynecol 2001;44(2): 316–326.

3. Divakar H. Asymptomatic uterine fibroids. Best Pract Res Clin Obstet Gynaecol 2008;22 (4):643–654.

4. Wilcox LS, Koonin LM, Pokras R, Strauss LT, Xia Z, Peterson HB. Hysterectomy in the Unit-ed States, 1988-1990. Obstet Gynecol 1994; 83(4):549–555.

5. Townsend DE, Sparkes RS, Baluda MC, McClelland G. Unicellular histogenesis of uterine leiomyomas as determined by elec-trophoresis by glucose-6-phosphate dehydro-genase. Am J Obstet Gynecol 1970;107(8): 1168–1173.

6. Murase E, Siegelman ES, Outwater EK, Per-ez-Jaffe LA, Tureck RW. Uterine leiomyomas: histopathologic features, MR imaging findings, differential diagnosis, and treatment. Radio-Graphics 1999;19(5):1179–1197.

7. Ciavattini A, Di Giuseppe J, Stortoni P, et al. Uterine fibroids: pathogenesis and inter-actions with endometrium and endomyome-trial junction. Obstet Gynecol Int 2013;2013: 173184.

Page 16: Management of Uterine Fibroids - CIRwebcir.org/.../2016/4_noviembre/rsna/management_uterine_fibroids.pdf · icantly improves symptoms related to uterine fibroids in 85%–90% of patients

690 radiology.rsna.org n Radiology: Volume 280: Number 3—September 2016

STATE OF THE ART: Management of Uterine Fibroids Silberzweig et al

after uterine artery embolization for leio-myomas. Obstet Gynecol 2002;100(5 Pt 1): 873–880.

63. Chamsuddin A, Achou R, Loufti E, Kanj V. The use of intrathecal combination of an-algesics for control of procedural and post-procedural pain in uterine fibroids. J Vasc Interv Radiol 2014;25(3):S66.

64. Verma SK, Bergin D, Gonsalves CF, Mitchell DG, Lev-Toaff AS, Parker L. Sub-mucosal fibroids becoming endocavitary following uterine artery embolization: risk assessment by MRI. AJR Am J Roentgenol 2008;190(5):1220–1226.

65. Spies JB, Allison S, Flick P, et al. Spherical polyvinyl alcohol versus tris-acryl gelatin mi-crospheres for uterine artery embolization for leiomyomas: results of a limited random-ized comparative study. J Vasc Interv Radiol 2005;16(11):1431–1437.

66. Yu SC, Lok I, Ho SS, Tong MM, Hui JW. Comparison of clinical outcomes of tris-acryl microspheres versus polyvinyl alcohol microspheres for uterine artery emboliza-tion for leiomyomas: results of a randomized trial. J Vasc Interv Radiol 2011;22(9):1229–1235.

67. Smeets AJ, Nijenhuis R, van Rooij WJ, et al. Embolization of uterine leiomyomas with polyzene F-coated hydrogel microspheres: initial experience. J Vasc Interv Radiol 2010; 21(12):1830–1834.

68. Nijenhuis RJ, Smeets AJ, Morpurgo M, et al. Uterine artery embolisation for symp-tomatic adenomyosis with polyzene F-coat-ed hydrogel microspheres: three-year clini-cal follow-up using UFS-QoL questionnaire. Cardiovasc Intervent Radiol 2015;38(1): 65–71.

69. Mu Y WY, Li M, Hu Y, Hao Z. Comparison of efficacy of different embolic agents on uterine leiomyoma. Clin Exp Obstet Gynecol 2016;43(1):114–118.

70. Smeets AJ, Nijenhuis RJ, Boekkooi PF, Vervest HA, van Rooij WJ, Lohle PN. Is an intrauterine device a contraindica-tion for uterine artery embolization? A study of 20 patients. J Vasc Interv Radiol 2010;21(2):272–274.

71. Subramanian S, Spies JB. Uterine artery embolization for leiomyomata: resource use and cost estimation. J Vasc Interv Radiol 2001;12(5):571–574.

72. Goldberg J, Bussard A, McNeil J, Diamond J. Cost and reimbursement for three fibroid treatments: abdominal hysterectomy, ab-dominal myomectomy, and uterine fibroid embolization. Cardiovasc Intervent Radiol 2007;30(1):54–58.

