Akila N. Viswanathan
ANATOMY
The uterus is a hollow, thick-walled, pear-shaped, muscular organ located in the pelvis above the vagina, behind the bladder, and in front of the rectum (Fig. 69.1). On average, it is approximately 7 to 8 cm long, 5 to 7 cm wide, and 2 to 3 cm thick. The uterus is divided into the uterine corpus superiorly and the uterine cervix inferiorly, with the most superior part of the corpus also known as the fundus and the middle portion of the corpus known as the body. The fundus is located superior to the line joining the entrance of the fallopian tubes. The body of the uterus is enclosed between layers of the broad ligament and is freely mobile. The regions of the body where the fallopian tubes enter are called the cornua. The most inferior, slightly constricted, portion of the uterus is called the isthmus or lower uterine segment (LUS). The cervix rests inferior to the LUS.
The uterus is usually bent anteriorly (anteflexed) between the cervix and the uterine body. The entire uterine-cervix structure is normally bent anteriorly (anteverted) in the pelvis. The uterus is frequently posteriorly retroverted, especially in older women who have a small uterus. The wall of the uterus has three layers: the outer serosal layer; the middle myometrium, which is approximately 12 to 15 mm of muscle through which the main blood vessels and nerves flow; and the inner coat called the endometrium.
The cervix measures approximately 3 by 3 cm and is predominantly a fibrous organ. The cervix is divided into an upper or supravaginal portion, above the ring containing the endocervical canal, and the vaginal portion, projecting in the vaginal vault. Central in the rounded vaginal region is the external os, bounded by the anterior and posterior lips of the cervix, extending inward to the internal os, the endocervical canal, and endometrial canal.
The uterus is partially covered by peritoneum in its fundal portion and posteriorly; its anterior and lateral surfaces are related to the bladder and the broad ligaments, respectively. It is attached to the surrounding structures in the pelvis by two pairs of ligaments—the broad and the round ligaments. The broad ligament is a double layer of peritoneum extending from the lateral margin of the uterus to the lateral wall of the pelvis. It contains the fallopian tubes. The two layers of peritoneum forming the broad ligament enclose the parametrium as it reaches the uterus. Inferiorly, the broad ligament follows the plane of the pelvic floor and ends medially in the upper portion of the vagina.1
The round ligament, a band of smooth muscle and connective tissue that contains small vessels and nerves, extends forward horizontally from its attachment in the anterolateral portion of the uterus to the lateral pelvic wall. The cord ascending from the lateral wall of the true pelvis crosses the pelvic brim and extends laterally to reach the abdominoinguinal ring, through which it leaves the abdomen to traverse the inguinal canal and terminates in the superficial fascia.
The uterosacral ligaments are paired supports for the lower uterus, extending from the uterus to the sacrum and running along the recto-uterine-peritoneal fields.1 The cardinal ligaments, also called transverse cervical ligaments (Mackenrodt’s), are thickened connective tissue and fascia arising at the upper lateral margins of the cervix and inserting into the fascial covering of the pelvic diaphragm.
The uterus including the uterine cervix has a rich lymphatic network (Fig. 69.2) that drains principally into the paracervical lymph nodes; from there it goes to the external iliac (of which the obturator nodes are the innermost component) and the hypogastric lymph nodes. The pelvic lymphatics drain into the common iliac and the para-aortic lymph nodes. Lymphatics from the fundus pass laterally across the broad ligament continuous with those of the ovary, ascending along the ovarian vessels into the para-aortic lymph nodes. Some of the fundal lymphatics also drain into the common iliac lymph nodes. The main artery supplying the uterus is the uterine artery, which originates from the anterior division of the hypogastric artery.
EPIDEMIOLOGY
Over the last 80 years, the morbidity and mortality of locally advanced invasive cervical cancer has dramatically declined in the United States and Europe due to effective screening and treatment of preinvasive lesions.2 In the United States 50% of women who develop cervical cancer have never been screened and another 10% have not been screened within the previous 5 years. However, in the last decade, incidence rates of invasive carcinoma have remained relatively constant. The American Cancer Society estimates that approximately 12,200 new cases of invasive carcinoma of the cervix arise in the United States and about 4,200 deaths will occur per year, in addition to >60,000 cases of carcinoma in situ.3
Worldwide, cervical cancer remains the most common gynecologic cancer and the third-most-common malignancy in women, with over 500,000 women globally developing this tumor and 233,000 dying of the disease every year.4 Unfortunately, it often affects young women, resulting in loss of the ability to bear future children. The larger societal impact from the death of young women in the prime of life and motherhood has not been measured.
In developing countries, cervical cancer is the leading cause of cancer-related death.2 Cervical cancer is more common in areas where women have less access to screening, including parts of Asia, Africa, and Central and South America. Whether regional differences in predisposition to developing cervical cancer exist is debated because it is impossible to adequately correct for unknown and known confounders, such as socioeconomic status, access to health care, parity, smoking, presence of other infections, immune status, and other factors affecting host immunity such as nutritional status.5
FIGURE 69.1. Anatomy of the pelvis. (Asset provided by the Anatomical Chart Company Lexington, SC.)

Human Papilloma Virus
Estimates indicate that >90% of cervical cancers are related to the presence of human papilloma virus (HPV) and are contracted via sexual intercourse.6 HPV is a small, double-stranded DNA virus; HPV 16 and 18, as well as a long list of other, less frequent subtypes, including but not limited to HPV 31, 33, 35, 39, 45, 51, 52, 56, and 58, have been well characterized as causative agents for cervical cancer, with some geographic variation.7 The HPV genome integrates into the host cell chromosomes in cervical epithelial cells and codes for six early and two late open reading frame proteins, of which three (E5, E6, and E7) alter cellular proliferation. Two viral genes, E6 and E7, are typically expressed in HPV-positive cervical-cancer cells. The E6 protein inactivates the major tumor suppressor p53; this causes chromosomal instability, inhibits apoptosis, and activates telomerase. The E7 protein affects the retinoblastoma protein (Rb), resulting in a loss of regulation of the cell’s proliferation and immortalization.8
Although a high prevalence of HPV exists worldwide, peaking at ages 25 to 35 years, <15% of exposed women develop persistent infection that results in dysplasia,9 whereas the majority of women clear the infection within 2 years.10 Cervical cancer may develop 10 to 20 years after initial exposure to HPV. Social factors related to cervical cancer include those associated with HPV transmission, such as early age of first intercourse; a history of multiple sexual partners; a male partner with a history of multiple sexual partners; a large number of pregnancies;11,12 and a history of sexually transmitted disease, including gonorrhea, chlamydia,13 herpes simplex virus II, and/or human immunodeficiency virus (HIV).14 A higher incidence of cervical cancer exists among women whose spouses are known or suspected to have had higher exposure to HPV15 or whose partners have a history of penile carcinoma.16,17Whether circumcision may be protective to women is controversial18 because circumcision may be a surrogate for unknown factors related to HPV transmission.19
Chemical,20 hormonal, or other carcinogens21 may be implicated in cervical cancer. An association between cervical carcinoma and oral contraceptive use has been reported but is considered controversial.22 Prenatal exposure to diethylstilbestrol (DES) is linked to the development of clear-cell adenocarcinoma, although the overall incidence is small (0.14 to 1.4 per 1,000 DES-exposed women).23–25 Cigarette smoking may increase the risk of cervical cancer.26 However, passive smoking may not be an independent factor in the absence of active smoking.27 A review of >50 studies considers smoking a cofactor for HPV infection and carcinogenesis,28 although one study does not confirm this.15 Current smoking (relative risk [RR] = 1.55) and younger age at HPV exposure (RR = 1.75) are considered risk factors among HIV-positive women.29 Intrauterine device use may decrease cervical cancer risk, potentially through an increase in cellular immunity triggered by the device.30
HPV Vaccination
The quadrivalent human papillomavirus recombinant vaccine for HPV types 6, 11, 16, and 18, first approved in the United States in 2006 for girls and women ages 9 to 26 years, is now available for boys ages 9 to 26 years, with the goal of eradicating HPV-related gynecologic, penile, anal, and oropharyngeal cancers. A second vaccine with strong immunogenicity to HPV types 16 and 18, approved for girls 9 to 25 years old, is more frequently administered in Europe. Although its development is a major advance in the prevention of cancer, vaccine implementation has been hindered worldwide by cost and access. With an increase in understanding and availability, the hope is that all children will be given a vaccine covering all subtypes in the future.
FIGURE 69.2. A: Lymph vessels and lymph nodes of the cervix and the body of the uterus. (Asset provided by the Anatomical Chart Company, Lexington, SC.) B: Three-dimensional reconstruction of location of pelvic and common iliac lymph nodes outlined on computed tomography scans in patients with carcinoma involving the distant vagina. Treatment portal is shown. C: The incidence of cervical cancer increased slightly in the U.S. from 2005–2010.

TABLE 69.1 INCIDENCE OF PELVIC NODE METASTASES IN CARCINOMA OF THE UTERINE CERVIX

TABLE 69.2 METASTASES TO PARA-AORTIC LYMPH NODES IN CARCINOMA OF THE UTERINE CERVIX

NATURAL HISTORY AND PATTERNS OF SPREAD
Squamous cell carcinoma of the uterine cervix usually originates at the squamous columnar junction (transformation zone) of the endocervical canal and the portio of the cervix.
Cellular transformation follows a stepwise progression from normal to higher levels of dysplasia. Of patients diagnosed with cervical intraepithelial neoplasia (CIN) type 1, 60% have regression of the lesion, and of those with CIN2, 40% regress. Higher levels of dysplasia are more likely to progress to cancer, particularly in the presence of cofactors such as smoking or impaired immunity. Although progression typically takes 10 to 20 years,31,32 in some instances a rapid development of carcinoma may be associated with aggressive disease.
The development of a malignant phenotype results from cells that break through the basement membrane of the epithelium and invade the cervical stroma. Invasion may result in spread to pelvic lymph nodes or other, more distant sites.33 If the cells are detected at this stage by a Papanicolaou (Pap) or thin preparation test, appropriate minimally invasive therapy may suffice. However, if the lesion progresses, it may present as a superficial ulceration or exophytic tumor in the ectocervix or with extensive infiltration of the endocervix. If untreated, the tumor may spread to the adjacent vaginal fornices, paracervical or parametrial tissues,34 or adjacent organs, including the bladder, the rectum, or both. Landoni et al.35 studied 230 patients with clinical stages IB and IIA tumors treated with radical hysterectomy with pelvic lymphadenectomy and noted that the tumor spread endocervically equally in all directions. Tumor extension into the vesicocervical ligament (anterior parametrium) was noted in 23% of cases, into the uterosacral ligaments (posterior parametrium) and the rectovaginal septum in approximately 15%, and into the parametria in 28% to 34% of cases. Paracervical extension was related to the depth of stromal invasion, tumor size, lymphatic invasion, and presence of lymph node metastasis.
Approximately 10% to 30% of patients with carcinoma of the uterine cervix have extension into the lower uterine segment and the endometrial cavity.36 Decreased survival rates and a greater incidence of distant metastases were reported by Perez et al.36 and Chao et al.37 in patients with stromal endometrial invasion or replacement of normal endometrium by cervical carcinoma. Regional lymphatic or hematogenous spread may occur and increases with stage, although dissemination does not always follow an orderly sequence, and occasionally a small primary tumor may be seen infiltrating the pelvic lymph nodes, invading the bladder or rectum, or metastasizing distantly.
Both adjacent parametrial and pelvic lymph nodes may be involved. Girardi et al.38 analyzed 359 radical hysterectomy specimens and found positive parametrial nodes in 280 patients (78%); the incidence of positive nodes was 11.4% in stage IB and 21.5% in stage IIB disease. With negative parametrial nodes, only 26% of patients had positive iliac lymph nodes, whereas 81% of patients with positive parametrial lymph nodes also had pelvic-node metastases. These data underscore the need to irradiate the parametrial tissues or carry out a complete bilateral pelvic lymphadenectomy with a radical hysterectomy in patients with invasive cervical carcinoma.
Spread of carcinoma of the cervix may progress to the obturator lymph nodes, considered a medial group of the external iliac chain, to other external iliac nodes, and to the hypogastric lymph nodes. From these, there may be tumor metastases to the common iliac or para-aortic lymph nodes.39 The incidence of metastasis to pelvic or para-aortic lymph nodes for various stages of the disease is listed in Tables 69.1 and 69.2. In one study, pelvic lymph nodes were dissected in 225 patients with cervical carcinoma treated with radical hysterectomy; positive pelvic nodes were identified in 13 of 91 women (14.2%) with stages IB and IIA, 16 of 81 (19.8%) with stage IIB, and 11 of 40 (28%) with stage IIIB disease.40 The most commonly involved groups were the parametrial, obturator, external iliac, and common iliac nodes (Fig. 69.3). Para-aortic lymph nodes were involved in 3 of 91 patients (3.3%) with stage IB or IIA tumors 4 cm or less and in 5 of 38 patients (13.1%) with stage IIB or III disease.
Spread through the venous plexus and the paracervical veins resulting in hematogenous dissemination, though infrequent, is relatively common with more advanced stages. In an analysis of 322 patients in whom distant metastases developed, the most frequently observed metastatic sites were the lung (21%), para-aortic lymph nodes (11%), abdominal cavity (8%), and supraclavicular lymph nodes (7%).41 Bone metastases occurred in 16% of patients, most commonly to the lumbar and thoracic spine (Table 69.3). Spinal epidural compression from metastatic tumor, often involving lumbar segments, can occur rarely,42 and metastasis to the brain and the heart have been reported, although it is unusual to have spread to the brain without evidence of pulmonary metastases already present,43,44 even for small-cell carcinoma of the cervix.45
FIGURE 69.3. Distribution of pelvic node metastases in 14 patients with stages IB to IIA cervical cancer, tumor size <4 cm (A), and 38 patients with locally advanced cervical cancer treated with neoadjuvant chemotherapy (B). (From Benedetti-Panici P, Maneschi F, Scambia G, et al. Lymphatic spread of cervical cancer: an anatomical and pathological study based on 225 radical hysterectomies with systematic pelvic and aortic lymphadenectomy. Gynecol Oncol 1996;62:19–24; with permission from Elsevier.)

PAP SMEAR SCREENING
The American College of Obstetrics and Gynecology guidelines published in 200946 state that Pap smear screening should begin at age 21 years and continue every 2 years until age 30 years; then, if there are three normal consecutive Pap smears and no history of CIN2, CIN3, DES exposure, or HIV infection and the woman is not otherwise immunocompromised, screening should be every 3 years. Women who have had a hysterectomy for benign reasons and have no history of high-grade squamous intraepithelial lesion may discontinue testing. Co-testing of the Pap smear with an HPV DNA test is appropriate for low-risk women older than age 30 years. If negative, rescreening is not required sooner than 3 years. Women who have been treated for CIN2 or CIN3 need annual screening for at least 20 years. Those who have had a hysterectomy and a history of CIN2/CIN3 should continue to undergo screening with annual pelvic exams.
When obtaining the Pap smear, special attention should be directed to not using a lubricating agent (warm water on the speculum will suffice), to obtaining good “scrapings” from the cervix and vaginal posterior fornix (without blood), and to using a small brush to obtain an endocervical sample. The patient should be instructed not to cleanse with a douche before the examination, and, if indicated, specimens should be obtained to check for trichomonas. If the cytologic smear shows atypia or mild dysplasia (class II), it should be repeated no sooner than 2 weeks after the initial test to allow representative cellular exfoliation to occur. Guidelines for reporting results of cervical and vaginal cytology were promulgated in 1988. The Bethesda system eliminated the classes of Pap cytology. The correlation between the cytologic diagnosis and subsequent histologic examination is >90%.47 This system was modified in 1991 and in 2001.48
TABLE 69.3 CARCINOMA OF THE UTERINE CERVIX (MALLINCKRODT INSTITUTE OF RADIOLOGY 1959–1986): ANATOMIC SITE OF FIRST METASTASIS

TABLE 69.4 DIAGNOSTIC WORKUP FOR CARCINOMA OF THE UTERINE CERVIX

CLINICAL PRESENTATION
Cervical cancer in the United States is most frequently identified during routine gynecologic examination. Intraepithelial or early invasive carcinoma of the cervix may be detected by cytologic smears before symptoms appear; Pap smear, colposcopy and biopsies, and HPV testing have high specificity and sensitivity. Visible lesions present with an exophytic mass or a barrel-shaped cervix due to an endocervical lesion. Patients may present complaining of metrorrhagia (intermenstrual bleeding), menorrhagia (heavier menstrual flow), or postcoital bleeding. If chronic bleeding occurs, the patient may complain of fatigue or other symptoms related to anemia.
In cases with more advanced disease, bowel obstruction, renal failure, foul-smelling serosanguinous or yellowish vaginal discharge, pelvic pain, flank and/or leg pain, rectal bleeding, obstipation, dysuria, hematuria, or persistent edema of lower extremities due to lymphatic/venous blockade by pelvic sidewall disease may occur. Pain in the pelvis or hypogastrium may be caused by tumor necrosis or associated pelvic inflammatory disease. In patients with pain in the lumbosacral area, the possibility of para-aortic lymph node involvement with extension into the lumbosacral roots or hydronephrosis should be considered.
DIAGNOSTIC WORKUP
When a patient presents with an abnormal smear or if abnormal squamous cells of undetermined significance (ASCUS) are detected but HPV status is negative, follow-up in 1 year is recommended. If the second smear reveals ASCUS, regardless of HPV status, colposcopy is recommended. When both ASCUS and HPV are present or adenocarcinoma in situ or a squamous intraepithelial lesion is identified, directed biopsies at the time of colposcopy should be carried out. Endocervical curettage may be performed except in pregnant women. If the biopsy results are negative, the procedure should be repeated in 6 months, and, if they are positive, a conization should be performed.
Patients who present with a clinically visible lesion should be jointly evaluated by the radiation and gynecologic oncologists. After obtaining a careful clinical history and performing a general physical examination, with attention to the inguinal and supraclavicular (nodal) areas, abdomen, and liver, a careful pelvic examination should be carried out with as little discomfort to the patient as possible without compromising the thoroughness of the evaluation.49Pelvic examination should include inspection of the external genitalia, vagina, and uterine cervix, a rectal examination, and bimanual palpation of the pelvis. Pelvic examination under anesthesia is a universally accepted component in the evaluation and clinical staging of patients in order to provide a pain-free examination that allows a clearer estimation of parametrial or sidewall tumor extension. In countries in which magnetic resonance imaging (MRI) is available, this may be used to assist with assessing tumor extension beyond the lower cervix, and in many institutions it has replaced the examination under anesthesia. Cystoscopy or rectosigmoidoscopy should be performed in all patients with symptoms consistent with presence of a fistula of the urinary or lower gastrointestinal tract, patients with clinical stage IIB, III, or IVA disease who cannot undergo an MRI, or patients with an MRI suspicious for bladder or bowel invasion. The diagnostic procedures for carcinoma of the cervix are presented in Table 69.4.
Conization/Loop Excision
Conization involves a conical removal of a large portion of the ectocervix and endocervix. Cold knife cone biopsy specimens should always be obtained with a scalpel or other appropriate instrument. At least 50% of the endocervical canal should be removed without compromising the internal sphincter. Curettage of the remaining endocervical canal should be carried out.
Conization must be performed in the following situations: no gross lesion of the cervix is noted and an endocervical tumor is suspected; the entire lesion cannot be seen with the colposcope; diagnosis of microinvasive carcinoma is made on biopsy; discrepancies are found between the cytologic and the histologic appearances of the lesion; or the patient is not reliable for all necessary follow-up. With careful selection of patients who have a negative positron emission tomography (PET) scan and an MRI with a central lesion <2 cm in width, knife conization with lymphadenectomy may be considered for fertility preservation. Laser conization and loop diathermy excision are frequently done in an office setting as an alternative to conization; loop excision is less expensive and more reliable than laser conization.
Biopsy
Multiple punch biopsies of a grossly visible lesion should be adequate to confirm the diagnosis of invasive carcinoma. Specimens should be obtained from any suspect area and from all four quadrants of the cervix and from any suspect areas in the vagina. It is important to obtain tissue from the periphery of the lesion with some adjoining normal tissue; biopsy specimens from central ulcerated or necrotic areas may not be adequate for diagnosis. Dilation and curettage is not required if the biopsy confirms a diagnosis of invasive disease.
TABLE 69.5 COMPUTED TOMOGRAPHY AND POSITRON EMISSION TOMOGRAPHY IN THE EVALUATION OF PARA-AORTIC NODES

Laboratory Studies
For invasive carcinoma, patients should have the following laboratory studies: complete peripheral blood evaluation, including hemogram, white blood cell count, differential and platelet count; blood chemistry profile, with particular attention to blood urea nitrogen and creatinine; liver function values; and urinalysis.
Imaging Studies
In countries where three-dimensional (3D) imaging is not routinely available, patients with cervical cancer should have a chest radiograph to assess for lung metastases and an intravenous pyelogram (IVP) to determine whether hydronephrosis is present. The IVP in many countries has been replaced by computed tomography (CT) scan of the pelvis and abdomen with intravenous (IV) contrast material or by PET/CT scan. In places where CT is not available, patients with stages IIB, III, and IVA disease who have symptoms in the colon and rectum may benefit from a barium enema. A skeletal survey may be performed to determine whether bone metastases are present. Historically, pedal lymphangiography was used to assess lymph node involvement in the pelvic or para-aortic nodes with mixed results.50,51 The number of physicians trained to perform lymphangiography has declined in the United States, and instead PET scan is preferred.
Since the 1990s, the use of CT scanning has rapidly increased worldwide. A CT provides diagnostic information about the presence of metastases, enlarged lymph nodes, and the primary tumor. On a CT scan, the cervical tumor may be seen as an enlarged, irregular, hypoechoic cervix or as a mass with ill-defined margins. Parametrial regions appear dense when involved, and uterosacral involvement may be seen. Lymph nodes appear enlarged, with most >1 cm on axial dimension considered pathologic. The overall accuracy of CT scanning in staging cervical cancer ranges from 63% to 88%.50,52 In the detection of lymph node abnormalities, the overall accuracy of conventional CT scanning is 77% to 85%, with sensitivity of 44% and specificity of 93%.53
In order to correlate radiographic and surgical findings, Camilien et al.54 reported on 61 patients with carcinoma of the cervix who had both preoperative CT scans and exploratory laparotomy; results showed that 75% of the enlarged pelvic lymph nodes on CT contained metastases, and 97% of patients with negative nodes on CT scan had pathologically negative findings (specificity of 97%). However, histologically positive pelvic nodes were often missed on CT scan (sensitivity of 25%). The CT scan is more valuable in evaluation of the para-aortic lymph nodes (specificity of 100% and sensitivity of 67%).
Several studies evaluated the role of CT or PET/CT with regard to para-aortic nodal detection (Table 69.5). Heller et al.55 conducted a prospective evaluation of 320 patients with stages IIB to IVA carcinoma of the cervix entered into a Gynecologic Oncology Group (GOG) protocol in which preoperative CT scan, lymphangiography, and ultrasonography of the aortic area were performed. Para-aortic node dissection was done in patients with negative staging studies. Lymphangiography, CT scan, and ultrasonography had false-negative frequencies for pelvic lymph node evaluation of 14.2%, 25%, and 30%, respectively. The sensitivity was 79% for lymphangiography, 34% for CT scan, and 19% for ultrasonography, and the specificity ratings were 73%, 96%, and 99%, respectively. Ultrasonography, therefore, is not reliable in preoperative detection of lymph node metastases, but it has limited value in evaluating extrauterine tumor involvement. Ultrasound has a primary role in assisting with intracavitary brachytherapy applicator insertion and may detect uterine perforation, allowing for proper positioning, which is critical for adequate dosing and affects survival.56,57 PET has a higher sensitivity than CT and higher specificity than MR in detecting bone metastases.58
Magnetic Resonance Imaging
MRI is frequently used for the initial assessment of the cervical tumor and of extracervical tumor extension,59 often in lieu of an examination under anesthesia. MRI is contraindicated in patients with pacemakers, cochlear implants, metallic prostheses, metallic fragments from prior accidents, or large vascular clips. On T2-weighted images, a cervical cancer may be seen as a mass of intermediate to high signal intensity, usually of greater intensity than the fibrocervical stroma. On T1-weighted images, tumors are usually isointense with the normal cervix and may not be seen60 but can increase in intensity with the administration of IV contrast. Abnormal, irregular cervical margins, prominent parametrial strands, exocentric parametrial enlargement, and loss of parametrial fat planes on T1-weighted images or high signal in the parametria or cardinal/uterosacral ligaments on T2-weighted images are indicative of more extensive tumors.52,59,61 Parametrial tumor may be identified as brighter regions on T2-weighted images when compared to the low signal intensity of the cervix and uterine ligaments.
A comparative evaluation of pretreatment tumor staging and volume as assessed by examination under anesthesia (EUA), transrectal ultrasonography (TRUS), and MRI in 60 patients with invasive carcinoma of the cervix was reported by Hawnaur et al.62 TRUS and MRI assigned the same tumor stage in only 30% of patients, and EUA and MRI agreed on tumor stage in an additional 27%. In cases of disagreement, the MRI staging correlated better with outcome than TRUS or EUA. Sixty-two percent of patients with enlarged lymph nodes on pretreatment MRI either died or had tumor recurrence or metastases. MRI was superior to both TRUS and EUA in assessing the full extent of bulky tumors and lymph node enlargement.
Postema et al.63 compared MRI with pelvic examination (including under general anesthesia in selected patients) and surgicopathologic findings in 103 patients with invasive cervical carcinoma. MRI was better at identifying extracervical tumor spread, but it had more false-positive results. The pelvic examination led to correct treatment decisions in 89% of patients. In a study by Hansen et al.,64 clinical assessment (done according to International Federation of Gynecology and Obstetrics [FIGO] recommendations) was superior to low-field MRI with contrast enhancement in staging cervical cancer in 95 women who had both within 2 weeks after clinical diagnosis; the clinical staging correctly classified 57 patients (accuracy, 92%) compared with 52 for MRI (accuracy, 84%).
A prospective study by the American College of Radiology Imaging Network compared clinical examination, CT, and MRI.53 MRI was significantly better than clinical examination or CT for detecting uterine-body involvement or measuring tumor size,65 but no method was accurate at evaluating the cervical stroma. MRI was significantly better66 at detecting the tumor and parametrial involvement. MRI also somewhat increased detection of involved lymph nodes.53
The tumor is less likely to be as visible on MRI for adenocarcinoma cases, compared to squamous cell cancer. Haider et al.67 evaluated 56 patients with adenocarcinoma involving the cervix using MRI and noted that 42 (75%) had a visible mass. Kodaira et al.68 reviewed records of 84 patients with stage II cancer evaluated by MRI. The 5-year disease-free survival (DFS) rate of patients with maximal tumor size (Dmax) of ≥50 mm was significantly lower than that for patients with Dmax < 50 mm (46% vs. 88%; p < .0001).
Ebner et al.69 reviewed MRI findings in 12 women with recurrent pelvic tumors and 10 with a fibrotic mass (confirmed by laparotomy or biopsy in 21 patients). They were able to differentiate between the two processes accurately in most instances. However, it is highly desirable to confirm abnormal or suspect lymph node radiographic findings with CT-guided fine-needle aspiration biopsies.
Corn et al.70 evaluated endorectal coil MRI in 18 patients with stages IB to IIIB cervical carcinoma; in 7 patients, tumors were a higher stage by endorectal coil MRI because of proximal vaginal involvement or the combination of proximal vaginal involvement and parametrial extension. Compared with those who had a dark or intermediate signal, patients with bright signal characteristics tended to present with earlier stages, were less likely to have anemia, and were more likely to have complete response to external-beam radiation.
Radiation-induced changes detected over the course of radiation may predict local recurrence and survival. Mayr et al.71 studied 34 patients with cervical cancer of various FIGO stages who underwent 1.5-T MRI before and after radiation therapy. Tumor volumetry (3D measurements) based on T2-weighted images quantified the tumor regression rate. Sequential tumor volumetry using MR imaging may be a very effective measure of the responsiveness of cervical cancer to irradiation. MRI dynamic contrast enhancement during the first 2 weeks of radiation therapy may provide early prediction of tumor regression rate. In 7 patients, tumor regression rates ranged from 2% to 15.2% per day and correlated positively with changes in both peak and mean tumor enhancement (p < .01). Hatano et al.72 evaluated MRI in 42 patients with advanced cervical cancer treated with external-beam irradiation and high–dose-rate (HDR) brachytherapy. In biopsies performed immediately after radiation therapy (RT), no residual cancer was found in 36 patients (86%). The simultaneous MRI study demonstrated no high-signal intensity on T2-weighted images in 28 patients (75%). A high–signal-intensity area was observed in 14 patients, and this disappeared 3 months after RT in 8 patients with a negative biopsy. The sensitivity, specificity, and accuracy of MRI tumor response studies at 3 months after radiation therapy were 100%. MRI studies performed after 30 Gy of external-beam irradiation and 3 months after all radiation therapy predicted local tumor control. Similar studies were published by Gong et al.73 and van de Bunt et al.74 Furthermore, MRI is useful in providing accurate target volume definition in brachytherapy treatment planning (Fig. 69.4).75
FIGURE 69.4. A: Magnetic resonance imaging (MRI) at diagnosis showing an enlarged cervical tumor. B: MRI with tandem and ring brachytherapy applicator in place showing dramatic shrinkage of the tumor after concurrent chemotherapy with external beam radiation.

Positron Emission Tomography
PET scanning is increasingly used in the evaluation of patients with malignant neoplasia, including invasive cervical cancer, using 2-[18F]-fluoro-2-deoxy-D-glucose (FDG). Rose et al.76 observed uptake in 91% of the primary tumors in 32 patients with locally advanced carcinoma of the cervix. Squamous cell carcinoma is more often FDG avid than is adenocarcinoma. Compared with surgical staging, PET scanning had a sensitivity of 75% and a specificity of 92% in detecting para-aortic metastasis.77 PET-CT provides highly accurate localization of focal radiotracer uptake, which significantly improves the diagnostic accuracy compared with PET or CT alone. Diagnostic PET images may be fused with simulation CT images to ensure accurate radiation dose coverage of the target and any PET-avid lymph nodes (Fig. 69.5). Care must be taken in interpretation because physiologic FDG excretion into the urinary bladder may result in false-positive assessment of the primary tumor, and ureters may be contoured as lymph nodes; therefore, tracing the ureters and complete bladder voiding prior to imaging are recommended.
Grigsby et al.77 compared CT and FDG-PET scanning for lymph node staging in 101 patients with carcinoma of the cervix. CT demonstrated abnormally enlarged pelvic lymph nodes in 20 patients and para-aortic lymph nodes in 7, whereas PET demonstrated abnormal FDG uptake in pelvic lymph nodes in 67, in para-aortic lymph nodes in 21, and in supraclavicular lymph nodes in 8. The 2-year progression-free survival rate, based solely on para-aortic lymph-node status, was 64% in CT-negative and PET-negative patients, 18% in CT-negative and PET-positive patients, and 14% in CT-positive and PET-positive patients (p < .0001). The most significant prognostic factor for progression-free survival was the presence of positive para-aortic lymph nodes on PET imaging (p= .025). Among 76 patients with no abnormal FDG uptake, the 2-year survival rate was 86%, with persistent abnormal uptake in 40%; there were no survivors among patients who developed new sites of abnormal uptake.78 In a follow-up study of 152 patients, the authors reported a 5-year cause-specific survival rate of 80% in 114 patients without abnormalities on posttherapy FDG-PET versus 32% in 20 patients with persistent uptake, and no survivors among 18 patients with new sites of abnormal uptake.79 In another study, Grigsby et al.80 noted in 208 patients a close correlation between radiation doses, number and size of positive lymph nodes and outcome (treatment failures and survival; Fig. 69.6). Hope et al.81 performed FDG-PET scans in 58 patients with cervical carcinoma who had an endometrial biopsy or dilatation and curettage; 36 (64%) had pathologic endometrial invasion (EI). Pelvic lymph node metastasis was more commonly detected in this group than in patients without EI (70% vs. 23%; p < .001), as were para-aortic and supraclavicular nodal metastasis (30% vs. 0%; p = .006). Furthermore, 2-year survival rates were 78% versus 58%, and overall survival rates were 92% versus 65%, respectively (p = .047). Lin et al.,82 using FDG-PET in 32 patients with cervical carcinoma, observed a reduction in physiologic tumor volume of 50% occurring within 20 days from the initiation of radiation therapy.
Kidd et al.83–87 demonstrated that maximum standardized uptake value (SUV max) is an independent predictor of death from cervical cancer and is associated with persistent disease. Similarly, the SUV of the pelvic node predicts pelvic disease recurrence. Survival rates are worse with supraclavicular nodal PET positivity; para-aortic lymph node metastases portend a survival rate between those of pelvic node and supraclavicular positivity. The detection of supraclavicular metastases on FDG-PET at diagnosis and its relationship to clinical outcome for 186 cervical cancer patients was reported by Tran et al.88 Fourteen patients (8%) had abnormal FDG uptake in left supraclavicular lymph nodes without palpable disease, confirmed on biopsy; 6 were treated with palliative intent, and 7 received definitive irradiation and concurrent chemotherapy. The median overall survival was 7.5 months; all patients developed distant metastases. After external-beam radiation is finished, an FDG-PET scan provides important information about posttreatment uptake and is prognostic with regard to outcome.89–92
FIGURE 69.5. A fusion of a diagnostic positron emission tomography (PET) scan with simulation computed tomography delineates the hypermetabolic lymph nodes, allowing for radiation dose escalation. In this case, the entire para-aortic chain received 45 Gy, followed by a sequential boost to the PET-avid node with a 7-mm margin to approximately 65 Gy.

FIGURE 69.6. Cause-specific survival in patients with carcinoma of the cervix correlated with pelvic lymph node status on posttreatment 2-[18F]fluoro-2-deoxy-D-glucose/positron emission tomography. (From Grigsby PW, Singh AK, Siegel BA, et al., Lymph node control in cervical cancer. Int J Radiat Oncol Biol Phys 2004;59:637–638; with permission from Elsevier.)

STAGING
The FIGO staging system is based on clinical evaluation (inspection, palpation, colposcopy); roentgenographic examination of the chest, kidneys, and skeleton; and endocervical curettage and biopsies. Lymphangiograms, arteriograms, imaging findings, and laparoscopy or laparotomy findings should not be used for clinical staging. The 2009 FIGO staging system for cervical cancer has one modification from the previous version: Stage IIA has been divided into stage IIA1, with tumors invading into the upper vagina but ≤4 cm in size, and stage IIA2, with tumors >4 cm in size.93
Patients with hydronephrosis or a nonfunctioning kidney ascribed to extension of the tumor are classified as stage IIIB regardless of the pelvic findings. Other prognostic factors, such as endometrial extension of cervical carcinoma, stromal invasion, lymphatic/vascular permeation, and involvement of the lateral parametrium (as opposed to the medial parametrium) in stage IIB, are not included in the staging system. Suspected invasion of the bladder or rectum should be confirmed by biopsy. Bullous edema of the bladder and swelling of the mucosa of the rectum are not accepted as definitive criteria for staging. For a lesion to be classified as stage IIIB based on tumor extension without hydronephrosis, the tumor should extend to the lateral pelvic wall, although fixation is not required.
A parallel TNM staging system is published by the American Joint Committee on Cancer;94 however, this system requires nodal staging, which is not feasible in many settings, and radiologic nodal information is not available to most patients with cervical cancer worldwide. The FIGO system remains the standard staging system, given the lack of 3D imaging to determine nodal status in countries with the highest incidence of cervical cancer. All histologic types should be included. When there is a disagreement regarding the staging, the earlier stage should be recorded (Table 69.6 and Fig. 69.7).
FIGURE 69.7. Diagrammatic representation of various anatomic stages of carcinoma of the uterine cervix, according to the International Federation of Gynecology and Obstetrics classification. Stage IIA has been divided into stage IIA1, with tumors invading into the upper vagina but ≤4 cm in size, and stage IIA2, with tumors >4 cm in size.

PATHOLOGIC CLASSIFICATION
More than 90% of tumors are squamous cell carcinoma. Approximately 7% to 10% are classified as adenocarcinoma, and 1% to 2% are the clear-cell mesonephric type. Squamous cell (or epidermoid) carcinoma is composed of cores and nests of epithelial cells arranged randomly; cells show central keratinization with pearls and sometimes necrosis. Nonkeratinizing tumors may be seen. Electron microscopy may show desmosomes and tonofilaments. Squamous cell carcinomas are divided into three types: large-cell keratinizing, nonkeratinizing, and small-cell carcinomas. They are subdivided according to the degree of differentiation into well, moderately, or poorly differentiated.
Verrucous carcinoma is a variant of a very well differentiated squamous cell carcinoma that characteristically has a tendency to recur locally but not to metastasize.95 Mitotic activity is very low. It may be difficult to discriminate verrucous carcinoma from a giant condyloma with cytologic atypia or from a well-differentiated invasive squamous carcinoma. Microscopically, verrucous carcinoma is exophytic, with an undulating, hyperkeratotic surface; the deep margin is composed of large, bulbous masses that invade along a wide front in a “pushing” fashion.
Adenocarcinoma arises from the cylindrical mucosa of the endocervix or the mucus-secreting endocervical glands. Mucinous is the most common subtype of adenocarcinoma. This endocervical adenocarcinoma may form mucosal glands lined by high columnar cells and produce tubular folds oriented in many directions. In another subtype, cells resemble those of the intestines; the epithelium tends to be pseudostratified and may contain goblet cells. The third variant is the signet-ring cell adenocarcinoma, which is rare and usually mixed with the endocervical or intestinal patterns.
Endometrioid carcinoma is the most common cell type of endocervical adenocarcinoma; the cells resemble those of the endometrium, and the presence of intracytoplasmic mucin in some cells may be seen in a substantial proportion of tumors. The World Health Organization recommends that endometrioid or endocervical types of adenocarcinoma be graded according to their architecture, based on the degree of gland formation.96
Sometimes it is difficult to differentiate a primary endocervical adenocarcinoma from an endometrial tumor. Drescher et al.97 described a higher incidence of involvement of the uterine corpus and the regional lymph nodes in 21 patients with adenocarcinoma compared with a similar number of patients with squamous cell carcinoma. Chao et al.98 and Contag et al.99 described the use of microarray analysis for gene profiling (cDNA/RNA) to understand the molecular features of these tumors, which could aid in their classification. HPV has been identified in some subtypes of adenocarcinoma of the cervix.100
Adenoma malignum is a rare form of cervical cancer that is difficult to diagnose, and often highly malignant and refractory to treatment.101 Adenoma malignum is associated with Peutz-Jeghers syndrome and has an ominous natural history, with few reported cures.102 Adenosquamous carcinoma is also relatively rare (2% to 5%) and consists of intermingled epithelial cell cores with squamous features and glandular structures. The squamous component is frequently nonkeratinizing. If the squamous component is benign metaplasia, the tumor is called adenoacanthoma.
Glassy-cell carcinoma (1% to 2%) is considered a poorly differentiated adenosquamous tumor; it is rare and highly malignant. Survival is poor after surgery or irradiation. Ulbright and Gersell,103 in five cases of glassy-cell carcinoma evaluated by light and electron microscopy, described both glandular and squamous differentiation. Littman et al.104 reported only 4 of 13 patients, the majority with stage II disease, surviving 5 years (6 had extrapelvic failures). Piura et al.105 reported on 5 patients with cervical glassy-cell carcinoma, 3 with stage IB1 disease. All 3 patients were alive without disease 4, 12, and 18 months after diagnosis.
Adenoid cystic carcinoma is a rare variant of adenocarcinoma of the cervix (<1%), with an appearance similar to its counterparts in the salivary gland or the bronchial tree.106 The tumor is composed of nests and nodules of small carcinoma cells with a few characteristic cribriform patterns. Immunohistochemical findings for type IV collagen and laminin reveal intercellular cylinders composed of basement membrane material in the solid area without a cribriform pattern. They are locally aggressive and prone to metastasize.107
Clear-cell carcinoma (mesonephric), not related to DES exposure, comprises approximately 2% primary cervical adenocarcinomas and is believed to arise in mesonephric remnants.24 These tumors are submucosal, composed of clear and “hobnail” cells, and may grow in a tubular, glandular, papillary, or solid pattern. They appear at any age, with one-third occurring in women younger than 30 years of age. The clear cell is characterized by a voluminous cytoplasm filled with glycogen and the hobnail cell by single-cell apical projections into the neoplastic lumina. These tumors tend to be deeply positioned, with the bulk of the lesion on the stroma forming tubular structures, diffusely infiltrating the cervical stroma.
Cervical malignant mixed Müllerian tumors, compared with their counterparts in the corpus, are more commonly confined at presentation and may have a better prognosis. Clement et al.108 described the clinicopathologic features with mixed Müllerian tumors of the cervix in nine patients. Gross examination revealed polypoid or pedunculated masses that invaded the cervical wall in 50% of the hysterectomy specimens. On microscopic examination, five tumors contained basaloid carcinoma or squamous cell carcinoma and four contained adenocarcinoma. In seven tumors, the sarcomatous component was homologous, usually resembling fibrosarcoma or endometrial stromal sarcoma, and two tumors contained heterologous sarcomatous elements.
Small-cell carcinoma of the cervix, according to some authors, arises from endocervical argyrophilic cells or their precursors, multipotential neuroendocrine cells; however, some small-cell tumors do not contain morphologic evidence of neuroendocrine origin. Nuclear molding, absence of nucleoli, cell necrosis, and high mitotic activity are common. One-third to one-half stain positively for neuroendocrine markers such as chromogranin, serotonin, synaptophysin, or somatostatin.45 In the majority of patients, the cervical stroma is extensively infiltrated by single small, round cells.109 Lymphatic and vascular invasion are significantly more common in small-cell carcinomas (noted in 58% of patients with stage IB disease; 40% of these patients had lymph-node metastases at the time of radical surgery).110 HPV 18 has been detected in the majority of these tumors.111
Van Nagell et al.,112 in an analysis of 25 patients, noted a 5-year survival rate of 54% for all stages of small-cell carcinoma, compared with 68% for matched large-cell nonkeratinizing squamous cell and 74% for keratinizing squamous cell carcinomas. Viswanathan et al.45 studied 21 patients. All were confirmed after central pathology review to stain positively for chromogranin, synaptophysin, or CD56. The median time to first relapse from the initiation of treatment was 8.4 months. No patient had brain metastases as the sole site of first recurrence. However, 2 patients developed brain metastases concurrently with lung metastases. The overall survival rate was 29% at 5 years; none of the patients who had disease more extensive than stage IB1 or clinical evidence of lymph node metastases survived their disease.
Basaloid carcinoma or adenoid-basal carcinoma, an extremely uncommon tumor, is characterized by nests or cords of small basaloid cells, prominent peripheral palisading of cells in the tumor nests, no significant stromal reaction or capillary space invasion, and an infiltrating growth pattern. Some authors have suggested a slow growth pattern with limited local invasiveness and low probability of lymph node metastases.113 Prognosis is excellent.114
Primary sarcomas of the cervix have been occasionally described (e.g., leiomyosarcoma, rhabdomyosarcoma, stromal sarcoma, carcinosarcoma).115 Malignant lymphomas, primary or secondary in the cervix, have been sporadically reported. They should be treated like other lymphomas.116 Melanoma of the cervix is similarly extremely rare and difficult to cure despite attempts at radical surgery. Metastasis of distant tumors to the uterine cervix is rare (about 4% of all tumors) and should be considered in the differential diagnosis. Metastases to the cervix from the breast, ovary, and kidney have been reported.117–119
PROGNOSTIC AND PREDICTIVE FACTORS
Patient-Related Factors
Age
According to some reports, age is not a prognostic factor in carcinoma of the cervix.120 Other authors noted decreased survival in women younger than 35 or 40 years,121 who have a greater frequency of poorly differentiated tumors. In contrast, two European studies showed improved outcome for younger patients.122 This apparent contradiction may be explained by an analysis by Rutledge et al.,123 who showed an interaction between age and stage in the relative hazard plots for 250 patients younger than 35 years of age and matched control subjects. Mitchell et al.124 evaluated 398 patients with stage I to III cervical carcinoma treated with radiation therapy. Patients were divided into nonelderly (35 to 69 years of age; n = 338) and elderly (≥70 years of age; n = 60) groups. Comorbid conditions in the elderly resulted in diminished ability to undergo intracavitary brachytherapy. Although the 5-year actuarial disease-free and cause-specific survival (CSS) rates were comparable in the two groups, tumor recurrence and death from cervical cancer were more common beyond 5 years in the elderly group.
Race/Socioeconomic Status
Several authors noted a correlation between racial or socioeconomic characteristics of patients and outcome of therapy. Mundt et al.125 examined factors affecting outcome in 316 African American and 94 white patients undergoing RT for cervical cancer. With a median follow-up of 72.4 months, African Americans had a trend toward poorer 8-year cause-specific survival rates (47.9% vs. 60.6%; p = .10) compared with white patients. Factors correlating with poor outcome, including lower hemoglobin (Hb) levels during RT (p = .001), lower median income (p = .001), and less frequent intracavitary brachytherapy (p = .09), were more likely to be present in the African American group. Multivariate analysis demonstrated that race was not an independent prognostic factor after controlling for differences in patient, tumor, and treatment factors. In a report on 452 white and 124 African American women with stage II or III cancer of the cervix treated with RT alone, Grigsby et al.,126 observed 5-year CSS rates for stage II of 66% and 61% (p = .56) for those with stage II and of 38% and 47% (p = 0.34) for those with stage III disease, respectively. Overall survival rates for stage II for the two racial groups were different (60% and 51%, respectively; p = .02) and may be related to non–cancer-related comorbidity factors.
Brooks et al.127 evaluated 1,009 patients with invasive carcinoma of the cervix: 606 white, 354 African American, and 5% “other” races. African Americans were more likely to have Medicaid or to be uninsured (44% vs. 23%; p = .001) and were more likely to be admitted for an emergency or for a cancer-related complication (p = .036), to have comorbid illness (p = .001), to be admitted for a transfusion (p = .01), or to be treated with radiation rather than surgery (p = .001). Racial differences existed in patterns of admission, type of therapy, and severity of illness.
Moreover, in an analysis of the 1994 Patterns of Care study of 471 cases of squamous cell carcinoma treated in the United States and a randomly selected 215 additional cases from 17 institutions that admitted >40% minority patients, women who lived in low-income neighborhoods, who had only Medicaid coverage, or who were treated at large academic or minority-rich institutions tended to have a poorer initial performance status, higher-stage or bulky central tumor, and a lower pretreatment hemoglobin level.128
General Medical Factors
Anemia and Tumor Hypoxia
Although stage, tumor volume, histologic type of the lesion, and vascular or lymphatic invasion are known to affect the prognosis of patients with cervical carcinoma, hemoglobin levels may also contribute to patient prognosis. Many radiation oncologists routinely administer red blood cell transfusions (RBCTs) to correct anemia before treatment with radiation therapy. This may have a generally favorable effect on the patient’s sense of well-being and energy level, and an impact on tumor radiosensitivity. Typically patients receive transfusion to maintain hemoglobin levels >12 to 12.5 g/dL.
Hirst129 emphasized that in animal tumor models the opportunity to affect radiosensitivity by blood transfusion is transient. Blood transfusion is in general beneficial to the anemic patient with cancer, but it must be given as soon as possible before the first radiation dose to maximize its effects. Accounting for both the normal pulmonary and peripheral circulation and parallel flow through tumor tissue, Kavanagh et al.130 calculated that decreasing hemoglobin–oxygen affinity should render a quantitatively greater decrease in radiobiologically hypoxic regions than what has been measured after the use of transfusions alone.131
Investigators have reported worse outcomes for patients whose tumors have either a median partial pressure of oxygen (PO2) level, measured using polarographic needle electrodes for direct tumor-tissue oxygen measurements, of <10 mm Hg,132 or a high percentage of PO2 measurements <5 mm Hg.133,134 Comparisons of intratumoral oxygen measurements before and after external-beam radiation therapy have usually indicated a trend toward improved oxygenation after radiation therapy,135,136 but the significance of posttreatment measurements is unclear.137 Hypoxic tumors are more likely to recur locoregionally than well-oxygenated tumors regardless of whether surgery or radiation therapy is the primary local treatment.132
Haensgen et al.138 analyzed 70 patients with stage IIB to IVA cervical cancer treated with EBRT and brachytherapy. In vivo oxygenation was measured with an Eppendorf probe, and patient hemoglobin levels were recorded. Patients with a hemoglobin level of <11 g/dL had a 3-year survival rate of 27%, compared with 62% for those with a hemoglobin level of ≥11 g/dL (p = .006). Combining hypoxia and tp53 allowed stratification of subgroups with differing 3-year survival rates: 79% for tp53 (n = 10) and 47% for tp53 without hypoxia (n = 44).
A randomized trial reported by Bush139 on 132 patients with stage IIB to III cervical cancer required the control arm to receive transfusions only if the hemoglobin level dropped to <10 g/dL, whereas the experimental arm had to maintain the hemoglobin level ≥12.5 g/dL. The results suggested an improved outcome for patients in the experimental arm who received transfusion; however, there was not a statistically significant difference in outcome between treatment arms when compared using an intent-to-treat analysis.140 Second, the randomization was not stratified according to the potentially confounding influence of tumor size. Finally, the thresholds for transfusion were based on anemia during therapy, not the initial hemoglobin. Thomas141 reviewed the Canadian experience and found that in 605 eligible patients with cervical cancer, 25% received blood transfusions, most frequently when Hb was <100 g/L. On multivariate analysis, baseline Hb was not a significant prognostic factor, but average weekly nadir during radiation therapy was significant, with those with values >120 g/L having lower incidences of local relapse and distant metastasis and a better 5-year survival rate.
Dunst et al.142 showed that pretreatment anemia had a significant impact on 3-year relapse rates (6% in 20 patients with Hb of >13 g/dL, 15% in 47 with Hb between 11 and 13 g/dL, and 67% in 20 with Hb of <11 g/dL). The 3-year survival rate was 38% in patients with poorly oxygenated tumors, compared to 68% in patients with higher Po2 (p = .02). Munstedt et al.,143 in a study of 183 patients who received adjuvant RT after radical surgery, noted that those with Hb of <11 g/dL had lower recurrence-free and overall survival rates, primarily in a subgroup of women who had inadequate surgery.
In a retrospective review of >600 patients treated at seven different cancer centers in Canada, Grogan et al.144 observed that the patients who maintained an average weekly hemoglobin level of >12 g/dL with or without transfusions had a significantly higher 5-year survival rate than patients with lower average weekly hemoglobin levels, regardless of the hemoglobin at presentation.
Kapp et al.145 reported on 204 patients who received RBCT during RT when Hb level was <11 g/dL. Patients whose Hb was corrected (18.5%) had outcomes similar to those of nontransfused patients. However, nonresponders to RBCT had decreased tumor control and survival rates. Vaupel et al.,146 in a review of published data, concluded that maximum oxygenation of tumors is expected with Hb in the range of 12 to 14 g/dL for women and that higher Hb levels may not be better.
Recombinant human erythropoietin is not routinely recommended as an alternative means of sustaining or raising hemoglobin levels during radiation therapy. Thrombotic complications147 and the lack of any survival benefit148mitigate the utility of this as a therapeutic intervention.
Other Medical Factors
Jenkin and Stryker149 observed a higher incidence of pelvic recurrences and complications in patients with arterial hypertension (diastolic pressure of >110 mm Hg). Kapp and Lawrence150 reported on 398 patients; patients with temperatures of >101°F had a higher incidence of distant metastases and a lower survival rate. In patients with cervical cancer screened for HIV and treated with RT, Campbell et al.151 observed a 4.2% positive HIV rate. These patients had more-advanced tumors. The duration of remission was shorter than in the HIV-negative group. RT had no effect on the HIV titers. Women who are HIV positive or have acquired immunodeficiency syndrome associated with in situ or invasive carcinoma of the cervix are at a higher risk for tumor recurrence after treatment and death as a consequence of the malignant process.152,153
Evidence continues to mount that increasing levels of plasma micronutrients are associated with a decreasing risk of cervical cancer. Increasing serum lycopene and α- and γ-tocopheral levels and higher intake of dark green and deep yellow vegetables and fruit were significantly inversely associated with cancer.154–156 Women should be counseled to eat a well-balanced diet, particularly those at high risk for developing cervical cancer.
Tumor Factors
HPV Subtype
HPV 16 and 18 are the most frequent HPV subtypes worldwide. Studies have reported a higher risk of lymph node and other distant metastases with HPV 18 compared to HPV 16.157,158 Wang et al.159 studied 1,010 patients with cervical cancer after radiotherapy between 1993 and 2000. The HPV genotypes were determined by a gene chip that can detect 38 types of HPV. A total of 25 genotypes of HPV were detected in 992 specimens, of which 8 types that predominated were HPV16, 58, 18, 33, 52, 39, 31, and 45. Two high-risk HPV species were identified: α-7 (HPV18, 39, 45) and α-9 (HPV16, 31, 33, 52, 58). Risk groups determined included the high-risk group, which consisted of patients without HPV infection or those infected with the α-7 species only. The medium-risk group included patients coinfected with the α-7 and α-9 species.
Tumor Volume
There is a close correlation between depth of stromal invasion, tumor size, and incidence of parametrial and pelvic node metastases and survival in patients with cervical cancer.160,161 In a study of women treated with radical hysterectomy, the 5-year disease-free survival rate was 90% in 181 patients with stage IB1 (≤4 cm) and 72.8% in 48 patients with stage IB2 disease (p = .02).162
Toita et al.,163 in a review of 70 patients with stage IIB and IIIB carcinoma of the uterine cervix treated with RT alone, reported no significant correlation of 5-year DFS with size of the cervical tumor <60 mm (70% to 85%); however, in patients with tumor ≥60 mm, the 5-year DFS was 28.6%. Piver and Chung164 showed a greater incidence of lymphatic and distant metastasis and lower survival rates in patients with bulky and barrel-shaped stage IB and IIA tumors treated by radical hysterectomy. In addition, a higher incidence of pelvic recurrences and distant metastases and a decreased survival rate were reported by Fletcher,165 Eifel et al.,166 and Perez et al.167 in patients with larger tumors treated with irradiation. In stages IB and IIA, higher radiation doses improved local tumor control.168,169
Furthermore, Leveque et al.,170 in patients with stage I to II adenocarcinoma of the cervix treated with RT alone or combined with radical surgery, noted that FIGO stage and pelvic node involvement were the most important parameters influencing overall survival. Silver et al.,171 in 93 patients with stage I adenocarcinoma of the cervix, described patient age and tumor grade as significant prognostic variables for survival (p < .01 and .01, respectively); tumor size was significant (p < .01) for survival and progression-free survival.
In contrast, Grigsby et al.,172 in patients with stage IB and IIA carcinoma of the cervix treated with preoperative irradiation and radical or conservative hysterectomy, observed no correlation of tumor volume with outcome. The 5-year pelvic failure rates for stage IB were 16% for tumors <3 cm and 9% for larger tumors (p = .90) and for stage IIA were 22% for tumors <3 or >3 cm (p = .75).
Several retrospective studies demonstrated decreased survival and a greater incidence of distant metastases in patients with endometrial extension of a primary cervical carcinoma (endometrial stromal invasion or replacement of the endometrium by tumor only).36 Grimard et al.,173 on the other hand, confirmed these findings only in patients with stage IB tumors but not in more advanced stages. Similar findings were noted by Noguchi et al.174 Patients without uterine body invasion had a 5-year survival rate of 92.4%, compared with 53.8% in patients with invasion.
Perez et al.,169 in an update of a previous report,167 reviewed 1,499 patients (stages IA to IVA) treated with definitive irradiation (combination of external-beam irradiation plus two intracavitary insertions to deliver doses of 70 to 90 Gy to point A). There was a close correlation between tumor size and extent and pelvic tumor control, incidence of distant metastasis, and disease-free survival in all stages.
Margin Status After Radical Hysterectomy
In addition to the known high risk factors of positive margins, positive parametrial spread, and/or positive lymph nodes and the intermediate risk factors of depth of stromal invasion, lymphovascular invasion, and tumor size, small series have indicated the significance of close margin status.175,176 Viswanathan et al.177 studied 284 patients after radical hysterectomy (RH). The crude rates for any recurrence were 11%, 20%, and 38% for patients with negative (≥1 cm), close (>0 and <1 cm), and positive margins, respectively. Postoperative RT decreased the rate of local recurrence (LR) from 10% to 0% for negative, 17% to 0% for close, and 50% to 25% for positive margins. The significant predictors of decreased relapse-free survival on univariate analysis were the depth of tumor invasion (hazard ratio [HR] = 2.14/cm increase, p = .007), positive margins (HR = 3.92, p = .02), tumor size (HR = 1.3/cm increase, p = .02), lymphovascular invasion (HR = 2.19, p = .03), and margin status (HR = 0.002/increasing millimeter from cancer for those with close margins, p = .03).
Histologic Grade
Most reports have shown no significant correlation of survival or tumor behavior with the degree of differentiation of squamous cell carcinoma or adenocarcinoma of the cervix.178–180 Alfsen et al.178 analyzed 417 adenocarcinomas and 88 other non–squamous cell carcinomas of the cervix; on multivariate analysis, small-cell histology, corpus infiltration, vascular invasion, and positive lymph nodes were significant prognostic variables. Although Reagan and Fu181 demonstrated prognostic value of histologic differentiation in patients treated with irradiation, Crissman et al.182 failed to observe a correlation between histologic parameters and patient survival. In the era of chemoradiation, Monk et al.183 showed no significant impact of histology or grade on survival in postoperative cervical cancer patients with other high-risk features.
Treatment Duration
In patients treated with radiation therapy, overall treatment time should be as short as possible, and any planned or unplanned interruptions or delays should be avoided. Timely integration of external-beam and intracavitary irradiation in patients with carcinoma of the uterine cervix is an important factor in improving pelvic tumor control (Fig. 69.8).184 Several studies described lower pelvic tumor control and survival rates in invasive carcinoma of the uterine cervix when the overall time in a course of irradiation is prolonged.120,185–187 Chatani et al.,188 in 216 patients with stage IIB to III cervical carcinoma treated with a combination of external-beam and HDR brachytherapy, noted that overall treatment time was the most highly significant factor for local tumor control in multivariate analysis (p = .0005). For relapse-free survival, stage classification (p = .0001), overall treatment time (p = .0035), and hemoglobin level (p = .0174) were the three most important prognostic factors; there was no relationship between treatment time and late complications.
Fyles et al.185 reported approximately 1% loss of tumor control per day of prolongation of treatment time beyond 30 days in 830 patients with cervical carcinoma treated with irradiation alone. Lanciano et al.,187 in an analysis of 837 patients with squamous cell carcinoma of the cervix from the Patterns of Care Study who were treated with irradiation and received doses of 66 Gy or greater, described a 4-year actuarial in-field recurrence increase from 6% to 20% when total treatment time increased from 6 weeks or fewer to 10 weeks (p = .0001); this translated into significantly decreased survival. Girinsky et al.,186 in 386 patients with stage IIB or III carcinoma of the cervix, also observed that the 10-year local recurrence–free survival rate decreased when overall treatment time exceeded 52 days. A 1.1% loss of pelvic tumor control per day was also observed in their regression analysis.
Perez et al.,189 in 1,330 patients treated with definitive irradiation, noted a major impact of prolongation of treatment time on pelvic tumor control in stages IB, IIA, and IIB. In stage III, although the rate of pelvic failure was higher with prolongation of treatment time, the difference was not statistically significant. There was also a strong correlation between overall treatment time and survival. Regression analysis confirmed previous reports that prolongation of overall treatment time resulted in an increased failure rate of 0.59% per day in stage IB and IIA and 0.86% per day in stage IIB disease. Performance of all intracavitary insertions within 4.5 weeks from initiation of irradiation yielded lower pelvic failure rates (8.8% vs. 18% in stage IIB tumors; p ≤ .01).
FIGURE 69.8. Pelvic failure rate correlated with length of treatment in stages IB (A) and IIA (B) carcinoma of the uterine cervix. (From Perez CA, Grigsby PW, Castro-Vita H, et al. Carcinoma of the uterine cervix: I. Impact of prolongation of treatment time and timing of brachytherapy on outcome of radiation therapy. Int J Radiat Oncol Biol Phys1995;32:1275–1288; with permission from Elsevier.)

Biomarkers
Several studies assessed various biologic markers to determine whether they are prognostic; however, the majority suffered from not having multivariate analyses to determine whether these may be valid independent factors. Noordhuis et al.,190 in a systematic review of 42 studies with 82 cell-biologic markers, reported that on univariate analysis, 34 biologic markers showed a relation with survival, 27 of which were independently associated with survival.
Angiogenesis and Hypoxia
Angiogenesis—the formation of new blood vessels—relies on the presence of proangiogenic growth factors, such as vascular endothelial growth factor (VEGF). As new blood vessels form, they deliver nutrients and oxygen to the cancerous cells. As tumors expand, the newly formed blood vessels may no longer reach the central portions of the tumor, and a hypoxic core develops. Hypoxia-inducible factor (HIF-1α) and HIF-2α regulate the response to hypoxic stress; HIF-1α but not HIF-2α has been associated with poor disease-free survival.191–194 The HIF-2α/CD68 ratio was correlated with poor disease-free survival.194 One study found that VEGF decreased overall survival.195,196Loncaster et al.,195 in a retrospective study of 100 patients, found that VEGF expression in tumor biopsies in advanced carcinoma of the cervix was associated with a poor prognosis. Level of thymidine phosphorylase,196 which increases hypoxic conditions, similarly was associated with poor outcomes. Nitric oxide synthase197 and carbonic anhydrase (CA) may be prognostic for a poor outcome. In particular, CA9 is related to poor disease-free survival.198,199 CA12, in contrast, was related to metastasis-free survival.200
Microvessel count is higher in patients with cervical neoplasia than in control patients and higher in patients who experience posttreatment recurrences. Obermair et al.,201 in 166 patients with stage IB cervical cancer, observed a 5-year survival rate of 89.7% in 102 patients whose tumors had a microvessel density of 20 per field or less and 63% in 64 patients whose tumors had a microvessel density of >20 per field (log rank p < .0001). In a multivariate Cox model, microvessel density, lymph node involvement, tumor size, and the application of radiation therapy were independent prognostic factors for survival. Similar findings were reported by Cooper et al.202
Flow Cytometry Studies on DNA and Growth Fraction
Some authors noted no significant difference in recurrence rates between patients with diploid or aneuploid tumors. Kristensen et al.,203 in a study 465 patients with invasive carcinoma of the uterine cervix on whom DNA index and S-phase fraction studies were performed, observed that neither ploidy level nor S-phase fraction had prognostic significance. Others204 noted more relapses in tumors with an S-phase rate of 20% or greater.
Apoptosis
Whether apoptotic markers might be of importance in cervical cancer is unclear, given heterogeneity in the data. Morphologic studies have been negative, but some studies evaluating apoptotic protein expression such as that of Bcl-2 and p63 show association with poor disease-free survival.205,206
Ohno et al.207 studied 20 patients before and after administration of 9 Gy and found an increase in apoptotic cell index and Bax protein. Wootipoom et al.,205 in 174 patients with cervical cancer, noted Bax, Bcl-2, and p53 expression in 68.4%, 25.9%, and 77.6% of the cases, respectively. Bax expression was associated with better survival, whereas Bcl-2 expression was associated with poor survival. Jain et al.208 also found that neither Bcl-2 nor p53 expression was an independent predictor of outcome in locally advanced cervical cancer.
The p53 gene controls entry into the S phase of the cell cycle. Mukherjee et al.209 analyzed radioresistant cervical cancer cases and found that 15% had positivity for Bcl-2 and p21 proteins and 34% showed mutant p53 protein. None of the radiosensitive tumors were positive for these proteins. Seventy-five percent of the radiosensitive tumors were positive for the Bax antibody, whereas 81% of the radioresistant tumors were negative for Bax (p < .01). Kainz et al.,210 in a study of 109 surgically treated patients, and Ebara et al.,211 in 46 patients with stage IIIB squamous cell carcinoma of the cervix treated with RT alone, noted no significant difference in outcome when correlated with p53 protein expression.
Cell Cycle and Cellular Oncogenes
Cerciello et al.,212 in 40 patients with stage IIA to IIIB cervix cancer treated with RT without chemotherapy, obtained biopsies before and after five fractions of RT. They observed significant changes in the cell cycle of cervical cancer, indicating intact G2/M checkpoint function, leading to the expectation that targeting compounds interfering with G2/M transition may enhance the effect of irradiation on cervix cancer.
In patients with carcinoma of the uterine cervix treated with irradiation, Tsang et al.,213 observed that the most significant factors for disease-free survival were large tumor size (p = .01), low hemoglobin (p = .01), labeling index (LI) flow cytometry (disease-free survival, 67% for LI < 7%; 33% for LI ≥ 7%; p = .03), and potential doubling time (Tpot; 66% for Tpot > 5 days, 35% for Tpot ≤ 5 days; p = .04). For small tumors (<6 cm in diameter), either a high LI (>7%) or a high apoptotic index (>1%) was associated with poorer disease-free survival. West et al.214 evaluated the intrinsic radiosensitivity of 145 tumor biopsies from patients with cervical carcinoma (in vitro survival fraction at 2 Gy using a clonogenic assay). Diploid tumors tended to be more radioresistant than aneuploid tumors (p = .07).
The p27/Kip1 gene inhibits a variety of cyclin-dependent kinase complexes and regulates cell growth. Oka et al.215 studied 202 biopsy specimens obtained from 77 patients with squamous cell carcinoma of the cervix before and during RT for expression of p27 and p53 proteins. A high p27 LI before radiation therapy was associated significantly with good disease-free and metastasis-free survival rates. A high p53 LI before irradiation was associated with poor overall survival.
Both specific point mutations and amplification of ras genes have been noted. Overexpression of the ras gene p21 product is associated with a poor prognosis and increased frequency of lymph node involvement.216 Although loss of heterozygosity of the c-Ha-ras gene in squamous cell carcinomas was not associated with advanced-stage disease, mutations were associated with a poor prognosis. In contrast, mutations of the Ki-ras gene have been detected in a small percentage of cervical adenocarcinomas but have not been significantly associated with stage, grade, or survival.217,218
The c-myc oncogene is amplified from 3 to 30 times in approximately 20% of squamous cell carcinomas and is more frequent in high-stage compared with low-stage tumors. Overexpression of c-myc has been associated with a worse clinical outcome.219,220
Gadd45 belongs to the class II family of DNA damage-inducible genes, and its role in DNA repair has been proven in many experimental models. Santucci et al.,221 in 14 patients with cervical cancer, found a correlation between the lack of gadd45 induction and a clinical response to irradiation (both local tumor control and disease-free survival) when a dose ranging from 18 to 25 Gy was delivered to the pelvis.
CD 34 is an antigen present in hemopoietic progenitor cells and is a sensitive marker for endothelial cells. In 62 patients with cervical cancer evaluated by Vieira et al.,222 CD 34 reactivity and higher microvessel density were associated with squamous cell carcinoma. CD 109 is a cell surface protein that was found to be expressed in cervical cancer more than in endometrial adenocarcinoma.223
Cytokeratin Markers and the Epidermal Growth Factor Receptor Pathway
Altered expression of c-ercB-2 (HER2) protein was shown to have prognostic significance in adenocarcinoma but not in squamous cell carcinoma of the cervix.224 HER1, as well as coexpression of epidermal growth factor receptor and HER2, has been associated with poor disease-free survival.225 PTEN mutations have also been associated with poor prognosis.226
In 80 patients with carcinoma of the cervix, expression of cytokeratin 10 and 13 and involucrin was found in 24%, 64%, and 53%, respectively.227 There was no difference in the expression of cytokeratin or involucrin between patients with positive or negative lymph nodes, although in the lymph node–positive group, survival was higher in patients lacking cytokeratin 13 expression (p = .02).
Squamous Cell Carcinoma Antigen and Carcinoembryonic Antigen
Tsai et al.,228 in 117 patients with adenocarcinoma of the cervix, 28 of whom had preoperative carcinoembryonic antigen (CEA) levels of >5 ng/mL, noted a correlation with larger tumor size, deeper cervical invasion, and lymphovascular invasion (p < .001). A Spanish study of 96 patients with invasive carcinoma of the cervix and 7 with intraepithelial neoplasia showed elevated CEA levels in 33%, CA 19.9 in 32%, and CA 125 in 21.5% of patients.229 Specificity for each tumor marker was 98%. Increased CEA and CA 19.9 levels were found with more advanced stages of the disease and in patients with adenocarcinoma compared with squamous cell carcinoma. At follow-up, all cases of progressive tumor or recurrence were detected by elevation of one of the three antigens. Specificity during follow-up was 92% for CEA and CA 125 and 92.6% for CA 19.9.
In a study of 272 patients with invasive carcinoma of the cervix with 1,053 samples, Bolli et al.230 noted an elevation of squamous cell carcinoma antigen (SCC-Ag) before treatment in 53% of 103 patients, increasing with advancing tumor stage at diagnosis. In 70 patients with recurrent tumor, 81% had elevated SCC-Ag. Ngan et al.231 also identified elevation of serum SCC-Ag in 62% of 308 women with carcinoma of the cervix. Posttreatment SCC-Ag levels were raised in 69 patients (22.4%), and this was associated with a <5% 5-year survival rate, in contrast to 87% in women with normal SCC-Ag levels.
Hong et al.,232 in 401 patients with stage I to IV squamous cell carcinoma of the cervix treated with RT, noted that the preirradiation SCC-Ag level strongly correlated with disease stage. A persistently elevated SCC-Ag level 3 months after RT was a stronger predictor for treatment failure than residual induration by pelvic examination, and it was associated with a higher incidence of distant metastasis.
Similarly, Micke et al.,233 in 141 patients with cervical cancer treated with RT, noted that the pretherapy serum level of SCC-Ag was elevated in 72% (>2 ng/mL). Patients with a SCC-Ag of <7.2 U/mL had better tumor response than those with higher levels. After RT, 98% of patients in complete remission and 87% of those in partial remission had a serum level below the cutoff. In recurrent tumors, 82% of patients had a significant increase in serum levels before clinical manifestation of relapse (≤0.001). Hong et al.,234 in 1,031 patients with squamous cell cervical cancer treated with RT with or without chemotherapy, noted that independent risk factors for local relapse were advanced stage and age <45 years; 5-year local relapse-free survival was higher (90%) if squamous cell carcinoma antigen was <2. This antigen may be a useful marker in the prognosis of patients with carcinoma of the uterine cervix. Huang et al.235,236 noted that SCC-Ag and CEA were predictive of para-aortic node failure.
Epstein-Barr Virus, Transforming Growth Factor,
β-Integrin, and Other Markers
Activity of Epstein-Barr virus antigen–specific killer T cells and shedding of Epstein-Barr virus were evaluated in 55 patients with carcinoma of the cervix.237 Activity was decreased in patients with cervical carcinoma compared with control patients; it became increasingly lower as the clinical stage of the disease advanced, and activity after treatment was clearly related to patient survival. These data may indicate an imbalance in local immunity against viral infection and impairment of T-cell immunity in patients with advanced cervical carcinoma.
In 79 patients undergoing radiation therapy for carcinoma of the cervix, pretreatment transforming growth factor-β1 (TGF-β1) levels were a significant prognostic factor for survival and local tumor control. There were weak significant correlations of TGF-β1levels with disease stage and the levels of circulating tumor markers (CA 125).238 Hazelbag et al.239 also assessed TGF-β1 and plasminogen activator inhibitor (PAI-1) expression in 108 specimens of cervical carcinoma and noted that TGF was not associated with worse prognosis, whereas PAI-1 was.
Gruber et al.,240 in biopsies of 82 patients with cervical cancer, found that β3-integrin was expressed in 50 (61%) and correlated it with higher incidence of locoregional recurrences and decreased survival.
Cyclooxygenase-2
Gaffney et al.,241 in 24 patients with carcinoma of the cervix treated with RT, observed that 5-year overall survival rates for tumors with low versus high COX-2 values were 75% and 35%, respectively (p = .021). COX-2 staining intensity was found to correlate positively with tumor size (p = .022).
Kim et al.242 screened 84 patients with stage IIB squamous cell and 21 with adenocarcinoma cervical cancer and found COX-2 expression more frequently in the adenocarcinoma group (57% vs. 24%; p = .007). The 5-year survival rate was 83% for COX-2–negative and 57% for COX-2–positive patients, regardless of histologic subtype (p = .001). Pyo et al.243 also showed that expression of COX-2 and coexpression of COX-2 and thymidine phosphorylase were correlated with high locoregional recurrence and lower survival. Moreover, Kang et al.,244 in 84 patients with cervix adenocarcinoma, observed a higher incidence of lymph node metastasis and decreased survival with elevated expression of COX-2.
Hormonal Receptors
Suzuki et al.245 investigated the expression of estrogen receptors and progesterone receptors (PgRs) in biopsy specimens from cervical tumors before RT in 44 patients with cervical adenocarcinoma and 22 with adenosquamous cell carcinomas. Staining for estrogen receptors or PgR was positive in 12 patients (19%). The estrogen receptor status did not correlate with the local tumor control, or disease-free, or cause-specific survival. The disease-free survival rate of PgR-positive patients was significantly higher than that of PgR-negative patients (p = .044), but PgR status was not statistically significant in relation to 5-year cause-specific survival or local tumor control.
FIGURE 69.9. Risk of developing acute ovarian failure, defined as ovarian failure within 5 years, stratified by age and radiation dose to the ovary. (From Wo JY, Viswanathan AN. Impact of radiotherapy on fertility, pregnancy, and neonatal outcomes in female cancer patients. Int J Radiat Oncol Biol Phys2009;73:1304–1312; with permission from Elsevier.)

TECHNIQUES USED FOR TREATMENT
Preinvasive Disease
Patients with CIN may be candidates for observation or treatment. CIN1-2 has a spontaneous regression rate in 1 to 3 years of >50%,246 and therefore observation may be an appropriate course. For patients with persistent dysplasia, cryotherapy has a very low complication rate and is highly successful but may be less effective than laser ablation for high-grade dysplasia.247 Cold knife conization (CKC) may be used for diagnostic and therapeutic intent, with side effects of bleeding, cellulitis, cervical stenosis, or loss of cervical competence. Loop electrosurgical excision procedure (LEEP) has become popular, although bleeding and stenosis may occur; pregnancy outcomes may be better with LEEP than with CKC.248–250
Invasive Disease
Surgical Techniques
Simple Conization
In patients with minimal invasion, no parametrial involvement, and small tumor size, simple conization with lymphadenectomy has been reported. Although only 36 patients were studied, after a median follow-up of 66 months, only 1 pelvic nodal relapse was observed.251
Radical Trachelectomy
Described in the 1960s,252 trachelectomy entails removal of the cervix entirely. Radical trachelectomy also removes the parametrial tissue. As a means of fertility preservation in early-stage cervical-cancer patients,253 trachelectomy should be combined with preoperative PET imaging to confirm no nodal involvement and with MRI to confirm no endocervical canal extension of tumor into the uterus.254 A lapraroscopic lymphadenectomy should accompany the trachelectomy to confirm no nodal involvement. A nonabsorbable cerclage is placed around the uterine isthmus. Radical abdominal trachelectomy also may result in successful fertility preservation.255 Selection criteria for a trachelectomy include those patients requesting fertility sparing surgery, age <40 years, stage IA1, IA2, or IB1 with no nodal involvement detected on MRI or PET scan, squamous cell or adenocarcinoma with a lesion <2 cm, no lymphovascular invasion on initial biopsy, and no upper endocervical involvement. The main site for recurrence is central pelvis (average 5% risk), at the cervico-uterine junction, or in the adjacent parametrial tissue.
Types of Hysterectomy
Several types of hysterectomy are used in the management of carcinoma of the uterine cervix (Table 69.7).256
Total (extrafascial) abdominal hysterectomy (class I) consists of removal of the cervix and adjacent tissues, as well as a small cuff of the upper vagina in a plane outside the pubocervical fascia. There is minimal disturbance of the ureters and the trigone of the bladder. This may be the surgical treatment of choice for stage IA1 cervical cancer.
The use of a radical hysterectomy for cervical cancer was first described by Wertheim in 1912.257 In a modified radical extended hysterectomy (class II), the cervix and upper vagina are removed, including paracervical tissues, and the ureters are dissected in the paracervical tunnel to their point of entry into the bladder. Because the ureters are unsheathed and retracted laterally, parametrial and paracervical tissue can be safely removed medial to the ureter. This operation is performed with a lymphadenectomy. This is the most common surgical approach selected for stage IA2 cervical cancer.
Radical abdominal hysterectomy (class III) with bilateral pelvic lymphadenectomy consists of a wider resection of the parametrial tissues to the pelvic wall, with dissection of the ureters and mobilization of the bladder, as well as of the rectum to allow for more extensive removal of tissues. This approach was described by Meigs in 1944.258 In addition, a vaginal cuff of at least 2 to 3 cm is always included in the procedure, as well as the uterosacral ligaments.256 A bilateral pelvic lymphadenectomy is usually carried out. This operation is often referred to as the Wertheim or Meigs procedure. The extended radical hysterectomy (class IV) includes complete dissection of the ureter from the vesicouterine ligament, sacrifice of the superior vesicle artery, and removal of the upper three-fourths of the vagina. Due to the high rate of fistula and significant morbidity, it is rarely used.256 In 2007, after a consensus conference in Japan, a new classification system was released based only on the lateral extent of the resection.259 Four levels of hysterectomy are described (A to D) and four levels of lymphadenectomy (1 to 4). The levels of lymph node dissection include the internal and external iliac (level 1), common iliac and presacral (level 2), aortic inframesenteric (level 3), and aortic infrarenal (level 4). The levels of primary tumor resection include the following:
Type A: Extrafascial hysterectomy, with minimum resection of the paracervix medial to the ureter and minimal vaginal resection <1 cm, without removal of the paracolpos.
Type B: Modified radical hysterectomy with partial resection of the vesicouterine and uterosacral ligaments, unroofing of the ureter, transection of the parametrial tissue at the ureter, and removal of at least 1 cm of the vagina. This type is divided into B1, without removal of paracervical lymph nodes, and B2, with removal of lateral paracervical nodes.
Type C: Classic radical hysterectomy, variant in which the entire uterosacral and vesicouterine ligaments are removed, 1.5 to 2 cm of vagina with paracolpos is excised, and neuronal preservation is critical.
Type D: Includes the complete radical hysterectomy and resects tissues to the pelvis sidewall, including the hypogastric (internal iliac) vessels, and exposes the sciatic nerve (type D1). Type D2 also removes the fascial and lateral muscles, called the laterally extended endopelvic resection.
Pelvic Exenteration
Pelvic exenteration has been used for en masse removal of the pelvic viscera for recurrent carcinoma of the cervix. Modern radiation therapy with concurrent chemotherapy results in high complete response rates and has made residual extensive disease a rare indication for exenteration. Patients with adjacent organ invasion are given a course of radical concurrent chemoradiation, followed by interstitial brachytherapy, with exenteration reserved for salvage.260–262 This operation, which is not done as a palliative procedure, consists of a radical hysterectomy, pelvic lymph-node dissection, and removal of the bladder (anterior exenteration), removal of the rectosigmoid colon (posterior exenteration), or both (total exenteration). The ileum or sigmoid has been the usual means of achieving urinary diversion. Because some patients have a pelvic recurrence after radiation therapy, the bowel is used for the urinary conduit. Proof that there is no fixation to the pelvic wall and no extension of disease beyond the pelvis is mandatory. Metastases outside the pelvis, including those in para-aortic lymph nodes or any viscera, are absolute contraindications to the procedure. Bilateral ureteral obstruction secondary to tumor is also a relative contraindication.263 Patients with sacroiliac or hip pain or leg edema rarely benefit from this procedure and should be excluded on a clinical basis.
TABLE 69.6 FIGO STAGING OF CARCINOMA OF THE UTERINE CERVIX

TABLE 69.7 TYPES OF ABDOMINAL HYSTERECTOMY

TABLE 69.8 CARCINOMA OF THE UTERINE CERVIX: SURVIVAL AFTER STAGING LAPAROTOMY

Pretreatment Surgical Nodal Assessment
Exploratory laparotomy and nodal staging to evaluate the presence of metastases to the pelvic or para-aortic nodes may provide diagnostic information but has not had a demonstrated impact on survival (Table 69.8). In a prospective evaluation of 290 patients with carcinoma of the cervix,264 para-aortic node metastases were found in 19 of 58 patients (32.8%) with clinical stage IIB and 19 of 61 (31.1%) with stage IIIB disease. A number of studies compared the significance of para-aortic nodal metastases with other clinical and surgical findings with regard to progression-free survival and overall survival.265 In 626 patients treated on GOG randomized studies, the relative risk associated with positive para-aortic nodes was 11.0 for time to recurrence and 6.2 for survival time. In addition to the significant increase in risk of regional relapses, patients with para-aortic nodal metastasis are more likely to have extrapelvic failure.266
Cosin et al.267 reviewed 266 patients with locally advanced cervical carcinoma who underwent extraperitoneal pelvic and para-aortic lymphadenectomy before RT. Patients were divided into four groups: group A had negative lymph nodes; B, resected, microscopic lymph node metastases; C, macroscopically positive lymph nodes that were resectable at the time of surgery; and D, unresectable lymph nodes. Lymph node metastases were detected in 50% of patients. All patients received pelvic external-beam radiotherapy (EBRT; pre–intensity-modulated radiation [IMRT] era) and brachytherapy; patients with lymph-node metastases received extended-field irradiation. Five- and 10-year disease-free survival rates were similar for all patients in groups B and C. All patients in group D recurred. There was a 10.5% incidence of severe radiation-related morbidity and a 1.1% incidence of treatment-related deaths.
The potential benefit of surgical nodal debulking followed by irradiation has been studied.268 Potish et al.269 noted that more than half of the patients with advanced cervical cancer with grossly positive pelvic nodes that were debulked survived, compared with none with unresectable lymph nodes, findings closely paralleling those of Inoue and Morita270; however, these studies pre-dated the ability to implement dose escalation with IMRT to the involved nodes. Nonetheless, surgical debulking of nodes decreases the dose of radiation required to these regions. The use of high-energy photon beams and limitation of the tumor dose to extended volumes in the para-aortic region for those with completely resected but positive nodes to 45 to 50 Gy decrease the probability of complications. IMRT also enhances the sparing of adjacent abdominal normal tissues.271,272 However, if node dissection is not feasible or would result in a treatment delay, patients may successfully be managed with IMRT with dose escalation to approximately 60 to 65 Gy to the grossly enlarged lymph nodes.80,273,274, 275
Complication risk may be high (Table 69.9), particularly when patients are treated with postresection RT. One study showed an 11.5% incidence of major primarily small-bowel complications with transperitoneal compared with 3.9% in the extraperitoneal lymphadenectomy group (p = .03).276 Transperitoneal lymphadenectomy should be avoided.277,278
Due to a high rate of complications, preirradiation laparotomy was discontinued at the MD Anderson Cancer Center. The status of the lymph nodes is investigated with lymphangiography and verified when possible with percutaneous transabdominal needle biopsy. Wharton et al.279 reported on 120 patients who had preirradiation celiotomy; 16 had fatal complications, and 32 had major intestinal complications. Most patients with positive lymph nodes died with distant metastasis. More recently, PET/CT followed by biopsy may be a useful approach. In one series of 60 patients without evidence of para-aortic lymph node involvement on preoperative CT or MRI, patients underwent a preoperative PET/CT followed by surgery. Twenty-six patients had a negative PET/CT, of whom 3 (12%) had positive nodes pathologically. Of the 27 with positive pelvic but negative para-aortic nodes on PET/CT, 6 (22%) had pathologically positive para-aortic nodes, indicating a sensitivity of 36% and a specificity of 96%. In another study, 125 patients had a PET/CT followed by para-aortic lymphadenectomy.280 Seventeen percent had positive para-aortic metastases, 66% of whom had a negative PET/CT. The sensitivity and specificity of PET/CT were 33% and 94%, respectively. Morbidity of the surgery was 7%, and there was no delay in initiating chemoradiotherapy. A Cochrane overview of pretreatment surgical para-aortic lymph-node assessment in stage IB2-IVA cervical cancer identified one randomized trial of 61 women that favored CT or MRI over surgical staging.281,282 Alternatively, para-aortic node sampling may be performed through a laparoscopic approach; the procedure is well tolerated, recovery is prompt, and yield is adequate.283,284
Aside from para-aortic nodes, supraclavicular metastases may rarely be involved. Perez-Mesa and Spratt285 found no supraclavicular node metastasis in 73 consecutive patients with various stages of cervical carcinoma. Manetta et al.286 also did not detect scalene node metastasis in 24 patients with recurrent carcinoma of the cervix evaluated for exploration and possible pelvic exenteration. There is no indication for removal of the supraclavicular nodes.
TABLE 69.9 COMPLICATION RATE FOR PRETHERAPY SURGICAL STAGING