73. Moss JG, Cooper KG, Khaund A, et al. Randomised comparison of uterine artery embolisation (UAE) with surgical treat-ment in patients with symptomatic uterine fibroids (REST trial): 5-year results. BJOG 2011;118(8):936–944.

49. Committee on Gynecologic Practice, Ameri-can College of Obstetricians and Gynecol-ogists. ACOG Committee Opinion: uterine artery embolization. Obstet Gynecol 2004; 103(2):403–404.

50. Pelage JP, Cazejust J, Pluot E, et al. Uter-ine fibroid vascularization and clinical rele-vance to uterine fibroid embolization. Radio-Graphics 2005;25(Suppl 1):S99–S117.

51. Gomez-Jorge J, Keyoung A, Levy EB, Spies JB. Uterine artery anatomy relevant to uter-ine leiomyomata embolization. Cardiovasc Intervent Radiol 2003;26(6):522–527.

52. Lai AC, Goodwin SC, Bonilla SM, et al. Sex-ual dysfunction after uterine artery emboli-zation. J Vasc Interv Radiol 2000;11(6):755–758.

53. Worthington-Kirsch RL. Uterine artery em-bolization: state of the art. Semin Intervent Radiol 2004;21(1):37–42.

54. Torre A, Paillusson B, Fain V, Labauge P, Pelage JP, Fauconnier A. Uterine artery em-bolization for severe symptomatic fibroids: effects on fertility and symptoms. Hum Re-prod 2014;29(3):490–501.

55. Razavi MK, Wolanske KA, Hwang GL, Sze DY, Kee ST, Dake MD. Angiographic classification of ovarian artery-to-uterine artery anastomoses: initial observations in uterine fibroid embolization. Radiology 2002; 224(3):707–712.

56. Pelage JP, Laurent A, Wassef M, et al. Uter-ine artery embolization in sheep: compar-ison of acute effects with polyvinyl alcohol particles and calibrated microspheres. Radi-ology 2002;224(2):436–445.

57. White AM, Banovac F, Yousefi S, Slack RS, Spies JB. Uterine fibroid embolization: the utility of aortography in detecting ovarian artery collateral supply. Radiology 2007; 244(1):291–298.

58. Golowa YW, Jagust M, Cynamon J. The utility of cone-beam computed tomography (CBCT) for the assessment of adequate uterine fibroid embolization (UFE). J Vasc Interv Radiol 2014;25(3):S68–S69.

59. Siskin GP, Beck A, Schuster M, Mandato K, Englander M, Herr A. Leiomyoma infarction after uterine artery embolization: a pro-spective randomized study comparing tris-acryl gelatin microspheres versus polyvinyl alcohol microspheres. J Vasc Interv Radiol 2008;19(1):58–65.

60. Patel K, Sakhi P, Soni K, Taori A, Patel A. Uterine artery embolisation as primary therapy for the treatment of symptom-atic uterine fibroids. Schol J Appl Med Sci 2013;1(6):873–877.

61. Song YG, Jang H, Park KD, Kim MD, Kim CW. Non spherical polyvinyl alcohol versus gelatin sponge particles for uterine artery embolization for symptomatic fi-broids. Minim Invasive Ther Allied Technol 2013;22(6):364–371.

62. Spies JB, Spector A, Roth AR, Baker CM, Mauro L, Murphy-Skrynarz K. Complications

36. Goldfarb HA. Myolysis revisited. JSLS 2008;12(4):426–430.

37. Pansky M, Cowan BD, Frank M, Hampton HL, Zimberg S. Laparoscopically assisted uterine fibroid cryoablation. Am J Obstet Gynecol 2009;201(6):571.e1–571.e7.

38. Seidman MA, Oduyebo T, Muto MG, Crum CP, Nucci MR, Quade BJ. Peritoneal dis-semination complicating morcellation of uterine mesenchymal neoplasms. PLoS One 2012;7(11):e50058.

39. Hagemann IS, Hagemann AR, LiVolsi VA, Montone KT, Chu CS. Risk of occult malig-nancy in morcellated hysterectomy: a case series. Int J Gynecol Pathol 2011;30(5):476–483.

40. Parker WH, Fu YS, Berek JS. Uterine sar-coma in patients operated on for presumed leiomyoma and rapidly growing leiomyoma. Obstet Gynecol 1994;83(3):414–418.