Sentinel Lymph Node Biopsy
Sentinel lymph node (SLN) studies show mixed results, with a 20% false-negative rate and a 50% incidence of other pelvic metastases.287 However, the method allows for aborting surgery and pursuing RT instead. Sentinel node biopsy in patients with cervical cancer is used in several institutions; a seven-center prospective study analyzed 139 patients and detected 454 sentinel lymph nodes. Intraoperative examination did not detect micrometastasis or isolated tumor cells. Other techniques, such as molecular assays, may improve SLN assessment in the future.288,289 The SENTICOL study290 injected 145 stage IA1 to IB1 cervical-cancer patients with combined technetium-99 and Patent Blue. The sensitivity (92%) and lack of false-negative results were favorable, but SLN biopsy was reliable only when SLNs were detected bilaterally.
Ovarian Transposition
In premenopausal women, surgery and radiation may directly impact gonadal function. For women with early-stage cervical cancer who undergo a radical hysterectomy, ovarian function may be preserved. In patients who require postoperative radiation, transposition of ovarian tissue outside of the radiation field may help to preserve ovarian function, although even low doses to the ovaries may cause acute ovarian failure (Fig. 69.9). In women who require radical radiation to the uterus and ovarian tissue, counseling before treatment begins with a fertility expert in reproductive endocrinology may be of benefit if an intervention such as egg retrieval is desired. Care must be taken to ensure that any potential intervention does not delay the initiation of curative treatment. In addition, women with early-stage cervical cancer have an approximately 1% risk of ovarian metastasis with squamous cell carcinoma and 5% risk with adenocarcinoma. Therefore, women should be counseled on the potential risks of recurrence with ovarian preservation.291
In a survey of 124 patients who had undergone radical hysterectomy and lymphadenectomy with ovarian transposition, 68 responders were premenopausal at the time of surgery. Six of 30 women (20%) with ovarian preservation experienced early hormonal failure (5 had one ovary, and 1 patient had both preserved).292 Combined-modality therapy affects ovarian function more than operation alone.293 Anderson et al.294 noted that only 4 of 24 patients (17%) with ovarian transposition who received postoperative pelvic irradiation had continued ovarian function. Feeney et al.295 reported on 132 patients on whom lateral ovarian transposition was performed at the time of radical hysterectomy; 28 patients received postoperative pelvic irradiation. Fourteen of 28 patients (50%) who received pelvic irradiation had evidence of ovarian failure, in contrast to 3 of 104 patients (2.9%) on whom ovarian transposition was performed, without postoperative irradiation. Buekers et al.296 also evaluated ovarian function in 102 patients with cervical cancer, 83 of whom underwent radical hysterectomy and 19 of whom had a staging laparotomy, all with ovarian preservation (80 included ovarian transposition); 26 patients received postoperative radiation therapy. After ovarian transposition without RT, 98% of patients retained ovarian function for a mean of 126 months, with menopause at a mean age of 45.8 years. When ovarian transposition and RT were added, 41% retained ovarian function for a mean of 43 months and experienced menopause at a mean age of 36.6 years.
Morice et al.297 reported on 107 patients treated for cervical cancer with radical hysterectomy and lymphadenectomy, 104 of whom (98%) had ovarian transposition to the paracolic gutters performed. Preservation of ovarian function was achieved in 100% for patients treated exclusively by surgery, 90% for those treated by postoperative vaginal brachytherapy, and 60% for patients treated by postoperative EBRT and vaginal brachytherapy.
Ovarian transposition or oophoropexy has been performed using laparoscopy, achieving continued hormonal function in 68% (8 of 11) and 50% (3 of 6) of the patients.298 Mean follow-up was 8.5 years, and mean radiation absorbed dose to the displaced ovaries was 26 Gy. Stockle et al.299 performed laparoscopic lateral ovarian transposition during staging lymphadenectomy in 11 patients with carcinoma of the cervix treated with brachytherapy (11 cases), EBRT (9 cases), and chemotherapy (2 cases). Ovarian preservation was achieved in 30% of the cases. Age was the most predictive factor for ovarian function preservation. Pahisa et al.300 performed laparoscopic ovarian transposition on 29 FIGO stage IB1 cervical-cancer patients. After a mean follow-up of 44 months, ovarian function was preserved in 93% of the nonirradiated and 64% of the irradiated patients.
Radiation Therapy Techniques
Since the early 1900s, radiation has been used in the curative management of cervical cancer, with a combination of external-beam and brachytherapy resulting in the highest survival rates. Over the ensuing 100 years, treatment-planning techniques have evolved, as has the equipment used for treatment. Several methods have been developed to aid with conformality and normal-tissue sparing. External irradiation is used to treat the whole pelvis. Structures treated include the uterus and cervix or, in the postoperative cases, the tumor bed, the vagina, the parametrial tissue, and the pelvic lymph nodes, including the internal, external, and common iliac nodes. In selected cases the para-aortic lymph nodes may be treated. In patients with locally advanced disease, in addition to external-beam radiation, treatment of central disease (cervix, vagina, and medial parametria) relies heavily on dose given with intracavitary sources through brachytherapy. The techniques described apply, with some individualization, to most patients with locally advanced cervical carcinoma.
External-Beam Irradiation
External-beam treatments may be routinely administered to cervical cancer patients with stages IB2 to IVA in a curative fashion. Patients with stages IA to IB1 may be considered for external-beam treatment if they are deemed inoperable or prefer to avoid surgery. Patients with stage IVB disease may receive palliative radiation to the pelvis for selected indications such as to stop vaginal bleeding, relieve pain, or alleviate urethral obstruction from extrinsic compression. External-beam radiation covers the primary cervical tumor, treats any adjacent parametrial or uterosacral, uterine, or vaginal extension, and, most important, addresses microscopic disease present in pelvic lymph nodes. In treatment of invasive carcinoma of the uterine cervix, it is important to deliver adequate doses of irradiation not only to the primary tumor, but also to the pelvic lymph nodes to maximize tumor control.
The initiation of external-beam radiation typically precedes brachytherapy. Although brachytherapy may be interdigitated with external-beam treatment, based on the desire to minimize the duration of treatment, the brachytherapy dose to the normal tissues may be better optimized after maximal tumor shrinkage; therefore, many institutions prefer to wait until the completion of 45-Gy treatment before initiating brachytherapy for patients with large tumors.
Patient Positioning
Patients may be positioned in either the supine position for stability or the prone position on a belly board. The prone position aids in shifting small bowel out of the pelvis. In patients who have had a hysterectomy, small bowel may drop into the pelvic area. For those patients treated for cervical cancer with an intact cervix, the small bowel often lies superior to the uterus and above the pelvic brim, creating less need to shift the bowel out of the pelvis. For patients receiving IMRT, due to stability of the pelvis, the supine position is typically preferred with immobilization devices surrounding the pelvis to ensure minimal motion during treatment. IV contrast may be helpful to localize the pelvic vessels for contouring but is not routinely used in most centers. Oral contrast delineates the small bowel. Rectal contrast and placement of a Foley catheter for bladder contrast are not considered necessary in the majority of cases that use CT simulation because the outer wall of these normal-tissue structures can be contoured without contrast on CT.
Plain X-Ray Simulation
If CT is not available, simple plain film simulation may be performed. The standard plain radiographic simulation to the pelvis with x-rays, typically using opposed anterior–posterior:posterior–anterior (AP-PA) fields, results in comprehensive coverage of all pelvic regions. Due to the lack of visible soft-tissue detail, contrast may be placed using barium in the rectum, a vaginal tube in the vagina, and/or a wire marker over surgical scars. The superior border is set at the L4-5 interspace in order to cover the common iliac lymph nodes and the lateral borders 1.5 to 2 cm from the pelvic brim, and the inferior border covers at least the obturator foramen (Fig. 69.10). More commonly in patients with large tumors, the inferior border extends to the ischial tuberosities. When there is vaginal involvement, the entire length of this organ should be treated down to the introitus. It is very important to identify the distal extension of the tumor at the time of simulation by placing a radiopaque clip or bead on the vaginal wall or inserting a small fiducial marker in the vagina. When the tumor involves the distal half of the vagina, the portals should be modified to cover the inguinal lymph nodes because of the increased probability of metastases (see Fig. 69.2).
For the lateral field borders, in both postoperative and intact cervix settings, the posterior border must be set in such a way that the entire sacrum is covered because the uterosacral ligaments are at high risk for harboring microscopic extension. The uterosacral ligaments insert onto the sacrum, and therefore the posterior block should ensure coverage of the entire sacrum. The anterior border on the lateral field should be set at a vertical line anterior to the pubic symphysis, since the external iliac lymph nodes must be covered.
For patients with para-aortic nodal involvement, simple plain film simulation followed by AP-PA treatments to the para-aortic nodal chain may overdose the kidneys, spinal cord, and small bowel. Dose escalation to para-aortic nodes to approximately >45 Gy is not feasible with AP-PA fields, given potential bowel complications. The use of four fields, including AP-PA and two lateral fields, is implemented as an alternative to AP-PA alone as a way to reduce some of the dose to the anterior small bowel. Patients receive oral barium approximately 30 minutes before the simulation to ensure blockage of as much small bowel as feasible superiorly. The superior border covers the renal hilum, often at the T12-L1 interspace, and the inferior borders cover the obturator foramen, unless there is distal vaginal or inguinal node involvement. For the para-aortic portion of the field, the anterior border rests 2 cm in front of the vertebral body or enlarged nodes as contoured, and posteriorly the border bisects the mid-vertebral body. The pelvic portion mimics that described for the four-field pelvic setup. The use of lateral fields allows a decrease in dose to the small bowel, but care must be taken to include all structures of interest.301–303
Three-Dimensional Conformal Treatment Planning
CT simulation allows direct assessment of the pelvic vessels and by adjacent location the para-aortic and pelvic nodes. Oral contrast is beneficial to identify the small bowel. Cerrobend customized blocks or multileaf collimator blocking is used on each field to block the radiation to selected areas, including the skin, muscle, soft tissue, anterior small bowel, and portions of the anus and lower rectum (Fig. 69.11).
The superior border is set based on the CT-visualized bifurcation of the common iliac nodes into the external and internal iliac nodes, which may lie as high as the L3-4 interspace. If patients have positive pelvic nodes based on PET imaging, the superior border may be shifted to either the superior border of the common iliac nodes or the superior aspect of the renal hilum to treat the para-aortic nodes. In postoperative cases in which the patient has had an extensive surgical staging, the superior border may be reduced to the L5-S1 interspace. Similar to plain x-ray simulation, in patients with vaginal involvement, the inferior border is extended to cover 2 cm below the lowest extent of disease, which may lie in the vulvar tissue, and in such cases the inguinal lymph nodes are treated, resulting in a wider AP field.
On the lateral fields, the anterior border covers the front of the pubic symphysis. Bonin et al.,304 in a review of 22 patients on whom detailed anatomic mapping of the anatomy of the pelvic lymph nodes was carried out by lymphangiography, concluded that if the criteria for adequacy of standard pelvic fields as defined in prior clinical trials were applied (anteroposterior: 1.5-cm margin on the pelvic rim; lateral field anterior edge is a vertical line anterior to the pubic symphysis and posterior border), 10 patients (45%) would have had inadequate nodal coverage in the irradiation fields. The incompletely irradiated lymph nodes were in the lowest lateral external iliac group. However, CT simulation with contouring may prevent omission of these nodes.
For the lateral borders in postoperative and intact cervix cases, posterior coverage of the entire sacral hollow is imperative. Zunino et al.305 reviewed the appropriateness of radiation therapy box technique for cancer of the cervix in 35 sagittal MRIs and 10 lymphangiograms. If the posterior border were to be placed at the S2-3 interspace, for 50% of the patients with FIGO IB and in 67% with stage IIA disease, the posterior border of the lateral field would not adequately encompass the planning target volume (PTV). In stage IIB, the posterior border was inadequate in eight patients (42%). In patients with stage IIB and IVA disease, the PTV was not encompassed. Furthermore, Knocke et al.306 used standard simulator planning guided by bony landmarks for pelvic irradiation in 20 patients with primary cervical carcinoma, stages I to III, using a four-field box technique. After defining the PTV with a 3D planning system, they compared the field configuration of the simulator planning with a second one based on the defined PTV. They evaluated the ability of the PTV to encompass the treatment volume (International Commission on Radiation Units and Measurements [ICRU]). Planning by simulation resulted in 1 geographic miss, and in 10 more cases the coverage of the PTV by the treatment volume was inadequate.
Finlay et al.307 contoured pelvic blood vessels on CT scans as surrogates for lymph nodes in 43 patients and found this to be more accurate than bony landmarks for field delineation. In total, 95% of patients planned with conventional fields had inadequate coverage of some portion of lymph node coverage, whereas in 56% additional normal tissue was treated that did not require radiation. Therefore, most centers implement 3D simulation when feasible. Taylor et al.308 used MRI to outline the pelvic lymph nodes in 20 patients and noted that with margins of 10 mm, nodal coverage was 94% and with 15 mm, 99%; with a modified 7-mm margin they ensured 99% nodal coverage with less volume of small bowel at risk.
FIGURE 69.10. A: Anteroposterior simulation film of the pelvis illustrating portals used for external irradiation. The 15 by 15 cm portals at source-to-skin distance are used for stage IB (broken line), and 18 by 15 cm portals are used for more advanced disease (solid line). This allows better coverage of the common iliac lymph nodes. The distal margin is usually placed at the bottom of the obturator foramina. B: Diagram of pelvic portals used in external irradiation of carcinoma of uterine cervix. Standard portal for stage IB tumors is outlined (solid line indicated as A). When the common iliac nodes are to be covered, the upper margin is extended to the L4–5 space (indicated in section B). If there is vaginal tumor extension, the lower margin of the field is drawn at the introitus (indicated in section C).

FIGURE 69.11. Anterior–posterior and lateral digitally reconstructed radiograph simulation film of the pelvis, illustrating portals used for external irradiation with the patient in the prone position on a belly board to minimize small-bowel dose. Pelvic lymph nodes are indicated in their approximate position. The uterus, cervix, and vagina are contoured to ensure adequate coverage.

In the postoperative setting, van den Berg et al.,309 using 47 lymphangiograms and 15 CT scans, asked radiation oncologists (n = 17) to define the clinical target volume (CTV) and PTV and to delineate on simulation films the RT treatment portals to be used after a radical hysterectomy with lymph node dissection for stage IB or IIA cervical carcinoma with positive iliac lymph nodes. Large variations were observed in the portals used and in treatment techniques. From the digitized films, it appeared that in 50% of the cases the defined PTV was not covered adequately. Furthermore, 71% of the treatment plans would not cover the lateral borders of the reference PTV sufficiently. Thus, there is a need for careful adherence to standardized guidelines.
Treatment techniques may have an effect on outcome. Yamazaki et al.310 compared 34 patients with cervical cancer treated with irregularly shaped four-field whole-pelvis radiation therapy using CT simulation and 40 patients receiving whole-pelvis EBRT with parallel-opposed fields in a nonrandomized study of postoperative radiation therapy consisting of 50 Gy in 25 fractions in 6 weeks. With a mean follow-up of 60 months, the actuarial 5-year pelvic tumor control was 94% with the two-field technique and 100% for the irregularly shaped four-field technique. The incidence of grade 2 or 3 bowel complications in the irregularly shaped–technique group (2.9%, 1 of 34) was significantly lower than that in the two-field–technique group (17.5%, 7 of 40; p < .05).
Burnett et al.311 described a prosthetic silicone plastic device that is filled with saline and Renografin for x-ray visualization (capacity between 750 and 1,500 mL) to conform to the pelvis and exclude the small bowel from the irradiated volume. The device remains in place throughout the radiation therapy course and is removed through a small incision after draining the contents of the prosthesis. Seven devices had been placed to date of the report. In the postoperative period, there was one pulmonary embolism. All seven patients completed planned radiation therapy. The devices have been removed with no adhesions to the prosthesis.
Intensity-Modulated Radiation
IMRT was developed using the techniques required for inverse planning. That is, one starts with the necessary dose around the target then works backward to develop the requisite beam intensities. IMRT spatially modulates the intensity of the beam using the motion of multileaf collimators. Because of the increasing use of intensity-modulated or image-guided radiation therapy (IMRT/IGRT) in the treatment of patients with gynecologic malignancy, there is growing emphasis on imaging the pelvic anatomic structures, including lymph nodes, for treatment planning.312 IMRT may reduce the amount of small bowel and bone marrow that receives the full dose of radiation.
The use of IMRT has been standardized in the postoperative setting but remains a topic of debate for intact cervix cases. What constitutes adequate margins in the intact cervix setting continues to be a matter of concern, given significant organ motion during treatment. Uncertainties in the definition of target volumes arise using 3D techniques. Bladder-filling and rectal-filling changes require accurate definition of margins for the PTV.313 Beadle et al.314found that mean maximum changes in the center of the cervix were 2.1, 1.6, and 0.82 cm in the superior–inferior, anterior–posterior, and right–left lateral dimensions, respectively. Mean maximum changes in the perimeter of the cervix were 2.3 and 1.3 cm in the superior and inferior, 1.7 and 1.8 cm in the anterior and posterior, and 0.76 and 0.94 cm in the right and left lateral directions, respectively. Haripotepornkul et al.315 found in 10 women with locally advanced cervical cancer that within and between radiation treatments, cervical motion averages approximately 3 mm but may be up to 18 mm in any given direction. The mean intrafractional movements in cervical seed positions in the lateral, vertical, and AP directions were 1.6 mm (standard deviation [SD] ± 2.0), 2.6 mm (SD ± 2.4), and 2.9 mm (SD ± 2.7), respectively, with a range from 0 to 15 mm for each direction. The mean interfractional movements in the lateral, vertical, and AP directions were 1.9 mm (SD ± 1.9), 4.1 mm (SD ± 3.2), and 4.2 mm (SD ± 3.5), respectively, with a range from 0 to 18 mm for each direction. Tyagi et al.316 show that a uniform CTV planning treatment volume margin of 15 mm would not encompass the cervical CTV in 32% of fractions. With IMRT, there is a need for continual replanning (at least every other week), given rapid tumor regression and internal-organ motion.317–319
FIGURE 69.12. Intensity-modulated radiation therapy treatment plan for external irradiation of pelvic lymph nodes while sparing organs at risk.

After a CT scan for simulation, the images are brought into a treatment-planning workstation. Contouring the gross tumor volume (GTV) may include the uterus, cervix, and/or vagina. Lim et al.320 had 19 experts in gynecologic radiation oncology contour a case of locally advanced cervical cancer on axial magnetic resonance images of the pelvis. Substantial Simultaneous Truth and Performance Level Estimation (STAPLE) agreement sensitivity and specificity values were seen for GTV delineation (0.84 and 0.96, respectively) with a kappa statistic of 0.68 (p < .0001). Agreement for delineation of cervix, uterus, vagina, and parametria was moderate. The greatest variability in physician contouring was in the parametrial tissue.320
The CTV for the pelvic lymph nodes was based on the Radiation Therapy Oncology Group (RTOG) atlas for the female postoperative pelvis.321 An example of dose distribution achieved with supine IMRT pelvic irradiation is illustrated in Figure 69.12. Fiducial markers may be placed in the apex of the vagina for identification on CT scan and show up to 3.5 cm of vaginal cuff motion during treatment.322 Therefore, for postoperative patients, the vagina is contoured using a full-bladder CT scan fused to an empty-bladder CT scan to account for vaginal mobility due to differences in bladder filling.323 This vaginal target volume has been referred to as an integrated target volume (ITV). The expansion of the CTV and/or ITV to the PTV is necessary, although given the movement of the uterus, the exact amount of margin is a matter of debate. Generous margins of approximately 2 to 3 cm are considered, particularly in the regions of the uterus and cervix or in the postoperative case around the ITV vagina. Dose constraints required for an optimal IMRT plan have not been standardized. In the RTOG postoperative clinical trial 0921 using IMRT,324a PTV of 7 mm around the nodal contours is recommended, and the dose is prescribed to cover 97% of the vaginal PTV and nodal PTV. A volume of 0.03 cc within any PTV should not receive >110% of the prescribed dose. No more than 0.03 cc of any PTV will receive <93% of its prescribed dose. Any contiguous volume of 0.03 cc or larger of the tissue outside the vaginal/nodal PTVs must not receive >110% of the dose prescribed to the vaginal/nodal PTV; for normal tissues the small/large bowel (30% of the entire bowel volume must not receive >40 Gy), rectum/sigmoid (60% of the rectosigmoid volume must receive ≤40 Gy), bladder (35% of the bladder volume must receive ≤45 Gy), and femoral head (15% of the femoral head volume must receive <35 Gy) constraints are being tested in RTOG 0921. Careful attention must be paid to all normal-tissue organ motion because the bladder and rectum may have 3- to 5-cm shifts due to filling changes in a short time frame.
For patients that have had a diagnostic PET or MRI before the simulation, these images may be registered to create a fused data set for contouring, particularly when a nodal boost is required. This allows the physician to visualize areas of PET enhancement or tumor volume, as seen on the MRI, onto the simulation films. Grigsby and colleagues325 use a PET-defined target volume contoured with a metabolically active tumor specified at the 40% threshold. Normal-tissue structures, including the rectum, sigmoid, bladder, and small bowel, are routinely contoured for patients treated with IMRT who will be undergoing a nodal boost in order to limit the dose received primarily to the small bowel. Based on an overview of published data, the absolute volume of small bowel receiving ≥15 Gy should be held to <120 cc when possible to minimize severe acute toxicity if delineating the contours of bowel loops themselves. Alternatively, if the entire volume of peritoneal space in which the small bowel can move is delineated, the volume receiving >45 Gy should be <195 cc when possible.326 For the rectum, dose–volume constraints selected as a conservative starting point that have not yet been validated for 3D treatment planning include V50 < 50%, V60 < 35%, V65 < 25%, V70 < 20%, and V75 < 15%.327 No dose constraint for external beam planning for the bladder could be identified, although the limits for prostate cancer may be adopted for gynecologic IMRT, including a dose constraint of no more than 15% of the volume to receive a dose >80 Gy, no more than 25% of the volume to receive a dose >75 Gy, no more than 35% of the volume to receive a dose >70 Gy, or no more than 50% of the volume to receive a dose >65 Gy.328
Imaging may guide more accurate definition of lymph nodes.308 Portelance et al.272 carried out IMRT as well as conventional planning with two- and four-field techniques in 10 patients. Prescription was 45 Gy in 25 fractions to the uterus and the pelvic and para-aortic lymph nodes. All IMRT plans were normalized to obtain a full coverage of the cervix with the 95% isodose curve (Fig. 69.13A). The volumes of small bowel receiving the prescribed dose (45 Gy) with IMRT para-aortic–only technique were, with four fields, 11%; with seven fields, 15%; and with nine fields, 13.5% (Fig. 69.13B). These dose distributions were all significantly better than with two-field or four-field conventional techniques (p < .05.) Ahmed et al.329 arrived at similar conclusions in five patients with para-aortic node metastasis, and they demonstrated the feasibility of escalating the dose to 60 Gy while sparing the kidneys, spinal cord, small bowel, and bone marrow. Heron et al.,330 in a study of 10 patients, showed that with IMRT there was a reduction of 52% in the small-bowel volume receiving >30 Gy and a decrease of 66% for the rectum and 36% for the bladder compared with 3D radiation therapy. D’Souza et al.,331 in 10 patients, also noted a reduction of small-bowel volume (33%) with IMRT compared with four-field pelvic RT; however, small volumes of bowel received 55 to 60 Gy with the IMRT plans. Positioning the patient prone on a belly board was shown to reduce the volume of small-bowel irradiated. However, patients on a belly board may have large daily anatomic shifts that make prone IMRT unreproducible.332
Brixey et al.333 and Lujan et al.334 also used IMRT planning to spare the bone marrow of patients with gynecologic tumors. Brixey et al.,333 in 36 patients, noted no significant difference in hematologic toxicity with IMRT or conventional RT alone; however, in patients receiving chemotherapy, less grade 2 white blood cell toxicity was observed with IMRT (31.2% vs. 60%, respectively).
Hasselle et al.335 report on 111 patients treated with multiple different approaches, including 22 treated with postoperative IMRT, 8 with IMRT followed by intracavitary brachytherapy and adjuvant hysterectomy, and 81 with IMRT followed by planned intracavitary brachytherapy. Median follow-up time was 27 months. Acute and late grade 3 toxicity or higher was 2% (95% confidence interval [CI], 0% to 7%) and 7% (95% CI, 2% to 13%), respectively. Guerrero et al.336 proposed using an IMRT simultaneous integrated boost (SIB) as an alternative to conventional whole-pelvis irradiation and used the linear quadratic equation to calculate equivalent uniform dose in multiple plans. However, a report by Kavanagh et al.337 found that accelerated fractionation caused an unacceptably high rate of complications.
Although IMRT has dosimetric advantages over conventional RT, IMRT exposes a greater amount of normal tissues to lower irradiation levels, which has the potential to increase the incidence of radiation-induced second cancers,338 a phenomenon already described with conventional RT techniques.339
FIGURE 69.13. A: Axial views of intensity-modulated radiation therapy dose distribution. B: The functional volume of small bowel, rectum, and bladder receiving ≥45 Gy with intensity-modulated radiation therapy or conventional techniques when 100% of the target volume (uterus) receives ≥95% of the prescription dose (45 Gy). (From Portelance L, Chao KSCC, Grigsby PW, et al. Intensity-modulated radiation therapy [(IMRT)] reduces small bowel and bladder doses in patients with cervical cancer receiving pelvic and para-aortic irradiation. Int J Radiat Oncol Biol Phys 2001;51:261–266; with permission from Elsevier.)

Image-Guided Radiation Therapy
Although not always available, some institutions on protocol have attempted daily cone-beam CT imaging for in-room image-guided RT (IGRT).340 Particularly in cases with a large, mobile uterus, such as is seen in young women, if an extended field is used and there is concern that the uterus may be out of the field, this may be instituted. In clinics where IMRT is used for locally advanced cervical cancer, the priority should be to treat with wide margins, so that the field mimics that of a 3D conformal (four-field) plan.316
Stereotactic body radiotherapy (SBRT) uses highly conformal treatments with large fraction sizes and in selected cases has been considered for a nodal boost of an isolated para-aortic node, although care must be taken to treat the entire para-aortic chain to 45 Gy with IMRT or 4 Field (4F) prior to considering an SBRT boost in order to ensure eradication of adjacent micrometastatic disease.341 SBRT should not be used instead of brachytherapy, given the significant increase in normal-tissue doses with SBRT compared to brachytherapy.342
Midline Shielding in AP-PA Portals and Use of a Parametrial Boost
Depending on the institution and brachytherapy dose administered, midline shielding with rectangular or specially designed blocks has been traditionally used for a portion of the external beam dose delivered with the AP-PA ports.302 Midline blocks may be individualized, based on the point A isodose line or a rectangular block of approximately 4-cm width. In one series, overall survival and incidence of chronic complications were not related to the type of shielding.343 However, in the era of three-dimensional brachytherapy planning, the use of a midline block has been questioned because it may result in tumor underdosing while still contributing significant dose to the bladder, sigmoid, and rectum.344
Several institutions reported placing a midline block after a full course of external-beam treatment to the pelvis in order to boost the parametria or nodes for patients with persistent disease after approximately 45 to 50 Gy. When parametrial tumor persists, 50 to 60 Gy may be delivered to the parametria, with reduced anteroposterior–posteroanterior portals (8 by 12 cm for unilateral and 12 by 12 cm for bilateral parametrial coverage). However, careful estimation of dose to the small bowel, sigmoid, and rectum is needed. In the modern era, the use of highly conformal boosts with 3D planning allows an increase in normal-tissue sparing. Contours on CT of the parametrial and nodal region allow more precise tailoring of dose. For patients with enlarged nodes, when available, IMRT techniques may be best at providing conformal dose escalation to 54 to 65 Gy.
Similarly, with 3D brachytherapy and computerized optimization as available with high HDR or pulse–dose-rate (PDR) brachytherapy, the physician may cover the adjacent parametria using large enough fraction sizes that an additional external-beam boost is not needed. When one prescribes HDR brachytherapy, the per-fraction nodal dose is approximately 25% of prescription. In one study the per-fraction dose to the pelvic lymph nodes by HDR brachytherapy, when the high-risk clinical target volume (HR-CTV) received 5.5 Gy per fraction, was 1.4 Gy per fraction. Therefore, HDR brachytherapy may obviate the need for a parametrial boost, given the high per-fraction dose to the parametria and pelvic sidewall.344
In a comparison of three different approaches, Fenkell et al.345 reported on parametrial boost with midline shielding in six patients with locally advanced cervical cancer (IIB to IIIB) treated with definitive chemoradiotherapy and MRI-guided brachytherapy. A three-phase plan was modeled: 45-Gy (1.8 Gy per fraction) four-field box, 9-Gy (1.8 Gy per fraction) midline-shielded anteroposterior/posteroanterior fields (MBB), and intracavitary MRI-guided brachytherapy boost of 28 Gy (7 Gy per fraction). Midline shields 3, 4, and 5 cm wide were simulated for each patient. Brachytherapy and MBB plans were volumetrically summed. After a 4-cm MBB, HR-CTV D90 remained <85 Gy in all cases (mean, 74 Gy; range, 64 to 82 Gy). Bladder, rectum, or sigmoid D2ccincreased by > 50% of the boost dose in four of six patients. The authors concluded that a midline block may not be beneficial in patients receiving 3D image-planned brachytherapy with adequate optimization of dose to the tumor and away from the normal tissues.
FIGURE 69.14. Extended-field intensity-modulated radiation therapy for external irradiation of uterine cervix and pelvic and para-aortic lymph nodes. Dose–volume histogram values for the spinal cord, kidneys, para-aortic nodes, and pelvic node boost are shown for a patient who had an unresectable 4-cm pelvic lymph node.

FIGURE 69.15. A, B: Example of treatment plan with intensity-modulated radiation therapy for irradiation of para-aortic lymph nodes. C: Dose–volume histogram illustrating sparing of left kidney and small intestine (Plan 7A, solid line; Plan 7B, dashed line). CTV, clinical target volume. (From Mutic S, Malyapa RS, Grigsby PW, et al. PET-guided IMRT for cervical carcinoma with positive para-aortic lymph nodes—a dose-escalation treatment planning study. Int J Radiat Oncol Biol Phys 2003;55:28–35; with permission from Elsevier.)

Para-Aortic Lymph Node Irradiation
If para-aortic node metastases are enlarged or suspected to harbor disease, patients are treated with 45 to 50 Gy to the para-aortic area plus a sequential 5- to 10-Gy boost to enlarged lymph nodes through reduced lateral or rotational portals.275 If feasible, 3D planning with IMRT treatment is preferred to spare normal tissues, superiorly covering above the level of the renal hilum or the highest extent of disease and inferiorly covering 2 cm below the lowest extent of disease (Fig. 69.14).
The use of IMRT has allowed dose escalation to para-aortic nodes, particularly unresectable nodes. Clinical reports show excellent control of disease with dose escalation, with one report demonstrating an 85% 2-year nodal control rate after IMRT with a median dose of 63 Gy.346 Esthappan et al.271 described a technique using CT and FDG-PET to treat the para-aortic lymph nodes (50.4 and 59.4 Gy) with IMRT (Fig. 69.15). Acceptable dose distribution of the target volumes and sparing of the stomach, liver, and colon were achieved. Sparing of the spinal cord was dependent on the number and arrangements of the beams, as it was for the small bowel, sparing of which was limited because of overlap with the target volume. Adjusting the number of beams and prescription parameters minimally improved kidney sparing.
When only conventional techniques are available, the para-aortic lymph nodes are irradiated either with an extended field, often using a four-field approach, that includes both the para-aortic nodes and the pelvis or through a separate portal (Fig. 69.16).302,347 This may be done as one long field, or, in cases requiring extended distance, one may instead choose to separate the pelvic and para-aortic fields. This requires a “gap calculation” between the pelvic and para-aortic portals to avoid overlap and excessive dose to the small intestines. The upper margin of the field is at the T12-L1 interspace and the lower margin at L5-S1. The width of the para-aortic portals (in general, 8 to 10 cm) can be determined by CT scans, MRI, lymphangiography, FDG-PET scans, or IV pyelography outlining the ureters. The spinal cord dose (T12 to L2–3) should be kept to <45 Gy by interposing a 2-cm-wide 5–half-value-layer shield on the posterior portal (usually after 40-Gy tumor dose) or using lateral ports and limiting the kidney dose to <18 Gy. A technique using four isocentric fields weighted 2:1 anteroposterior–posteroanterior over lateral portals and 1.8-Gy fractions was described by Russell et al.348 to deliver high-dose therapy (56 to 61 Gy). Kodaira et al.349 evaluated a four-field para-aortic irradiation technique with 10-MV photons (mean, 50.4 Gy) in 97 patients with cervical cancer. The 5-year cause-specific survival rate was 32.2%. Grade 1 or 2 stomach and duodenum sequelae developed in 26.8%, grade 2 sequelae of small bowel in 3.1%, and grade 2 sequelae of bone in 3.1%. Rates of toxicity with IMRT may be lower.275
FIGURE 69.16. Extended field simulated with conventional plain films for external irradiation of pelvic and para-aortic lymph nodes.

FIGURE 69.17. Example of isodose curves of 4F box irradiation of the pelvis with high-energy photons and high-dose-rate brachytherapy optimized to cover the tumor and minimize dose to the bladder and rectum.

Beam Energies
For IMRT, 6-MV energy is used to provide the most homogeneous dose. However, in conventional irradiation, because of the thickness of the pelvis, high-energy photon beams (10 MV or higher) are especially suited for this treatment. They decrease the dose of radiation delivered to the peripheral normal tissues (particularly bladder and rectum) and provide a more homogeneous dose distribution in the central pelvis. With lower-energy photons (60Co or 4- to 6-MV x-rays), higher maximum doses must be given, and more complicated field arrangements should be used to achieve the same midplane tumor dose (three-field or four-field pelvic box or rotational techniques) while minimizing the dose to the bladder and rectum and to avoid subcutaneous fibrosis (Fig. 69.17).350 Biggs and Russell351 noted that the presence of a metallic prosthesis when using lateral fields or a box pelvic irradiation technique may result in a dose decrease of approximately 2% for 25-MV x-rays and average increases of 2% for 10-MV x-rays and 5% for 60Co. Allt352 and Johns,353 in an update of a randomized study, reported better pelvic tumor control and survival and fewer complications in 65 patients with stages IIB and III cervical carcinoma treated with 23-MV photons compared with 61 treated with external irradiation with 60Co in addition to brachytherapy in both groups. In contrast, Holcomb et al.350 compared outcome of 195 patients with stages IIB and IVA cervical carcinoma treated with 60Co radiation therapy (group 1) and 53 treated with linear accelerators (group 2). There was no significant difference in overall survival, although there was a trend toward increasing pelvic recurrence in the 60Co group (50.8%) compared with group 2 (35.8%; p = .08). Mixed-beam external radiation with neutrons and photons resulted in unacceptably high toxicity rates and is not recommended.354 Similarly, carbon-ion therapy was reported but resulted in major intestinal complications.355
Hyperfractionated or Accelerated Hyperfractionated Radiation Therapy
MacLeod et al.356 reported on a phase II trial of 61 patients with locally advanced cervical cancer treated with accelerated hyperfractionated radiation therapy (1.25 Gy administered twice daily at least 6 hours apart to a total pelvic dose of 57.5 Gy). A boost dose was administered with either low–dose-rate (LDR) brachytherapy or EBRT to a smaller volume. Thirty patients had acute toxicity that required regular medication. One patient died of acute treatment-related toxicity. The overall 5-year survival was 27%, relapse-free survival was 36%, and actuarial local tumor control was 66%. There were eight severe late complications observed in seven patients, who required surgical intervention (actuarial rate of 27%). Five patients also required total hip replacement.
Another study reported on 30 patients with stage II or III cervical cancer randomized to receive either hyperfractionation (15 patients) or conventional fractionation (15 patients).357 At 5 years, 2 patients in the hyperfractionation group and 8 patients in the conventional treatment group had recurrent tumor (p = .04). Delayed bowel complications (grade 2 and 3) occurred in 9 women in the hyperfractionation group and 2 patients in the conventional group (p = 0.0006).
RTOG 88-05 conducted a phase II trial of hyperfractionation (1.2 Gy to the whole pelvis twice daily at 4- to 6-hour intervals, 5 days per week) with brachytherapy in 81 patients with locally advanced carcinoma of the cervix. Total dose to the whole pelvis was 24 to 48 Gy, followed by one or two LDR intracavitary applications to deliver 85 Gy at point A and 65 Gy to the lateral pelvic nodes. Grigsby et al.358 updated the results and noted that external irradiation was completed in 71 cases (88%). The 5-year cumulative rate of grade 3 and 4 late effects for patients with stage IB2 or IIB tumors was 7% and at 8 years was 10%, and with stage III or IVA disease, it was 12% at 5 years. The absolute survival was 48% at 8 years, and disease-free survival was 33%. Comparison with historical control patients treated on other RTOG studies showed equivalent rates of pelvic tumor control, survival, and grade 3 and 4 toxicities at 3, 5, and 8 years, respectively.
Calkins et al.359 assessed the toxicities of multiple–daily-fractionated (twice-daily, 1.2-Gy fractions) whole-pelvis radiation plus concurrent chemotherapy for locally advanced carcinoma of the cervix. In the first study (GOG 8801), for 38 patients, hydroxyurea was given orally (80 mg/kg to a maximum of 6 g) at least 2 hours before irradiation, twice every week. In the second study (GOG 8901), for 30 patients, cisplatin and fluorouracil (5-FU) were used concomitantly with RT. Acute toxicity was primarily enteric and appeared to be dose related. The maximum tolerated dose of whole-pelvis radiation that could be delivered in a hyperfractionated setting with concomitant chemotherapy was 57.6 Gy in 48 fractions, followed by brachytherapy.
Thomas et al.360 conducted a four-arm study in which 234 women with bulky stage IB to IVA cervical cancer were randomized to receive either standard RT (EBRT and brachytherapy to deliver 90 Gy to point A) with or without a 4-day infusion of 5-FU (1 g/m2) on days 1 to 5 and 22 to 25, or partially hyperfractionated RT with or without the same chemotherapy regimen. The partially hyperfractionated regimen delivered two fractions, 6 hours apart, on the first 4 days of treatment, coinciding with the infusion of 5-FU. The addition of 5-FU did not improve pelvic tumor control (37% to 75% at 5 years) or overall survival. However, this study closed without reaching target patient accrual. A concomitant boost technique was reported by Kavanagh et al.337 but had an unacceptably high rate (8 of 20 patients) of late complications.
GENERAL MANAGEMENT
Carcinoma in Situ
Patients with persistent high-grade carcinoma in situ are usually treated with a total abdominal hysterectomy with or without a small portion of the upper vagina removed. The decision to remove the ovaries depends on the age of the patient and status of the ovaries. Occasionally, when the patient wishes to have more children, carcinoma in situ may be treated conservatively with a therapeutic conization,361 laser therapy, or cryotherapy.362 This approach should be judiciously selected when the extent of tumor allows it and the patient is reliable for continued follow-up.32 Conization microscopic margins are critical in decision making regarding a conservative approach or proceeding with a hysterectomy. A therapeutic hysterectomy can be performed 6 weeks after the conization.
Irradiation may be useful for the treatment of carcinoma in situ, particularly in patients with strong medical contraindications to surgery or when there is extension of the lesion to the vaginal wall or multifocal carcinoma in situ in both the cervix and the vagina.302,363 In a group of 26 patients with carcinoma in situ treated at Washington University with intracavitary brachytherapy alone (approximately 5,000 milligram-hours [mgh], 45 Gy to point A with LDR) with tandem and ovoids, no recurrences were recorded.364 Ogino et al.365 used HDR brachytherapy in 14 patients with grade 3 cervical and 6 with grade 3 vaginal intraepithelial neoplasia (3 with microinvasion) and 6 with recurrent cervical intraepithelial neoplasia after hysterectomy. Seventeen patients were treated with HDR brachytherapy alone and 3 in combination with EBRT without surgery. The mean dose of HDR brachytherapy was 26.1 Gy (range, 20 to 30 Gy) prescribed at point A for intact uterus, or at 1 cm superior to the vaginal apex or 1 cm beyond vaginal mucosa for lesions of the vaginal stump. At mean follow up of 90 months, 14 patients were alive and 6 had died from intercurrent disease; none had recurrent disease. Rectal bleeding occurred in 3 patients and subsided spontaneously. Moderate and severe vaginal reactions were noted in 2 patients in whom the treatment included the entire vagina.
Invasive Disease
Based on available resources globally and the stage of disease, a debate continues among those who advocate radical surgery,302,366,367 those who favor radiation, and those who favor chemoRT for the treatment of carcinoma of the uterine cervix. Patients should be treated with close collaboration between the gynecologic oncologist and the radiation oncologist, and an integrated team approach should be vigorously pursued. In countries with access to RT facilities and financial resources to supply chemotherapy, the use of concurrent chemoRT represents the accepted standard for patients with stage IB2 to IVA cervical cancer. For earlier-stage patients, the use of either surgery or chemoRT is recommended. The most recent survey of patterns of radiotherapy practice again documented a rise in the use of cisplatin-based concurrent chemoRT in patients with advanced stages, from 63% in 1999 to 74% in 2007.154,368,369 Moreover, Barbera et al.370 also reported a significant increase in the use of chemoRT in Canada after the U.S. National Cancer Institute Bulletin on the subject.371
Carcinoma of the Cervix Inadvertently Treated with a Simple Hysterectomy
Occasionally, a simple or total abdominal hysterectomy is performed and invasive carcinoma of the cervix is incidentally found in the surgical specimen. In general, extrafascial abdominal hysterectomy is not curative because the paravaginal or paracervical soft tissues and vaginal cuff are not removed. Furthermore, it may be technically difficult to perform an adequate radical operation after previous simple hysterectomy. If only microinvasive carcinoma is found when a total or extrafascial hysterectomy with a wide cuff is performed, no additional therapy is necessary. For lesions with deeper stromal invasion, at most one or two vaginal intracavitary insertion(s) to deliver a 65-Gy LDR mucosal dose (or 7 Gy é six fractions prescribed at the vaginal surface, or 5 Gy at 0.5 cm é six fractions with HDR brachytherapy) to the vault are sufficient. If a less comprehensive resection was carried out, it is critical that these patients receive radiation therapy immediately with or without concurrent chemotherapy, depending on the risk factors present pathologically when their postoperative status allows it because the prognosis is worse if postoperative irradiation is not administered.
In patients with fully invasive tumor, therapy consists of approximately 40 to 45 Gy to the whole pelvis with cylinder brachytherapy to the vaginal vault for an approximately 60-Gy mucosal dose. If there is gross tumor present in the vaginal vault or parametrium, the dose to the whole pelvis should be 45 Gy with concurrent weekly cisplatin chemotherapy, followed by an additional parametrial dose of 10 to 20 Gy. An intracavitary insertion should be performed. If there is gross residual tumor, an interstitial implant should be carried out to selectively increase the dose to this volume.
Several studies have reported results of postoperative external-beam irradiation after conservative surgery (Table 69.10). Andras et al.372 reported on 148 patients, 90 of whom were available for 10-year evaluation, who were divided into five groups, depending on tumor extent when therapy was instituted. The majority of patients were treated with 50-Gy total-pelvis irradiation (with 10-Gy parametrial boost through reduced fields), at times combined with vaginal vault intracavitary irradiation. Eight major complications were noted in 148 patients.
Ampil et al.373 described results in 44 patients receiving postoperative irradiation after hysterectomy for stage IB and IIA carcinoma of the uterine cervix (15 patients treated with radical hysterectomy). Their 5-year results were 80% local tumor control and 63% overall survival. In 3 patients treated with intracavitary vaginal cuff irradiation only, 2 had tumor control.
Green and Morse374 reported 9 of 30 patients (30%) surviving 5 years after definitive radiation therapy for treatment of invasive cervical carcinoma after simple hysterectomy. The same authors noted that 14 of 32 patients retreated with another surgical procedure, usually a Wertheim hysterectomy, died within 5 years. They pointed out that the 5-year cure rate was 30% in patients treated within 1 year after the hysterectomy but was only 16% in those treated after 1 year. Thus, the time at which the patient is treated and the volume of tumor are important prognostic factors.
Crane and Schneider375 described results in 18 patients treated with RT (with or without brachytherapy) for invasive carcinoma of the cervix discovered after simple hysterectomy. The 10-year actuarial local tumor control was 88%, and the overall survival rate was 93%. Huerta Bahena et al.,376 in 59 patients with carcinoma of the cervix incidentally found in simple hysterectomy specimens (27 with gross residual tumor) who were treated with postoperative RT, reported a 3-year survival of 59%; factors affecting prognosis included gross residual tumor, time between hysterectomy and irradiation >6 months, RT doses < 50 Gy, and histologic tumor type.
Munstedt et al.377 reported on 119 patients who received postoperative RT after radical hysterectomy and 80 who received it after simple hysterectomy. There was a trend toward better survival in the radical hysterectomy group, but the authors concluded that postoperative RT is a good treatment in patients with invasive cervical cancer who undergo a simple hysterectomy. In another report of 105 patients with invasive cervical carcinoma found in inadequate surgery specimens treated with postoperative RT, 5-year pelvic tumor control was 72% and the survival rate was 55%. Late rectal toxicity was 19%, bladder toxicity was 4.8%, and small-bowel toxicity was 14.3%.378
In a series of 147 patients treated at Asan Medical Center in Korea, 48 patients with stage IA1 lesions did not receive further treatment. Another 99 patients had stage IA2 to IIA lesions incidentally identified. Of these, 26 received no further therapy, 44 had either radiation or chemoradiation, and 29 had a radical parametrectomy. For patients with stage IA1 disease who were observed, 0% relapsed, whereas 35% of patients with stage IA2 to IIA disease who were observed suffered from a recurrence.379 Either radical parametrectomy or radiation is required for patients with stage IA2 or higher cancer after a simple hysterectomy. However, these treatments increase the risk of side effects,380 including for those patients receiving robotic parametrectomy.381
TABLE 69.10 RESULTS OF POSTOPERATIVE EXTERNAL-BEAM IRRADIATION AFTER CONSERVATIVE HYSTERECTOMY IN EARLY-STAGE CARCINOMA OF THE CERVIXA

Stage IA
The definition of microinvasive (stage IA) carcinoma of the cervix includes invasive carcinoma diagnosed only by microscopy. Conization is mandatory for a more accurate diagnosis. According to Kolstad,382 lesions <1 mm in depth can be treated with conization, provided all margins are tumor free and continued careful follow-up is instituted. Raspagliesi et al.383 used margins of 8 to 10 mm as guidelines for clearance in conization. Smaller margins or lymphovascular invasion in addition to depth of invasion were prognostic factors for recurrence.
Tumor volume in the stroma may be a more reliable criterion than depth of invasion to arrive at a definition of stage IA. Vascular space involvement does not impact stage. Depth of invasion and tumor confluence have been identified as prognostic factors that should be taken into consideration in the planning of therapy.384
Early invasive carcinoma of the cervix (stage IA2) is usually treated with a total abdominal or modified radical hysterectomy or in some cases with simple conization251 or radical trachelectomy.385 Inoperable patients may be treated with intracavitary radioactive sources alone with 6,500 to 8,000 mgh, 60 to 75 Gy to point A, in two LDR insertions, respectively, or with the equivalent dose using HDR brachytherapy, approximately 10 fractions of 5 Gy per fraction. In 47 patients with microinvasive carcinoma treated at Washington University—20 with intracavitary therapy only and 27 patients with combined external irradiation and intracavitary brachytherapy—only 1 patient had a pelvic recurrence and distant metastases 10 years later; the 5-year disease-free survival rate was 96%.366
When the depth of penetration of the stroma by tumor is <3 mm, the incidence of lymph node metastasis is 1% or less, and a lymph node dissection or pelvic external irradiation is not warranted.302,366 With more extensive lesions, a Wertheim radical hysterectomy with pelvic lymphadenectomy is the preferred treatment. Tumor control with all treatment methods is >95%, with patients eventually dying of intercurrent disease. Gadducci et al.386 treated 30 patients with conization and 82 with total and 54 with radical hysterectomy; the recurrence rates were 10%, 4.9%, and 9.3%, respectively. None of 67 patients subjected to lymphadenectomy had positive pelvic nodes. In 98 patients with adenocarcinoma of the cervix, none of 48 with depth of invasion (DOI) of ≤5 mm had involved parametria or positive nodes, in contrast to 6 of 36 (16%) with DOI of >5 mm.387
Vaginal trachelectomy (removal of the cervix) and laparoscopic lymphadenectomy have been used to treat young patients with microinvasive carcinoma to preserve fertility. The overall incidence of central recurrence is approximately 5%.364 Webb et al.388 analyzed lymph node status and survival rates of women with microinvasive cervical adenocarcinoma (FIGO stages IA1 and IA2) from the Surveillance, Epidemiology, and End Results (SEER) database between 1988 and 1997. Among reported cases, 131 had stage IA1 and 170 had stage IA2 disease. Simple hysterectomy was done in 54 women with IA1 and in 64 women with IA2 disease and radical hysterectomy in 50 and 83 women, respectively. Only 1 of 140 women who had lymphadenectomy had a single positive lymph node. There were 4 tumor-related deaths (1 with IA1 and 3 with IA2 disease). The survival rate was 98.7%.
Stages IB to IIA
The choice of definitive irradiation or radical surgery for stage IB and IIA carcinoma of the cervix remains controversial, and the preference for one procedure over another depends primarily on the impact on the patient’s fertility and on the institution, the gynecologic oncologist or radiation oncologist involved, the general condition of the patient, and characteristics of the lesion. An operation has been preferred by some in young women to preserve the ovaries, attempting to prevent premature menopause. However, in some reports294 ovarian function preservation has been observed in only 50% to 60% of surgically treated patients not receiving irradiation. Postmenopausal patients may have a survival benefit with chemoradiation and avoid the operative risks. When therapeutic results in invasive carcinoma of the cervix are evaluated, a direct comparison of surgically treated or irradiated patients is fraught with many uncertainties, including patient selection, reporting of surgical cases using staging determined by laparotomy findings, and different treatment techniques.389 In particular, in the modern era when concurrent chemoradiation is known to be superior to radiation alone, a direct comparison of chemoradiation versus surgery in early-stage cervical cancer is needed.
Surgery provides an opportunity for a thorough pelvic and abdominal evaluation. However, surgical staging has not been shown to improve overall patient survival.279,390 Kupets et al.391 assessed the value of debulking large nodes and concluded that the incremental overall benefit by stage was small. Delgado et al.161 described a GOG study in which 1,125 patients were registered before surgery; 80 were ineligible after strict pathology review, and an additional 129 patients were explored, but the hysterectomy was abandoned because of intraoperative complications in 49 patients or extent of disease beyond the uterus in 80 patients. In the era of MRI, surgeons may rely on imaging findings to screen for operability. The impact of patient selection in results of surgical series was illustrated by Whitney and Stehman,392 who evaluated the frequency with which intended radical hysterectomy for cervical cancer is abandoned and the outcomes for those selected patients. In 1,127 patients with stage IB carcinoma of the cervix entered on GOG Protocol 49, 98 women (8.7%) were found at surgery to have extrauterine disease, and the proposed radical operation was abandoned. Subgroups of patients with extrapelvic disease31 and pelvic extension,387 including grossly positive pelvic nodes,160 other pelvic implants,393 and gross serosal extension,109 were identified. Sixty-three (93%) patients subsequently underwent pelvic radiation therapy and brachytherapy. Para-aortic fields were added for 8 patients who were found to have positive para-aortic nodes. The disease-free survival was shorter for patients whose radical procedure was abandoned than for those patients who underwent radical hysterectomy.
The important contribution of external-beam irradiation to improve pelvic tumor control in larger lesions has been documented. Hamberger et al.,394 in 151 patients with stage IA or IB lesions <1 cm in diameter treated with intracavitary therapy alone to high doses (8,640, 9,340, and 13,680 mgh), noted no failures in 41 patients with stage IA disease, and only 4 of 93 patients (4%) with stage IB, small-volume disease. However, 3 of 17 patients (18%) with more extensive stage IB lesions, treated with intracavitary therapy only, had regional failures. Only 3 of 151 patients (0.2%) had grade 3 complications.
Volterrani and Lombardi395 reported 5-year survival of 82.6% in 23 patients with occult stage IB carcinoma of the cervix treated with intracavitary 226Ra only (7,500 mgh), in contrast to only 64.8% with larger stage IB tumors and 50% with stage II. Unfortunately, the authors did not report the exact location of the failures. It is obvious that intracavitary therapy alone is grossly inadequate to irradiate larger primary tumors, including stage IB1.
With EBRT and brachytherapy without chemotherapy, the usual 5-year survival rate for stage IB is 86% to 92% and for stage IIA is approximately 75%.396 Late toxicity was observed in 1% to 2% of patients. Concurrent chemotherapy significantly improves survival, including in stage IB to IIA disease. In RTOG 90-01, for the subset of 272 stage IB to IIA patients, the 8-year overall survival was 55% with RT alone versus 78% with concurrent chemoradiation (p < .001).397
Randomized Studies: Surgery Versus Radiation
Few randomized trials have compared the results of radical hysterectomy with definitive RT, and none have compared surgery to chemoradiation. Outcome between radiation alone versus surgery is comparable. Newton398 and Roddick and Greenlaw399 reported, in prospectively randomized studies, equivalent survival and pelvic recurrence rates in patients with stage IB and IIA carcinoma of the uterine cervix treated with a radical hysterectomy or irradiation alone. Landoni et al.35 published results of a prospective, randomized trial of radiation therapy versus surgery; 469 women with stage IB and IIA cervical carcinoma were referred for treatment and 343 were randomized (172 to surgery and 171 to radiation therapy). Postoperative irradiation was delivered after surgery for women with surgical stage pT2b or greater, <3 mm of cervical stromal invasion and cut-through margins or positive pelvic nodes. Scheduled treatment was delivered to 169 and 158 women, respectively; 62 of 114 women with cervical diameters of <4 cm and 46 of 55 women with >4 cm received radiation therapy. After a median follow-up of 87 months (range, 57 to 120 months), 5-year overall and disease-free survival rates were nearly identical in the surgery and radiation therapy groups (83% and 74%, respectively); recurrent disease developed in 86 women: 42 (25%) in the surgery group and 44 (26%) in the radiation therapy group (Fig. 69.18). Forty-eight patients (28%) in the surgery group had severe morbidity, compared with 19 (12%) in the radiation therapy group (p = .0004; Table 69.11). The combination of surgery and radiation therapy had the worst morbidity, especially urologic complications.
Of note, no randomized study has compared chemoRT to radical hysterectomy, although chemoRT has a significant survival advantage over RT alone for patients with stage IB to IIA cervical cancer.400 In a meta-analysis looking at the value of adjuvant cisplatin-based chemotherapy after radical hysterectomy, radiation therapy, or both for patients with stage IA2, IB1, or IIA cervical cancer, three randomized clinical trials were evaluated.400 Two of the three trials showed a significant benefit compared to adjuvant chemotherapy concurrent with radiation, with a reduced risk of death (HR = 0.56, 95% CI = 0.36 to 0.87). No benefit was seen when chemotherapy was given prior to radiotherapy.
Neoadjuvant chemotherapy plus surgery does not improve survival over surgery alone.401 In a meta-analysis of six trials, although there was an improvement in progression-free survival with neoadjuvant chemotherapy (HR = 0.76, 95% CI = 0.62 to 0.94, p = .01), this did not translate into an overall survival benefit. Another meta-analysis reviewed 18 trials with locally advanced cervical-cancer patients treated with neoadjuvant chemotherapy before radiation or surgery or both and excluded concurrent chemoradiation trials; it showed significant heterogeneity and no conclusive results.402
FIGURE 69.18. Overall actuarial survival of patients with carcinoma of the cervix randomized to treatment with radical surgery or radiation therapy according to treatment group and cervical diameter. (From Landoni F, Maneo A, Colombo A, et al. Randomised study of radical surgery versus radiotherapy for stage IB-IIA cervical cancer. Lancet1997;350:535–540; reprinted with permission from Elsevier.)

TABLE 69.11 RANDOMIZED TRIAL OF RADICAL SURGERY OR IRRADIATION IN STAGES I TO II CERVICAL CANCER: RELAPSES AND MORBIDITY

Nonrandomized Studies Comparing Surgery to Radiation
Keilbinska et al.,403 in a long-term study of 792 women treated with irradiation and 789 women treated with hysterectomy and/or irradiation for stage I cervical carcinoma, found no difference in survival, general health, incidence of recurrent carcinoma, or appearance of second primary malignancies. Piver et al.404 treated 103 women with stage IB cervical carcinoma with either radical hysterectomy and pelvic lymphadenectomy (if tumor was <3 cm in greatest diameter) or irradiation (tumor of >3 cm or medically inoperable). The 5-year disease-free survival rate was 92.3% for the surgical group and 91.1% for the radiation therapy group. Equivalent overall 5-year survival rates were noted. Einhorn et al.,405 in a nonrandomized study, observed a 100% 5-year survival rate in 49 patients with stage IB disease receiving combined therapy in comparison with 81% in 64 patients treated with irradiation alone. No difference was observed in 25 patients with stage IIA tumor treated with combined therapy and 40 patients treated with irradiation alone (5-year survival rate, 75%).
Perez et al.406 reported on a prospectively randomized study of 118 patients with stage IB or IIA carcinoma of the uterine cervix in which patients were treated with RT alone or irradiation and surgery (20 Gy to the whole pelvis, one intracavitary insertion for 5,000 to 6,000 mgh, followed by a radical hysterectomy with pelvic lymphadenectomy 2 to 6 weeks later). In stage IB, the 5-year tumor-free survival was 80% and 82% (p = .23), respectively, and in stage IIA it was 56% and 79%, respectively (p = .13). The incidence of grade 2 or 3 complications for radiation alone was 13.8% and with preoperative irradiation and surgery was 11%. Subsequently, Perez et al.407 described results in 415 patients with stage IB or limited stage IIB treated with preoperative or postoperative irradiation and surgery. The 10-year cause-specific survival rate for patients with stage IB nonbulky tumors treated with irradiation alone or irradiation combined with surgery was 84% with either modality. With bulky tumors (>5 cm), the 10-year rates were 61% and 68%, respectively (p = .5). For patients with stage IIA nonbulky tumors, the 10-year cause-specific survival rates were 66% and 71%, respectively, and with bulky tumors, 69% and 44%, respectively (p = .05). In patients with stage IIB nonbulky tumors treated with irradiation alone or combined with surgery, the 10-year cause-specific survival rates were 72% and 65%, respectively. In stage IB and IIA disease after a hysterectomy and lymphadenectomy (even combined with irradiation), patients with metastatic lymph nodes have survival rates that are approximately 50% of those of patients with negative nodes.408
Randomized Trials of Postoperative Radiation Therapy or Chemoradiation After Radical Hysterectomy
Patients who have undergone radical hysterectomy with no preoperative radiation therapy are considered for postoperative chemoradiation therapy if they have high-risk prognostic factors, which include positive pelvic lymph nodes, as are patients with negative nodes who have microscopic positive margins of resection or parametrial involvement.409
Patients with any two of deep stromal invasion, vascular/lymphatic permeation, and large tumor size are candidates for postoperative radiation.410 These patients have an intermediate risk of failure.161 Whether to add concurrent chemotherapy to postoperative radiation in the intermediate risk group is being tested in an accruing randomized trial; many institutions routinely implement chemoRT for intermediate-risk patients. Song et al.411 reported a 20-year experience in stage IB to IIA cervical-cancer patients with intermediate-risk factors (two or more of deep stromal invasion, lymphovascular invasion, and large tumor size) received postoperative RT or chemoradiation. Chemoradiation significantly decreased pelvic recurrence and distant metastases; there was no difference in acute or chronic grade 3 and 4 gastrointestinal side effects.
One randomized study showed improved recurrence-free survival with postoperative pelvic irradiation (46 to 50.4 Gy in 23 to 28 fractions) after radical surgery in the presence of positive pelvic nodes or node-negative high-risk factors in women with stage IB cervical cancer treated by radical hysterectomy and pelvic lymphadenectomy. There were 277 eligible patients with at least two of the following risk factors: greater than one-third stromal invasion, capillary lymphatic space involvement, and large clinical tumor diameter; 137 patients were randomized to pelvic radiation therapy and 140 to no further treatment. The results were updated by Rotman et al.;412 24 (17%) patients in the irradiation group and 43 (30.7%) in the no-further-treatment group had cancer recurrences. In the radiation therapy group 27 patients died of cancer, and in the no-further-treatment group 40 died from cancer. There was a statistically significant reduction in risk of recurrence in the irradiation group, with recurrence-free rates at 2 years of 88% versus 79% for the irradiation and no-further-treatment groups, respectively. Overall survival difference did not reach statistical difference (p = .074; Fig. 69.19). Severe or life-threatening (GOG grade 3 or 4) adverse effects occurred in 9 patients (6.6%) in the radiation therapy group and 3 (2.1%) in the observation group. A meta-analysis of trials including stage IB1 to IIA cervical cancer found that women who received postoperative radiation had a significantly lower risk of disease progression at 5 years (RR = 0.6, 95% CI = 0.4 to 0.9). The risk of serious adverse events was not significantly higher if women received radiotherapy rather than no further treatment, possibly because the rate of adverse events was low.413
Sundfør et al.414 conducted a randomized study in which 122 patients with stage IIA and 20 patients with stage IIB cervix cancer were treated with intracavitary radium followed by either radical pelvic surgery including lymphadenectomy (group A, 72 patients) or EBRT (40 Gy) to the pelvis (group B, 70 patients). Postoperative RT (40 to 50 Gy) was given to patients in group A found to have node metastasis at operation. Fourteen patients in group A and 23 in group B died of recurrent cancer. The 10-year survival was 84% and 69%, respectively.
FIGURE 69.19. Recurrence-free survival (A) and overall survival (B) of patients with stage IB carcinoma of the cervix, correlated with treatment method. (From Rotman M, Sedlis A, Piedmonte MR. A phase III randomized trial of postoperative pelvic irradiation in patients with stage IB cervical carcinoma with poor prognostic features: follow up of a Gynecologic Oncology Group study. Int J Radiat Oncol Biol Phys 2006;65:169–176; with permission from Elsevier.)

Nonrandomized Trials of Postoperative Radiation Therapy or Chemoradiation After Radical Hysterectomy
Snijders-Keilholz et al.415 described results in 233 women who underwent radical hysterectomy for stage I or IIA cervical carcinoma; 156 were treated with surgery alone, and 77 received adjuvant radiation therapy for tumor-related high-risk prognostic factors. The most important prognostic factor for survival and disease-free survival was pelvic lymph node positivity; additional factors were depth of invasion and positive surgical margins. Twelve patients recurred after surgery alone, all in the pelvis (100%). Of the 23 recurrences after surgery and adjuvant radiation therapy, 13 were in the pelvis (56%; p = .003). Ten patients with poor prognostic factors and negative nodes received adjuvant radiation therapy, and none of these patients recurred. The incidence of severe gastrointestinal radiation-related side effects was 2%. The incidence of lymphedema of the leg was 11%, which was similar to that in the surgery-alone group.
Garipagaoglu et al.416 investigated prognostic factors in 100 patients with stage IB or IIA cervical carcinoma treated with radical hysterectomy and postoperative irradiation. The 5-year overall survival, disease-free survival, and pelvic tumor control rates were 83.6%, 82.8%, and 91.8%, respectively. Pelvic lymph node metastasis (p = .008), interval between surgery and irradiation (p = .001), overall radiation therapy time (p = .007), and tumor size (p = .028) were significant factors for pelvic tumor control, as well as for overall survival.
Lahousen et al.417 reported on a GOG prospectively randomized, multicenter trial in which patients with stage IB or IIB cervical cancer treated with radical hysterectomy who had pelvic lymph node metastases or vascular invasion randomly received adjuvant chemotherapy (400 mg/m2 carboplatin and 30 mg bleomycin), external pelvic radiation therapy, or no further treatment. After a median follow-up of 4.1 years (range, 2 to 7 years), there were no statistically significant differences (p = .9539) in disease-free survival rates among the three treatment arms, suggesting that adjuvant chemotherapy or radiation does not improve survival or recurrence rates in high-risk patients with cervical cancer after radical hysterectomy.
González González et al.418 reported that in 89 patients with stage IB or IIA cervical cancer with positive lymph nodes receiving postoperative irradiation, the 5- and 10-year survival rates were 60% and 51%, respectively. By comparison, 43 patients with negative lymph nodes had a survival rate of 85%. In the surviving patients, there were 4 gastrointestinal and 7 genitourinary severe complications requiring surgical correction. In 4 patients, asymptomatic stenosis of the ureters was detected by IV pyelography performed routinely every year.
Bianchi et al.,419 in 60 patients receiving external irradiation for pelvic node metastasis after radical hysterectomy, observed a 65% 5-year survival rate. In contrast, in 15 patients who refused postoperative irradiation, only 3 (20%) survived 5 years.
Chatani et al.420 reported on 128 patients with stage IB to IIB carcinoma of the cervix who underwent radical hysterectomy with bilateral pelvic lymphadenectomy and postoperative EBRT. The 5-year local and distant failure rates were, respectively, 2% and 12% for negative nodes, 23% and 25% for one positive node, and 32% and 57%, for two or more positive nodes (p = .0029 and .0051, respectively). The 5-year cause-specific survival rates were 90%, 59%, and 42%, respectively (p = .0001). The most common complication was lymphedema of the lower extremity, experienced by half of the patients (42% at 5 years and 49% at 10 years).
Uno et al.421 evaluated results of postradical hysterectomy irradiation in 98 patients with stage IB to IIB cervical cancer; all of the patients had at least one pathologic risk factor for pelvic recurrence. The 5-year overall survival was 82%. There were pelvic recurrences in 5 cases and distant metastases in 15 cases. The 5-year overall survival rates for patients with or without pelvic lymph node metastasis were 76% and 89%, respectively (p = .018).
Kinney et al.422 compared results of therapy in 82 patients with stage IB or IIA carcinoma of the cervix found to have pelvic lymph node metastases at Wertheim hysterectomy and bilateral lymphadenectomy without additional adjuvant therapy with 103 similar patients who received 50 Gy to the pelvis after surgery. The 5-year survival rate was 72% for the surgery-only patients and 64% for the group receiving adjuvant irradiation. The incidences of pelvic recurrence were 67% and 27%, respectively. The lack of impact on overall survival in the irradiated patients is most likely related to a higher incidence of distant metastases, which may be a reflection of shorter survival time and more high-risk patients. In 117 patients treated with radical hysterectomy and pelvic lymphadenectomy, histologically proven nodal metastatic disease was detected in 51 patients (44%; squamous cell in 35 and nonsquamous in 16). Nodal involvement was bilateral in 24 patients (47%).423 Para-aortic lymph node dissection was performed in 14 patients, and 5 had tumor involvement. Postoperative pelvic irradiation was administered to 29 of 51 patients (51.2 Gy, two fractions). Extended fields to the para-aortic area were used in 6 patients. The 5-year survival rates were 33% for the group receiving irradiation and 50% for the nonirradiated group. Only 1 patient treated with postoperative irradiation had a pelvic failure, in contrast to 7 patients not irradiated.
Inoue and Morita270 described results in 72 patients treated with extended-field irradiation after radical surgery for nodal metastases with stages IB (37 patients), IIA (6 patients), and IIB (29 patients) cervical cancer. The median dose to para-aortic lymph nodes was 43.5 Gy and to the pelvis was 45 Gy. The 5-year disease-free survival rates were 72% in 61 patients with squamous cell carcinoma and 27% in 11 patients with non–squamous cell carcinoma. The 10-year disease-free survival rates were 88% for 22 patients with one positive node, 67% for 15 patients with two or three positive nodes, 64% for 16 patients with four to 17 positive nodes, and 20% for 10 patients with unresectable lymph nodes. Nineteen severe complications occurred in 17 patients; 5 were attributed to surgery, 5 to irradiation, and 9 to both modalities. Four patients (5%) died of severe complications. Another 6 patients (8%) underwent major abdominal surgery for rectovaginal and ureterovaginal fistulas.
Mitsuhashi et al.424 described an analysis of 108 patients with carcinoma of the cervix treated with postoperative EBRT to the pelvis followed by intravaginal cone boost with electron beam to the vaginal cuff. The 5-year cause-specific survival rates were 89% for 89 patients undergoing elective radiation therapy and 56% for 19 patients undergoing salvage irradiation (p < .001). Recurrent tumors at the vaginal cuff were observed in only 2 patients in the elective irradiation group. Vesicovaginal fistula developed in 4 patients; only 1 patient had grade 2 rectal complications.
A Japanese group treated 189 stage IIB cervical cancer patients; 95 had a radical hysterectomy followed by adjuvant RT, and the other 94 patients had RT alone.425 There was a significant increase in grade 3 to 4 late toxicities in the surgery group, 24% versus 10%, p = .048. Therefore, RT is preferable, and in the modern era, chemoRT provides a superior outcome and should be administered to all stage IIB patients. The same group reported on 55 stage IA2 to IIB patients with multiple pelvic lymph nodes positive treated either with pelvic RT and concurrent chemotherapy or extended-field RT. Overall survival significantly improved with concurrent chemotherapy (p = .03).426
Lee et al.427 retrospectively compared 201 stage IB1 to IIB cervical cancer patients who had a radical hysterectomy with pelvic lymph node dissection followed by adjuvant concurrent weekly cisplatin to triweekly combination chemotherapy. With a median follow-up of 52 months, the weekly cisplatin group had the same therapeutic effect with less toxicity. The 5-year disease-free and overall survival were 82% and 81%, respectively, for patients treated with weekly cisplatin chemotherapy versus 74% and 79% for those treated with triweekly combination chemotherapy (p = NS). Leukopenia, neutropenia, thrombocytopenia, anemia, and hepatopathy were significantly more common in the triweekly combination chemotherapy group.
Postoperative External-Beam Radiation Dose
When metastatic pelvic lymph nodes are present, treatment has consisted of 45 Gy to the whole pelvis delivered with a four-field technique with concurrent weekly cisplatin. If gross residual disease is present, dose escalation to 54 to 65 Gy, depending on small-bowel dose limits (for example, D5cc < 55 Gy), may be considered with a sequential IMRT nodal boost.80 Patients with positive common iliac or para-aortic node metastases should receive 45 Gy to the entire para-aortic region with the superior border covering the renal hilum, with consideration of a boost to the tumor bed. If gross residual nodal disease is left, a nodal boost up to 65 Gy with IMRT is particularly suited to treat these patients.271,275
In patients for whom postoperative irradiation is indicated for deep stromal invasion in the cervix or close or positive surgical margins, an alternative is to deliver 45-Gy pelvic external irradiation in combination with an intracavitary insertion (LDR, PDR, or HDR) and LDR equivalent dose of 65 Gy to the vaginal mucosa, using colpostats or a cylinder.428 At some institutions, external irradiation alone (50 Gy to the midplane of the pelvis) with a four-field box technique has been used. Hong et al.176 recommended, for node-negative patients with high-risk factors, to irradiate only the low pelvis (median dose, 50 Gy), which resulted in a reduction of grade 3 small bowel morbidity (3 of 149 = 2%) in comparison with patients treated to the whole pelvis (6 of 79 = 8%). Five-year disease-specific survival was 84% and 86%, respectively.
Postoperative Intracavitary High–Dose-Rate Brachytherapy
Depending on the extent of surgical resection of the vagina, the vaginal cuff may be at risk for recurrence. There is no clear agreement on the indications for vaginal brachytherapy after radical hysterectomy, although adjuvant vaginal brachytherapy for cervical cancer is most commonly used as a boost after EBRT. Based on the American Brachytherapy Society guidelines, vaginal cuff boost should be considered in patients with a less-than-radical hysterectomy, close or positive margins, large or deeply invasive tumors, parametrial or vaginal involvement, or extensive lymphovascular invasion.428 Consideration of postoperative vaginal intracavitary brachytherapy after external-beam therapy is recommended for patients with carcinoma at the vaginal margin of resection.302 If parametrial margins were close or positive, defined by either surgical clips or in the region of the surgical tumor bed, an external-beam dose of at least 54 Gy for close margins and higher for positive margins is recommended.
In patients receiving postoperative irradiation, extreme care should be exercised in designing treatment techniques, including intracavitary insertions; because of the surgical extirpation of the uterus, the bladder and rectosigmoid may be closer to the radioactive sources than in the patient with an intact uterus. Furthermore, vascular supply may be affected by the surgical procedure, and adhesions can prevent mobilization of the small-bowel loops that may be fixed in the pelvis. HDR brachytherapy after surgery is particularly suited for patients with cervical cancer because it prevents the prolonged immobilization required for LDR brachytherapy. In some patients at higher risk for parametrial tumor or lymph node metastases, HDR brachytherapy is combined with external-beam pelvic irradiation.
Hart et al.429 described results in 83 patients who received postoperative RT for early-stage cervical cancer with positive surgical margins, positive pelvic or para-aortic lymph nodes, lymphovascular space invasion, or deep stromal invasion or for disease discovered incidentally at simple hysterectomy. Twenty-eight patients were treated with LDR brachytherapy with or without EBRT and 55 with EBRT to the pelvis and HDR intracavitary. Of these 83 patients, 66 were evaluable (20 LDR and 46 HDR patients). Mean follow-up time was 101 months for the LDR group and 42 for the HDR group. The 5-year disease-free survival rates were 89% and 72%, and local tumor control rates were 90% (18 of 20) and 89% (41 of 46), respectively. Three of 20 patients (15%) receiving LDR and 4 of 46 (9%) receiving HDR experienced grade 2 or 3 late treatment-related complications. No patient in either group had grade 4 or 5 complications.
Busch et al.430 studied the outcome of 68 patients with cervical carcinoma; 48 were treated with radical hysterectomy and, because of risk factors, with postoperative RT (group 1), and 20 patients (group 2) were pretreated with standard hysterectomy and then admitted to the hospital for postoperative radiation therapy of the whole pelvis. Postoperative pelvic RT consisted of 39.6 Gy (box technique) and 6-Gy external-beam therapy to the pelvic lymph nodes, sparing the midline, plus two HDR applications (7.5 Gy each). Survival, locoregional tumor control, and metastatic disease rates were nearly identical in both groups. Patients with positive lymph nodes had a worse prognosis (75% 3-year survival rate).
Atkovar et al.431 described results in 126 patients treated with postoperative irradiation (median of 50 Gy in 5 weeks); 37 received vaginal cuff HDR brachytherapy (three fractions of 8 to 10 Gy at 5 mm, weekly). Overall and disease-free survival and locoregional tumor control rates were 71%, 69.9%, and 78.1%, respectively. Grade 2 and 3 complications developed in 5.5% of patients. Survival was the same in 67 patients treated with total abdominal hysterectomy and bilateral salpingo-oophorectomy and in 59 patients treated with radical hysterectomy and pelvic lymphadenectomy.
Stages IB2 to IVA: Chemoradiation
Patients with stages IB2 to IVA tumors are treated with irradiation including external beam and brachytherapy combined with concurrent chemotherapy. Numerous reports have been published on the concomitant use of irradiation and cytotoxic agents (hydroxyurea, cisplatin, and 5-FU, in some trials combined with mitomycin C) administered to obtain a radiosensitizing effect.432–434 Cisplatin is one of the most active cytotoxic agents in squamous cell carcinoma of the uterine cervix.435 When cisplatin and irradiation are used concomitantly, substantial enhancement of cell killing is observed. Coughlin and Richmond433 and Douple436 suggested two mechanisms for radiation enhancement by cisplatin: (a) in hypoxic or oxygenated cells, free radicals with altered binding of cisplatin to DNA are formed at the time of irradiation, and (b) interaction inhibits repair of sublethal damage.
It is important, however, that patients complete the full course of 45 Gy with, ideally, five to six weekly doses of cisplatin or two doses of cisplatin and 5-FU every 3 weeks. In a study of 41 patients who had weekly biopsies while receiving RT and chemotherapy for cervix cancer, increased tumor cell proliferation and accelerated repopulation was observed within 2 weeks from the initiation of therapy. Patients with a sustained yield and high S-phase fraction for 2 or more weeks were at increased risk for tumor progression.437
Green et al.,438 in a search of medical databases for randomized trials of cervical cancer that compared RT with or without concurrent chemotherapy, identified 19 trials comprising 4,580 randomized patients, and they were the subjects of the meta-analysis. Concomitant chemotherapy and radiation improved tumor control and overall survival (RR = 0.71; p < .0001) and progression-free survival (RR = 0.61; p < .0001). The benefit was maximal in early-stage (I and II) disease. The absolute survival benefit was 12%. Patients receiving chemoirradiation had a higher incidence of grade 3 or 4 hematologic and gastrointestinal toxicities.
Patients with stage IVA disease (bladder and/or rectal invasion) can be treated either with higher doses of external radiation to the whole pelvis with concurrent chemotherapy followed by intracavitary or interstitial insertions (total dose to point A with LDR brachytherapy about 90 Gy) and additional parametrial irradiation, or with pelvic exenteration.439 Niibe et al.,440 in an analysis of 179 patients with stage IIIB adenocarcinoma, suggested that an optimal dose for large tumors was a total biological effective dose of >100 Gy.
A prospective single-arm trial, RTOG 0116, treated patients with positive para-aortic or high common iliac lymph nodes with extended-field radiation combined with cisplatin chemotherapy followed by brachytherapy.441 IMRT was not allowed. The trial showed that this regimen is feasible, but the late grade 3 and 4 toxicity rate was 40%. There was no reduction in acute toxicity with the addition of amifostine.442 The use of extended-field IMRT has been shown to reduce the risk of gastrointestinal toxicity in two retrospective series reports443,275 and may be of benefit to patients with positive common or para-aortic lymph nodes requiring extended-field radiation with concurrent chemotherapy.
In countries or circumstances where a wait time exists and patients cannot immediately start on concurrent chemoradiation, induction chemotherapy may be considered. An alternative to cisplatin in this situation may be concurrent nedaplatin, which did not cause any nephrotoxicity in an analysis of 104 patients.444
Randomized Trials of Chemoradiation
Results from several cooperative oncology groups demonstrated that cisplatin-based chemotherapy, when given concurrently with RT, prolongs survival in women with locally advanced cervical cancers (Table 69.12), as well as in women with stage I to IIA disease who have metastatic disease in the pelvic lymph nodes, positive parametrial disease, or positive surgical margins at the time of primary surgery.445
The GOG conducted randomized Protocol 85, in which patients with carcinoma of the cervix, a clinical stage of IIB to IVA, and negative para-aortic nodes were treated with external pelvic irradiation (51 Gy) combined with 30 Gy to point A with LDR brachytherapy.446 One hundred twenty- seven patients received 5-FU (IV infusion, 1 g/m2 for 4 days) and cisplatin (50 mg/m2 IV) on days 1, 29, and 30 to 33, and 191 patients received hydroxyurea (80 mg/kg orally twice weekly). With a median follow-up for survivors of 8.7 years, the 5-year survival rate in the cisplatin/5-FU arm was 60%, compared with 47% for women in the hydroxyurea arm.
TABLE 69.12 DETAILS OF THE TREATMENT PROTOCOLS OF THE FIVE RANDOMIZED TRIALS THAT FORMED THE BASIS OF THE NATIONAL CANCER INSTITUTE ANNOUNCEMENT

After completion of GOG 85, the group opened GOG 120447,448 for the same patient population, which was a three-arm randomized trial comparing irradiation plus hydroxyurea versus irradiation plus weekly cisplatin versus irradiation plus hydroxyurea, cisplatin, and 5-FU. In 526 evaluable patients with a median follow-up for survivors of 106 months, the 5- and 10-year survival rates for women in both the weekly cisplatin and irradiation arm and the irradiation, 5-FU, and cisplatin arm were 60% and 53%, respectively, compared with 40% and 34% in the hydroxyurea and irradiation arm (p ≤ .01). Overall survival was also significantly better in the two patient groups receiving cisplatin. Hematologic toxicity was greater in the group treated with the three drugs compared with cisplatin or hydroxyurea alone.
The RTOG conducted a randomized study of 389 patients with stage IB to IIA of >5 cm, proven positive pelvic lymph nodes, or stage IIB to IVA carcinoma of the cervix in which patients were treated with either pelvic and para-aortic irradiation (best arm of RTOG Protocol 79-20) or pelvic irradiation and three cycles of concomitant chemotherapy with cisplatin (75 mg/m2) and 4-day infusion of 5-FU (1,000 mg/m2 per day).449 Results were updated by Eifel et al.397 With a median follow-up of 6.6 years for 228 survivors, the 8-year overall survival rate for women on the irradiation and cisplatin/5-FU arm was 67% versus 41% in the irradiation-only arm (p < .0001). Disease-free survival rates were 66% and 36%, respectively. There were no significant differences in late complications in the treatment groups.
Southwest Oncology Group 8797 was a study for women with FIGO stage IA2, IB, or IIA carcinoma of the cervix with metastatic disease in the pelvic lymph nodes, positive parametrial involvement, or positive surgical margins at the time of primary radical hysterectomy with total pelvic lymphadenectomy. Patients had confirmed negative para-aortic lymph nodes; if the para-aortic lymph nodes were not sampled, the patients had confirmed negative common iliac lymph nodes. One hundred twenty-seven patients were randomized to treatment with pelvic EBRT with 5-FU infusion and cisplatin, and 116 were treated with irradiation alone. The 3-year survival for women on the adjuvant cisplatin/5-FU and RT arm was 87%, compared with 77% for women on the pelvic irradiation arm.409 The difference was statistically significant. An updated analysis with 5.2-year median follow-up reported 5-year overall survival of 80% versus 66%, favoring postoperative chemoradiation in high-risk patients.183
In GOG 123, 369 women were enrolled. One hundred eighty-three women with bulky (≥4 cm) stage IB carcinoma of the cervix with negative pelvic and para-aortic nodes radiographically or surgically determined were randomized to be treated with pelvic EBRT and brachytherapy, followed by extrafascial hysterectomy, and 186 received EBRT and brachytherapy with weekly cisplatin (40 mg/m2; total dose not to exceed 70 mg/week) followed by extrafascial hysterectomy.450 In an updated analysis with median follow-up of 101 months451 the 6-year progression-free survival rate for women treated with irradiation and cisplatin was 71%, compared with 60% for those treated with RT alone, after adjusting for age and tumor size (p < .004). The unadjusted 6-year overall survival rates were 78% and 64%, respectively (p < .015).
The results of randomized trials using concurrent chemoradiation are summarized in Table 69.13.452 Curtin et al.453 completed a small phase III trial in which 89 patients with high-risk stage IB or IIA undergoing radical hysterectomy and pelvic node dissection were randomized to be treated with postoperative cisplatin/bleomycin alone (44 patients) or combined with pelvic RT (45 patients). There were 9 and 10 recurrences, respectively, and survival was equivalent.
On the other hand, Pearcey et al.454 reported on a Canadian randomized study in which 127 patients with stage IB to IIA of >5 cm or IIB carcinoma of the cervix were randomized to be treated with cisplatin (40 mg/m2 weekly) and RT, and 126 patients were treated with RT alone (50.4 Gy to the pelvis combined with brachytherapy). With a median follow-up of 65 months, the 5-year survival rates were 59% and 56%, respectively (p = .43). There was a somewhat greater incidence of significant late morbidity in the RT-alone group (12% vs. 6%; p = .08). Possible explanations for the discrepancy in results between the five U.S. trials371 and the Canadian study were analyzed by Lehman and Thomas.445 Some theories include that a higher percentage of early-stage patients were accrued, who therefore had less of a difference in survival, given that the baseline survival rate for both arms was quite high; and that treatment time was short for both arms, again minimizing the difference in improving survival in the chemotherapy arm. This was the smallest of the randomized chemoradiation trials, and although the hazard ratio was reduced, given the factors equalizing the two arms, a larger number of patients would have possibly shown a significant difference.
A 2005 update of a meta-analysis of concomitant chemotherapy and radiation therapy found 24 trials and concluded that chemoradiation improves overall survival and progression-free survival, whether or not cisplatin was used, with absolute benefits of 10% and 13% respectively.455 Similarly, a 2008 meta-analysis of the 13 trials that compared chemoradiotherapy to radiation found there was a 6% improvement in 5-year survival with concurrent chemoradiation (hazard ratio 0.81, p < .001). The effect was attributed to a reduction in both local and distant recurrence. Chemoradiation increased acute hematologic and gastrointestinal toxicity, but no confirmation was made about a difference in late toxicity.456
TABLE 69.13 RANDOMIZED STUDIES OF CONCURRENT CHEMOIRRADIATION IN CERVICAL CARCINOMA