41. Takamizawa S, Minakami H, Usui R, et al. Risk of complications and uterine malig-nancies in women undergoing hysterectomy for presumed benign leiomyomas. Gynecol Obstet Invest 1999;48(3):193–196.

42. Laparoscopic uterine power morcellation in hysterectomy and myomectomy: FDA safety communication. U.S. Food and Drug Administration Web site. http://www.fda.gov/medicaldevices/safety/alertsandnotices/ucm393576.htm. Published April 17, 2014. Accessed July 9, 2014.

43. Power morcellation and occult malignancy in gynecologic surgery: a special report. Amer-ican Congress of Obstetricians and Gyne-cologists Web site. http://www.acog.org/Resources-And-Publications/Task-Force-and-Work-Group-Reports/Power-Morcella tion-and-Occult-Malignancy-in-Gynecologic-Surgery. Published May 2014. Accessed July 9, 2014.

44. Lethaby A, Shepperd S, Cooke I, Farqu-har C. Endometrial resection and ablation versus hysterectomy for heavy menstrual bleeding. Cochrane Database Syst Rev 2000 (2):CD000329.

45. Hald K, Noreng HJ, Istre O, Kløw NE. Uterine artery embolization versus lapa-roscopic occlusion of uterine arteries for leiomyomas: long-term results of a random-ized comparative trial. J Vasc Interv Radiol 2009;20(10):1303–1310; quiz 1311.

46. Holub Z, Mara M, Kuzel D, Jabor A, Mas-kova J, Eim J. Pregnancy outcomes after uterine artery occlusion: prospective multi-centric study. Fertil Steril 2008;90(5):1886–1891.

47. Ravina JH, Herbreteau D, Ciraru-Vigneron N, et al. Arterial embolisation to treat uter-ine myomata. Lancet 1995;346(8976):671–672.

48. Goodwin SC, Vedantham S, McLucas B, For-no AE, Perrella R. Preliminary experience with uterine artery embolization for uterine fibroids. J Vasc Interv Radiol 1997;8(4): 517–526.

Page 17: Management of Uterine Fibroids - CIRwebcir.org/.../2016/4_noviembre/rsna/management_uterine_fibroids.pdf · icantly improves symptoms related to uterine fibroids in 85%–90% of patients

Radiology: Volume 280: Number 3—September 2016 n radiology.rsna.org 691

STATE OF THE ART: Management of Uterine Fibroids Silberzweig et al

100. Hamoda H, Tait P, Edmonds DK. Fatal pul-monary embolus after uterine artery fibroid embolisation. Cardiovasc Intervent Radiol 2009;32(5):1080–1082.

101. Manyonda I, Sinthamoney E, Belli AM. Controversies and challenges in the mod-ern management of uterine fibroids. BJOG 2004;111(2):95–102.

102. Mara M, Maskova J, Fucikova Z, Kuzel D, Belsan T, Sosna O. Midterm clinical and first reproductive results of a randomized controlled trial comparing uterine fibroid embolization and myomectomy. Cardiovasc Intervent Radiol 2008;31(1):73–85.

103. Pron G, Mocarski E, Bennett J, Vilos G, et al. Pregnancy after uterine artery emboliza-tion for leiomyomata: the Ontario multicen-ter trial. Obstet Gynecol 2005;105(1):67–76.

104. Lynn JG, Zwemer RL, Chick AJ, Miller AE. A new method for the generation and use of focused ultrasound in experimental biology. J Gen Physiol 1942;26(2):179–193.

105. Fry WJ, Fry FJ. Fundamental neurological research and human neurosurgery using in-tense ultrasound. IRE Trans Med Electron 1960;ME-7:166–181.

106. Tempany CM, Stewart EA, McDannold N, Quade BJ, Jolesz FA, Hynynen K. MR im-aging-guided focused ultrasound surgery of uterine leiomyomas: a feasibility study. Ra-diology 2003;226(3):897–905.

107. Roberts A. Magnetic resonance-guided fo-cused ultrasound for uterine fibroids. Semin Intervent Radiol 2008;25(4):394–405.

108. Fennessy FM, Tempany CM, McDannold NJ, et al. Uterine leiomyomas: MR imaging-guided focused ultrasound surgery—results of different treatment protocols. Radiology 2007;243(3):885–893.