In the United States, weekly cisplatin has become the preferred approach, with less toxicity than an every 3-week regimen and the increased likelihood of completing the treatment on schedule. The number of completed cycles of weekly treatment was shown by Nugent et al.457 to be predictive of survival. One hundred eighteen patients with locally advanced cervical cancer (stages IB2 to IVA) were treated with combination weekly cisplatin (40 mg/m2) and radiation between 2003 and 2007. Thirty percent of patients completed fewer than six cycles of chemotherapy. In multivariate analyses, the number of chemotherapy cycles was independently predictive of progression-free survival (PFS) and overall survival (OS). Patients who received fewer than six cycles of cisplatin had a worse PFS (HR = 2.65; 95% CI = 1.35 to 5.17; p = .0045) and OS (HR = 4.47; 95% CI = 1.83 to 10.9; p = .001). Advanced stage, longer time to RT completion, and absence of brachytherapy were also associated with decreased OS and PFS (p < .05). Similar results were found when analysis was conducted using a breakpoint of at least but not less than five chemotherapy cycles. The authors concluded that aggressive supportive care to minimize missed chemotherapy treatments may improve survival after chemoradiation. A retrospective review458 questioned whether cervical cancer patients should receive cisplatin 20 mg/m2 é 5 days every 21 days concomitant with RT, or weekly 40 mg/m2 weekly concomitant with RT, given that an advantage with regard to both acute toxicity and progression-free survival was seen in the 5-day regimen.
High-risk patients may benefit from adjuvant chemotherapy after chemoRT. A randomized trial of 515 cases of stages IIB to IVA cervical carcinoma treated with concurrent gemcitabine plus cisplatin followed by adjuvant gemcitabine and cisplatin compared to standard concurrent cisplatin with radiation showed a significant 3-year progression-free survival benefit 74% versus 65% (p = .03), as well as one in overall survival (HR = 0.68, 95% CI = 0.49 to 0.65).459 Grades 3 and 4 toxicity was higher in the extended-chemotherapy arm, including 2 deaths. Ongoing trials comparing “outback” chemotherapy continue to accrue patients.
Alternatives to Concurrent Cisplatin-Based Chemoradiation
In GOG Protocol 165, patients with stages IIB to IVA cervical cancer received either radiation therapy and concurrent weekly cisplatin (40 mg/m2) or radiation therapy and a protracted venous infusion (PVI) of 5-FU. Lanciano et al.460 reported that the study was prematurely closed after an interim analysis showed a failure rate 35% higher and would not result in improved DFS with PVI 5-FU/RT compared with weekly cisplatin.
Lorvidhaya et al.461 reported on 673 patients with predominantly stages IIB and IIIB disease randomized to receive either irradiation alone or combined with chemotherapy administered in an adjuvant, concurrent, or adjuvant and concurrent schedule. Concomitant chemotherapy consisted of mitomycin C (10 mg/m2) given on days 1 and 30 and oral 5-FU (300 mg/m2 per day) given on days 1 to 14 and 42 to 56. Adjuvant chemotherapy consisted of three cycles of oral 5-FU (200 mg/day) given for 4 weeks, with a 2-week rest every 6 weeks. With a median follow-up of 25 months, there was a statistically significant improvement in disease-free survival for all patients who received chemotherapy/RT, regardless of the timing of administration of the chemotherapy. However, the authors did not state the radiation dose delivered with brachytherapy, the total dose prescribed to point A, or the overall treatment time. In the absence of this information, the adequacy of the radiation therapy cannot be evaluated, and we cannot assume that the results of this study apply to all patients treated with irradiation.
In a randomized trial comparing monthly fluorouracil and cisplatin versus weekly cisplatin concurrent with pelvic radiation, Kim et al.462 enrolled 158 stages IIB and IVA patients. With a median follow-up of 39 months, the acute grades 3 and 4 hematologic toxicity were significantly worse in the fluorouracil/cisplatin arm, 43% versus 26% (p = .04). The trial was not powered to detect a survival difference, and no difference in the overall or progression-free survival was noted.
Tseng et al.463 published results of a study in which patients with advanced carcinoma of the cervix were randomly assigned to either RT alone or concurrent chemotherapy (cisplatin, vincristine, and bleomycin every 3 weeks for a total of four courses) and RT. After a median follow-up of 46.8 months, the disease-free survival and actuarial survival rates were 51.7% and 61.7% in the concurrent group and 53.2% and 64.5% in the RT group, respectively (p = .27). Treatment-related toxicity was higher with the combination therapy compared with irradiation alone (36.7% vs. 17.7%; p = .02).
Several small phase II studies have been performed showing no advantage to weekly paclitaxel over weekly cisplatin464 and too high toxicity with concomitant cisplatin–paclitaxel.462 Concurrent weekly carboplatin alone has been shown in many studies to be feasible,466 including in the elderly; it is also an alternative in patients that have an elevated creatinine. Docetaxel and carboplatin concurrent with radiation was also found to be a feasible regimen.467
The GOG carried out a trial of irradiation with either concomitant hydroxyurea (HOU) or a placebo in patients with stage IIIB or IVA cervix cancer.468 The study was criticized because patients were not surgically staged, half of the 190 patients were not evaluable, and radiation doses were low.302 Piver et al.469 published an update of a study of 130 patients (13 with para-aortic lymph node metastasis), 75 of whom were surgically staged. Of 66 patients who underwent surgical staging, 33 received hydroxyurea and 33 a placebo in combination with irradiation. Of the patients who did not have surgical staging, 27 received hydroxyurea and 37 received placebo. The 2-year survival was higher in the HOU group. Symonds et al.,470 in a review of seven randomized trials, found no evidence to support the use of hydrea with RT in cervix cancer.
In larger randomized trial by the GOG reported by Stehman et al.,471 296 surgically staged patients with stage IIB to IVA disease and negative para-aortic nodes were randomized to irradiation plus either hydroxyurea (139 patients) or misonidazole (157 patients). Survival was not statistically different between the regimens, with 33.8% deaths in the hydroxyurea group and 38.9% deaths in the misonidazole group (p = .25). Failure limited to the pelvis occurred in 18% of patients in the hydroxyurea group and 23.6% in the misonidazole group. Of note, in a randomized RTOG trial of patients with stage III disease, Leibel et al.472 and Overgaard et al.473 reported lower survival in patients receiving misonidazole than in the patients treated with irradiation alone.
Grigsby et al.474 published results of an RTOG study in which 120 patients with carcinoma of the cervix were randomized to receive irradiation alone or combined with misonidazole. The 5-year progression-free survival was 22% and 29%, respectively. These findings are similar to those reported by Overgaard et al.,473 who, in a randomized study of 331 patients with carcinoma of the cervix treated with either misonidazole or a placebo and irradiation, found no significant difference in local tumor control (50% vs. 54%), disease-free survival (47% vs. 46%), or crude survival (39% vs. 45%).
Nonrandomized Studies of Chemotherapy and Irradiation
Numerous preliminary reports have been published on results of neoadjuvant/concomitant use of cisplatin and 5-FU, with or without mitomycin C, combined with irradiation to treat patients with locally advanced or recurrent carcinoma of the cervix.475
Trials with Cisplatin, 5-Fluorouracil, or Both
Perez and Grigsby476 reported on 58 patients with locally advanced carcinoma of the cervix treated with concurrent 5-FU/cisplatin and irradiation and compared the results with 257 patients with similar stages treated with irradiation alone during the same period. Pelvic tumor control and disease-free and cause-specific survival were comparable. The incidence of rectal and bladder fistula was 7% in the chemoirradiation group and 4% with irradiation alone (p = .61).
Park et al.434 described results in 113 patients with high-risk invasive cervical carcinoma treated with cisplatin and 5-FU. For adenocarcinoma, doxorubicin (45 mg/m2 IV) was added. The patients subsequently received radiation therapy (not described in the publication). For patients with stage I or II tumors >4 cm, the 5-year survival rate was 78.3% with chemoirradiation and was 48% for 77 patients treated with RT alone (p < .01). For stages III and IV the rates were 69.1% and 57.4%, respectively. Toxicity with combined chemoirradiation was not significantly enhanced compared with irradiation alone.
Sardi et al.477 reported results of three courses of cisplatin, vincristine, and bleomycin (days 1 to 3) at 10-day intervals combined with RT in 205 unselected patients with stage IB cervix cancer (tumors >2 cm) who were divided at random into two groups treated with surgery and RT or neoadjuvant chemotherapy, surgery, and irradiation. After 67 months, no difference in survival was seen in patients with tumors 2 to 4 cm in both groups (77% for control patients vs. 82% with neoadjuvant chemotherapy), but statistically significant differences were seen in bulky tumors (>4 cm): 61% versus 80% in favor of neoadjuvant chemotherapy.
Souhami et al.478 treated 50 patients with bulky, locally advanced carcinoma of the cervix with a combination of weekly cisplatin (30 mg/m2) concurrent with RT. At 44 months, the actuarial survival rate was 65%, the total pelvic failure rate was 26%, and the distant metastasis rate was 24%. The incidence of late gastrointestinal toxicity was high, with 10 rectal ulcers (4 colostomies required for severe bleeding), 2 rectovaginal fistulas, and 2 small-bowel obstructions.
Park et al.479 treated patients with stages I and II carcinoma of the cervix >4 cm with RT alone or concurrent or sequential chemoradiation with cisplatin and 5-FU. The 30-month survival rates were 100% with concurrent chemoirradiation, 89.5% with sequential treatment, and 79.5% with irradiation alone (p < .05).
Lee et al.480 treated 40 women with cervix cancer using 50-Gy EBRT and brachytherapy; in 25 cases, three concurrent cycles of cisplatin/5-FU were given, and in 15 cases, six cycles of consolidation chemotherapy were given. There was no difference in 2-year survival between the two groups (98% to 100%). Grade 2 or greater hematologic toxicity was more frequent in the consolidation patients.
Grigsby et al.,481 in a prospective study of 65 patients with cervical cancer and node negative on FDG-PET treated with RT alone (15 patients) or combined with concurrent weekly cisplatin (50 patients), noted a 5-year cause-specific survival of 78% and 74%, respectively. Severe complications included 1 rectovaginal fistula and 1 rectal stricture in the concurrent chemotherapy/RT group and 1 chemotherapy-related death.
Trials with Carboplatin
Katanyoo et al.466 reported 148 patients with stages IIB to IVA cervical cancer treated with concurrent weekly carboplatin (100 mg/m2 or area under the curve 2) for a median of six cycles and radiation. Among the 142 responders, 36 experienced recurrences: pelvic recurrences in 7 (4.7%), distant failure in 25 (16.9%), and both pelvic and distant in 4 (2.7%). The 2- and 5-year progression-free survival rates were 75.1% and 63.0%, respectively, with the corresponding 2- and 5-year overall survival rates of 81.9% and 63.5%. No grade 3 or 4 hematologic and nonhematologic toxicities were observed during treatment in any patients. Late grade 3 to 4 gastrointestinal or genitourinary toxicities were 10.1% and 0.7%, respectively.
Cetina et al.482 looked at the use of weekly carboplatin in 59 elderly, diabetic, and/or hypertensive stage IB2 to IIIB cervical cancer patients. All patients completed radiation and 80% received five of six planned cycles, with 83% reaching a complete response. With a median follow-up of 20 months, 33% relapsed, and the 3-year overall survival rate was 63%. Although the regimen is safe and tolerable, it may have reduced efficacy compared to weekly cisplatin.
Trials with Mitomycin C or Tirapazamine
Mitomycin C acts as an alkylating agent and inhibits DNA and RNA synthesis. Activation of mitomycin C is increased in hypoxic conditions, and thus it acts as a hypoxic radiosensitizer. Interstitial pneumonitis and pulmonary fibrosis are usually related to the dose of drug. Use of IV dexamethasone before administration of the drug may prevent pulmonary toxicity.
Christie et al.432 described results in three groups of patients with stages IIB and III carcinoma of the cervix treated with pelvic irradiation and an intracavitary insertion combined with chemotherapy. Group A (64 patients) received 5-FU infusion during the first and last weeks of irradiation combined with mitomycin C (10 mg/m2 IV). Group B (29 patients) received 5-FU without mitomycin C, and group C (84 patients) received irradiation alone. With median follow-up of 7.2 years, the 5-year survival rates were 56%, 32%, and 36%, and the local tumor control was 73%, 53%, and 50%, respectively. Toxicity was greater in group A (36% grade 3 and 4) compared with the 5-FU and irradiation group (14%) and the irradiation-alone group (20%).
Roberts et al.483 reported on a trial in which 160 patients with locally advanced cervical cancer were randomized to receive RT alone (82 patients) or RT with concomitant mitomycin C (78 patients). The 4-year actuarial survival was 72% and 56%, respectively (p = .13), and the local recurrence-free survival rate was 78% and 63%, respectively (p = .11). There were no treatment-related deaths. No excess in nonhematologic toxicity has been observed with combined mitomycin C and irradiation.
Tirapazamine, a radiation sensitizer with selective cytotoxic effect on hypoxic cells, was combined with cisplatin in 56 patients with recurrent or metastatic cancer. After six cycles given every 21 days, 4 complete and 13 partial responses were noted. Overall 6-month survival was 56%. Better response was seen in patients who had not received radiosensitizing chemotherapy previously.484
Besides the usual hematologic and pelvic toxicity described in many of these studies with chemoradiation, Wun et al.,485 in a retrospective analysis of 75 patients with gynecologic cancer who received erythropoietin and chemotherapy/RT, noted that 17 had upper- or lower-extremity thrombosis, in contrast to 2 of 72 who did not receive erythropoietin. Of note, Anders et al.,486 in a review of the literature, reported that 6 of 128 patients (4.7%) treated with chemotherapy/RT without erythropoietin developed grade 4 or 5 thrombosis toxicity.
Trial with Bevacizumab Concurrent with RT
RTOG 0417 treated patients with once-weekly cisplatin (40 mg/m2) chemotherapy and standard pelvic radiotherapy and brachytherapy. Bevacizumab was administered at 10 mg/kg intravenously every 2 weeks for three cycles. A total of 49 patients were evaluable. The median follow-up was 12.4 months (range, 4.6 to 31.4 months). There were no treatment-related serious adverse events. There were 15 (31%) protocol-specified, treatment-related adverse events within 90 days of treatment start; the most common were hematologic (12 of 15; 80%). Eighteen (37%) occurred during treatment or follow-up at any time.487
Trial with Epidermal Growth Factor Receptor Inhibition Concurrent with RT
Nogueira-Rodrigues et al.488 reported a phase I study administering escalating doses of erlotinib (50/100/150 mg) combined with cisplatin (40 mg/m2, weekly, five cycles) and radiotherapy (external beam, 4,500 cGy in 25 fractions, followed by four fractions/600 cGy weekly of brachytherapy) in squamous cell cervical carcinoma patients, stages IIB to IIIB. Fifteen patients were enrolled, 3 at dose level (DL) 50 mg, 4 at DL 100 mg, and 8 at DL 150 mg. Three patients did not complete the planned schedule. One patient at DL 100 mg withdrew informed consent due to grade 2 rash; at DL 150 mg, 1 patient presented with Raynaud’s syndrome and had cisplatin interrupted, and another patient presented with grade 4 hepatotoxicity. The latter was interpreted as dose-limiting toxicity, and a new cohort of 150 mg was started. No further grade 4 toxicity occurred. Grade 3 toxicity occurred in 6 cases: diarrhea in 3 patients, rash in 2 patients, and leukopenia in 1 patient. Treatment did not lead to limiting in-field toxicity.
Intra-Arterial Chemotherapy
Intra-arterial infusion of chemotherapeutic agents in cervical carcinoma was used for some years based on the distinct arterial supply to the tumor-bearing area. Unfortunately, the responses have been uncommon and short, and the toxicity and complication rates have been significant.489
Onishi et al.490 evaluated intra-arterial cisplatin through catheters inserted into both internal iliac arteries in cervix carcinoma. Patients were randomized into a concurrent intra-arterial infusion of cisplatin with RT (18 patients) or RT alone (15 patients). Five-year overall survival rates were 44.4% and 50%, respectively. In the group receiving intra-arterial infusion, grade 3 or 4 late bowel complications were seen in 44% and grade 3 or 4 myelosuppression in 33%, significantly more than in the RT group.
Neoadjuvant Chemotherapy
Thomas491 summarized the rationale and potential limitations of neoadjuvant chemotherapy in carcinoma of the cervix. Although response rates to the chemotherapy are between 30% and 85%, none of the studies showed an advantage for pelvic tumor control or survival.492–499 Colombo et al.,500 in a review of the literature, concluded that the role of neoadjuvant chemotherapy followed by radiation and by concomitant chemotherapy or by surgery is controversial because no significant advantages in survival or local control have been shown, and receiving upfront chemotherapy may compromise immune status and the patient’s ability to receive definitive treatment with radiation or surgery.
Souhami et al.501 randomized 107 patients with stage IIIB carcinoma of the cervix to treatment with irradiation alone or combined with bleomycin, vincristine, mitomycin, and cisplatin. The overall 5-year survival rate for the neoadjuvant-treated patients was 23%, in contrast to 39% for those treated with irradiation alone (p = .02). Locoregional and distant failure rates were similar in both groups.
Kumar et al.502 reported a randomized trial in which 94 patients with carcinoma of the cervix were treated with chemotherapy (two cycles of bleomycin, ifosfamide-mesna, and cisplatin) followed by RT, and 90 patients were treated with irradiation alone. In the chemotherapy/RT group, 32-month survival was 63% for stage IIB and 50% for stage III, and in the RT group, the rate was 59% for stage IIB and 27% for stage IIIB tumors (differences not statistically significant). There was no difference in radiation-induced toxicity between the two groups.
In a Swedish study,498 47 patients with carcinoma of the cervix were randomized to be treated with irradiation alone (64.8 Gy, 1.8-Gy fractions) and 47 with a combination of three cycles of cisplatin and 5 days of 5-FU administered every third week, followed by the same pelvic irradiation. The 5-year disease-free survival rates were 70% with chemoirradiation and 57% with irradiation alone (p = .07). The incidences of pelvic recurrence were 60% and 47%, respectively, and for distant metastasis they were 19% and 35%, respectively. Two patients in the chemoirradiation and 1 in the irradiation-alone group died as a consequence of therapy.
Response to Chemotherapy Alone for Patients with Metastatic or Recurrent Disease
Cisplatin has been combined with other cytotoxic agents. Long et al.503 conducted a randomized study comparing methotrexate, vinblastine, doxorubicin, and cisplatin (MVAC), cisplatin/topotecan, or cisplatin alone in patients with advanced cervical cancer. The MVAC arm was closed after 4 deaths in 63 patients. In 294 patients assigned to the other arms, response rate was 27% for cisplatin/topotecan and 13% for cisplatin alone, with median survival of 9.4 and 6.5 months, respectively.
Paclitaxel, a natural product found initially in the bark of the western yew tree, produces depolymerization and irreversible bundling of tubulin in the cell. It has been shown to have a radiosensitizing effect and may also be considered for patients with metastatic disease. Rose et al.504 reported on a phase II study of 44 patients in which the starting dose was paclitaxel (135 mg/m2, maximum 170 mg/m2) infused over 24 hours, followed by cisplatin (75 mg/m2) every 21 days. Forty patients (90.9%) had received prior radiation therapy. A median of six courses of chemotherapy was given. Of the 41 assessable patients, 5 (12.2%) had a complete response and 14 (34.1%) had a partial response. Vinorelbine is a semisynthetic derivative of vinblastine. In a phase II trial in patients with prior irradiation, a 28% response rate was observed.505 Other trials used the drug as neoadjuvant chemotherapy; in 42 patients, 2 complete and 17 partial responses (45%) were observed.506,507
Irinotecan and topotecan are camptothecin derivatives whose cytotoxic mechanism is believed to target topoisomerase I.508 An international phase II trial reported a similar 21% response rate in patients predominantly with prior irradiation (1 complete and 8 partial responses among 42 patients).509
Gemcitabine, a nucleoside analogue, showed a 4.5% partial response and 36% stable disease in 22 patients.510 In combination with cisplatin, it was evaluated in 32 women with previously treated cervix cancer (initial dose 800 mg/m2 on days 1 and 8, then every 28 days); there were 7 (22%) partial responses and 12 stable disease responses.511
A phase II trial of docetaxel and gemcitabine showed an overall response rate of 21%. With a median survival of 7 months, 39% were alive at 1 year. Doxetaxel combined with carboplatin has been shown to have a 25% response rate.467
A four-arm comparison of cisplatin/paclitaxel versus cisplatin/vinorelbine, cisplatin/gemcitabine, or cisplatin/topotecan found that the standard arm of cisplatin/paclitaxel remained the best option for patients with stage IVB, recurrent or persistent cervical carcinoma.512
In a phase II GOG study of cetuximab 400 mg/m2 initial dose followed by 250 mg/m2 weekly until disease progression or prohibitive toxicity, 38 patients (15%) had no progression for at least 6 months. The median OS was 7 months. When cetuximab was combined with cisplatin, no additional benefit beyond cisplatin was identified.513
Ifosfamide, paclitaxel, and carboplatin as a triple regimen showed a 33% objective complete or partial response, with an overall median survival of 10 months.514 Antiangiogenic tyrosine kinase inhibitors pazopanib and lapatinib were tested in 230 patients with stage IVB persistent/recurrent cervical carcinoma. An improvement in progression-free and overall survival was seen with pazopanib.515
Studies with Radiation Alone
For patients unable to tolerate concurrent chemotherapy or in countries where chemotherapy is not readily available, radiation alone may be used as an alternative to chemoradiation. Mendenhall et al.516 analyzed 1211 patients treated with radiation alone with a minimum follow up of 3 years. In patients with Stage IB and IIA disease there was no significant correlation between doses to these points and pelvic tumor control. In Stage IIB doses of less than 6000 cGy to point A correlated with a high pelvic failure rate (8 of 12, 66.7%) in contrast to doses of 6000 to 9000 cGy (61 of 261, 23.4%) or higher than 9000 cGy (10 of 74, 13.5%) (P 0.01). In Stage III the pelvic failure rate with doses below 6000 cGy to point A was 72% (18 of 25) compared to 39% (71 of 180) for 6000 to 9000 cGy or 35% (27 of 77) with doses above 9000 cGy (p ≤ 0.01).
Thoms et al.517 reported on 363 patients with bulky endocervical carcinoma treated with curative intent (246 with irradiation alone and 117 with irradiation and surgery); 10-year survival was 45% and 64%, respectively. In a subset of 48 patients with similar tumors treated with irradiation alone and 45 treated with irradiation and surgery, the 10-year survival rates were comparable, and the pelvic tumor control rates were 90% and 87%, respectively.
Eifel et al.168 evaluated 1,526 patients, of whom 371 had tumors 6 cm or greater. There were biases in treatment selection, but a statistically significantly higher 10-year survival rate was noted in patients treated with irradiation and surgery (64% vs. 45%). Tumor diameter was highly significant as a prognostic factor, and the authors concluded that only patients with lesions >8 cm in diameter benefited from adjuvant hysterectomy. In the same study, 98 patients with stages IB and IIB bulky endocervical carcinomas (≥6 cm in diameter) were treated with RT alone. Twenty-four patients received <6,000 mgh of intracavitary treatment, and 73 received higher doses. Despite having somewhat more favorably treated tumors, patients who received <6,000 mgh had a higher rate of pelvic recurrence at 5 years (33%) than those who received higher doses (16%; p = .03). Actuarial 5-year survival rates were 44% and 60% for low- and high-dose groups, respectively (p = .14).
Kim et al.518 assessed the prognostic factors for pelvic tumor control in 40 patients with FIGO stage IB or IIA carcinoma and 25 patients with stage IIB carcinoma classified as barrel shaped (i.e., at least 5 cm in diameter) treated with curative intent. Seventy-two percent were treated with RT alone and 28% with RT and extrafascial hysterectomy. The extent of tumor regression after external-beam radiation therapy correlated with the likelihood of local tumor control (p = .02). For patients treated with radiation therapy alone, increased brachytherapy dose was associated with better local tumor control. The 10-year overall and cause-specific survival rates were 53% and 68%, respectively, and did not differ significantly between treatment groups.
Paley et al.519 reported on 57 patients with barrel-shaped (mean diameter, 5 to 9 cm) cervical carcinoma treated with preoperative EBRT and BT (mean dose to point A, 79.6 Gy) followed by extrafascial hysterectomy 6 to 8 weeks later. Residual disease was present in 35 (61%) of the hysterectomy specimens; tumor sterilization correlated significantly with the mean dose to point A (p = .016). Ninety-five percent of the patients with negative specimens remained clinically free of disease at their last follow-up versus 31% of those with residual disease (p < .001).
The GOG and RTOG conducted a randomized phase III clinical trial in which 282 patients with carcinoma of the cervix measuring 4 cm or greater (exophytic or barrel shaped) were treated with either external-beam and intracavitary irradiation or a slightly lower dose of intracavitary irradiation and the same pelvic EBRT followed by an extrafascial hysterectomy.520 The survival rates were 61.4% for irradiation alone and 64.4% for the combined irradiation and surgery group. The incidence of recurrences was 43.3% in the irradiation group compared to 34.5% with combined therapy (p = .081). The incidence of local recurrences was 25.8% and 14.4%, respectively. The incidence of grade 3 and 4 sequelae of therapy was 10.5% and 9.8%, respectively. Thus, the addition of hysterectomy to standard irradiation did not significantly affect survival, although there was a small reduction in the local recurrence rate. When combined therapy is used, the dose of irradiation delivered to the lymph nodes, the time of the operation, and the pathologic examination of the specimens are critical in determining the presence of postirradiation residual tumor.
Perez et al.36 noted that in patients with primary carcinoma of the uterine cervix who had endometrial stromal invasion or tumor only in the curettings, the addition of a hysterectomy did not improve the survival rate because most of the patients failed at distant sites.
For stage IIB tumors treated with irradiation alone, the 5-year survival rate is 60% to 65%. The pelvic failure rate ranges from 18% to 39%. In an analysis of the Patterns of Care Study in 157 patients who had stage IIB disease, Coia et al.521 reported a better 4-year survival rate (67% and 54%) and in-field tumor control rate (78% and 68%) in patients with unilateral versus bilateral parametrial involvement, respectively. Similarly, in a review of 1,178 patients with stage IIB disease, the 5-year survival rates were 70% with medial parametrial and 58% with lateral parametrial involvement (p = .004).522 Kim et al.,523 in patients with stage IIB disease, found a correlation of point A dose and incidence of pelvic failures.
In stage IIIB carcinoma, the 5-year survival rates range from 25% to 48%, and pelvic failure rates range from 38% to 50%.165,524 Hanks et al.,525 reporting on the Patterns of Care Study, noted a 28% probability of 5-year survival in patients with stage III carcinoma of the cervix treated in a large number of facilities in the United States versus 60% survival in selected large centers (extended survey). Later, Komaki et al.526 reported a significant increase in local pelvic tumor control (69%) in patients with stage III carcinoma of the cervix treated in 1983, compared with 37% and 49% in earlier periods (p = .03). The 5-year survival rate increased from 25% to 47% (p = .02). The improvement in pelvic tumor control may be associated with higher external-beam doses but more likely is related to the substantial increase in the percentage of patients receiving brachytherapy (96%) and more careful dosimetry and dose calculations for intracavitary therapy. They noted a decrease in major complications from 15% in the 1973 and 13% in the 1978 Patterns of Care Surveys to 7% in 1983. Montana et al.527 reported that calculation of doses to the bladder and rectum were performed in 80% and 76% of patients, respectively, in the 1983 survey, which may also have resulted in decreased toxicity.
Arthur et al.,528 in 89 patients with stage IIIB carcinoma of the cervix treated with external irradiation and brachytherapy, observed a locoregional tumor control rate of 22.5% and a disease-free survival rate of 15% in 16 patients treated with 78 Gy or lower doses to point A, in comparison with 53% and 47%, respectively, in 24 patients receiving higher doses.
Horiot et al.529 reported the results of a French cooperative study of 1,383 patients with invasive carcinoma of the uterus treated with irradiation alone following the MD Anderson Hospital treatment guidelines. Survival and locoregional tumor control were similar in both groups, except in stage III, in which the pelvic and central failure rates were lower in the French patients, may be because of different tumor volumes or socioeconomic factors. Major urinary complications were noted in 2% of the patients. Grade 3 bowel complications occurred in 3% of the patients with stages I and IIA disease and in 7% of patients with stages IIB and III disease. Barillot et al.530 updated the results in 642 patients; the analysis was divided into three periods: 1970 to 1978 (use of standard prescriptions), 1979 to 1984 (implementation of individual adjustments), and 1985 to 1994 (systematic individual adjustments). There was a significant reduction of the external radiation dose (>40 Gy in 47% of patients before 1979 vs. 36% after 1984), use of parametrial boost (55% vs. 39%), use of vaginal cylinder (28% vs. 11.5%), and combined intracavitary and external irradiation volume (842 vs. 503 cm3 on average). The 5-year actuarial toxicity rates were as follows: grade 2, 23.5%; grade 3, 10%; and grade 4, 3%. The three main predictive factors for rectal and bladder sequelae were increased external radiation dose, higher dose rate at reference points, and whole-vagina brachytherapy.
Marcial et al.531 described results of a randomized trial in 301 patients with stages IIB, III, and IVA carcinoma of the uterine cervix treated with split-course irradiation (10 fractions of 2.5 Gy, five weekly doses up to 25 Gy, followed by a rest period of 2 weeks, and an additional 25 Gy delivered in the same manner) or continuous irradiation (30 fractions of 1.7 Gy daily, five times per week, total dose 51 Gy) combined with LDR brachytherapy for 30 Gy to point A. There was no significant difference in tumor control, acute or late complications, or survival in the two groups.
In patients with stage IVA disease, the 5-year survival rates range from 18% to 34%, and pelvic failures from 60% to 80% after definitive irradiation.260 Million et al.261 reported 18 of 53 patients (34%) with bladder involvement surviving without disease after definitive irradiation, results comparable with those obtained with exenteration. Kramer et al.260 reported on 48 patients with stage IVA carcinoma of the cervix treated with definitive RT. Patients with minimal parametrial involvement had a 5-year survival rate of 46%, compared with only 5% for those with extensive parametrial tumor. The major complication rate was 22%, consisting mostly of vesicovaginal fistula in 5 patients.
Crozier et al.532 described equivalent 5-year survival rates after salvage pelvic exenteration (37% in 35 patients with adenocarcinoma and 39% in 70 patients with squamous cell carcinoma). In the adenocarcinoma group, 14 of 22 patients, and in the squamous cell-carcinoma group, 14 of 30 patients had distant metastases after pelvic exenteration.
Combination of Irradiation and Surgery
Preoperative Chemoirradiation
Bulky endocervical tumors and the so-called barrel-shaped cervix have a higher incidence of central recurrence, pelvic and para-aortic lymph node metastasis, and distant dissemination.533 In the setting of plain x-ray, point-based planning for brachytherapy, because of the inability of intracavitary sources to encompass the entire tumor in a high-dose volume, larger doses of external radiation to the whole pelvis or extrafascial hysterectomy, or both, have been advocated to improve therapeutic results.4 Alternatively, the use of 3D planning for the brachytherapy component significantly improves survival,534 and the ability to dose escalate due to more precise dose delivery improves local control.535 In rare cases with large residual after 45 Gy external beam, an extrafasical hysterectomy may be considered 6–12 weeks after completion of preoperative irradiation (45 Gy to the whole pelvis and one intracavitary LDR insertion for 5,500 mgh, delivering approximately 50 Gy to point A, with a total dose to point A of 70 Gy, or the equivalent dose in HDR, approximately three fractions of 6 Gy per fraction). Higher doses of irradiation alone yield equivalent pelvic tumor control and survival rates.168,302
Keys et al.450 reported on 183 women with bulky (≥4 cm) stage IB carcinoma of the cervix with negative pelvic and para-aortic nodes radiographically treated with pelvic EBRT and brachytherapy, followed by extrafascial hysterectomy, or EBRT and brachytherapy (to 70 Gy) with weekly cisplatin followed by extrafascial hysterectomy. A significant survival advantage was seen with the combination of concurrent chemoradiation. However, in comparison to other trials, the survival was not significantly improved with the addition of a hysterectomy. The use of PET scanning to determine residual disease allows selection of patients who may be appropriate candidates for a hysterectomy after completion of external-beam treatment.90 Therefore, patients should receive definitive doses of chemoradiation, with hysterectomy reserved for salvage in patients with either gross residual disease or PET-positive disease that is biopsy proven at 3 months after completion of radiation. A subsequent reanalysis including GOG 123 and GOG 71 analyzed 464 patients allocated to pelvic radiation (75 Gy, n = 291) plus hysterectomy or to pelvic radiation (75 Gy) and cisplatin (40 mg/m2, n = 176) plus hysterectomy. A benefit to chemoradiation was seen for patients who had a poor response.536
Morice et al.537 reported a randomized trial of 61 patients treated with adjuvant hysterectomy versus none after EBRT with concurrent weekly cisplatin and vaginal brachytherapy (15 Gy to intermediate-risk CTV) for stage IB2 or II cervical cancer. Hysterectomy increased the number of deaths, with an 11% nonsignificant survival advantage in the no-hysterectomy arm (86% vs. 97%). As a result of this trial, routine adjuvant hysterectomy is no longer practiced for patients who have no residual disease at 6 weeks after chemoradiation.
Motton et al.538 retrospectively reviewed 171 patients treated with chemoradiation followed by simple extrafascial hysterectomy or extended hysterectomy. There was no difference in survival or complication rate based on type of surgery. Leguevaque et al.539 reported on 111 patients treated with or without adjuvant hysterectomy after chemoradiation; there was no advantage to overall survival, but there was a significant difference in recurrence rates.
Touboul et al.540 reported on toxicities for 150 patients with stages IB2 to IVA cervical cancer treated at Institut Gustave Roussy with extrafascial versus modified radical hysterectomy after chemoradiation. After a median follow-up of 3.6 years, 15% had grade 2 or greater side effects, including lymphedema, ureteral fistula, bowel fistula, iliac and vessel rupture. There were 2 postoperative deaths. Modified radical versus extrafascial hysterectomy had a higher odds ratio (OR) for complications of 2.4 (p = .04), as did the presence of residual disease.
Elective Para-Aortic Lymph Node Irradiation
Rotman et al.412 updated results of an RTOG randomized study of 337 patients with stage IIB carcinoma of the uterine cervix with no clinical or radiographic evidence of para-aortic lymphadenopathy who, in addition to standard pelvic irradiation, were randomized to electively receive or not 45 Gy to the para-aortic region (1.6- to 1.8-Gy fractions). The 10-year survival rate was 55% for patients receiving elective para-aortic irradiation and 44% for those treated to the pelvis only (p = .02). The locoregional tumor control rate was similar (69% in the para-aortic node–irradiated group and 65% for the pelvis-irradiated group). The 10-year grade 4 or 5 (major) complication rate was 8% in the group receiving para-aortic irradiation, compared with 4% in patients treated with pelvic irradiation alone (p = .06).
A similar randomized study was reported by Haie et al.541 and the European Organization for Research and Treatment of Cancer (EORTC) on 441 patients with cervical carcinoma, including stage III, who had no evidence of para-aortic lymph node involvement. In the study group, the para-aortic area either received or did not receive 45 Gy with external-beam irradiation. No statistically significant difference was found between the two treatment arms with regard to local tumor control, distant metastases, or survival. However, the incidence of para-aortic and distant metastases without pelvic failure was significantly higher in patients receiving pelvic irradiation alone. The incidence of small-bowel injury was 0.9% in the pelvic irradiation group and 2.3% in the pelvic plus para-aortic irradiation group. A severe complication rate of 9% was observed in patients receiving para-aortic irradiation, compared with 4.8% in those treated to the pelvis only.
Sood et al.542 treated 54 patients with cervix cancer using extended fields (45 Gy) and HDR brachytherapy; 44 received concurrent cisplatin (20 mg/m2 per day for 5 days during week 1 and 4 and once after the second HDR insertion). During a median follow-up of 28 months, 6 patients had died. The 3-year local tumor control was 100% and 85%, respectively. Late toxicity was 10% and 6%, respectively.
Huang et al.235 assessed 758 patients and found that 38 (5%) and 42 (6%) had isolated and nonisolated para-aortic lymph node (PALN) recurrences after a median follow-up of 50 months (range, 2 to 159 months), respectively. The 3-year and 5-year overall survival rate after PALN recurrence was 35% and 28%, respectively, with those with isolated recurrences faring better than those with a nonisolated recurrence (p < .001). An SCC-Ag level of >40 ng/mL (p< .001), advanced parametrial involvement (score 4 to 6; p = .002), and the presence of pelvic lymphadenopathy (p = .007) were independent factors associated with PALN relapse on multivariate analysis. This group subsequently identified pretreatment CEA of ≥10 ng/mL as an additional risk factor of PALN relapse after definitive concurrent chemo-radiation therapy (CCRT) for SCC of the uterine cervix in patients with pretreatment SCC-Ag levels of <10 ng/mL.236 The role of prophylactic para-aortic radiation, particularly in patients with large tumor size, parametrial involvement, or involved pelvic lymph nodes, must be carefully weighed against the potential toxicities of para-aortic radiation.
METASTASES TO PARA-AORTIC LYMPH NODES
Para-aortic lymph node metastases are frequently combined with distant dissemination but are clinically apparent in only 10% to 20% of patients who have recurrences.
Varia et al.543 reported on GOG Protocol 125, in which 87 patients with biopsy-confirmed para-aortic lymph nodes from cervical cancer were treated with extended-field irradiation (45 Gy in 1.5-Gy fractions) and higher doses to the pelvis (approximately 80 Gy to point A) in combination with 5-FU and cisplatin. The 3-year progression-free survival rate was 33%, and the overall survival rate was 39%. Grades 3 and 4 hematologic toxicity were noted in 13 patients (15%) and chronic proctitis in 3 (3.5%), and 4 patients (4.6%) required surgery for rectal complications.
Nelson et al.390 reported on 104 patients with stages II and III cervical carcinoma who had exploratory laparotomy and para-aortic lymph node biopsies; 12.5% of patients had stage IIA disease, 14.9% had stage IIB disease, and 38.4% had stage III disease had para-aortic lymph node metastases. They were treated with 60 Gy to the para-aortic region. Within 4 years, 50% of these patients had distant metastases, and only 1 out of 13 was alive. There was no significant increase in complications in the patients receiving para-aortic irradiation (39% and 32%). The authors concluded that the main goal of exploratory laparotomy and para-aortic lymph node biopsy is to define the extent of disease.
Lovecchio et al.544 noted a 50% 5-year survival rate in 36 patients with stages IB and IIA cervical carcinoma who had histologically confirmed para-aortic lymph node metastases treated with RT (including 45 Gy to the para-aortic lymph nodes). Fourteen of 31 evaluable patients had pelvic recurrences (12 combined with distant metastases). Unfortunately, the authors did not specify how many patients had para-aortic recurrences, although they reported 4 abdominal failures.
Stryker and Mortel545 determined survival after extended-field treatment of para-aortic lymph node metastasis plus brachytherapy or pelvic boost in 35 patients; 5-year survival was 41.7% for 12 patients with microscopic para-aortic lymph node metastasis and 26.1% for 23 patients with grossly enlarged lymph nodes. Three patients (8.6%) had grade 4 morbidity.
Grigsby et al.546 reviewed 43 patients with cervical cancer and biopsy-proven positive para-aortic lymph nodes treated with external irradiation to the pelvis and para-aortic regions (45 to 50 Gy) combined with brachytherapy. The 5-year overall survival rate was 32%, and the cause-specific survival rate was 49%. Tumor recurrence occurred in 20 patients (3 in the pelvis, 9 in pelvis and distant metastasis, and 8 in distant metastasis only). Severe grade 3 complications occurred in 2 patients (1 had an enterovaginal fistula and the other had radiation myelitis).
Hacker et al.,547 in 437 patients with invasive cervical carcinoma, 222 of whom were treated with radical hysterectomy and lymphadenectomy, identified 34 in whom resection of bulky pelvic or para-aortic lymph nodes was carried out without a complete lymphadenectomy. Thirty-three patients received pelvic external irradiation, and 28 combined pelvic and para-aortic extended-field irradiation (50.4 Gy in 1.8-Gy fractions using a four-field technique). Four cycles of cisplatin were administered to 23 patients. The 5-year survival was 80% in patients with pelvic and common iliac nodes and 48% in those with positive para-aortic lymph nodes. Serious long-term morbidity occurred in 6 patients (18%). Radiation enteritis was observed in 5 patients, leading to small-bowel obstruction necessitating resection.
Grigsby et al.548 evaluated twice-daily external irradiation to the pelvis and para-aortic nodes (1.2 Gy at 4- to 6-hour intervals, 5 days/week) combined with brachytherapy and concurrent chemotherapy in 29 patients with carcinoma of the cervix and positive para-aortic lymph nodes. EBRT doses were 24 to 48 Gy to the whole pelvis, 12 to 36 Gy parametrial boost, and 48 Gy to the para-aortics, with additional boost to a total dose of 54 to 58 Gy to known metastatic para-aortic sites. One or two LDR brachytherapy applications were performed to deliver a total dose of 85 Gy to point A. Cisplatin (75 mg/m2, days 1 and 22) and 5-FU (1,000 mg/m2 per 24 hours for 4 days, days 1 and 22) were given for two or three cycles. Hyperfractionated external radiation therapy was completed in 86% (25 of 29). Radiation therapy toxicity was grade 2 in 34%, grade 3 in 21%, and grade 4 in 28%. An unacceptably high rate (31%, 9 of 29) of grade 4 nonhematologic toxicity was recorded. With a median follow-up of 18.9 months, at 2 years the overall survival rate was 47%, and the probability of locoregional failure was 49%.
Malfetano et al.549 treated 67 patients with carcinoma of the cervix (44 with stage IIB and 23 with stage III disease) with cisplatin (1 mg/kg up to 60 mg weekly) and extended-field radiation therapy, including the para-aortic nodes, and brachytherapy; 75% were alive without evidence of disease with a mean follow-up of 47.5 months.
Chou et al.550 treated 19 patients with isolated para-aortic lymph node metastasis from cervix cancer, 14 of them with chemoradiation, 4 with chemotherapy, and 1 with irradiation alone. Seven of the 14 patients receiving chemoradiation survived.
Goodman et al.551 compiled survival statistics on patients with para-aortic lymph node metastasis and found an average 5-year survival rate of approximately 40% (Table 69.14).
TABLE 69.14 RESULTS OF EXTENDED-FIELD IRRADIATION FOR PARA-AORTIC LYMPH NODE METASTASES