109. Shen SH, Fennessy F, McDannold N, Jolesz F, Tempany C. Image-guided thermal ther-apy of uterine fibroids. Semin Ultrasound CT MR 2009;30(2):91–104.

110. Li S, Wu PH. Magnetic resonance image-guided versus ultrasound-guided high-inten-sity focused ultrasound in the treatment of breast cancer. Chin J Cancer 2013;32(8): 441–452.

111. Wang W, Liu W, Zhou J, Xu J, Gai L, Huang C. Treatment of symptomatic uterine leio-myomata with high intensity focused ultra-sound: a preliminary clinical study. Chinese Journal of Ultrasonography 2002;3:161–163.

112. Venkatesan AM, Partanen A, Pulanic TK, et al. Magnetic resonance imaging-guided volumetric ablation of symptomatic leiomyo-mata: correlation of imaging with histology. J Vasc Interv Radiol 2012;23(6):786–794.e4.

113. Ruhnke H, Eckey T, Bohlmann MK, et al. MR-guided HIFU treatment of symptomatic uterine fibroids using novel feedback-regu-lated volumetric ablation: effectiveness and clinical practice. Rofo 2013;184(10):983–991.

comes from the FIBROID Registry. Obstet Gynecol 2008;111(1):22–33.

88. Coyne KS, Margolis MK, Murphy J, Spies J. Validation of the UFS-QOL-hysterecto-my questionnaire: modifying an existing measure for comparative effectiveness re-search. Value Health 2012;15(5):674–679.

89. Spies JB, Coyne K, Guaou Guaou N, Boyle D, Skyrnarz-Murphy K, Gonzalves SM. The UFS-QOL, a new disease-specific symptom and health-related quality of life question-naire for leiomyomata. Obstet Gynecol 2002; 99(2):290–300.

90. Hehenkamp WJ, Volkers NA, Birnie E, Reekers JA, Ankum WM. Symptomatic uterine fibroids: treatment with uterine artery embolization or hysterectomy—re-sults from the randomized clinical Emboli-sation versus Hysterectomy (EMMY) Trial. Radiology 2008;246(3):823–832.

91. Gupta JK, Sinha A, Lumsden MA, Hickey M. Uterine artery embolization for symp-tomatic uterine fibroids. Cochrane Database Syst Rev 2012;5:CD005073.

92. Spies JB, Ascher SA, Roth AR, Kim J, Levy EB, Gomez-Jorge J. Uterine artery emboliza-tion for leiomyomata. Obstet Gynecol 2001; 98(1):29–34.

93. Walker WJ, Pelage JP. Uterine artery em-bolisation for symptomatic fibroids: clinical results in 400 women with imaging follow up. BJOG 2002;109(11):1262–1272.

94. Pron G, Bennett J, Common A, et al. The Ontario Uterine Fibroid Embolization Trial. Part 2. Uterine fibroid reduction and symptom relief after uterine artery embolization for fi-broids. Fertil Steril 2003;79(1):120–127.

95. Pelage JP, Guaou NG, Jha RC, Ascher SM, Spies JB. Uterine fibroid tumors: long-term MR imaging outcome after embolization. Radiology 2004;230(3):803–809.

96. Chrisman HB, Saker MB, Ryu RK, et al. The impact of uterine fibroid embolization on re-sumption of menses and ovarian function. J Vasc Interv Radiol 2000;11(6):699–703.

97. Siskin GP, Shlansky-Goldberg RD, Good-win SC, et al. A prospective multicenter comparative study between myomectomy and uterine artery embolization with poly-vinyl alcohol microspheres: long-term clin-ical outcomes in patients with symptom-atic uterine fibroids. J Vasc Interv Radiol 2006;17(8):1287–1295.

98. Mara M, Fucikova Z, Maskova J, Kuzel D, Haakova L. Uterine fibroid embolization versus myomectomy in women wishing to preserve fertility: preliminary results of a randomized controlled trial. Eur J Obstet Gy-necol Reprod Biol 2006;126(2):226–233.

99. Stokes LS, Wallace MJ, Godwin RB, Kundu S, Cardella JF; Society of Interventional Ra-diology Standards of Practice Committee. Quality improvement guidelines for uterine artery embolization for symptomatic leiomy-omas. J Vasc Interv Radiol 2010;21(8):1153–1163.

74. Miller DL, Balter S, Cole PE, et al. Radia-tion doses in interventional radiology pro-cedures: the RAD-IR study: part I. overall measures of dose. J Vasc Interv Radiol 2003; 14(6):711–727.

75. Tse G, Spies JB. Radiation exposure and uterine artery embolization: current risks and risk reduction. Tech Vasc Interv Radiol 2010;13(3):148–153.

76. Nikolic B, Spies JB, Lundsten MJ, Abbara S. Patient radiation dose associated with uterine artery embolization. Radiology 2000;214(1): 121–125.

77. Hovsepian DM, Siskin GP, Bonn J, et al. Quality improvement guidelines for uterine artery embolization for symptomatic leio-myomata. J Vasc Interv Radiol 2009;20(7 Suppl):S193–S199.

78. Gonsalves C. Uterine artery embolization for treatment of symptomatic fibroids. Se-min Intervent Radiol 2008;25(4):369–377.

79. Katsumori T, Akazawa K, Mihara T. Uterine artery embolization for pedunculated subse-rosal fibroids. AJR Am J Roentgenol 2005; 184(2):399–402.

80. Sato K, Yuasa N, Fujita M, Fukushima Y. Clinical application of diffusion-weighted imaging for preoperative differentiation be-tween uterine leiomyoma and leiomyosarco-ma. Am J Obstet Gynecol 2014;210(4):368.e1–368.e8.

81. Chan D, Anthwal S, Prabhudesai V. Assess-ing the efficacy, complications, and re-inter-vention incidence following uterine fibroid embolization in patients with large fibroids. J Vasc Interv Radiol 2014;25(3):S8.

82. Smeets AJ, Nijenhuis RJ, van Rooij WJ, et al. Uterine artery embolization in patients with a large fibroid burden: long-term clini-cal and MR follow-up. Cardiovasc Intervent Radiol 2010;33(5):943–948.

83. Brandis A, Shlansky-Goldberg R, Trerotola S, Stavropoulos SW, Mondschein J, Panaro J. Uterine artery embolization in women with megauterus. J Vasc Interv Radiol 2014;25(3): S66–S67.

84. Lohle PN, De Vries J, Klazen CA, et al. Uterine artery embolization for symptom-atic adenomyosis with or without uterine leiomyomas with the use of calibrated tris-acryl gelatin microspheres: midterm clinical and MR imaging follow-up. J Vasc Interv Ra-diol 2007;18(7):835–841.

85. Siskin GP, Tublin ME, Stainken BF, Dowling K, Dolen EG. Uterine artery embolization for the treatment of adenomyosis: clinical response and evaluation with MR imaging. AJR Am J Roentgenol 2001;177(2):297–302.

86. Jung DC, Kim MD, Oh YT, Won JY, Lee Y. Prediction of early response to uterine ar-terial embolisation of adenomyosis: value of T2 signal intensity ratio of adenomyosis. Eur Radiol 2012;22(9):2044–2049.

87. Goodwin SC, Spies JB, Worthington-Kirsch R, et al. Uterine artery embolization for treatment of leiomyomata: long-term out-

Page 18: Management of Uterine Fibroids - CIRwebcir.org/.../2016/4_noviembre/rsna/management_uterine_fibroids.pdf · icantly improves symptoms related to uterine fibroids in 85%–90% of patients

692 radiology.rsna.org n Radiology: Volume 280: Number 3—September 2016

STATE OF THE ART: Management of Uterine Fibroids Silberzweig et al

MR-guided focused US for symptomatic uter-ine fibroids: review of 138 patients with an average follow-up of 2.8 years. J Vasc Interv Radiol 2014;25(10):1506–1512.

138. Hindley J, Gedroyc WM, Regan L, et al. MRI guidance of focused ultrasound therapy of uterine fibroids: early results. AJR Am J Roentgenol 2004;183(6):1713–1719.

139. Hesley GK, Felmlee JP, Gebhart JB, et al. Noninvasive treatment of uterine fibroids: early Mayo Clinic experience with magnetic resonance imaging-guided focused ultrasound. Mayo Clin Proc 2006;81(7):936–942.