CARCINOMA OF THE CERVICAL STUMP
A supracervical hysterectomy, which removes the uterus and leaves the cervix behind, may be performed for benign conditions of the uterus. The use of subtotal hysterectomy has declined, given the persistent risk of cervical cancer arising in the remnant tissue and the difficulty of managing cancer of the cervical stump.
It is important to divide carcinoma of the cervical stump into true, when the first symptom occurs 3 or more years after subtotal hysterectomy, or coincidental, when the symptoms are noticed before the third postoperative year. This separation is important because the prognosis for true carcinoma of the stump is significantly better than for coincidental lesions, in which carcinoma was probably present when the hysterectomy was performed.302
The natural history and patterns of spread of carcinoma of the cervical stump are similar to those of the cervix in the intact uterus. The diagnostic workup, clinical staging, and basic principles of therapy are the same. Treatment also follows similar paradigms. When surgery is indicated for early stage I tumors, it may be more difficult because of the previous surgical procedures and the presence of adhesions in the pelvis. With radiation therapy, external-beam treatments are identical, although small bowel may be adherent to the superior portion of the cervix due to scar tissue, and using the prone position, as well as having the patient maintain a full bladder for treatment, may aid in attempting to move the small bowel out of the radiated field. Whole-pelvic radiation is recommended, with the superior border set at the level of the bifurcation of the common iliac nodes as determined by a CT simulation, or at the L4–5 interspace for those planned with plain film radiographs. A dose of 45 Gy in 1.8-Gy fractions over 5 weeks is the most commonly recommended dose, with concurrent weekly cisplatin at 40 mg/m2 given for five doses. For brachytherapy, if >2 cm of the endocervical canal remain, it is best to insert a short tandem surrounded by ovoids, a ring, or, if indicated due to vaginal extension or lateral extension, interstitial catheters. For low–dose-rate insertions, as many sources as technically feasible should be inserted in the remaining cervical canal. For HDR brachytherapy, the tandem should extend from the cervical os superiorly to the full extent of the canal. When there is no opportunity to insert any sources in the cervical canal, an interstitial implant to bring the tumor dose to approximately 80 to 90 Gy for brachytherapy after completing a standard dose of 45 Gy to the whole pelvis is recommended while monitoring the radiation dose to the organs at risk (OAR). The use of external-beam radiation alone is not recommended due to the significant mobility of the cervix due to bladder motion, the low dose tolerance of the surrounding small bowel, and the high dose of radiation necessary for cure in patients with bulky disease. Total dose (external and LDR intracavitary brachytherapy) to the upper vaginal mucosa should not exceed 150 Gy, and tolerance doses to small volumes (D2cc) of the bladder (90 to 100 Gy) or rectum and sigmoid (70 to 75 Gy) should be carefully monitored.
The 5-year survival rate for carcinoma of the cervical stump treated with irradiation is similar to that reported for patients with carcinoma of the intact uterus.552,553 The anatomic sites of failure and the incidence of recurrences are similar to those of patients in whom the uterus is intact. Distant metastases also follow the same distribution. In 253 patients with carcinoma of the cervical stump treated at MD Anderson Cancer Center, median survival was 203, 140, and 32 months for stages I, II, and III, respectively.554 Kovalic et al.552 reported on 70 patients with carcinoma of the surgical stump treated with irradiation; 16 also underwent a surgical procedure. The 10-year disease-free survival was 79% for stage IB, 66% for stage IIB, and 39% for stage IIIB disease. The pelvic failure rates were 10%, 9%, and 50%, respectively. Major gastrointestinal complications were noted in 9% of patients and urinary complications in 3.8%. The results are comparable with those seen in patients treated for invasive carcinoma of the cervix with an intact uterus.
Hannoun-Levi et al.555 published results in 77 patients treated for carcinoma of the cervical stump. Treatment consisted of a combination of EBRT and brachytherapy, and, in a few cases, patients underwent surgery or interstitial brachytherapy. Three-year pelvic tumor control was achieved in 59 of 77 patients (76.6%); tumor control probabilities were 77%, 73.7%, and 56% in patients with stage I, II, and III tumors, respectively. Late complications were grade 2 in 5 patients (6.5%); grade 3 in 1 patient (1.3%), and grade 4 in 2 patients (2.6%).
Hellstrom et al.556 published a retrospective study of 145 patients treated for carcinoma of the cervical stump, representing 2.2% of all cervical cancers. Three control cases to each case were matched from the cohort of cases with cervical carcinoma with intact uterus. The dose of irradiation from the intracavitary application given to the stump cancers was lower than for comparable cases with intact uterus. Long-term prognosis for squamous cell carcinoma of the uterine stump was comparable to that of the ordinary cervical carcinomas. Stump adenocarcinomas had a worse prognosis compared with adenocarcinoma of the intact uterus (p < .07) and with squamous cell carcinoma stump (p= .05). The complication rate was higher for stump cancer cases compared with that for cervical cancers with an intact uterus.
Because of the close proximity of the bladder, rectum, and small intestine to the intracavitary sources and to the often higher doses of external-beam irradiation given to the whole pelvis, complications are somewhat more frequent than in carcinoma of the cervix with an intact uterus. Care in brachytherapy treatment planning, including the use of 3D-based treatment planning to minimize dose to the normal tissues, should be considered.
SMALL-CELL CARCINOMA OF THE CERVIX
Small-cell carcinoma of the cervix, like its counterparts in the lung and other anatomic locations, has a high proliferation rate and marked propensity for regional lymph node and distant metastases. Miller et al.557 demonstrated that all small-cell carcinomas of the cervix are aneuploid, compared with only 30% of large-cell nonkeratinizing squamous carcinomas. The incidence of lymphatic vascular space invasion is 80% to 90%, and that of lymph node metastases has been reported to be 40% to 67%.558 These patients must be evaluated in conjunction with a medical oncologist; the workup should include bone marrow aspiration biopsy of the iliac crest and other tests to rule out metastatic spread. Furthermore, the basic therapy should include a combination of cytotoxic agents with pelvic EBRT and intracavitary brachytherapy to doses similar to those used in squamous cell carcinoma, although some patients have been treated with radical surgery. If bleeding is present, prompt institution of radiation therapy with concurrent chemotherapy is necessary. Patients have extremely poor outcomes, with the only reported survivors having had triple-modality therapy of small tumors treated by radical hysterectomy, concurrent chemoRT, and adjuvant chemotherapy. Prophylactic cranial irradiation is not indicated because cervix cancer will most commonly spread first to lung and then to brain.
Patients with small-cell carcinoma of the cervix are treated with the same irradiation techniques as outlined for other histologic varieties of cervical carcinoma in combination with multiagent chemotherapy, including external beam radiation to 45 Gy, followed by nodal boost if PET-positive nodes are identified, followed by brachytherapy. The most frequently prescribed drugs are cisplatin and etoposide (VP-16) every 3 weeks.559 Hoskins et al.560 used a multimodality regimen of four cycles of cisplatin and etoposide with concurrent locoregional RT in 11 women with small-cell carcinoma of the cervix. The 3-year overall and failure-free survival rates were 28%. Four patients were alive in first remission; the remaining 7 died (2 from toxicity, 5 from cancer). Toxicity of therapy was significant, with 70% experiencing severe neutropenia; 40% were admitted to the hospital for emesis control.
Twelve patients with small-cell carcinoma of the cervix were treated with radical hysterectomy (5 received postoperative RT for lymph node metastases and 2 for close margins). With a mean follow-up of 73 months, the disease-free survival rate was 36.4%, compared with 71.6% for patients with non–small-cell carcinoma.558 Four of five patients who received postoperative irradiation died with pelvic recurrence, and 3 also had disseminated metastases. However, only those with small lesions or those who received adjuvant irradiation were cured.
Delaloge et al.561 reported only 2 of 10 patients with neuroendocrine small-cell carcinoma of the cervix surviving at 13 and 53 months after treatment, which included surgery, irradiation, and cisplatin/etoposide combination chemotherapy.
Boruta et al.562 reviewed results in 11 of their and 23 other patients with early-stage neuroendocrine cervical carcinoma identified by a Medline search. Lymphovascular space invasion was present in 21 of 27 patients (78%) (7 unknown), and 15 of 29 (52%) had lymph node metastases. Fifteen patients were treated with cisplatin/etoposide (PE), 7 with vincristine/doxorubicin/cyclophosphamide (VAC), 2 with alternating cycles of VAC and PE, and 10 with other chemotherapy regimens. Twenty women were treated with radiation therapy. The presence of lymph node metastases was a poor prognostic factor (p < .001). PE and VAC chemotherapy were associated with increased survival (p < .01).
ADENOCARCINOMA OF THE CERVIX
Squamous cell carcinoma accounts for 80% of cervical cancers, adenocarcinoma for 15%, and adenosquamous carcinoma for 3% to 5%. SEER data from 1972–2002 suggest that the incidence of cervical adenocarcinoma is rising, but, based on SEER data, cause-specific mortality is not significantly different than that for SCC.563 Adenocarcinoma has been linked to HPV 18, which has a higher rate of nodal and distant metastases than HPV 16.159 Despite a slower regression after irradiation, reflecting cellular kinetics and slow growth, no difference in tumor control or survival has been observed in adenocarcinomas compared with squamous cell carcinomas,564,565 although prognosis is related to clinical stage, volume of disease, and dose of irradiation.566 Because of the predilection for endocervical involvement in adenocarcinoma, a combination of irradiation and conservative hysterectomy has been advocated by some authors,567 although results are comparable with those obtained with irradiation alone.564 Given a lower toxicity profile with chemoRT, this is preferable to a planned course of RT alone followed by hysterectomy upfront. Patients who have residual disease after chemoRT may be candidates for adjuvant hysterectomy.
Grigsby et al.564 found no difference in 5-year DFS in patients with adenocarcinoma of the cervix (AC) compared with SCC treated with RT alone or combined with surgery. In contrast, Eifel et al.568 reported that overall 5-year survival rates for patients with SCC and AC were 81% and 72%, respectively (p < .01). Patients with AC had a maximum cervical diameter of <4 cm more often than did those with SCC (53% vs. 47%). For 903 patients with tumors of ≥4 cm, 73% of those with SCC survived ≥5 years, compared with only 59% of those with AC (p < .01). Although there was no significant difference in the rate of pelvic disease recurrence for patients with AC or SCC tumors of ≥4 cm (17% vs. 13%; p = .16), the rate of distant metastases was greater for patients with AC (37% vs. 21%; p < .01). For patients with tumors of ≥4 cm, prognosis was strongly correlated with tumor size (p < .01) and lymphangiogram findings (p < .01) but not with age (p = .58) or tumor morphology (exophytic vs. endocervical; p = .33); a trend toward better survival in 165 patients who underwent adjuvant hysterectomy (78% vs. 71%) was not significant (p = .09). Multivariate analysis confirmed a highly significant independent association between histology and survival; patients with tumors ≥4 cm in diameter that were AC had an estimated risk of death 1.9 times that of patients with SCC (p < .01).
Nakano et al.569 studied 58 patients with adenocarcinoma of the cervix treated with LDR or HDR brachytherapy and external pelvic irradiation. The 10-year survival rates for stages I, II, III, and IV were 85.7%, 60%, 27.6%, and 9.1%, respectively. The local tumor control rate with HDR treatment was 45.5%, significantly lower than with LDR (85.7%) or mixed–dose-rate treatments (72.7%). Kilgore et al.,565 in a study of 162 patients with adenocarcinoma compared with matched patients with squamous cell carcinoma, found that clinical stage and lesion size were the most important prognostic factors. In patients with stage I tumors, no significant difference in survival was found when they were treated with radical surgery, irradiation alone, or irradiation combined with hysterectomy. In contrast, Kjorstad et al.570 reported a worse 5-year survival rate in 102 patients with adenocarcinoma (51%) compared with that of 1,900 patients with squamous cell or other differentiated carcinomas (68%).
A Cochrane database review571 found only one randomized trial, the Landoni study, in which a small subgroup analysis that was not powered to detect a difference showed an apparent improvement with surgery, although the majority of patients required adjuvant RT, rather than chemoradiation, which increases the rates of toxicity. Adenocarcinoma predicts worse OS on multivariate analysis (HR = 2.68, 95% CI = 1.9 to 3.8). Primary chemoradiation is considered the standard regimen for patients with locally advanced cervical adenocarcinoma.
Huang et al.572 reported on 318 stage IB to IIB postoperative cervical cancer patients, 202 (63.5%) with SCC and 116 (36.5%) with AC/atypical squamous cells (ASC), treated by radical hysterectomy and adjuvant RT/concurrent chemo-RT (CCRT). The 5-year relapse-free survival rates for SCC and AC/ASC patients were 83.4% and 66.5%, respectively (p < 0 .001). Distant metastasis was the major failure pattern in both groups. After multivariate analysis, prognostic factors for local recurrence included younger age, parametrial invasion, AC/ASC histology, and positive resection margin; for distant recurrence they included parametrial invasion, lymph node metastasis, and AC/ASC histology. Compared with SCC patients, those with AC/ASC had higher local relapse rates for the intermediate-risk group but a higher distant metastasis rate for the high-risk group. Postoperative CCRT tended to improve survival for intermediate-risk but not for high-risk AC/ASC patients.
TREATMENT OF ELDERLY PATIENTS
Oguchi et al.573 reported on 23 patients 90 years of age or older treated for cervix carcinoma. Definitive radiation therapy was completed in 13 of the patients, and local tumor control at 6 months was attained in 9 patients. Palliative RT was completed in 7 of 11, and palliation was observed in 9 patients (81%). Seven patients were alive for 15 to 67 months. Fourteen patients died because of intercurrent disease or senility associated with active cancer and 2 because of senility without evidence of cancer. The 2-year overall and relapse-free survival rates were 30% and 21%, respectively.
CARCINOMA OF THE CERVIX AND PREGNANCY
The concurrent presence of carcinoma in situ or invasive carcinoma of the uterine cervix and pregnancy, although rare, poses a therapeutic dilemma to gynecologic and radiation oncologists. Reported incidence is approximately 1 to 10 per 10,000 pregnancies.574 In the United States, the incidence has decreased. Norstrom et al.,575 in Sweden, found that cervical cancer was diagnosed in 33 women in association with pregnancy (incidence, 11.1 cases per 100,000 deliveries and 7.5 per 100,000 pregnancies). Abnormal bleeding was the symptom that led to diagnosis in 54.5% of the women; 45.5% were asymptomatic but had an abnormal cervicovaginal cytologic test result (39.4%) or abnormal vaginal examination (6.1%) in association with pregnancy. During the follow-up, 1 of 12 women with cervix cancer in the first trimester, 4 of 12 in the third trimester, and 2 of 9 postpartum died of the disease. Primary surgery was used more frequently than radiation therapy.
For carcinoma in situ, if the pregnancy is allowed to reach full term, confirmation of the diagnosis by colposcopy and conservative management with monthly Pap smears constitutes the best approach. Conization has frequently been performed. Punch biopsies can be obtained, but the diagnostic accuracy is less reliable. As many as 50% of the patients have residual carcinoma in situ after delivery.
In patients with invasive carcinoma, the lesion is usually clinically apparent. Multiple punch biopsies are adequate to confirm the diagnosis. Management is individualized based on tumor size and stage, patient age, and desires of the patient (or couple) regarding the pregnancy. The majority of patients with cervical cancer diagnosed during pregnancy (approximately 75%) have stage I tumors.574,576,577
Women with tumors diagnosed early in pregnancy are often recommended to abort the fetus. Because there is a greater need to institute therapy as soon as possible, the accepted method of treatment in patients in the first 6 months of pregnancy is to carry out definitive surgery or radiation therapy, as indicated by the stage of the disease, with resultant loss of the fetus. An abortifacient may be administered before initiating radiation to ensure fetal demise and delivery of the placenta prior to initiation of treatment. The whole pelvis is irradiated (40 to 45 Gy in 4 to 5 weeks). However, in one series of 45 patients, 27% did not abort spontaneously and surgical evacuation was required;578alternatively, misoprostol may be administered as an alternative to surgical evacuation after failed spontaneous abortion.579 After this dose of radiation, careful evacuation of the uterus and LDR (or equivalent-dose HDR) brachytherapy may be performed under general anesthesia. If a radical hysterectomy is performed and positive pelvic lymph nodes are found, the usual postoperative irradiation, including external beam with or without intracavitary insertion, should be carried out.
If the woman refuses abortion, serial MRI scans at 2- to 3-month intervals to ensure no growth or spread to lymph nodes is recommended. Neoadjuvant chemotherapy may be considered in selected patients.580 Sorosky et al.581reported on eight pregnant women with stage I carcinoma of the cervix who had declined immediate therapy and followed until the late third trimester; a cesarean section–radical hysterectomy was performed, with delay in therapy ranging from 3 to 40 weeks during the pregnancies. There was no clinical progression of the disease with follow-up of 33 months.
When patients are diagnosed in mid-pregnancy (second trimester), consideration to keeping the pregnancy and treating with chemotherapy is given. A French series reported five cases between 2002 and 2009.582 Three patients received neoadjuvant chemotherapy; 1 patient died of cancer. A Chinese report described treatment of two patients with neoadjuvant paclitaxel and cisplatin as feasible.583
Occasionally in late pregnancy (final trimester), if tumors are small and an MRI confirms no lymph node involvement, definitive therapy is postponed until after imminent delivery. In a review of the literature intentional treatment delay was associated with a recurrence rate of 4%.362 Greer et al.584 noted that in 600 infants without congenital abnormalities, when stage IB cervical carcinoma was diagnosed during pregnancy and fetal survival was chosen, the neonatal mortality rate decreased from 30% when the fetus was delivered at 26 to 27 weeks to 2.7% when the fetus was allowed to mature to 34 to 35 weeks. In the third trimester of pregnancy, when the fetus may be salvaged, some gynecologic oncologists prefer a postpartum cesarean section, combined with a radical hysterectomy and lymphadenectomy followed by radiation of high risk features are present. However, some authors report that vaginal delivery has no detrimental effect on the prognosis.585
Patients who require high doses of pelvic irradiation should be counseled regarding the permanent loss of reproductive capability, not only because of ovarian ablation (which happens with doses of 8 Gy or higher), but as a consequence of radiation effects in the uterus.586,587
Survival is the same regardless of the trimester of the pregnancy in which definitive treatment is instituted. Creasman et al.585 reported on 48 patients treated by irradiation, 45 by irradiation followed by surgery, and 5 with radical hysterectomy alone. The survival was comparable with that of nonpregnant patients for similar stages. The survival rate for patients with stage I disease was comparable whether vaginal delivery was allowed or a cesarean section was performed (approximately 85% in stage I and 50% to 64% in stage II). In addition, the percentage of infants surviving (>80%) was the same in both groups.
Sood et al.587 performed a retrospective analysis of 26 women with cervical carcinoma diagnosed during pregnancy and treated primarily with radiation therapy (mean dose, 46.7 Gy) and LDR intracavitary radiation (mean dose, 56.5 Gy to point A). These cases were matched with 26 nonpregnant control patients based on age, histology, stage, treatment, and year of treatment. There were no statistically significant differences in recurrence rates or survival between the pregnant group and the control patients. Short-term toxicity was comparable in pregnant and nonpregnant patients. Long-term complication rates were 12% in pregnant patients and 27% in control patients, but this difference was not statistically significant. Most complications were likely related to radiation techniques (particularly the predominance of 60Co).
Sood et al.588 compared the prognosis of 56 women who had cervical cancer diagnosed during pregnancy and 27 who were diagnosed within 6 months after delivery. Control patients (cervical cancer diagnosed at least 5 years since last delivery) were matched one-to-one with cases based on age, histology, stage, treatment, and time of treatment. Among the postpartum women, 11 were treated with radical hysterectomies and 14 with radiation therapy, and 2 with stage IA1 disease were treated with vaginal hysterectomies. One of 7 patients who had cesarean sections had a local and distant recurrence. In contrast, 10 of 17 (59%) patients who delivered vaginally had recurrences (p = .04). In multivariate analysis, vaginal delivery was the most significant predictor of recurrence, followed by high tumor stage. Survival for patients diagnosed in the postpartum period was significantly worse than for control patients and for those diagnosed during pregnancy. The authors concluded that pregnant women with cervical cancer should be delivered by cesarean section.
Jones et al.589 published a survey by the American College of Surgeons that evaluated management of invasive cervical carcinoma in 161 pregnant patients; 86 were treated with surgery alone, 30 with radiation therapy alone, and 45 with a combination of the two modalities. Approximately one-third of patients were diagnosed in each trimester. The 5-year survival was 94.6% for patients diagnosed in the first trimester, 76.9% for the second, and 68.9% for the third. The prognosis of patients with invasive carcinoma of the cervix associated with pregnancy was similar to that of nonpregnant patients. There was no significant difference in 5-year survival between the patients delivered by cesarean section and by normal vaginal delivery.
Senekjian et al.590 reported no difference in survival or patterns of failure in 24 women who were pregnant at the time of diagnosis of clear-cell adenocarcinoma of the cervix and vagina compared with 408 who had never been pregnant. The 5- and 10-year actuarial survival rates were 86% and 68% for the pregnant patients and 87% and 79% for the patients who had not been pregnant, respectively.
The practice popularized 30 years ago of administering a “restraining dose of radium” and deferring definitive radiation therapy until delivery is carried out should be strongly rejected. Strauss591 reported 2 of 11 infants being born with microcephaly in addition to other complications such as alopecia, facial deformity, eye damage, and chromosomal abnormalities after this procedure.
BRACHYTHERAPY
After the discovery of radium in 1898 by the Curies, publications followed describing use of the first glass radium capsule in 1904 and a metal brachytherapy applicator for cervical cancer in 1905.592 The incorporation of brachytherapy after external-beam treatment arose from the recognition that tumor control probability correlated with radiation dose and cancer volume.593 Evidence confirms that brachytherapy as used for dose escalation after external-beam treatment for cervical cancer significantly improves survival.525,594–597 Therefore, brachytherapy is a standard part of the treatment of locally advanced (stages IB2 to IVA) cervical cancer after external beam radiation; brachytherapy alone may be used as primary treatment for selected cases with early-stage (stages IA to IB1) cervical cancer.598 The increasing complexity of brachytherapy administration, including the use of complex imaging, mandates implementation of careful quality assurance measures and a culture of open communication to ensure safe practices in the clinic.598
Dose Rate
The ICRU in its Report 38599 defines brachytherapy dose rate as follows: LDR, 0.4 to 2 Gy/hr; medium dose rate (MDR) or PDR, 2 to 12 Gy/hr; and HDR, >12 Gy/hr. For LDR, the most commonly used isotope is 137Cs, and for PDR and HDR it is 192Ir. The use of HDR has significantly increased in the United States, from 13% in the 1996–1999 Quality Research in Radiation Oncology (formerly known as the Patterns of Care) survey to 62% in the 2007–2009 survey.369,600 In other U.S., European, and Japanese surveys of gynecologic brachytherapy practitioners, approximately 85% state that they use HDR brachytherapy,601–602,603 whereas in Canada the reported use is 68% HDR, 10% PDR, and 23% LDR.604 The dose delivered per fraction for HDR and proportion of dose delivered with external beam versus brachytherapy vary substantially in different centers around the world.602
Most institutions in the United States have either LDR or HDR brachytherapy available. Larger centers may have LDR, PDR, and/or HDR. Overall, outcomes are similar regardless of dose rate, with the caveat that for HDR, 3D imaging should be incorporated to ensure coverage of the tumor, particularly for large-volume disease.605 Several studies demonstrate worse survival for stage IIIB cervical cancer treated with HDR due to initiating the treatment early in the course of external beam therapy,606 when the tumor mass was >4 cm and therefore a prescription to point A did not cover the tumor volume, or by not using 3D planning to cover large residual disease.607
TABLE 69.15 MEAN VALUES OF THE NUMBER OF FRACTIONS, DOSE/FRACTION TO POINT A (FOR HIGH DOSE RATE) AND TREATMENT TIME, DOSE RATE FOR LOW DOSE RATE (WITH STANDARD ERRORS) AND THE RATIO OF TOTAL DOSES

FIGURE 69.20. Relationship between number of high–dose-rate (HDR) fractions and late normal tissue effects. Solid lines show the increase in normal-tissue late effects as the number of HDR fractions used to treat cervical cancer decreases. Dotted lines indicate the maximum level of late damage calculated for conventional low–dose-rate brachytherapy of 70 Gy at 0.5 Gy/hour for 140 hours and an arbitrary level 5% above this. The intersection of the solid lines with the dotted lines indicates the number of HDR fractions needed to give equal late normal-tissue effects. In this model, the dose to late-responding tissues should be kept at 83% of the tumor dose when treating with four to six HDR fractions. (Modified from Fowler JF. The radiobiology of brachytherapy. In: Martinez AA, Orton CG, Mould RF, eds. Brachytherapy HDR and LDR. Leersum, Netherlands: Nucletron International BV, 1990;121–137; with permission from Elsevier.)

Biology of High–Dose-Rate Brachytherapy for Cervical Carcinoma: Equivalent Dose in 2 Gy and Biologically Effective Dose
To achieve tumor control using HDR equivalent to that with LDR brachytherapy, attention to the dose/fractionation schedule and to normal tissue doses is mandatory.608,609–610 The linear-quadratic (LQ) model provides calculation estimates of biologically equivalent dose taking into account dose rate, dose per fraction, and overall treatment time.611,612 For comparison of LDR to HDR, the LQ model doses are normalized to an equivalent dose in 2 Gy (EQD2).613 The α/β ratio is a critical component of the LQ model.614 For cervix cancer, an α/β ratio of 10 Gy is used for tumor and an α/β of 3 Gy is used for normal tissues,615 which may have inherent inaccuracies in generalizability,616 although an α/β ratio of 3 Gy for the rectum has been correlated with late rectal complications.617,618–619
Spreadsheets to assist with HDR EQD2 dose conversions are available from the American Brachytherapy Society (www.americanbrachytherapy.org/guidelines). The values derived are not actual doses but biologically effective ones that take into consideration dose rate and effect of fraction size.620
Although use of a detailed EQD2 conversion is preferred, an approximation of these values was proposed by Orton et al.,610 with an LDR-to-HDR reduction factor of 0.54 to 0.6 (Table 69.15), and by Patel et al.,607 with a similar correction factor of 0.58. These conversion factors are valid when three to five HDR fractions are used, but with a higher number of fractions (six to eight), the conversion factor is closer to 0.75. Therefore, use of the EQD2 worksheet is recommended for consistency.
The importance of adopting biologically based equivalent doses when switching from LDR to HDR brachytherapy is exemplified in a report by Newman621 on 115 patients treated with external irradiation (40 to 50 Gy) and manual afterloading cesium sources delivering 60 Gy to point A with a dose rate of 0.75 Gy/hour, or a Selectron device with 40-mCi sources, which delivered from 0.75 to 1 Gy/hour to point A. Because of the increased dose rate, the total intracavitary dose was reduced by 20%. Grade 3 genitourinary and gastrointestinal complications were observed in 3 of 87 patients (3.4%) treated with LDR, in contrast to 30 of 132 patients (22.7%) treated with the Selectron HDR sources. No significant differences in local tumor control and survival were found.
Figure 69.20 illustrates the late normal-tissue effect, which is proportional to log cell kill, and the relationship to the number of HDR treatment fractions.622 Each solid curve is calculated assuming the same log cell kill. Late damage rises sharply as the number of HDR fractions is decreased. When these curves are above the dashed lines that represent the maximum late effect of 70 Gy of LDR brachytherapy given at 0.5 Gy/hour, the risk of late complications increases. Displacing the bladder and rectum away from the HDR sources for the short duration of therapy may offset the radiobiologic disadvantage of using a few brachytherapy fractions.610
Brachytherapy Process: Preparation and Timing
Standard procedures for pretreatment evaluation, imaging, anesthesia use, and treatment duration should follow accepted general principles in the guidelines published by the American Brachytherapy Society in 2012.598,623,624Planning the course of brachytherapy should begin at the time of initial presentation. An examination at diagnosis assessing disease extension and tumor size should be recorded. Periodic examinations during external beam treatment should be performed to monitor response. For patients receiving concurrent chemoradiation, rapid shrinkage should be expected because treatment may regress the tumor to 70% to 80% of the pretherapeutic volume. Therefore, a clinical gynecologic examination at the time of brachytherapy is also important.625,626 Inserting radiopaque marker seeds (“fiducials”) at the time of diagnosis that mark the inferior, lateral, and superior extension of a vaginal tumor may aid in identifying regions requiring dose escalation with brachytherapy even after complete regression during external-beam treatment.
Treatment schedules integrating external-beam irradiation and brachytherapy were initially designed with regard to the disease stage and volume.627 For LDR, insertion of the applicator may be done after all external beam finishes, with the caveat that one or two insertions may be required, approximately 1 week apart. All treatment, including external-beam treatment and brachytherapy, should finish 8 weeks from the initiation of radiation.184
The optimal time–dose–fractionation scheme and the technique for remote-control afterloading intracavitary brachytherapy for cervical cancer have yet to be established through systematic clinical trials. For HDR or PDR brachytherapy, the applicator insertion and treatment may commence after external-beam treatment finishes, to ensure optimal geometry with normal tissues far from the applicator. Alternatively, the physician may choose to insert the applicator for treatment as early as during the second week of external radiation if the tumor is small enough, to minimize total treatment time. However, brachytherapy and external-beam treatments are not given on the same day.
FIGURE 69.21. Ultrasound used during intrauterine tandem insertion can ensure proper placement into the intrauterine canal and shorten overall procedure time. A: Ultrasound depicts that the tandem is placed in the posterior myometrium. B: Ultrasound directs the tandem into the intrauterine canal.

Applicator Selection
The most frequently used applicator for locally advanced cervical cancer patients worldwide is the tandem and ovoid applicator,602 which provides radiation dose covering the cervix, uterus, inner parametria, and approximately 1 to 2 cm of the upper vagina. The tandem and ring applicator has a slightly narrower dose distribution, but with HDR the dose distribution may be optimized and mimics that obtained with the tandem and ovoid applicator. For patients with a very narrow vagina due to stricturing, a tandem and cylinder applicator may be the only option available. However, this applicator provides insufficient dosing to the parametrial tissue and should be used with caution. For patients with large tumors with residual bulky disease after external-beam radiation or those with vaginal extension, fistulae, or pelvic sidewall invasion, a combination of tandem/ring or ovoid with interstitial applicator or tandem/interstitial applicator alone may be inserted.
Applicator Insertion
General guidelines from the American Brachytherapy Society should be followed.598 The patient receives appropriate anesthesia for pain management. In the lithotomy position, the perineum is sterilely prepped and draped. The Foley catheter is inserted into the bladder. The catheter is clamped and the bladder filled with 150 to 200 cc of saline if ultrasound (US) is used. US with a transabdominal probe can significantly speed up the insertion process and assist with accurate placement because the uterine canal is often clearly visible after instilling fluid in the bladder (Fig. 69.21). Transabdominal US can measure uterine width and height in patients who do not have large fibroids or tumor volumes that greatly distort uterine configuration.628 US does not define the target volume as clearly as MRI but may be used to assist with 3D-based planning when CT or MRI is unavailable.629 Transrectal US may assist with interstitial brachytherapy when other imaging modalities are not available. For interstitial insertions, CT or MRI may be used during the insertion process iteratively using real-time guidance to ensure proper tumor coverage and no inadvertent insertions into the rectum or bladder.630 After tandem and ovoid or tandem and ring applicator placement, vaginal packing covered with lubricant gel is placed in the vagina to separate the bladder and rectum, which also helps to maintain applicator position.
Applicator Position
An experienced brachytherapist has greater familiarity with applicator insertion and evaluation.368 Applicator position is a critical determinant of dose specification600,631 and pelvic control.57,632 As Fletcher533 emphasized, conditions for an adequate intracavitary insertion include the following:
1. The geometry of the insertion must prevent underdosing around the cervix.
2. Sufficient dose must be delivered to the paracervical areas.
3. Vaginal mucosal, bladder, and rectal tolerance doses must be respected.
For LDR Fletcher-Suit applicators, Potish et al.633 used linear least-squares regression to show that although there was a moderately good correlation between the milligram-hours and dose to point A, it was markedly affected by the position of the colpostats and the tandem. A review of plain films of 808 LDR intracavitary applications in 396 cervical cancer patients treated at MD Anderson634 quantified acceptable implant geometry. The median distance from the tandem to the sacrum was 4 cm, or one-third the distance from the pubis to the sacrum. The distance between the vaginal ovoids and cervical marker seeds was 7 mm, and the median distance between the tandem and the posterior edge of the ovoids was 50% of the ovoid length. In 92% of insertions, vaginal packing was posterior to or within 5 mm of a line that passed through the posterior edge of the ovoids, parallel to the tandem. The median doses to point A and rectal, bladder, and vaginal surface reference points were 87, 68, 70, and 125 Gy, respectively. Analysis of the LDR and HDR brachytherapy positions for 103 patients enrolled on RTOG trials 0116 and 0128 found that patients with unacceptable symmetry of ovoids to the tandem had a significantly higher risk of LR than patients in the acceptable group (HR = 2.67; 95% CI = 1.11 to 6.45; p = .03).57 Patients with displacement of ovoids in relation to the cervical os had a significantly increased risk of LR (HR = 2.50; 95% CI = 1.05 to 5.93; p = .04) and a lower DFS rate (HR = 2.28; 95% CI = 1.18 to 4.41; p = .01). Inappropriate placement of packing resulted in a lower DFS rate (HR = 2.06; 95% CI = 1.08Y3.92; p = .03).
FIGURE 69.22. Definition of point A shown on a tandem and ovoid applicator based on the American Brachytherapy Society 2012 Guidelines for Cervical Cancer.

Imaging After Insertion
For LDR brachytherapy, active sources may be inserted after the films have been reviewed and the position of the applicators judged to be satisfactory.57,634 Placing a small amount of contrast into the bladder and rectum before CT or plain film may clarify the location of these structures.
When a CT scan is obtained after applicator insertion, it also verifies proper placement (no perforation) and analyzes cervix and normal-tissue location in relationship to brachytherapy dose distribution.601 CT provides a reasonable estimate of the location of the uterus and cervix. The CT contours of the cervix overestimate the tumor contours compared to an MRI, although the additional width contoured on a CT may not be of detriment to the patient because cervical cancer tends to spread laterally along the parametrial tissues.635 CT depicts changes in the OAR related to tumor shrinkage, organ motion, and the location of the brachytherapy applicator in relation to the uterus. However, separating the OAR, such as the sigmoid or the small bowel from portions of the uterus or cervix, may be difficult on CT, given the lack of contrast. OAR dosimetry based on CT is similar to that based on MRI when optimized similarly.636 CT may not provide sufficient detail of the tumor if selected dose escalation is required, such as in cases with large residual tumors. In the vast majority of cases, however, it should suffice.637–638,639 The uterus and cervix cannot be distinguished as separate structures on a CT, whereas they can on an MRI. Therefore, CT-based contouring guidelines recommend delineating the entire cervix and uterus.635
In a multicenter study, MR imaging was significantly better than CT for tumor visualization and detection of parametrial involvement.66 Other advantages of MR include multiplanar capability and excellent soft-tissue contrast resolution. The strength of a magnet in an MR scanner is expressed by a unit of measurement referred to as a tesla (T). Higher field strength produces a better overall signal-to-noise ratio and more accurate imaging.
Regardless of whether a plain film radiograph, CT, or MR is obtained after insertion to aid treatment planning, in order to properly visualize the apparatus, radiopaque markers should be inserted to identify source position to aid with dosimetric planning. For MRI, identifying the applicator using either a radiopaque marker inserted into the applicator, or, for interstitial cases undergoing MR, a special 3-T MR sequence may be used to create artifact that allows the tip of the needle to appear as a balloon on the sagittal image and as a cross on the axial images.640
Dose Specification
Point A
Several methods for specifying dose in brachytherapy evolved over the twentieth century.
Due to the limited availability of 3D imaging worldwide and a 100-year history of plain film radiography, the majority of institutions internationally use point-based dosimetry based on the ICRU 38 nomenclature,599 defining point A rather than prescribing dose to a tumor volume.601 Other institutions use a system of milligram-hours, whereas others consider the volume of the region of interest. Based on the general principles guidelines for cervical cancer brachytherapy published by the American Brachytherapy Society and the ICRU report, the point A definition was updated in 2012.598 To determine point A, connect a line through the center of each ovoid or the lateralmost dwell position in the ring; extend this line superiorly along the radius of the ovoids (or ring), and then move an additional 2 cm superior along the tandem. From this point, extend out 2 cm on each side laterally on a line perpendicular to the tandem (Fig. 69.22). For tandem and cylinders, begin at the flange or cervical marking seed and move 2 cm superiorly along the tandem and then 2 cm laterally.
Three-dimensional image-based brachytherapy treatment planning precisely defines the tumor and aids with the precision of radiation dose delivery, which may reduce the dose to the normal tissues and reduce toxicity.641 In the era of increasing use of HDR brachytherapy, proper applicator placement and precise estimation of the location of the normal tissue is critical. When 3D imaging is available, point-based radiographic dosimetry has limited utility for the dose adaptation required for HDR brachytherapy because point A may overestimate or underestimate the tumor dose based on 3D imaging.642 Kim et al.643 found that dose to point A was significantly lower than the D90 for HR-CTV calculated using 3D image-based optimization. With imaging, one may visualize the tumor volume and conform dose to the volume which may result in the dose to point A being lower than the prescription 100% isodose line (covering a smaller tumor volume) or a higher-than-prescription dose to point A due to a large tumor volume extending beyond the boundaries of point A. The tumor coverage relies on tumor volume at the time of brachytherapy, with larger tumors requiring greater optimization to be adequately covered by the prescribed isodose line.642,644,645 The dose to point A should be reported to ensure consistency in terminology between centers.646
Accurate delineation of the tumor and OAR is critical for precise treatment planning (Fig. 69.23). Due to the rapid fall-off of dose, imprecise contouring can dramatically change dosing to normal-tissue structures. Formal contouring education programs reduce the variability of interobserver contours.647
Computed Tomography Imaging for Brachytherapy Contouring
A CT scan can define a CTV with the lateral borders of the cervix and any parametrial extension defined based on suspicious regions seen on the scan. CT-contouring guidelines should be followed.635 Uterosacral ligaments may be visualized on CT, and, if detected, they should be included in the CTV contours. No GTV may be identified on CT. The superior border of the cervix is not defined, but instead the entire tandem length is planned and the top dwell is optimized off the sigmoid to reduce bowel dose.
FIGURE 69.23. Tandem and ovoid implant showing the use of three-dimensional (magnetic resonance or computed tomography) imaging to adequately cover the high-risk clinical target volume (HR-CTV) while minimizing dose to the organs at risk (OAR)—the rectum, sigmoid, and bladder. Dose–volume histograms record the D2cc limits to the OAR. The dose at point A is recorded but varies between patients based on optimization of the HR-CTV and OAR.

Magnetic Resonance Imaging for Brachytherapy Contouring
For MR-based contouring, the Groupe Europeen Curietherapy-European Society for Therapeutic Radiation Oncology (GEC-ESTRO) guidelines625,626 delineate volumes for MR. The recommended volumes include the GTV, including all T2-bright areas of enhancement; the HR-CTV, which is the entire cervix, any regions of high to intermediate signal intensity in the parametria, uterus, or vagina, and any residual disease detected on clinical examination at the time of brachytherapy; and, the intermediate-risk clinical target volume (IR-CTV), which subtracts out the OARs but includes the tumor extension at the time of diagnosis, adding 1 cm to the HR-CTV volume. The IR-CTV defines regions with potential microscopic seeding of tumor cells (Fig. 69.24).648 Lang et al.,649 in a multicenter study, confirmed the feasibility of these recommendations, with total doses to point A from both BT and EBRT ranging from 85 to 91 Gy and to CTV from 69 to 73 cGy. Doses to organs at risk were comparable to those obtained with standard dosimetric methods, although they were more accurately determined with dose–volume histograms.
Dose–Volume Histogram Reporting
With MR-planned brachytherapy, the most common dose–volume parameters reported for target structures of the entire cervix and any residual disease at the time of brachytherapy, the HR-CTV, are D90, defined as the dose received by at least 90% of the target volume, D100, and V100, based on the GEC-ESTRO recommendations.650,651 The cumulative D90 equals the sum of D90 values from the individual fractions plus the dose from a homogeneous 3D conformal external-beam treatment. D100, the minimum target dose, may be more sensitive to inaccuracies in contouring and dose calculation. V100 assesses dose coverage of the whole target volume and is 100% when the entire target is covered by the prescribed dose. V150 and V200 are often reported in interstitial brachytherapy. One may report these for CT, although the dimensions of the target will differ significantly from the absolute dimensions on MR, and the CT contour of the HR-CTV will include the cervix, residual areas in the parametria or uterosacral ligaments, and part of the uterus because the superior border of the cervix is not visible.635 CT contours are more accurate if an MRI can be performed immediately before or with the first fraction of brachytherapy.652 In both CT and MRI, prescribed dose is based on the physician’s directive of the dose intended for the target volume, that is, the volume covered by the 100% isodose line, and point A should be recorded. Several institutions have validated the use of these guidelines with HDR, LDR, or PDR brachytherapy.645,652,653,654,655,656
With 3D imaging, one may define the surrounding normal tissue structures as the OAR, including the rectum, sigmoid, and bladder. With 2D imaging, the ICRU 38 report requires only reporting point estimates for the rectum and bladder because the sigmoid cannot easily be visualized.599 However, the ICRU bladder point may underestimate maximum doses to the OAR, in particular for the bladder657,658; it is less likely that rectal doses will be incorrectly estimated. Numerous publications correlate the ICRU point dose and the probability of late complications for bladder and rectum.659,660 Nevertheless, DVH metrics may provide a more reliable predictor of long-term complications.661 In a review of 50 patients treated with LDR or HDR brachytherapy who then had a CT for treatment planning, the closest point of the sigmoid was related to sigmoid dose but varied in proximity to the tandem up to 40% between fractions, with a median distance of 1.7 cm. No sigmoid toxicity was noted after a median follow-up of 31 months.
FIGURE 69.24. Diagrammatic representation of gross tumor volume (GTV) and clinical target volume (CTV) for three-dimensional treatment planning in carcinoma of uterine cervix. Coronal (A,C) and transverse (B,D) sections for limited (A,B) and advanced (C,D) disease (gray zones in left parametrium). (From Potter R, Haie-Meder C, Limbergen EV, et al. Recommendations from gynaecological (GYN) GEC ESTRO working group (II): Concepts and terms in 3D image-based treatment planning in cervix brachytherapy-3D dose volume parameters and aspects of 3D image-based anatomy, radiation physics, radiobiology. Radiother Oncol 2006;78:67–77; with permission from Elsevier.)

Kapp et al.662 analyzed 720 192Ir HDR applications in 331 patients with gynecologic tumors to evaluate the dose to normal tissues. CT-based dosimetry showed that the maximum doses to bladder and rectum were generally higher than those obtained from orthogonal films, with an average ratio of 1.44 for the bladder neck, 2.42 for the bladder base, and 1.37 for the rectum. The ratio of bladder-base dose to bladder-neck dose was 1.5 for intracervical and 1.46 for intravaginal applications. If conventional methods are used for dosimetry, the authors recommended that doses to the bladder base should be routinely calculated because single-point measurements at the bladder neck seriously underestimate the dose to the bladder. In addition, the rectal dose should be determined at several points over the length of the implant because of the wide range of anatomic variations.
Eich et al.,663 in 11 applications of HDR brachytherapy for cervical carcinoma, calculated doses to ICRU points on orthogonal radiographs, and the doses at rectum reference points were compared with in vivo measurements. The in vivo measurements were 1.5 Gy below the doses determined for the ICRU rectum reference point (4.05 ± 0.68 vs. 6.11 ± 1.63 Gy). The advantage of in vivo dosimetry is the possibility to determine rectal dose during radiation. The advantages of computer-aided planning at ICRU reference points are that calculations are available before radiation and they can be taken into account for treatment planning.
Pelloski et al.664 compared CT-based volumetric calculations and ICRU reference point radiation doses in 60 patients with cervix cancer treated with LDR brachytherapy. Of 118 insertions performed, 93 were evaluated, and the minimal doses delivered to the 2 or 3 cm of bladder or rectum (DBV2 and DRV2, respectively) were determined on a dose–volume histogram (DVH). They concluded that the ICRU dose was a reasonable surrogate for the DRV2 but not for the DBV2. Furthermore, these calculations may not be applicable to other treatment guidelines or intracavitary applicators.
Patil et al.665 found significant correlations between ICRU point doses to the bladder and rectum and volumetric doses, particularly the D2cc. However, there was significant variability, and they concluded that 3D imaging is essential to properly assess doses to the OAR.
Both CT- and MR-based OAR dosimetry report similar cumulative DVH parameters, including D2cc and D0.1cc. The D2cc is the minimum dose received by the most exposed 2-cm3 volume of the analyzed organ. For CT-based brachytherapy, contrast placed in the OAR may cause some artifact, resulting in some variation in contouring the wall of the organ. MR-based brachytherapy relies less on contrast because the organ wall may be more clearly visualized. Wachter-Gerstner et al.666 analyzed the correlation between dose–volume histograms for bladder and rectum and found that D2cc served as a good estimate for doses to the organ wall, whereas D5cc was less reliable because is changed based on filling status. Rectal wall thickness did not significantly affect D2cc.667
Based on CT-dosimetry, Koom et al.668 showed that more-severe rectal side effects (endoscopy score >2) occurred in patients with a higher D2cc. Seventy-one patients with FIGO stages IB to IIIB uterine cervical cancer had CT-based HDR intracavitary brachytherapy. The mean values of the DVH parameters and ICRU rectal point [α/β = 3] were significantly greater in patients with a score of >2 than in those with a score of <2 at 12 months after brachytherapy (ICRU, 71 Gy vs. 66 Gy [p = .02]; D0.1cc, 93 vs. 85 Gy [p = .04]; D1cc, 80 vs. 73 Gy [p = .02]; D2cc, 75 vs. 69 Gy [p = .02]). The probability of a score of >2 was significantly correlated with the DVH parameters and ICRU rectal point (ICRU, p = .03; D0.1cc, p = .05; D1cc, p = .02; D2cc, p = .02).
For MR-based dosimetry, Georg et al.661 tested the predictive value of dose–volume parameters for late effects of the rectum, sigmoid colon, and bladder using the D2cc, D1cc, and D0.1cc of these three OARs for 141 cervical cancer patients treated with tandem and ring HDR brachytherapy after EBRT. The mean D2cc values for bladder, rectum, and sigmoid were 95 ± 22, 65 ± 12, and 62 ± 12 Gy,, respectively. This study confirmed that D2cc was a predictor of late toxicity for the rectum and bladder. A rectoscopy study669 was done in 35 patients in which EQD2 (α/β = 3 Gy) of the D0.1cc, D1cc, and D2cc of the rectum was recorded. After a mean follow-up time of 18 months, telangiectasia was found in 26 patients (74%), and 5 had ulceration that corresponded to the D0.1ccof the anterior rectal wall. The D2cc was higher in patients with rectoscopy score of >3 compared to <3 (72 ± 6 vs., 62 ± 7 Gy; p < .001) and in symptomatic versus asymptomatic patients (72 ± 6 vs. 63 ± 8 Gy; p < .001). Based on these two studies, a dose limit of 70 to 75 Gy EQD2 is recommended for the rectal dose constraint.
For interstitial brachytherapy, in which a much longer portion of the anterior rectal wall is treated as part of the target volume, D2cc > 62 Gy predicted for late toxicity.670 The development of mucosal and clinical changes in the rectum seems to follow a clear dose effect and volume effect. For patients receiving interstitial brachytherapy who require treatment to the entire vaginal length, the dose to the rectum should be reduced as much as possible without compromising target coverage.
3D Treatment Planning for Pulse- and High–Dose-Rate Brachytherapy
Treatment planning for PDR and HDR brachytherapy can be accomplished by a variety of techniques. Treatment planning for LDR is covered in a separate chapter. For cervical cancer, customized optimization of source loading for each HDR insertion is recommended (Fig. 69.25), given significant changes in tumor and OAR dosimetry between fractions.627,671 Himmelmann et al.672 described individualized computer treatment optimization of source position and the dwell time for each position. Customized planning does increase the time needed for planning and requires experience on the part of the physics and dosimetry staff.673
CT-Based Treatment Planning
Fellner et al.674 compared treatment planning for cervical carcinoma based on CT sections and 3D dose computations or, when these techniques were not available, dose evaluation based on orthogonal radiographs. The CT-based planning provides information on target and organ volumes and dose–volume histograms. The radiography-based planning provides dimensions and doses only at selected points. For the study, 28 patients with 35 applications receiving HDR treatment with 192Ir were investigated. For a dose prescription of 7 Gy at point A, 83% (44 cm3) of the CTV received at least 7 Gy.
Gebara et al.675 estimated the external, internal, and common iliac dose rates using 3D CT-based dose calculations in tandem and ovoid brachytherapy in 30 patients with carcinoma of the uterine cervix treated with LDR brachytherapy using a CT-compatible Fletcher-Suit-Delclos device. Thirty-six implants were performed, and the authors concluded that the point B dose is similar to the maximum common iliac nodal dose. With HDR brachytherapy, the dose to the pelvic lymph nodes is approximately 25% of the per-fraction dose.344
Dewitt et al.,676 in 15 patients with cervical cancer, defined target and organs at risk for planning of HDR brachytherapy and established guidelines for volume and dose constraint parameters using image-guided inverse treatment planning.
Careful assessment of the quality of brachytherapy and dose distributions is critical. Suyama et al.677 analyzed the minimal radiation dose to the peripheral area of the cervix in relation to local tumor failure using CT images taken at the time of intracavitary brachytherapy in 80 patients with carcinoma of the cervix. After CT scanning, isodose curves were superimposed on the CT images. Histograms of both the minimum percentage peripheral dose and the dose to the cervical area showed significant correlation in the local tumor control and local failure groups (p < .001).
With HDR intracavitary applicators the use of a rectal retractor has been shown to substantially reduce the rectal dose.610 Lee et al.,678 in a study of 15 patients, found that this reduction was significant only in the subgroup who received >70% of the prescription dose (p < .05).
Wanderas et al.679 reviewed data from 19 patients (72 fractions) retrospectively. Standard library plans were compared to individually optimized plans using a Fletcher HDR applicator. For standard treatment planning, the tolerance dose limits were exceeded in the bladder, rectum, and sigmoid in 26%, 4%, and 15% of the plans, respectively. This was observed most often for the smallest target volumes. The individualized planning of the delivered treatment gave the possibility of controlling the dose to critical organs to below certain limits. The dose was still prescribed to point A. An increase in target dose coverage was achieved when additional individual optimization was performed while keeping the dose to the OARs below predefined limits. Relatively low average target coverage was seen, however, especially for the largest volumes.
MR-Based Treatment Planning
Basic principles of MR imaging during brachytherapy have been described.680 Several institutions have reported the dosimetric advantages of MR-based brachytherapy, with reduction in OAR doses with optimization.645,654,681–683Tanderup et al.681 showed that point A dose was a poor surrogate of HR-CTV dose, and MR-based planning improved target coverage and reduced OAR dose. Starting with a standard plan is important for consistency because relative uniformity of the dose distribution should be maintained.684
A comparison of MR to ultrasound was reported by Van Dyk et al.629 for 71 patients, showing comparability between the two modalities in terms of target volume and rectal point dose. Mahantshetty et al.685 similarly confirmed the feasibility of US for institutions that do not have easy access to MR or CT.
Recommended Doses
Stage IA (microinvasive) tumors are treated with intracavitary therapy only. LDR dose is approximately 60 Gy in one insertion or 75 to 80 Gy in two insertions to point A, or with HDR an equivalent dose, with one or two fractions per week. This may be given in approximately 5 Gy per fraction for 10 fractions or other regimens based on normal-tissue exposure.
For stages IB to IVA cervical cancer in the United States, the most common EBRT dose treats the elective pelvic nodes to 45 to 50 Gy given in 1.8 Gy per fraction.598 Some institutions instead use lower doses of whole-pelvis external irradiation (20 to 40 Gy) in addition to parametrial doses to complete 50 to 60 Gy to the involved parametrial tissues or nodal regions for more advanced stages. Brachytherapy follows, with a goal EQD2 dose of 80 to 90 Gy to point A or to the HR-CTV. For LDR, intracavitary treatment for approximately 4,000 to 5,000 mgh (36 to 50 Gy to point A at 60 cGy/hour) is given, depending on the tumor volume and stage and age of the patient. Fyles et al.137identified FIGO stage as the most significant prognostic factor in 965 patients with invasive carcinoma of the cervix, followed by dose of irradiation to point A and overall time of radiation therapy. The 10-year survival rate was 62% in 743 patients receiving doses to point A of 85 Gy or higher, in contrast to 53% for 222 patients receiving lower doses.
For HDR, one study found that a dose to the HR-CTV of >87 Gy resulted in a local recurrence rate of 4% compared to 20% for D90 < 87 Gy when the tumor was >5 cm and using an HDR tandem/ring or tandem/ring/interstitial approach. They concluded that local control rates of >95% can be achieved for patients with a poor response after EBRT if D90 for the HR-CTV is 87 Gy or higher.535 The IR-CTV intended dose should be approximately 60 Gy EQD2. In the United States the most common regimen uses five fractions (5 to 6 Gy per fraction), with two fractions per week 24 to 48 hours between fractions.
FIGURE 69.25. A: Coronal view of a CT planned tandem and ring applicator with standard HDR loading (to point A). B: Tandem and ring treatment plan optimized to maximize tumor coverage and minimize the dose to the organs at risk (OAR) including the rectum (brown), sigmoid (blue) and bladder (yellow). C: Sagittal CT of the standard plan at point A. D: Sagittal CT image of the optimized plan showing the reduction in OAR dose. E: Cervical stump cancer showing an MR-planned tandem and interstitial brachytherapy implant without HDR optimization. F: Optimized tandem and interstitial plan cover the posterior border of the cervical stump while minimizing dose to the bladder and rectum.