140. Kohi MP, Coakley F, Jacoby A, et al. A pilot randomized placebo-controlled trial of MR guided focused ultrasound for symptomatic uterine fibroids. J Vasc Interv Radiol 2014; 25(3):S68.

141. Registry for Leiomyoma. International ef-ficacy of focused ultrasound. http://www.fibroidrelief.org/registry/. Accessed October 12, 2014.

142. Morita Y, Ito N, Ohashi H. Pregnancy following MR-guided focused ultrasound surgery for a uterine fibroid. Int J Gynaecol Obstet 2007;99(1):56–57.

143. Hanstede MM, Tempany CM, Stewart EA. Focused ultrasound surgery of intramural leio-myomas may facilitate fertility: a case report. Fertil Steril 2007;88(2):497.e5–497.e7.

144. Gavrilova-Jordan LP, Rose CH, Traynor KD, Brost BC, Gostout BS. Successful term pregnancy following MR-guided focused ul-trasound treatment of uterine leiomyoma. J Perinatol 2007;27(1):59–61.

145. Bouwsma EV, Gorny KR, Hesley GK, Jensen JR, Peterson LG, Stewart EA. Magnetic res-onance-guided focused ultrasound surgery for leiomyoma-associated infertility. Fertil Steril 2011;96(1):e9–e12.

146. Rabinovici J, David M, Fukunishi H, et al. Pregnancy outcome after magnetic resonance-guided focused ultrasound surgery (MRgFUS) for conservative treatment of uterine fibroids. Fertil Steril 2010;93(1):199–209.

147. O’Sullivan AK, Thompson D, Chu P, Lee DW, Stewart EA, Weinstein MC. Cost-ef-fectiveness of magnetic resonance guided focused ultrasound for the treatment of uterine fibroids. Int J Technol Assess Health Care 2009;25(1):14–25.

148. Zowall H, Cairns JA, Brewer C, Lamping DL, Gedroyc WM, Regan L. Cost-effective-ness of magnetic resonance-guided focused ultrasound surgery for treatment of uterine fibroids. BJOG 2008;115(5):653–662.

149. Iversen H, Lenz S, Dueholm M. Ultrasound-guided radiofrequency ablation of symptom-atic uterine fibroids: short-term evaluation of effect of treatment on quality of life and symptom severity. Ultrasound Obstet Gyne-col 2012;40(4):445–451.

126. Park MJ, Kim YS, Rhim H, Lim HK. Tech-nique to displace bowel loops in MRI-guided high-intensity focused ultrasound ablation of fibroids in the anteverted or anteflexed uterus. AJR Am J Roentgenol 2013;201(5): W761–W764.

127. Zhou M, Chen JY, Tang LD, Chen WZ, Wang ZB. Ultrasound-guided high-intensity focused ultrasound ablation for adenomyo-sis: the clinical experience of a single center. Fertil Steril 2011;95(3):900–905.

128. Kim KA, Yoon SW, Lee C, Seong SJ, Yoon BS, Park H. Short-term results of magnetic resonance imaging-guided focused ultra-sound surgery for patients with adenomyo-sis: symptomatic relief and pain reduction. Fertil Steril 2011;95(3):1152–1155.

129. Zhang X, Li K, Xie B, He M, He J, Zhang L. Effective ablation therapy of adenomyo-sis with ultrasound-guided high-intensity focused ultrasound. Int J Gynaecol Obstet 2014;124(3):207–211.

130. Rabinovici J, Inbar Y, Eylon SC, Schiff E, Hananel A, Freundlich D. Pregnancy and live birth after focused ultrasound surgery for symptomatic focal adenomyosis: a case re-port. Hum Reprod 2006;21(5):1255–1259.

131. Okada A, Morita Y, Fukunishi H, Takeichi K, Murakami T. Non-invasive magnetic resonance-guided focused ultrasound treat-ment of uterine fibroids in a large Japanese population: impact of the learning curve on patient outcome. Ultrasound Obstet Gyne-col 2009;34(5):579–583.

132. Hijnen NM, Elevelt A, Grüll H. Stability and trapping of magnetic resonance imag-ing contrast agents during high-intensity focused ultrasound ablation therapy. Invest Radiol 2013;48(7):517–524.

133. Zhang L, Chen WZ, Liu YJ, et al. Feasibil-ity of magnetic resonance imaging-guided high intensity focused ultrasound therapy for ablating uterine fibroids in patients with bowel lies anterior to uterus. Eur J Radiol 2010;73(2):396–403.

134. Rabinovici J, Inbar Y, Revel A, et al. Clin-ical improvement and shrinkage of uterine fibroids after thermal ablation by magnetic resonance-guided focused ultrasound sur-gery. Ultrasound Obstet Gynecol 2007;30(5): 771–777.

135. Park MJ, Kim YS, Rhim H, Lim HK. Safety and therapeutic efficacy of complete or near-complete ablation of symptomatic uterine fibroid tumors by MR imaging-guided high-intensity focused US therapy. J Vasc Interv Radiol 2014;25(2):231–239.

136. Gorny KR, Woodrum DA, Brown DL, et al. Magnetic resonance-guided focused ultrasound of uterine leiomyomas: review of a 12-month outcome of 130 clinical pa-tients. J Vasc Interv Radiol 2011;22(6):857– 864.

137. Gorny KR, Borah BJ, Brown DL, Woodrum DA, Stewart EA, Hesley GK. Incidence of additional treatments in women treated with

114. Hesley GK, Gorny KR, Woodrum DA. MR-guided focused ultrasound for the treatment of uterine fibroids. Cardiovasc Intervent Ra-diol 2013;36(1):5–13.

115. Trumm CG, Stahl R, Clevert DA, et al. Mag-netic resonance imaging-guided focused ul-trasound treatment of symptomatic uterine fibroids: impact of technology advancement on ablation volumes in 115 patients. Invest Radiol 2013;48(6):359–365.

116. Morita Y, Takeuchi S, Hikida H, Ohashi H, Ito N. Decreasing margins to the uterine se-rosa as a method for increasing the volume of fibroids ablated with magnetic resonance-guided focused ultrasound surgery. Eur J Obstet Gynecol Reprod Biol 2009;146(1): 92–95.

117. Hudson SB, Stewart EA. Magnetic reso-nance-guided focused ultrasound surgery. Clin Obstet Gynecol 2008;51(1):159–166.

118. Mikami K, Murakami T, Okada A, Osuga K, Tomoda K, Nakamura H. Magnetic res-onance imaging-guided focused ultrasound ablation of uterine fibroids: early clinical ex-perience. Radiat Med 2008;26(4):198–205.

119. Stewart EA. The FIRSTT: comparing MRg-FUS (MR-guided focused ultrasound) versus UAE (uterine artery embolization) for uter-ine fibroids. http://clinicaltrials.gov/show/NCT00995878. Published October 13, 2009. Updated April 22, 2014. Accessed July 9, 2014.

120. Zhao WP, Chen JY, Zhang L, et al. Feasi-bility of ultrasound-guided high intensity focused ultrasound ablating uterine fibroids with hyperintense on T2-weighted MR imag-ing. Eur J Radiol 2013;82(1):e43–e49.

121. Smart OC, Hindley JT, Regan L, Gedroyc WG. Gonadotrophin-releasing hormone and magnetic-resonance-guided ultrasound sur-gery for uterine leiomyomata. Obstet Gyne-col 2006;108(1):49–54.

122. Kim YS, Kim BG, Rhim H, et al. Uterine fibroids: semiquantitative perfusion MR im-aging parameters associated with the intra-procedural and immediate postprocedural treatment efficiencies of MR imaging-guided high-intensity focused ultrasound ablation. Radiology 2014;273(2):462–471.

123. Kim YS, Lim HK, Kim JH, et al. Dynamic contrast-enhanced magnetic resonance imag-ing predicts immediate therapeutic response of magnetic resonance-guided high-intensity focused ultrasound ablation of symptomatic uterine fibroids. Invest Radiol 2011;46(10): 639–647.

124. Funaki K, Fukunishi H, Funaki T, Kawakami C. Mid-term outcome of magnetic reso-nance-guided focused ultrasound surgery for uterine myomas: from six to twelve months after volume reduction. J Minim Invasive Gynecol 2007;14(5):616–621.

125. Arleo EK, Khilnani NM, Ng A, Min RJ. Fea-tures influencing patient selection for fibroid treatment with magnetic resonance-guided focused ultrasound. J Vasc Interv Radiol 2007; 18(5):681–685.