DVH constraints for both PDR and HDR are 90 Gy (EQD2) for bladder and 70 to 75 Gy (EQD2) for both rectum and sigmoid as minimal doses to the most exposed D2cc of the OAR. There are no generally accepted constraints for the 0.1-cm3 level. Given the rapid regression of the tumor and the dramatic change in the location and size of the normal tissues, it is recommended to replan and determine the doses to the OAR with each fraction if the patient is treated on an outpatient basis.671
Dose Fractionation in High–Dose-Rate Brachytherapy
The relationship between dose and fractionation for HDR and LDR intracavitary irradiation of stage I and II carcinoma of the cervix was examined by Arai et al.644 The dose rate at point A was 2 to 3 Gy/minute (120 to 180 Gy/hour) for HDR and 0.6 to 0.9 Gy/hour for LDR irradiation. Concurrent EBRT was given to the whole pelvis (23 to 30 Gy), followed by 25 to 30 Gy with central shielding, along with brachytherapy. The authors concluded that the optimal dose fractionation schedules for HDR brachytherapy were 28 ± 3 Gy in 4 to 5 fractions, 34 ± 4 Gy in 8 to 10 fractions, or 40 ± 5 Gy in 12 to 14 fractions at point A. Petereit and Pearcey,619 based on their preliminary results and published reports in the literature, recommend doses of 45 to 50.5 Gy in a 1.8-Gy/fraction external beam followed by HDR with either 5.5 or 6 Gy per fraction in the era before the standardization of chemoradiation. Since the implementation of concurrent chemoradiation, several institutions in the United States have standardized the use of 5.5 Gy for five fractions, given some concerns about rectal toxicity with 6 Gy per fraction.686
Chatani et al.687 described a study in which 165 patients with carcinoma of the cervix were randomized to a HDR brachytherapy point A dose of 6 Gy (group A) or 7.5 Gy (group B) per fraction, both combined with external irradiation. The 5-year local failure rate was 16% in both groups, and distant failure rates were 23% and 29%, respectively (p = .2955). Moderate to severe complications requiring treatment were comparable (6 patients, 7%) in the two groups.
Hama et al.688 compared the effectiveness and safety of once- versus twice-weekly HDR brachytherapy for cervical cancer in 124 patients treated with EBRT (50 Gy); 74 patients (group A) were treated with one HDR brachytherapy insertion weekly (three fractions of 7 Gy each to point A), and 50 patients (group B) were treated twice weekly (six fractions of 4.5 Gy each to point A). Overall survival rates were 65.2% and 65.3%, respectively (p = .96). Local recurrence-free survival rates were 69% for group A and 90% for group B (p < .001). The rate of grade 2 (moderate) and grade 3 (severe) complications was significantly lower for group B (6%) versus 32% in group A (p< .001).
Mayer et al.689 compared HDR BT in two schedules used to treat 210 patients with cervix cancer—one sequential (SRT), consisting of four fractions of 8 Gy followed by EBRT, and the other continuous (CRT), consisting of five fractions of 6 Gy one session per week integrated with EBRT (four fractions per week). Total dose to point A was 68 to 70 Gy. Progression-free survival was 71% with CRT versus 56% with SRT (p > 0.05). Late bladder and rectal morbidity were 13% in the CRT group and 25% in the SRT group (p = .037), related to the higher dose per fraction (8 Gy).
Nam and Ahn690 compared, in a randomized study of 46 patients, two schedules of HDR BT (10 fractions of 3 Gy or five fractions of 5 Gy) followed by a small BT boost to residual tumor in combination with EBRT (30.6 Gy to whole pelvis and 14.4 Gy to parametria with midline block). Three-year pelvic tumor control was 90% in both groups, and disease-specific survival was 90.5% and 84.9% (p = .64), respectively. Late grade 2 and greater bladder or rectal morbidity was 23.8% and 9.1%, respectively (p = .24).
Liu et al.,691 based on the linear-quadratic model, developed isoeffect tables to convert traditional LDR doses and number of fractions to point A to HDR brachytherapy; depending on dose rate, different dose values can be calculated for various fractionation schedules. They predicted that, using therapeutic gain ratio, similar results would be obtained with either brachytherapy modality with two to four fractions of LDR and four to seven fractions of HDR.
TABLE 69.16 HIGH–DOSE-RATE BRACHYTHERAPY DOSE AND FRACTIONATION REGIMENS WORLDWIDE

The optimal time–dose–fractionation scheme for HDR brachytherapy for cervical cancer has yet to be established. In an international survey from the Gynecologic Cancer Intergroup, 28 different fractionation regimens were used by international cooperative group members (Table 69.16); the most common was 6 Gy for five fractions after 45 Gy.602 The American Brachytherapy Society published recommendations for HDR brachytherapy for carcinoma of the cervix.623 Each institution should follow a consistent treatment policy, including complete documentation of treatment parameters and correlation with clinical outcome (pelvic tumor control, survival, and complications). The goals are to treat point A to at least a total LDR equivalent of 80 to 85 Gy for early-stage disease and 85 to 90 Gy for advanced-stage disease. The pelvic sidewall dose recommendations are 50 to 55 Gy for early lesions and 55 to 65 Gy for advanced ones. As with LDR BT, every attempt should be made to keep the bladder and rectal doses to <100 Gy and 75 Gy LDR-equivalent doses, respectively. Interstitial brachytherapy should be considered when the tumor cannot be optimally encompassed by intracavitary brachytherapy. Some suggested dose and fractionation schemes for combining the external-beam radiation therapy with HDR brachytherapy for each stage of disease have been presented by the American Brachytherapy Society, although they have not been thoroughly tested. The responsibility for the medical decisions ultimately rests with the treating radiation oncologist. Petereit and Pearcy,619 in a review of 24 HDR dose fractionation schedules published in the last three decades, found no dose relationship for either tumor control or late morbidity. Viswanathan et al.,602 for the Gynecologic Cancer Intergroup, found significant international variation in the HDR dose/fractionation regimens reported, but aside from Japan, where the ratio of HDR brachytherapy dose to external beam is higher, there was consistency in converted EQD2 doses administered.
TABLE 69.17 RANDOMIZED TRIAL RESULTS OF TOXICITY AND OVERALL AND DISEASE-FREE SURVIVAL COMPARING HDR AND LDR

Clinical Outcomes of Brachytherapy
Randomized Studies Comparing HDR to LDR Using Plain X-Ray Dosimetry
Four randomized trials (Table 69.17) and a meta-analysis summarizing the results of these have been published comparing HDR and LDR brachytherapy for carcinoma of the cervix.692,693 Teshima et al.694 reported on a prospective, randomized study of 430 patients with carcinoma of the uterine cervix treated with either LDR (171 patients) or HDR (259 patients) brachytherapy combined with external irradiation. Cause-specific and overall survival rates were comparable for each clinical stage with either modality, except for stage I overall survival. The conversion factor of total intracavitary dose from LDR to HDR was 0.5 to 0.53. With HDR, four fractions usually were delivered, and with LDR, two fractions. The incidence of pelvic failures was comparable in both groups. The incidence of grade 2 and 3 morbidity was somewhat higher in the HDR group (~10%) than in the LDR group (4%; p = .002).
Patel et al.607 published a randomized trial of 482 patients with invasive squamous cell carcinoma of the cervix. The overall local tumor control rate with LDR brachytherapy was 79.7%, compared with 75.8% with HDR. The 5-year survival rates were 73% with LDR and 78% with HDR in stage I, 62% and 64%, respectively, in stage II, and 50% and 43% in stage III. The only statistically significant difference was the incidence of overall rectal complications, which was 19.9% for LDR, compared with 6.4% for HDR. However, the incidences of more severe grade 3 and 4 complications were not significantly different (2.5% and 0.4%, respectively). Bladder morbidity was similar in both groups.
Hareyama et al.695 conducted a randomized study in 132 patients with stage II or IIIB cervical carcinoma treated with LDR or HDR BT and identical pelvic EBRT. The conversion factor from LDR to HDR was 0.588. The 5-year DSS with HDR for stages II and IIIB was 69% and 51%, respectively, and with LDR it was 87% and 60%, respectively. Pelvic tumor control for stage II and III was 89% and 73% with HDR and 100% and 70% with LDR, respectively, and grade 3 or greater morbidity was 10% and 13%, respectively (differences were not statistically significant).
Lertsanguansinchai et al.696 randomized 237 patients with cervical cancer to be treated with LDR (109 patients) or HDR (112 patients) brachytherapy and EBRT. Median follow-up was 40 and 37 months, respectively. Three-year pelvic tumor control was 89% and 86.4%, respectively, and relapse-free survival was 69% in both groups. Grade 3 or 4 morbidity was noted in 2.8% of LDR and 7.1% of HDR patients (p = .23).
A meta-analysis693 in-cluding these four trials reported a pooled RR for HDR versus LDR of 0.95 (95% CI = 0.79 to 1.15), 0.93 (95% CI = 0.84 to 1.04), and 0.79 (95% CI = 0.52 to 1.20) for 3-, 5- and 10-year overall survival rates and 0.95 (95% CI = 0.84 to 1.07) and 1.02 (CI = 0.88 to 1.19) for 5- and 10-year DSS rates. For local control rates the RR was 0.95 (95% CI = 0.86 to 1.05) and 0.95 (95% CI = 0.87 to 1.05) at 3 and 5 years, respectively. For bladder, rectosigmoid, and small-bowel complications, the RR was 1.33 (95% CI = 0.53 to 3.34), 1.00 (95% CI = 0.52 to 1.91), and 3.37 (95% CI = 1.06 to 10.72), respectively, indicating no significant differences except for increased small-bowel complications with HDR (p = .04). Of note, none of the randomized studies used 3D imaging to optimize away from normal tissues.
The use of 3D imaging to guide brachytherapy treatment planning has allowed optimization of HDR and PDR brachytherapy, thereby reducing the high per-fraction doses to the normal tissues that might potentially cause significant side effects. A retrospective comparison of LDR and HDR with pretreatment MRI used for tumor volume determination showed a significant reduction in complications with HDR when image-based planning was implemented.697 Similarly, the use of 3D imaging optimizes tumor coverage, which is critical with fractionated HDR therapy.
Prospective Data Using Point A Dosimetry
Haie-Meder et al.,698 in 204 patients with cervical cancer randomized to receive one of two preoperative LDR brachytherapy procedures (0.4 or 0.8 Gy/hour), noted similar local tumor control (93%) and overall survival (85%) rates at 2 years with either dose rate. Grade 3 late complications were observed in 7% of patients treated with 0.4 Gy/hour and in 13% of patients treated with 0.8 Gy/hour. There was 1 small-bowel obstruction in the 0.4-Gy/hour group (1%), in contrast with 5 (5%) in the 0.8-Gy/hour group. Vesicovaginal fistulas were observed in 2% and 4%, respectively.
A prospective study in Japan of stage I and II cervical cancer with tumors <4 cm (by T2 MRI) and no lymphadenopathy treated 60 patients with whole-pelvis EBRT 20 Gy/10 fractions with midline block followed by 30 Gy/15 fractions and HDR 24 Gy/4 fractions (at point A).699 The cumulative BED was 62 Gy (α/β = 10) at point A, lower than that reported by any other institution worldwide. Median tumor diameter was 28 mm (range, 6 to 39 mm). Median overall treatment time was 43 days. Median follow-up was 49 months (range, 7 to 72 months). Seven patients developed recurrences: 3 patients had pelvic recurrences (2 central, 1 nodal), and 4 patients had distant metastases. The 2-year disease-free and overall survival rates were 90% (95% CI = 82% to 98%) and 95% (95% CI = 89% to 100%), respectively. The 2-year late complication rates (according to RTOG/EORTC grade ≥ 1) were 18% (95% CI = 8% to 28%) for large intestine/rectum, 4% (95% CI = 0% to 8%) for small intestine, and 0% for bladder. No cases grade ≥3 were observed for genitourinary/gastrointestinal late complications.
The prospective French STIC trial534 reported patients treated with x-ray simulation compared to 3D-based planning. A total of 705 patients with stages IB to IIIB cervical cancer were enrolled. Toxicity and survival were significantly improved with 3D-based treatment planning. Plain film–based 2-year local control was 74% for patients treated with chemoradiation and LDR or PDR brachytherapy. Detailed results are shown in Table 69.18.
TABLE 69.18 CLINICAL OUTCOMES WITH THREE-DIMENSIONAL PLANNED BRACHYTHERAPY

Retrospective Data Using Point A Dosimetry for Low–Dose-Rate Brachytherapy
Fowler700 analyzed results in 270 patients with carcinoma of the cervix treated with either 75 cGy/hour from manually loaded cesium or 150 cGy/hour by remote afterloading. There was an increase in grade 3 late complications from 4% to 22%, in spite of a reduction of 20% in dose, implying a rather large difference in biologic effect between the two systems. The effect of the increased dose rate was also described by Leborgne et al.701 Linear-quadratic modeling was used to calculate biologically effective doses in the clinical protocols used. When the LDR was doubled, it was called MDR. The maximum ratios calculated for the biologic effective doses of 16 Gy at MDR to 20 Gy at LDR were 1.06 to 1.15, assuming α/β = 4 to 2 Gy, the latter being an unlikely extreme for rectal or urinary complications. The theoretically ideal dose reduction factors, calculated using the t1/2 values derived from the clinical data, are in the range of 24% to 29% instead of 20%.
Rodrigus et al.702 analyzed late complications in 143 patients with cervical cancer treated with two different brachytherapy schedules and external radiation. Seventy-seven patients had two intracavitary applications with a dose rate of 0.54 Gy/hour and 66 patients with that of 1.07 Gy/hour. Because of the expected increase in complications with the higher dose rate, the latter dose per application was reduced from 25 to 20 Gy. Late intestinal and urinary complications were scored in 49 of 77 and 46 of 68 patients, respectively. Actuarial estimates at 5 years showed 42% and 54.1% late intestinal complications and 16.9% and 24.1% late urinary complications, respectively. Thus, despite the dose reduction, there was a clear dose-rate effect on late morbidity. These studies emphasize the importance of the dose rate of brachytherapy in carcinoma of the cervix.
Rotmensch et al.703 compared the outcome in 140 patients with early-stage cervical cancer undergoing whole-pelvis radiation therapy with one versus two LDR intracavitary brachytherapy applications. The two groups had similar 5-year local tumor control (p = .83), disease-free (p = .23), and cause-specific (p = .29) survival. Late complications were similar in the two groups. These results support the use of a single LDR application in patients with early-stage disease undergoing definitive radiation therapy after 45-Gy external-beam pelvic irradiation.
In a retrospective analysis, Perez et al.,169 noted that in patients with cervical cancer treated with radiation therapy alone for stage IB tumors <2 cm in diameter, the pelvic failure rate <10% with LDR doses of 70 to 80 Gy to point A, whereas for larger lesions, even doses of 85 to 90 Gy resulted in 25% to 37% pelvic failure rates. In stage IIB with LDR doses of 70 Gy to point A, the pelvic failure rate was approximately 50%, compared with 20% in nonbulky and 30% in bulky tumors with doses >80 Gy. In stage III unilateral lesions, the pelvic failure rate was approximately 50% with 70 Gy or less to point A versus 35% with higher doses, and in bilateral or bulky tumors it was 60% with doses <70 Gy and 50% with higher doses.
A study from France reported on preoperative LDR followed by radical surgery with lymph node dissection for 257 patients with stages IB1, IIA, and IIB cervical cancer of <4 cm.704 Residual tumor was identified in 44% of patients, whereas 4.3% of patients had parametrial invasion and 17.9% of patients had lymph node involvement. Late complications of grade 2 occurred in 7.4% and of grade 3 in 2.7% of patients. Five-year actuarial overall survival and disease-free survival were 83% (CI = 78.3 to 87.5) and 80.9% (CI = 76.3 to 85.7), respectively. In multivariate analysis, lymph node involvement, parametrial involvement, and smoking factors significantly affected overall survival and disease-free survival rates.
Retrospective Results Using Point A with Pulse–Dose Rate Brachytherapy
Rogers et al.705 treated 52 patients with cervical carcinoma, 31 of whom had staging laparotomy before radiation therapy. Brachytherapy was interstitial in 18 patients and intracavitary in 28. The median EBRT pelvis dose was 45 Gy in 25 fractions. Median total doses were 75.8 Gy to the implant volume with interstitial and 84.1 Gy to the A points with intracavitary at a median dose rate of 0.55 Gy per pulse per hour. Six patients had laparotomy-documented para-aortic node involvement and received EBRT to this site (45 Gy in 25 fractions). Thirty patients received concomitant weekly cisplatin chemotherapy (40 mg/m2). With a median follow-up of 25 months, the actuarial 4-year disease-free survival rates were 66% for the entire group (100% for stage IB, 69% for stage II, 68% for stage III/IVA, and 43% in patients treated for recurrences after surgery). Grade 4 complications occurred in 2 patients (4.3%). One patient (2.2%) had a grade 3 complication (frequent hematuria), and 5 (10.9%) had grade 2 complications.
Kaneyasu et al.706 treated 419 patients with squamous cell carcinoma of the cervix from 1969 to 1999 with LDR or MDR. LDR required overnight admission, whereas MDR was given over approximately 5 hours on an outpatient basis. The 5-year overall survival rates for stages I, II, III, and IVA in the LDR group were, respectively, 78%, 72%, 55%, and 34% versus 100%, 68%, 52%, and 42% in the MDR group (not statistically different). The actuarial rates of late complications of grade 2 or greater at 5 years for the rectum, bladder, and small intestine in the LDR group were 11.1%, 5.8%, and 2.0%, respectively, whereas for the MDR group they were 11.7%, 4.2%, and 2.6%, respectively (not significantly different).
El-Baradie et al.707 published a prospective study in which 45 patients with carcinoma of the uterine cervix were randomly allocated to either HDR or MDR. The external-beam radiation dose was the same in the two groups. The point A dose rate correction factor from LDR to HDR was 0.53, and that from LDR to MDR 0.6. The 3-year survival and locoregional tumor control rates for both modalities were equivalent (respectively 62% and 67% for HDR and 68% and 74% for MDR). The rectal and bladder complication rates were the same in both groups (29% at 3 years). Tanaka et al.708 also compared HDR and MDR brachytherapy in 150 and 56 patients, respectively. The survival was equivalent in the two groups; grade 2 or greater late toxicity tended to be higher in the HDR group (14% vs. 6%, respectively).
Bachtiary et al.709 reported on 109 patients treated with LDR BT and 57 who received PDR BT. The 3-year overall survival and disease-free survival rates were 70% and 57% for the LDR group and 82% and 70% for the PDR group, respectively (p = .25 and .19). The 3-year probability rate for late grade 3 or worse toxicity was 7.4% for LDR BT patients and 7.6% for PDR BT patients (p = .69) and was 6.9% and 7.6%, respectively, for concurrent chemotherapy versus none (p = .69).
Rath et al.710 reported on 48 patients treated with PDR brachytherapy (ICRT) and pelvic irradiation. A single session delivered a dose of 27 Gy to point A by PDR (hourly pulse, 70 cGy). Ten patients had disease recurrence (5 each in stage IIB and stage IIIB). Eight patients had pelvic failure, 1 had bone metastases, and 1 had supraclavicular node metastases. Overall the grades III to IV late toxicity rate at 50 months was 6%. For the median follow-up period of 15 months, the actuarial recurrence-free survival in stages I to II was 82% and in stages III to IV was 78%.
Retrospective Results with Point A Using High–Dose Rate Brachytherapy
Many nonrandomized studies compared the results of HDR with those of historic or concurrent control patients receiving LDR at the same institution.608,627,672,711–713 HDR in patients with stage IIIB disease or large tumors must be used cautiously because the brachytherapy prescription dose should cover the tumor volume and avoid the normal tissues as much as possible (Table 69.19). Although it is generally recommended not to give concurrent chemotherapy on the day of HDR brachytherapy,598 several studies indicate that HDR does not increase toxicity in patients treated with chemoradiation (Table 69.20). Most studies used point A as a reference point, although the definition of point A may have differed from center to center.
A retrospective population-based cohort study of all uterine cervix cancer cases in Saskatchewan diagnosed between 1985 and 2001 had 107 LDR and 37 HDR cases with similar stage distribution. The 5-year cause-specific survival rate was 56% for HDR and 67% for LDR (p = .72). Acute toxicities were diarrhea (60%) and abdominal cramps (12.5%), and chronic toxicities were vaginal stenosis (5.5%) and small-bowel obstruction (4%).714
TABLE 69.19 STAGE III OVERALL SURVIVAL, PELVIC CONTROL, AND TOXICITY IN RETROSPECTIVE SERIES

TABLE 69.20 FRACTIONATION AND TOXICITY OF HIGH–DOSE-RATE AND CONCURRENT CHEMOTHERAPY

Petereit et al.606 reported on 191 patients receiving LDR brachytherapy and 173 receiving HDR brachytherapy with equivalent external-beam radiation therapy techniques. Pelvic tumor control and survival rates were comparable with the two techniques, except in stage III; in this subgroup, outcome was better with LDR brachytherapy, but this may have been related to a lower HDR equivalent dose administered. In an analysis of 198 patients treated with LDR brachytherapy, the 3-year survival rate was 66% versus 77% for 40 patients treated with HDR brachytherapy.715 Pelvic tumor control rates were 80% and 77%, respectively. The incidences of complications requiring hospitalization or surgery were 10% (20 of 198) and 2.5% (1 of 40), respectively.
Kapp et al.,716 in a study of 181 patients with FIGO stages IB to IV carcinoma of the cervix, documented that prognostic factors for patients treated with HDR are similar to those in previous series with LDR brachytherapy. In multivariate analysis, tumor size was the most powerful factor for pelvic tumor control and incidence of distant metastasis.
Ferrigno et al.717 carried out a retrospective study of 190 patients treated with LDR and 118 with HDR brachytherapy in combination with pelvic EBRT for cervical cancer. For stage I or II patients, there was no difference in outcome; however, in the stage III group local tumor control was 58% with LDR and 50% with HDR (p = .19), and DFS was 49% versus 37% (p = .03). At 5 years, rectal sequelae were 16% versus 8% (p =.03), bladder sequelae were 6% and 3% (p = .13), and small-bowel sequelae were 4.6% and 8.9% (p = .17).
Falkenberg et al.718 reviewed 160 patients, 103 treated with LDR and 57 treated with HDR from 1990 to 2000. Locoregional control was 78% for LDR and 76% for HDR (p = .96); overall survival was 60% for LDR versus 55% for HDR (p = .48) at 3 years. Late complications were reported in 2 HDR patients (3.5%) and 5 LDR patients (4.8%).
Orton et al.610 noted that dose per fraction of HDR brachytherapy significantly influenced toxicity. Morbidity rates were significantly lower for point A doses/fractions of 7 Gy or less for both severe (1.28% vs. 3.44%; p <.0001) and moderate plus severe injuries (7.58% vs. 19.51%; p <.001). The effect of dose/fractionation on cure rates was equivocal.
Kuske et al.719 described a method to improve target coverage and locoregional tumor control with HDR tandem and ovoid applications by which HDR endocavitary and interstitial brachytherapy are applied in the same session for tumors with a lateral expansion of 25 mm or more from the axis of the cervical canal. Seventy-six combined applications were given to 41 patients. With a follow-up average of 23 months, in stage IIB tumors, 3-year DFS was 75%. No severe early or persistent late complications were observed. Combined applicators with the tandem and ring with interstitial720 and tandem and ovoid with interstitial656,721 are now available for HDR brachytherapy.
Forrest et al.686 presented the results of 122 patients treated with EBRT followed by 6 Gy per fraction for five fractions of HDR. They reported a 2-year disease-free survival rate of 70% and grade 3/4 toxicity rate of 14% (13 patients). The median time to recurrence was 8 months (range, 2 to 22 months) and to toxicity was 10 months (range, 4 to 27 months). They concluded that the high toxicity of this regimen should prompt consideration of dose reduction in BT dose or use of 3D imaging to shape the dose. Several institutions in the United States now report 5.5 Gy é five fractions instead of 6 Gy per fraction for patients treated with chemoradiation. 601,602
Anker et al.722 treated 65 patients with HDR with 6 Gy é fractions, and 45 patients had the top dwells retracted as the tumor regressed. With a median follow-up of 24.5 months, the 3-year overall, disease-free distant metastases–free survival, and local control rates were 67%, 76%, 79%, and 97%, respectively. Acute and actuarial 3-year late grade 3 toxicity or greater occurred in 24.6% and 17% of patients, respectively.
Le Pechoux et al.723 treated 130 patients with cervical cancer with HDR brachytherapy (for stage I, 30 Gy in six weekly sessions) in combination with EBRT (50-Gy mean dose with midline shielding). Patients with more-advanced disease received four sessions of biweekly brachytherapy for a total dose of 18 to 24 Gy and external irradiation (20 to 30 Gy to the whole pelvis, 50 to 66 Gy to parametria with midline shielding). The 5-year survival rates were 82% for patients with stage IIB and 47% with stage IIIB disease. There were 4 rectovaginal or vesicovaginal fistulas and 1 case of proctitis requiring colostomy. Survival, local tumor control, and morbidity were equivalent in 76 patients treated with 6 Gy once a week and in 54 patients receiving twice-weekly brachytherapy of 5 Gy per session.
Hsu et al.724 dosed 92 patients with cancer of the cervix with HDR brachytherapy, six fractions of 7 Gy per fraction (42 Gy) at point A (HDR-6); 57 received four fractions of 8 Gy per fraction (32 Gy) at point A (HDR-4). A twice-daily program was used for all patients receiving HDR in two split courses. A historic control group of 259 patients was treated with LDR brachytherapy (40 Gy in two split courses). All patients received whole-pelvis external irradiation of 36 to 45 Gy (mostly 40 Gy) before brachytherapy. Five-year local tumor control rates were equivalent in the three groups (82%, 85.5% for HDR, and 89.5% for LDR). Five-year survival rates were also comparable (67.7%, 77.9%, and 74.1%, respectively). However, late complications were lower in the HDR-4 group, which received treatment more biologically equivalent to the LDR regimen, than in patients in the HDR-6 group (11% vs. 25.6%).
Selke et al.725 published results in 187 patients with primary carcinoma of the cervix treated with whole-pelvis irradiation (46 Gy) and HDR brachytherapy with a dose rate to point A of 1.6 Gy/minute, decreasing to approximately 0.8 Gy/minute at the end of the 5-year study. Three HDR fractions (8 to 10 Gy to point A per fraction) were concurrently administered with the last 2 to 3 weeks of external irradiation. The 5-year actuarial survival rates were 72% for stage IB, 65% for IIA, 66% for IIB, 66% for IIIA, and 45% for stage IIIB. With a median follow-up of 54 months, 23 patients had 25 complications; 13 (7.6%) were grade 3 or 4. Rectal complications were significantly higher in patients who received a total rectal dose of >54 Gy (p = .045).
Choi et al.726 treated 136 patients with carcinoma of the cervix with external-beam whole-pelvis irradiation (46 Gy in 23 fractions) and three weekly applications of HDR brachytherapy of 7 or 8 Gy per fraction to point A. The actuarial 5-year survival was 85% in stage IB, 64% in stage IIA, 70% in stage IIB, and 53% in stage IIIB. Grade 3 or higher complications occurred in 3% to 7% of the patients. The most significant determinants of severe rectal complications were the addition of a lower vaginal tandem (p < .01), uterine tandem length >5 cm, a total biologically effective dose to the rectum of >120 Gy, and stage III disease.
Kagei et al.727 reported on 217 patients with carcinoma of the cervix (71 patients with stage II and 146 with stage III disease) who received whole-pelvis EBRT (40 Gy in 20 fractions or 39.6 Gy in 22 fractions) and an additional 10 Gy in five fractions to the parametria followed by HDR brachytherapy. Cause-specific 5-year survival rates were 77% for stage II and 50% for stage III. Pelvic failure rates were 13% and 36%, respectively. The rates of severe (grade 4) late complications were 2% for the rectum, 1% for the small intestine or sigmoid colon, and 1% for the bladder.
Takeshi et al.728 treated 265 patients with stage III cervical carcinoma with external-beam radiation therapy (50.3 Gy) and intracavitary HDR brachytherapy (19.8 Gy). The 5-year overall survival, relapse-free survival, and locoregional event–free rates were 50.7%, 57.1%, and 71.2%, respectively. The 5-year incidence of major complications was 2.6% for bladder and 8.3% for rectum. The radiation dose in the subgroup with rectal complications was significantly greater than that in the subgroup without complications.
Wang et al.729 reported treatment results in 173 patients with cervical carcinoma treated with HDR brachytherapy and whole-pelvis irradiation (40 to 44 Gy in 20 to 22 fractions) followed by pelvic wall boost (6 to 14 Gy in three to seven fractions with central shielding). HDR brachytherapy delivered 7.2 Gy to point A in each of three applications 1 to 2 weeks apart. Five-year pelvic tumor control rates were 94%, 87%, and 72% for stages IIA, IIB to IIIA, and IIIB to IVA, respectively. Five-year actuarial survival rates were 79%, 59%, and 41%, respectively. Sixty-six patients (38%) had rectal complications, and 19 (11%) had bladder complications. The 5-year actuarial rectal complication rates were 15%, 4%, and 3% for grades 2, 3, and 4, respectively.
Lorvidhaya et al.730 reported the results in 1,992 patients with carcinoma of the cervix treated by external irradiation and HDR brachytherapy. There were 211 patients with stage IB, 225 with stage IIA, 902 with stage IIB, 14 with stage IIIA, 675 with stage IIIB, 16 with stage IVA, and 16 (0.8%) patients with stage IVB. With a median follow-up of 96 months, the actuarial 5-year disease-free survival rates were 70%, 59.4%, 46.1%, 32.3%, 7.8%, and 23,1%, respectively. The late complication rates (RTOG) for bowel and bladder combined were 7% for grade 3 and 1.9% for grade 4 complications.
Leborgne et al.731 described a 4-year pelvic control rate of 93% and a disease-free survival rate of 88% for 59 patients with stage IB to IIA disease. All were treated with 18 Gy to the whole pelvis and 22 Gy to the parametria combined with six HDR fractions (14 Gy/hour to point A) of 7 Gy to point A, two in each treatment day, with 6-hour interfraction intervals. The corresponding parameters for 29 patients with stage IIB disease were 79%, 75%, and 75%. The actuarial 4-year late grade 2 and 3 complication rate was 4.7%.
In 1,148 patients with squamous cell cervical cancer treated with external RT and HDR brachytherapy with 22 years median of follow-up, the 10-year pelvic tumor control was 93% for stage IB, 82% for stage II, and 75% for stage III.569 Cause-specific survival was 89%, 74%, and 59%, respectively. Major sequelae were 4.4% in the rectosigmoid, 0.9% in the bladder, and 3.3% in the small intestine. Nakano et al.732 subsequently presented a study of 210 patients with stage IIIB cervical cancer from eight Asian countries treated from 1996 to 1998 with radiation and brachytherapy. Though follow-up was difficult to obtain, the reported 5-year major complication rates were 6% in the HDR group and 10% in the LDR group. The 5-year overall survival rates were 51.1% in the HDR group and 57.5% in the LDR group.
Novetsky et al.733 presented data on 77 patients treated with external beam with concomitant cisplatin followed by two HDR brachytherapy fractions of 9 Gy each. Median follow-up was 3.5 years. The local control rate was 88% for stages IB2/II and 68% for stages III/IV. Grade 3/4 gastrointestinal acute symptoms occurred in 47%. Grade 3/4 late toxicities occurred in 5 (6%) patients. Patel et al.734 describe 104 cervical cancer patients treated with external beam and HDR, either 9 Gy for two fractions or 6.8 Gy for three fractions, each fraction 1 week apart. Median follow-up was 31 months. The 3-year actuarial local control was 81.35% with 9 Gy versus 65.18% with 6.8 Gy (p = .04). The 3-year actuarial risk of developing any grade 3 or worse late toxicity was 7.47% with 9 Gy and 3.57% with 6 Gy (p = 0.3).
Prospective Trial with CT or MR Compared to X-Ray
The clinical outcome results from institutions using CT- or MR-based treatment planning for cervical cancer brachytherapy are listed in Table 69.18. The French STIC trial733 collected data from 20 centers prospectively and stratified to 2D versus 3D (mainly with CT) brachytherapy. A total of 705 patients were treated with one of three arms: (a) brachytherapy followed by surgery (stage IB1, 165 patients); (b) EBRT plus chemotherapy, BT, then surgery (305 patients); or (c) EBRT plus chemotherapy and then BT (235 patients). For the 235 patients treated with concurrent chemoradiation and then brachytherapy, 2-year overall survival was 74% for 3D versus 65% for 2D (p = .27); disease-free survival was 60% versus 55% (p = .09); local regional relapse–free survival was 70% versus 61% (p = .001), and local-only relapse–free survival was 79% versus 74% (p = .003). Toxicity was reduced overall from 23% with 2D to 2.6% with 3D (p = .002); urinary from 9% in 2D to 1% with 3D (p = .02), gastrointestinal from 9% to 0% (p = 0.17), and gynecologic from 15% to 1% (p = .01).
Retrospective Comparison of CT to MR-Planned Brachytherapy
Three studies compared CT to MRI contouring for HDR tandem and ring brachytherapy. Wachter-Gerstner et al.735 compared MR-based plans in 15 patients to those derived with either CT or orthogonal films. CT and MR enabled higher dose to the target volume with similar OAR dosing. Viswanathan et al.635 compared CT contours to MR contours based on a standard set of guidelines; the CT contours were larger in width, but no other significant differences in DVHs were identified. A report by Eskander et al.636 of 10 patients had an MR for the first fraction only and showed that CT volumes had a greater length on the coronal plane, whereas MR images had a greater height on the sagittal plane. No differences were found in DVH parameters after optimization.636 Similar to the Eskander et al.636 study, using an MR for the first fraction and CT for subsequent fractions, Beriwal et al.652 treated 44 patients with 5- to 6-Gy per fraction HDR after EBRT. Ninety-three percent had a complete response by PET at 3 months. Of those with a CR, 2 had a local recurrence at 6 and 8 months. With a median follow up of 8 months (range, 2.5 to 38 months), 2-year local control, disease-specific, and overall survival rates were 88%, 85%, and 86%, respectively.
Retrospective Results with CT-Planned Brachytherapy
Potter et al.736 reported results in 189 patients treated with HDR brachytherapy and EBRT (48.6 to 50 Gy). Small tumors were treated with five to six fractions of 7 Gy at point A (25 Gy in the brachytherapy volume), which is isoeffective to 76 to 86 Gy at point A. Large tumors received three to four fractions of 7 Gy after 50 Gy of EBRT, which is isoeffective to 82 to 92 Gy at point A. Three-dimensional treatment planning for brachytherapy was based on conventional x-rays and in 181 of 189 patients on CT scan. The mean brachytherapy dose was 16.2 Gy at the ICRU rectum reference point and 14.4 Gy at the ICRU bladder point. Taking into account the dose for EBRT, the mean isoeffective dose at the ICRU rectum reference point was 69.9 Gy. After a mean follow-up of 34 months, the actuarial pelvic control rate was 78% and the late complication rate for grades 3 and 4 was 2.9% for bladder, 4% for bowel, 6.1% for rectum, and 30.6% for the vagina (shortening and obliteration).
CT-based clinical outcomes were reported by the Addenbrooks Hospital. Twenty-eight patients had HDR, 8 Gy é 3, CT-planned brachytherapy.737 The 3-year actuarial cancer-specific survival rate in this group was 81%, with a pelvic control rate of 96%. Five of the 28 patients died of para-aortic or other distant disease, 1 of them being the only one with local recurrence presenting as a malignant vesicovaginal fistula. In 24 patients, D90 ≥74 Gy was achieved. The only patient with local recurrence had D90 = 63.8 Gy, which was a 20% improvement over historical non–image-guided controls.
At Brigham and Women’s Hospital, 115 stages IB to IVA cervical cancer patients had CT-planned brachytherapy and were treated with 595 fractions of 5.5- to 6-Gy per fraction HDR brachytherapy.738 The 2-year local relapse rate was 6.9%. The 2-year disease-specific survival was 83%, and overall survival rate was 78%.
Retrospective Results with MR-Planned LDR Brachytherapy
An initial report of MRI during intracavitary gynecologic brachytherapy was published in 1992 from the University of Michigan by Schoeppel et al.739 Three patients had CT and MRI with their first of two intracavitary implants. A CT- and MR-compatible Fletcher applicator was used. CT could not distinguish the tumor with as much clarity as MR. Tardivon et al.740 at Institut Gustave Roussy (IGR) treated 10 patients with MR evaluation of the tumor during intracavitary brachytherapy for cervical and vaginal cancer and found that in 7 cases MR findings were concordant with clinical examination. MR was useful to determine the tumor/applicator relationship and distinguish the adjacent OAR.
A review was published of 39 patients treated at IGR with MRI-guided LDR brachytherapy in the preoperative setting.741 A total dose of 60 Gy to the IR-CTV was followed 6 weeks later by extrafascial hysterectomy and bilateral salpingo-oophorectomy with pelvic node dissection. Adjuvant chemoradiation was delivered to patients with pelvic lymph node involvement. After a median follow-up of 4.4 years (range, 2.6 to 6.6 years), there were no central recurrences; 1 local recurrence occurred in the lateral pelvis (2.6%). The 4-year actuarial overall and disease-free survival rates were 94% and 86%, respectively. The 2- and 4-year actuarial local relapse–free survival rates were 94% and 91%, respectively. Haie-Meder et al.655 subsequently published a series of 84 patients treated with LDR MR-planned brachytherapy after chemoradiation. With a median follow-up of 53 months (range, 31 to 79 months), the 4-year overall survival and disease-free survival rates were 57 (95% CI = 43 to 69) and 52% (95% CI = 40 to 64), respectively. Thirty-nine late complications occurred in 28 patients (33.3%): 13 bladder, 7 rectal, 5 small bowel, 4 urethral, 3 colic, 2 vaginal, 1 pelvic fibrosis, and 4 others. Four grade 3 delayed complications were observed, and no grade 4 complication occurred.
Retrospective Results with MR-Planned PDR or HDR Brachytherapy
With a 0.2-T MRI at the Medical University of Vienna, 145 patients with stage IB to IVA cervical cancer were treated with four fractions of 7-Gy HDR from 1998 to 2003.742 Complete remission was achieved in 138 patients (95%), with 7 patients having locally persistent or progressive disease in the central (n = 5) or noncentral (n = 2) pelvis. With a median follow-up of 40 months, the 4-year local control rate was 83%, compared to 63% for historical controls. A subsequent analysis of 156 patients treated from 2001 to 2008 with MR-based brachytherapy was reviewed.653 Local control was 98% for tumors 2 to 5 cm and 92% for tumors >5 cm. Overall survival, however, was 72% for tumors 2 to 5 cm and 65% for tumors >5 cm, indicating that despite the increase in local control with MR-based brachytherapy, death from distant metastases remains a problem in patients with large-volume cancer.
Investigators at the IGR reported on 45 patients treated between 2004 and 2006 with a tandem and mold technique using PDR brachytherapy and MR-based contouring.651 Until recently at IGR, surgery was often performed after brachytherapy if disease was suspected on clinical examination. A dose of ≥15 Gy (after EBRT) was prescribed to the IR-CTV. The dose to the HR-CTV was approximately 250% of the dose to the IR-CTV (i.e., 80 Gy to the HR-CTV). With a median follow-up of 26 months, the 2-year overall and disease-free survival rates were 78% and 73%, respectively. At Tata Memorial Hospital in India, 24 patients with squamous cell carcinoma were treated with MRI-based HDR. With a median follow-up of 12 months,743 2 patients had local failures.616 Other European centers645,654,656 and one Canadian center744 reported feasibility data for MR-based cervical cancer brachytherapy, showing a reduction in the normal-tissue toxicity rate. When implementing 0.5- to 1.5-T MR-based tandem/ring or tandem/ovoid brachytherapy with MRI, specific guidelines for MR use should be followed.680
Toxicities
Table 69.18 lists general toxicities in series using CT- or MR-planned brachytherapy. In the Medical University of Vienna series reporting patients treated from 2001 to 2008, 73% received concurrent cisplatin chemotherapy.653 A total of 11 grade 3 and 4 late events were recorded in 143 patients. With a median follow-up of 3.5 years, the actuarial grade 3 and 4 late morbidity at 3 and 5 years was respectively as follows: gastrointestinal, 4% and 4%; urinary, 2% and 3%; and vaginal, 1% and 3%. Two patients developed massive rectal bleeding requiring transfusions. Three patients required stoma (grade 4) for rectal wall ulceration, resulting in a fistula, a rectal perforation, and a rectovaginal fistula. Three patients developed grade 3 urinary frequency or urgency. Three patients experienced grade 3 or 4 coaptation of the vagina.
At IGR, of the 45 patients studied,651 23 and 2 developed acute grade 1 or 2 and grade 3 complications, respectively; 21 patients presented with delayed grade 1 or 2 complications. One other patient presented with a grade 3 vesicovaginal fistula. No grade 4 or greater complications, whether acute or delayed, were observed. In the IGR experience with LDR brachytherapy from 2000 to 2004, 39 late complications were reported; 13 bladder, 7 rectal, 5 small bowel, 4 urethral, 3 colic, 2 vaginal, 1 pelvic fibrosis, and 4 others. Grade 3 complications were 1 rectal, 2 bladder, and 1 urethral. Tan et al.737 reported on 28 patients treated with CT-guided brachytherapy for stage IB to IIIB cervix cancer. Their overall actuarial 3-year grade 3 and 4 morbidity rate was 14%,. Two patients had grade 3 abdominal pain and 1 had a colovaginal fistula. Overall, the data indicate that a potential reduction in morbidity appears to be a benefit of image-guided brachytherapy.
Template-Based Interstitial Brachytherapy
Interstitial implants with 226Ra, 137Cs needles, or 192Ir afterloading plastic catheters to limited tumor volumes are helpful in specific clinical situations. Indications include large residual bulky cervical tumors after external-beam treatment, residual tumor with sidewall invasion, vaginal extension, presence of a fistula and/or adjacent organ invasion, or a prior supracervical hysterectomy (Fig. 69.26). Syed-Neblett745 and Martinez746 perineal applicators are the most commonly selected. Methods for insertion have been described.598,747 A tandem should be inserted when a uterus is present.748 If the os is not visible, ultrasound guidance to determine the proper placement of the tandem is advised.749 A ring applicator modified to allow simultaneous insertion of interstitial needles750 and ovoid application with interstitial needles have been described.656
Traditionally, plain x-ray films are used for brachytherapy treatment planning. Determination of normal tissue doses and optimization is not feasible, and the risk of complications is high. In these cases, consideration of laparoscopic approaches is recommended.598 Syed et al.751 reported on 185 locally advanced cervical cancer patients treated with LDR interstitial brachytherapy from 1977 to 1997. Patients received external-beam treatment to 50.4 Gy, followed by interstitial brachytherapy to 40 to 50 Gy. Local control was 82%; 5-year disease-free survival rates were 65%, 67%, 49%, and 17% for patients with stage IB, II, III, and IV disease, respectively. Eighteen (10%) of the 185 patients developed RTOG grade 3 or 4 late complications.
Clinical outcomes using traditional techniques have been reported by several institutions. Thirty patients with stage IIB and 37 patients with stage III carcinoma received interstitial irradiation in the parametrium to supplement the dose delivered by external-beam treatment and intracavitary brachytherapy. Despite the fact that the patients treated with interstitial implant were in a high-risk group, local tumor control was comparable to that of patients treated with standard techniques.169 Pierquin et al.752 described locoregional recurrences in 6% of 53 patients with T1, 11% in 47 patients with T2, and 42% of 19 patients with T3 primary tumors of the uterine cervix treated with a combination of external-beam irradiation and the Creteil method for interstitial implantation of 192Ir sources in a plastic cervical-vaginal moulage and a uterine tandem. Prempree753 reported a 96% local tumor control rate and 61% 5-year disease-free survival rate in 23 patients with stage IIIB carcinoma of the cervix treated with a combination of external irradiation and intracavitary and interstitial implants to the parametrium. Overall, major complications were noted in 8% of the patients. Martinez et al.,746 using the Martinez Universal Perineal Interstitial applicator, treated 37 patients with advanced or recurrent carcinoma of the cervix and 26 with vaginal-urethral tumors. Doses of approximately 35 Gy were given, in addition to external irradiation (36 Gy to the whole pelvis and 14 Gy to the pelvic sidewall). They reported 6 local failures in the patients with cervical lesions and 5 in the group with vaginal-urethral tumors. The overall complication rate was 5.1%. Nag et al.754 reported on 31 patients with carcinoma of the cervix treated with external-beam radiation therapy and fluoroscopically guided interstitial brachytherapy. With a median follow-up of 36 months, 16 patients (51%) with cervical had local tumor control. The 5-year actuarial survival rate was 34%. Only 1 patient experienced grade 3 complications (2.5%).
FIGURE 69.26. A: Picture of anterior scout computed tomography showing a template-based interstitial brachytherapy application. B: Magnetic resonance imaging during interstitial needle insertion in a patient with stage IIIB cervical cancer ensures proper placement of the catheters adjacent to the tandem. The 100% isodose line is in yellow.

Recio et al.755 used laparoscopy at the time of interstitial brachytherapy in six patients with FIGO stages IIB to IVA cervical carcinoma after completion of whole-pelvis radiation; a total of 98 needles were inserted to deliver a median interstitial brachytherapy dose of 20 Gy. Eleven perforations in the pelvic peritoneum or bladder were identified during surgery in five of the six patients, leading to immediate repositioning of needles. No acute or short-term morbidity related to the procedure was noted.
Sharma et al.756 presented results on 42 patients treated from 2005 to 2007 in a prospective study of two weekly sessions of 10Gy, 1 week after finishing external-beam radiation. Median follow-up was 23 months. Delayed toxicity was 9%. The 3-year overall survival for all stages was 47% and the 3-year recurrence-free survival for stages IIB, IIIB, and IVA was 67%, 34%, and 20%, respectively. Sharma et al.757 also reported on the use of transrectal ultrasound to assist with insertion of the interstitial needles.
With image-based planning including either a CT637,638 or an MRI630 the physician evaluates the placement of the needles and may choose either to not treat specific catheters or to lower the dose given through catheters close to normal-tissue structures. An approximate 11% rate of bowel insertion and a long-term fistula rate of 4% to 10% have been reported in studies using CT for planning after insertion.637,638 When a physician has the facility to insert the applicator in a CT or MR suite while the patient is under anesthesia, an iterative process of image-guided needle insertion ensures proper placement of the catheters and prevents an inadvertent insertion into a surrounding normal-tissue structure, such as the rectum, sigmoid, or bladder.630
Dose optimization with either PDR or HDR may improve the normal-tissue doses for interstitial therapy for some patients. The University of Pittsburgh reported on 11 cervical cancer patients treated with CT-guided HDR interstitial brachytherapy (5 fractions of 3.5 Gy per fraction).758 From 1998 to 2004 interstitial brachytherapy was chosen for cases with distorted anatomy or extensive vaginal disease. The 5-year actuarial local control rate was 63%. No patient had acute grade 3 or 4 toxicity. Grade 3 or 4 late toxicity occurred in 1 patient, with a 5-year actuarial rate of 7%. Three patients had late grade 2 rectal toxicity, and 1 patient had grade 2 small-bowel toxicity.
Dimopoulos et al.720 reported on the use of tandem/ring with short interstitial needles and MR-planned HDR brachytherapy for 22 cervical cancer patients followed for a median of 20 months; no grade 3 or 4 toxicities were noted, and 1 patient had a local recurrence. Nomden et al.721 described the use of tandem/ovoid application with short interstitial needles for the second insertion with MR-planned PDR brachytherapy for 20 cervical cancer patients. They compared the first insertion with just a tandem and ovoid applicator to the second insertion, which included the addition of interstitial needles. There was an average increase in dose of 4.4 Gy (SD 2.3), with better coverage of the HR-CTV with the second insertion.
Mikami et al.759 analyzed needle applicator displacement in 10 patients treated with 30 Gy HDR in five fractions and found on daily CT scans an average of 1 to 2 mm of caudal displacement. Shifts of >3 mm were replanned. Shukla et al.760 presented data on 20 patients with cervical cancer treated with interstitial brachytherapy who underwent every-other-fraction CT imaging. The mean needle displacement was 2.5 (range, 0 to 7.4), 17.4 (range, 0 to 27.9), 1.7 (range, 0 to 6.7), 2.1 (range, 0 to 9.5), 1.7 (range, 0 to 9.3), and 0.6 mm (range, 0 to 7.8 mm) in cranial, caudal, anterior, posterior, right, and left directions, respectively. The mean displacement in the caudal direction was higher between days 1 and 2 than that between days 2 and 3 (13.4 vs. 3.8 mm; p = .01). Damato et al.,761 in a study of 10 patients treated with interstitial brachytherapy, found on average, that <1-cm displacements and deformations of the implant occurred over the course of treatment. The most significant dosimetric consequences were due to changes in organ filling rather than catheter shifts. Proper quality assurance methodologies should be in place to detect shifts that can potentially result in inadvertent insertion into normal tissue.
Brachytherapy in the Elderly
Magné et al.762 reported on 113 patients with median age of 76 years (range, 70.7 to 94.4 years) treated by conventional LDR BT as a part of their treatment. For rectal complications, grades 1/2, 3/4, and 5 (fatal) crude incidences were 19.4% (22 of 113), 1.8% (2 of 113) and 0.9% (1 of 113), respectively. Acute toxicity death occurred in 1 patient with major diarrhea associated with a hemodynamic shock. For small-bowel complications, grades 1/2 and 3/4 crude incidences were 3.5% (4 of 113) and 0.9% (1 of 113), respectively. For urinary tract complications, grades 1/2 and 3/4 crude incidences were 11.5% (13/113) and 2.7% (3/113), respectively. With a median follow-up of 3.1 years, 10 patients developed distant metastases, and 10 others had local relapses. The 3-year specific overall survival rate was 88.6% (95% CI = 77 to 92), and the corresponding disease-free survival rate was 81% (95% CI = 72 to 88). Age did not influence the effectiveness of BT in elderly patients, and BT should be considered whenever possible, even in elderly patients presenting with a cervix cancer.
TABLE 69.21 CARCINOMA OF THE UTERINE CERVIX: INCIDENCE OF CENTRAL/PELVIC RECURRENCES CORRELATED WITH METHOD OF THERAPY

Image-Guided Brachytherapy Versus External-Beam Boost
Studies of external-beam treatment as an alternative boost instead of brachytherapy demonstrate significantly inferior survival rates compared to those that use brachytherapy. The use of ultrasound enables tandem placement in most cases, even when the os cannot be identified, and should be attempted for difficult cases. Barraclough et al.763 reported on 44 patients with cervical cancer who did not receive brachytherapy and were treated with EBRT to 54 to 70 Gy via a three-dimensional conformal boost. After a median follow-up of 2.3 years, 48% relapsed, with 16 of 21 developing a central recurrence. The 5-year overall survival rate was 49%, which is much lower than for brachytherapy-treated patients.
The dosimetry of brachytherapy cannot be adequately mimicked by external-beam techniques. A treatment planning report compared inversely planned EBRT with photons (IMRT) and protons (IMPT) to 3D MRI-guided brachytherapy.764 EBRT was planned to deliver the highest possible doses to the PTV while respecting D2cc limits from brachytherapy, assuming the same fractionation. Volumes receiving 60 Gy (in equivalent dose in 2-Gy fractions) were approximately twice as large for IMRT compared with brachytherapy, and the high central tumor dose was lower than that seen with brachytherapy. Both IMRT and protons were inferior to 3D image-based brachytherapy.
With IMRT, there is a need for replanning due to rapid tumor regression317–319 and an increase in integral dose, with normal tissues throughout the pelvis receiving more radiation than with brachytherapy. Given the large movement and the increased dose to the normal tissues resulting in an increase in normal-tissue toxicity, highly conformal (IMRT, IGRT, SBRT) methods for boosting the cervix are not routinely recommended. Every effort should be made to use image guidance to insert a tandem into the uterus in order to provide adequate brachytherapy doses for all cervical cancer cases receiving radiation.
External-Beam Irradiation Alone
Occasionally, brachytherapy procedures cannot be performed because of medical reasons or unusual anatomic configuration of the pelvis or the tumor (i.e., extensive lesion, inability to identify the cervical canal). These patients may be treated with higher doses of external-beam irradiation alone, although survival is significantly worse than when brachytherapy is implemented, and normal-tissue toxicity is higher due to the excessive dose to the rectum and bowel. Therefore, every attempt to treat with brachytherapy should be made because brachytherapy moves with the patient and provides high regions of dose in the central regions of the tumor. With IMRT, a significantly higher normal-tissue dose is administered, and the desired high central regions of radiation cannot safely be administered.
Coia et al.,521 in an analysis of 565 patients with various stages of cervical carcinoma treated in the Patterns of Care Study, reported better survival (67%) and pelvic tumor control (78%) for patients treated with external irradiation and brachytherapy than for patients who had no intracavitary brachytherapy applications (36% 4-year survival rate and 47% in-field failure rate). Patients treated with two intracavitary applications had a higher 4-year survival rate (73%) and in-field tumor control rate (83%) than those receiving only one application (60% 4-year survival rate and 71% in-field tumor control rate).
Hanks et al.525 and Montana et al.524 reported a higher incidence of central pelvic recurrences in patients with stage III cervical carcinoma treated with external-beam therapy alone than in patients receiving brachytherapy in addition to external-beam irradiation (Table 69.21). The incidence of major complications was similar in both groups of patients.
Akine et al.765 treated 104 patients with carcinoma of the uterine cervix with external irradiation alone (anteroposterior–posteroanterior or four-field box techniques) because of inability to perform intracavitary brachytherapy. Average doses delivered were 50 Gy to the whole pelvis, followed by additional doses with reduced portals to deliver a total of 60.8 Gy in 6 weeks, 72.3 Gy in 7.5 weeks, or 80.5 Gy in 8 weeks, with a daily dose of 1.9 or 2 Gy. The local tumor control rate was 27% for stage II, 19% for stage III, and 15% for stage IVA disease. The 5-year survival rates were 36%, 17%, and 5%, respectively. Four patients had major complications (usually proctitis) that required surgical treatment, and 1 patient died of rectal bleeding. Eight of 23 patients treated with conformal therapy had control of the tumor and survived 5 years without major complications. Saibishkumar et al.766 treated 146 patients with cervix cancer with EBRT alone (60 to 66 Gy) because of unsuitability for brachytherapy; 5-year pelvic tumor control was 21.9% and DFS was 11.6%.
Cost-Effectiveness of LDR Versus HDR Brachytherapy
Wright et al.767 developed a questionnaire to elicit patient preference for two brachytherapy methods (one LDR or three HDR fractions and two HDR or five HDR fractions, assuming both methods to be isoeffective). The questionnaire was completed by 90 female staff members at their center, 18 previously treated patients, and 20 newly diagnosed patients. When both methods were assumed to be isoeffective, only 34% of the 38 patients preferred three HDR fractions to one LDR fraction. However, when HDR was assumed to be 2% more curative or 6% less toxic, 50% said they would prefer the HDR therapy. Both preference and strength of preference for LDR were significantly associated with a greater traveling distance for treatments. More studies on resource utilization768 are needed.
Alternative Isotopes
Californium has been proposed as an alternative to iridium as the radioactive isotope. Maruyama and Muir769 reported on 41 patients with stage IB cervix cancer treated with 40 to 50 Gy to the whole pelvis followed by a 5- to 15-Gy boost to the lateral pelvic wall and a single 252Cf-neutron brachytherapy insertion in approximately 8 hours. Nearly total tumor clearance was achieved in >90% of the patients; tumor regression was more rapid in the 252Cf group than in similar patients treated with 137Cs and the same external-beam irradiation dose.
FOLLOW-UP
After treatment, patients should be regularly followed by both the radiation and the gynecologic oncologist. Careful history taking and a complete physical and pelvic/rectal examination usually are performed every month for the first 3 months after completion of irradiation, every 3 months for the remainder of the first year, every 4 months the second year, every 6 months during the third through the fifth year, and yearly thereafter. The use of Pap smears for cervical and vaginal cytology as a follow-up study is controversial because of postirradiation cellular morphology that renders it difficult to distinguish postirradiation changes from residual or recurrent malignant cells.770,771 DNA analysis of postirradiation cytologic smears demonstrating atypia or dysplasia may provide ancillary information.772
Rintala et al.773 evaluated the reliability of cytologic analysis and atypia after radiation therapy in 89 patients treated for cervical carcinoma. A total of 697 Pap smears were taken; during the follow-up, 44 patients had recurrent disease, which was local in 17 (39%) cases. The rate of false-positive samples was only 3%. Radiation-induced atypia was detected in 28% of the Pap smears taken during the first 4 months after radiation therapy, and its incidence decreased thereafter. In 1,000 patients treated with either surgery or radiation therapy at the MD Anderson Cancer Center for stage IB cervical cancer posttreatment, Pap smears did not detect a single asymptomatic recurrence among 133 patients with recurrent disease.774
The presence of apparently viable tumor cells in the cytologic smear 3 months after irradiation should be evaluated with cervical biopsies, dilation and curettage, and careful examination under anesthesia, as indicated.
Complete blood counts and chemistry profile tests are obtained as clinically indicated. Chest radiography is commonly obtained on a yearly basis, usually for the first 5 years posttreatment, although its value to detect curable lung metastasis is not proven. If radiographs are consistently negative, obtaining them every other year thereafter may be sufficient.
Other imaging studies, such as CT, MRI, PET scanning, or bone scans, are obtained when clinically warranted. When persistent or recurrent tumor is suspected, biopsies should be obtained for histologic confirmation. If a biopsy is positive, immediate treatment should be instituted, as is discussed later.
Usually, hematometra after radiation therapy for cervical carcinoma is related to recurrent disease but occasionally may be related to estrogen replacement therapy, endometrial activity, or fibrosis and obliteration of the endocervix.775
TREATMENT OF RECURRENT CARCINOMA OF THE CERVIX
After Previous Surgery
Radiation may salvage approximately 50% of patients with localized pelvic recurrences after surgery alone. A combination of whole-pelvis external irradiation (45 to 50 Gy) with concurrent chemotherapy followed by interstitial brachytherapy is recommended. If the tumor lies outside of an accessible region for brachytherapy, dose escalation with conformal or IMRT techniques may be attempted, depending on the location of the tumor and the need to protect bowel, with at least 65 to 70 Gy necessary for adequate control. In the setting of recurrent disease, the total mucosal dose from the external and brachytherapy can approach 140 Gy to the upper vagina and 95 Gy to the distal vagina without a high risk.776 With interstitial brachytherapy, doses of 20 to 35 Gy are administered with single, double-plane, or volume implants, for a total tumor dose of approximately 80 Gy, depending on the extent of the tumor.
Larson et al.777 observed 27 recurrences (11%) in 249 patients treated with radical hysterectomy and pelvic lymphadenectomy for stage IB carcinoma of the cervix; 17 (63%) had tumor recurrence in the pelvis or vulva; the other 10 patients had recurrences outside the pelvis. Eight of 15 patients (53%) treated with irradiation for an isolated recurrence in the pelvis or vulvar region were tumor free between 10 and 126 months after treatment of the recurrence (median, 48 months).
Ijaz et al.778 reported on 50 patients treated with RT for an isolated pelvic recurrence of cervical carcinoma after radical hysterectomy; 7 patients were treated with palliative intent using hypofractionated RT. The remaining 43 patients were treated with curative intent, 33 with RT only and 10 with cisplatin-based chemoirradiation. The overall 5-year survival rate was 33% for all 50 patients, 39% for the 43 patients treated with curative intent, and 25% for patients with isolated sidewall recurrences treated with curative intent. Three patients experienced late treatment complications.
Hille et al.779 described results in 17 patients with recurrent cervix cancer (9 had a complete microscopically incomplete resection) treated with EBRT and brachytherapy to 50 to 65 Gy. The 5-year pelvic tumor control was 48%, and relapse-free survival was 24%.
After Definitive Irradiation
Reirradiation of previously irradiated patients must be undertaken with extreme caution. It is very important to analyze the techniques used in the initial treatment (beam energy, volume, doses delivered with external or intracavitary irradiation). In addition, the period of time between the two treatments must be taken into consideration because it is postulated that some repair of the initial damage may take place in the interval. In general, external irradiation for recurrent tumor is given to limited volumes (40 to 45 Gy, 1.8-Gy tumor dose per fraction, preferentially using lateral portals). Occasionally, intracavitary or interstitial irradiation can be used to treat relatively circumscribed recurrences.
Sommers et al.780 described the results of retreatment in 376 patients with recurrent carcinoma of the uterine cervix. Ninety-one patients received irradiation, mostly external (86.8%), occasionally combined with brachytherapy (7.7%) to control bleeding of central recurrences; brachytherapy alone was administered in 5.5% of patients. The usual dose for recurrent pelvic masses was 40 to 45 Gy, and for para-aortic lymph node metastases it was 45 to 50 Gy in 5 weeks. Other metastatic sites were treated with 35 to 40 Gy in 3 to 4 weeks. Pelvic exenteration was attempted in 23 patients, only 10 of whom were deemed to be operable (43.5%), but it was completed in only 7. The probability of 5-year survival after treatment for recurrence was 30% with combined surgery and external irradiation, 12% with surgery alone, and 4% with external irradiation alone. The 5-year survival rate for 10 patients who underwent pelvic exenteration was 16%. Only 1% of the untreated patients survived 5 years. Six of 140 patients (4.3%) experienced grade 2 or 3 complications.
Selected patients with limited pelvic recurrences not fixed to the pelvic wall and without evidence of extrapelvic metastases can be potentially salvaged by radical hysterectomy or pelvic exenteration. Coleman et al.781 described results in 50 patients who underwent radical hysterectomy for persistent (18 patients) or recurrent (32 patients) cervical cancer after primary radiation therapy. Lymph node metastases were identified in 5 of 39 patients (13%) in whom the lymph nodes were evaluated. The 5- and 10-year survival rates were 72% and 60%, respectively.
In 65 patients on whom pelvic exenteration was carried out at Memorial Sloan-Kettering Cancer Center, the 5-year survival rate was 23%.782 The operative mortality rate was 9.2%. The authors pointed out that the significant mortality and morbidity associated with this procedure preclude its use as palliative therapy.
Urinary diversion, either by nephrostomy or ileal bladder, may be of palliative value in patients with either recurrent carcinoma in the pelvis or complications. It must be kept in mind that diversion may prolong life but runs the risk of denying a terminally ill patient with cancer the oblivion and insensibility of uremia.
Kastritis et al.783 treated 200 patients with stage IV or recurrent cervix cancer with cisplatin-based chemotherapy; response rate was 43.5% in 142 patients with squamous cell and 53.5% in 58 patients with nonsquamous tumors (p= .79). Median survival was 11.57 and 19 months, respectively. Tinker et al.784 treated 25 women for recurrent cervical cancer with carboplatin–paclitaxel and noted a 20% cure rate and 20% progression rate, with median survival of 21 months. Brewer et al.511 in 32 women, all of whom had previous chemotherapy and 29 of whom had previous RT, used cisplatin and gemcitabine, with a progression rate of 22% and median time to progression of 3.5 months.
Para-Aortic Lymph Node Recurrences
Isolated recurrences in the para-aortic nodes after pelvic irradiation have been described in about 3% of patients, and some may be salvaged with aggressive therapy. The advent of IMRT makes treatment easier, with less morbidity.
Kim et al.785 treated 12 patients with isolated para-aortic lymph node metastasis with hyperfractionated RT (60 Gy in 1.2-Gy fractions twice a day) and concurrent cisplatin–paclitaxel. Fields extended from the superior plate of T12 to the lower plate of L5. Three-year survival was 19%. Grade 3 or 4 hematologic toxicity developed in two patients. Singh et al.786 detected 14 isolated para-aortic lymph node metastases in 816 patients previously treated with RT; these women were subsequently treated with RT to the para-aortic lymph nodes combined with concurrent chemotherapy. Seven patients survived 5 years.
In a review of 1,955 patients treated with RT for cervix cancer, Jhingran et al.,787 identified 120 patients with recurrent tumor above the pelvic fields. Initially, 10 had common iliac and 5 had para-aortic node involvement. In 104 patients, recurrences were immediately adjacent to the upper borders of the RT fields. In 15 patients treated with curative intent for the para-aortic lymph node recurrence, 5-year survival was 25%.
Intraoperative Irradiation
Intraoperative radiation therapy (IORT) has been used for treatment of locally advanced and recurrent carcinoma of the cervix, with 3-year survival rates of 8% to 21% as reported by Mahé et al.788 and Garton et al.,789 and a 5-year survival rate of 33% in 14 patients described by Kinney et al.423 Patient selection may have had an impact on the different results. Abe and Shibamoto790 noted that central recurrences, particularly in nonirradiated patients, and resection of the gross recurrent tumor in irradiated patients improve the benefit from IORT. Significant toxicity included peripheral nerve injury and ureteral stenosis (with doses >15 to 20 Gy).
IORT was used in 70 patients with pelvic recurrences in a European cooperative study.791 Complete tumor resection was carried out in 30 patients, partial in 37, and unspecified in 3. Sixty-five patients had electron beam therapy (12 to 25 MeV), with mean doses of 18 Gy (10 to 25 Gy) after gross complete resection and 19 Gy (10 to 30 Gy) after partial resection. The 3-year overall survival rate was 8%. Grade 2 or 3 toxicity was observed in 19/70 patients (27%), with 10 complications being related to IORT.
Martinez-Monge et al.792 reported a study of IORT in 26 patients with recurrent gynecologic tumors, some relapsing after full-dose radiation therapy (group 1) or after surgery (group 2). Cervical carcinoma was the initial tumor site of involvement in 18 patients. Treatment consisted of maximal surgical resection and IORT (10 to 25 Gy) to high-risk areas. Patients not previously irradiated also received external-beam irradiation (with or without chemotherapy) before or after surgery. There was 1 IORT-related incidence of motor neuropathy. The local tumor control rates were 33% and 77%; the 4-year actuarial survival for group 1 was 7%, and the 6-year actuarial survival rate for group 2 was 33%.
In another study, 42 patients with stages IIA to IVA cervical cancer initially received 50.4 Gy of pelvic external-beam radiation with concurrent cisplatin and 5-fluorouracil.793 Patients then underwent radical surgery 6 to 8 weeks later with IORT. The 5-year DFS and OS were 46% and 49%, respectively, which are inferior to reported results with standard concurrent chemoradiation followed by brachytherapy without surgery or IORT. Therefore, this regimen remains of questionable value in potentially curable patients.
URGENT BLEEDING AND PALLIATIVE IRRADIATION
Frequently, the radiation oncologist is faced with the challenge of treating a patient with stage IVB or recurrent carcinoma requiring palliation of pelvic pain or bleeding. Tumors respond rapidly to radiation, and bleeding usually resolves within a few days of treatment. If vaginal bleeding is the main concern, several possibilities may be effective. In a descriptive review of eight papers that presented palliative treatment data,794 five papers were found to report using 10 Gy per fraction, with the best resolution of bleeding and/or pain when each 10-Gy fraction was given at 3- to 4-week intervals for a total of three fractions. Alternatively, common palliative regimens used in other sites of the body, such as 4 Gy for five or six fractions or 3 Gy for 10 fractions, may be implemented. Patients who present with a new diagnosis may be treated with 3 to 4 Gy for two or three fractions, followed by standard 1.8 Gy to approximately 39.6 Gy and then brachytherapy. Alternatively, patients may receive 1 to 2 days of 1.8 Gy twice a day, switching to once-a-day treatment with 1.8 Gy per fraction after bleeding has stopped on day 2 or 3, completing treatment after 45 Gy and then commencing routine brachytherapy.
A single LDR intracavitary insertion with tandem and colpostats for approximately 6,000 mgh (55 Gy to point A) may be used for palliation. If irradiation was delivered previously, lower intracavitary doses should be prescribed (4,000 to 5,000 mgh). Grigsby et al.795 used two fractions of HDR brachytherapy with a ring applicator (once weekly) with control of bleeding in 14 of 15 patients.
Several high-dose fractionation schedules with external-beam radiation have been used. Spanos et al.796 reported on a phase II study of daily multifractionated split-course irradiation in 142 patients with recurrent or metastatic disease in the pelvis. Irradiation consisted of 3.7 Gy per fraction given twice daily for 2 consecutive days, repeated at 3- to 6-week intervals for a total of three courses, aiming at a total tumor dose of 44.4 Gy. Occasionally, this regimen was combined with an LDR intracavitary insertion (4,500 mgh), blocking the midline for the last 14.4-Gy external dose. Twenty-seven patients survived >1 year. There were only 2 recorded cases of grade 3 toxicity (lower gastrointestinal tract). This study was expanded to a phase III protocol randomizing 136 patients between a short (2 weeks) or a longer (4 weeks) rest period between the split courses of irradiation.797 There was a trend toward increased acute toxicity in patients with shorter rest periods (5 of 58 vs. 0 of 68; p = .07). Late toxicity was not significantly different in the two groups. Pelvic tumor response was comparable in both groups (34% vs. 26%). Spanos et al.798 reported a 6% complication rate in 290 patients treated in RTOG Protocol 85–02. No patient receiving <30 Gy experienced late toxicity. There was no significant difference in the incidence of complications for patients with a 2- or 4-week rest (p = .47).
IRRADIATION AND HYPERTHERMIA
Because of technical limitations in the delivery of adequate heat to large parts of the body such as the pelvis, the use of hyperthermia in the treatment of carcinoma of the uterine cervix has been rare. Hornback et al.799 described a nonrandomized study in which the combination of microwave hyperthermia (433 MHz) and irradiation resulted in improved pelvic tumor control (72%) in a group of 79 patients with stage IIIB carcinoma compared with previously irradiated historic control patients (53%). However, 5-year survival rates were comparable in both groups (22% to 30%).
Sharma et al.800 reported a 70% disease-free survival rate at 18 months in 20 patients with stage IIB or III carcinoma of the uterine cervix treated with a combination of irradiation and hyperthermia (13.5 MHz, 42°C to 43°C, 30 minutes before irradiation) in comparison with a 50% disease-free survival rate in 22 patients treated with irradiation alone. The grade 3 complication rate (8%) was similar in both groups.
Dinges et al.801 treated 18 patients with advanced carcinoma of the cervix with RT plus hyperthermia (in the first and fourth weeks, two regional hyperthermia treatments were applied). The acute toxicity was low and similar to that with RT alone. The local tumor control was 48% at 2 years.
Harima et al.802 evaluated radiation therapy or thermoirradiation (three sessions of hyperthermia) for stage IIIB cervical carcinoma; two groups of 20 patients each were randomly divided. A complete response was achieved in 50% (10 of 20) in the RT group versus 80% (16 of 20) in the thermoirradiation group (p = .048). The 3-year overall survival and disease-free survival rates for the patients who were treated with thermoirradiation (58.2% and 63.6%) were better than with RT (48.1% and 45%), but differences were not statistically significant. The 3-year local relapse-free survival rate of the patients who were treated with thermoirradiation (79.7%) was significantly better than that of the patients treated with irradiation alone (48.5%; p = .048). Thermoirradiation was well tolerated and did not add to either acute or long-term toxicity over radiation alone.
Vasanthan et al.803 reported on 110 patients with locally advanced cervix cancer randomized to treatment with RT alone or combined with hyperthermia (minimum five sessions, 60 minutes each, once weekly). Overall 3-year pelvic tumor control was 68.5% and survival was 73.2%, with no difference in either group, although survival was lower in the patients with stage IIB treated with hyperthermia. Acute toxicity was 18% (10 of 55) in the hyperthermia patients and 4% (2 of 55) with RT alone. Late toxicity was not different in the two arms.
A Cochrane database review identified six randomized, controlled trials published between 1987 and 2009 comparing RT versus combined hyperthermia and RT. The results show 74% of patients had stage IIIB cervical cancer. A significantly higher complete response rate (RR = 0.56, 95% CI = 0.39 to 0.79) and lower local recurrence rate (HR = 0.48, 95% CI = 0.37 to 0.63) and improved overall survival (HR = 0.67, 95% CI = 0.45 to 0.99) with no difference in acute or late grade 3 to 4 toxicity were seen for patients treated with combined therapy.804 Catheter-based ultrasound devices provide a method to deliver heat with HDR brachytherapy, but clinical results are not yet available.805
SIDE EFFECTS: SURGERY AND RADIATION
Descriptions of sequelae vary among institutions because toxicity-grading scales are not uniform and the scoring system for complications is not clearly stated in all reports. Surgical side effects alone depend on the extent of surgery and the amount of disease. Radical hysterectomy alone may cause long-term side effects such as urinary retention requiring chronic suprapubic catheter placement, sciatic nerve injury, postoperative seroma or hematoma formation, pelvic pain, or, when lymphadenectomy, is performed, life-long edema. Oophorectomy also may induce menopause; hysterectomy removes the ability to carry a pregnancy, and removal of a portion of the vagina may significantly change sexual function if vaginal shortening is severe.
With improved anesthesia, surgical techniques, and antibiotic therapy, the mortality rate for radical hysterectomy with pelvic lymphadenectomy has decreased to 1% or less. The most frequent sequela after radical hysterectomy is urinary dysfunction as a result of partial denervation of the detrusor muscle. Patients may have various degrees of loss of bladder sensation, inability to initiate voiding, residual urine retention, and incontinence.
In 375 patients treated with a modified radical hysterectomy for various gynecologic disorders, Magrina et al.,806 observed some form of postoperative (within 42 days of surgery) complications in 89 patients (24%). Patients who had a pelvic lymphadenectomy experienced a greater incidence of lower-extremity lymphedema than those who did not undergo this procedure. Preoperative or postoperative pelvic irradiation was a significant predisposing factor for urinary tract infection, lymphedema, and bowel obstruction in these patients compared with those who did not receive pelvic irradiation.
Some loss of defecatory urge associated with chronic rectal dysfunction was observed by Barnes et al.807 after radical hysterectomy. Manometric studies suggest a disruption of the spinal arcs controlling defecation.
Other complications include ureterovaginal fistula (the incidence of which has decreased to <3%), hemorrhage, infection, bowel obstruction, stricture and fibrosis of the intestine or rectosigmoid colon, and bladder and rectovaginal fistulas. Postsurgical complications are usually more amenable to correction than are late complications after irradiation.
When postoperative radiation therapy is given to selected patients, further complications of the additional therapy are expected. The main areas of side effects due to radiation are bowel, bladder, skin, and sexual function. Because of intestinal adhesions to denuded surfaces in the pelvis, enteric complications, such as obstruction, fistula, or dysfunction, were observed in 24% of patients reported by Fiorica et al.808 Other investigators, however, have reported no increase in the incidence of severe complications in patients treated with postoperative irradiation.387,821
Lower body mass index (BMI) is correlated with an increase in toxicity. A total of 404 patients with stage IB1 cervical cancer with positive lymph nodes or stage IB2 or higher were treated from 1998 to 2008. A BMI of <18.5 was associated with a decreased overall survival (HR = 2.37, p < .01). Grade 3 and 4 complications appeared to trend higher; overall, 17% versus 14%; specifically for fistula, 11% versus 9% (p = .05), for bowel obstruction, 33% versus 4% (p < .01), and for lymphedema, 5.6% versus. 1.2% (p= .0).809
Montz et al.810 evaluated bowel obstruction in 98 patients undergoing radical hysterectomy for a nonadnexal gynecologic malignancy. The incidence of small-bowel obstruction was significantly higher (p < .05) in patients who received concomitant radiation therapy (20%). None of these patients had recurrent disease at the time of small-bowel obstruction. Findings at surgery consisted of minimal incisional adhesions but extensive matted small-bowel loops adherent to the pelvic operative sites.
When irradiation is combined with surgery, the complication rate tends to be somewhat higher, particularly because of injury to the ureter or the bladder (ureteral stricture or ureterovaginal or vesicovaginal fistula).811 The dose of irradiation, technique, and type of surgical procedure performed are important in determining the morbidity of combined therapy. Jacobs et al.,812 in 102 patients with invasive cervical carcinoma treated with low-dose preoperative irradiation and radical hysterectomy with lymphadenectomy or high-dose preoperative irradiation and conservative extrafascial hysterectomy, noted a major complication rate of 5%. After combined treatment, some degree of lymphedema may be observed (30% to 40%).
A significant number of complications are associated with pretherapy staging laparotomy, particularly if irradiation (>55 Gy) is given to metastatic para-aortic lymph nodes. The incidence of complications is between 5% and 20%, depending on the extent of the para-aortic lymph node dissection, use of transperitoneal or retroperitoneal approach for the operation, and dose of irradiation given.279
TABLE 69.22 CARCINOMA OF THE UTERINE CERVIX: GRADE 2 SEQUELAE (WASHINGTON UNIVERSITY, 1959–1989)

TABLE 69.23 CARCINOMA OF THE UTERINE CERVIX: GRADE 3 SEQUELAE (WASHINGTON UNIVERSITY, 1959–1989)

Late Sequelae—Overall
The incidence of major late sequelae of radiation therapy for stages I and IIA carcinoma of the cervix ranges from 3% to 5% and for stages IIB and III between 10% and 15%. The most frequent major sequelae for the various stages are listed in Tables 69.22 and 69.23. Injury to the gastrointestinal tract usually appears within the first 2 years after radiation therapy, whereas complications of the urinary tract are seen more frequently 3 to 5 years after treatment.570,816 Pedersen et al.,813 in a review of morbidity of radiation therapy in 442 patients with cervical cancer stages IIB, III, and IVA, recommended that actuarial estimates rather than frequency of sequelae be reported to avoid underestimation of risks of late morbidity after radiation therapy in long-term survivors. In fact, Eifel et al.,811 in 1,784 patients with stage IB carcinoma of the cervix, noted that the greatest risk of sequelae is in the first 3 years after therapy. The risk of rectal complications declined after the first 2 years of follow-up to 0.6%/year, whereas the risk of major urinary tract complications for survivors continued at 0.3%/year, with a 20-year actuarial risk of major complications of 14.4%.
Montana et al.,524 Perez et al.,814 and Pourquier et al.815 noted a greater incidence of complications with higher doses of irradiation. Perez et al.816 and Pourquier et al.815 reported that with doses <75 to 80 Gy delivered to limited volumes, grade 2 and 3 complications in the urinary tract and rectosigmoid were approximately 5%. However, the incidence increased to >10% with higher doses of irradiation to these organs (Fig. 69.27). Doses >60 Gy were also correlated with a greater incidence of small-bowel injury (Fig. 69.28). The same analysis showed that patients who experienced sequelae of therapy had slightly better survival rates than patients without any complications. This was related to improved tumor control with higher doses of irradiation.816
Perez et al.814 quantitated the effect of total doses of irradiation, dose rate, and ratio of doses to bladder or rectum and point A on sequelae in 1,456 patients treated for cervical cancer with external-beam irradiation plus two LDR intracavitary insertions to deliver 70 to 90 Gy to point A. Median follow-up was 11 years. In stage IB, the frequency of grade 2 morbidity was 9%, and in grade 3 it was 5%; in stages IIA, IIB, III, and IVA, the frequency of grade 2 morbidity was 10% to 12% and that of grade 3 was 10%. The most frequent grade 2 urinary/rectal sequelae were cystitis and proctitis (0.7% to 3%). The most common grade 3 sequelae were vesicovaginal fistula (0.6% to 2% in patients with stage I to III tumors), rectovaginal fistula (0.8% to 3%), and intestinal obstruction (0.8% to 4%). In the bladder, doses <80 Gy correlated with a <3% incidence of morbidity, which was 5% with higher doses (p = .31). In the rectosigmoid, the incidence of significant morbidity was <4% with doses <75 Gy and increased to 9% with higher doses. For the small intestine, the incidence of morbidity was <1% with 50 Gy or less, 2% with 50 to 60 Gy, and 5% with higher doses to the lateral pelvic wall (p = .04). Multivariate analysis showed that dose to the rectal point was the only factor influencing rectosigmoid sequelae, and dose to the bladder point affected bladder morbidity.
In a review of the Patterns of Care Study, Lanciano et al.817 observed a 5-year actuarial rate of 14% for major late complications in 1,558 patients treated with irradiation for invasive carcinoma of the cervix. Women <40 years of age or with a history of prior surgery or laparotomy for staging had a greater incidence of significant morbidity (15% to 18% vs. 8% to 9%). In addition, EBRT dose per fraction of >2 Gy, paracentral doses of 85 Gy or greater, and lateral parametrial doses >60 Gy were independently associated with a higher complication rate.
Lee et al.,818 using 3-Gy fractions with EBRT, calculated the rectal point dose in the anterior wall at the level of the cervical os and noted that total higher BED (142.7 Gy) was associated with more frequent rectal sequelae compared with BED of <131 Gy.
Mitchell et al.124 evaluated 398 patients with stages I to III cervical carcinoma treated with radiation therapy. Patients were divided into nonelderly (35 to 69 years of age; n = 338) and elderly (≥70 years of age; n = 60) groups. The frequency and severity of acute and chronic sequelae were equivalent in both groups.
FIGURE 69.27. Incidence of moderate or severe genitourinary (A) or rectosigmoid (B) complications in patients with carcinoma of uterine cervix (all stages) treated with irradiation alone (external and brachytherapy). A greater frequency of complications is noted with maximum doses of >75 to 80 Gy to the bladder or rectum.

FIGURE 69.28. Incidence of moderate or severe complications in small intestine correlated with doses of irradiation.

Gastrointestinal Toxicity
When late radiation proctitis occurs, initial treatment is the same as for acute proctitis. If the symptoms and rectal bleeding persist, laser treatment of rectal telangiectasis or ulcers is frequently beneficial. Roche et al.819 treated six patients with hemorrhagic radiation-induced proctitis using outpatient intrarectal application of formaldehyde 4%. In four cases the bleeding ceased after the first formaldehyde application; two patients continued to bleed, but another application 3 weeks later definitively controlled the hemorrhage. There were no complications, such as burns or late stenosis of the deep layers of the rectum, and this technique was well tolerated. Rubinstein et al.820 and Seow-Choen et al.821 also reported treatment of radiation proctitis with a similar technique. Patients are sedated, a local anesthetic block is administered, and a sponge moistened with 4% formalin is applied for 4 minutes to each bleeding area of the rectum. Care is taken to protect the perianal skin from any caustic effects of the formalin.
Occasionally, a colostomy is necessary if conservation management fails. The importance of performing colonoscopy in patients with rectal bleeding to exclude other lesions in the colon, including polyps or cancer, is emphasized. If routine screening colonoscopy is not urgent, unless there is a medical reason, the colonoscopy may be postponed until 1 year after pelvic radiation to ensure no issues with poor wound healing, bleeding, or ulceration secondary to biopsy performed at the time of colonoscopy.
Anal incontinence is occasionally observed. This sequela must be assessed in light of a report by Nelson et al.,822 who in a survey of 6,959 nonirradiated patients, identified 153 (2.2%) who reported anal incontinence, without specific etiology. Thirty percent of incontinent subjects were >65 years of age, and 63% were women. Of those with anal incontinence, 36% were incontinent to solid feces, 54% to liquid feces, and 60% to gas.
Kim et al.823 investigated the effects of radiation on anorectal function using manometry in 24 patients with carcinoma of the uterine cervix who had late radiation proctitis. These data were compared with those from 24 age-matched nonirradiated female volunteers. Regardless of the severity of proctitis symptoms, 75% of irradiated patients exhibited abnormal manometric parameters for sensory or motor functions. Radiation damage to nerves and to the external sphincter muscle was considered to be an important cause of motor dysfunction.
Quilty824 noted a greater incidence of pelvic complications in patients treated with higher doses to the whole pelvis (40 to 50 Gy). The author commented that the intracavitary radium dose was not correlated with severe complications. Similar observations were made by Stryker et al.825 who recorded a 9% incidence of fistulas and a 14% incidence of grade 2 and 3 complications in 132 patients after delivery of 50 Gy or higher to the whole pelvis (1.8-Gy daily dose) combined with intracavitary insertion. They recommended that the whole-pelvis dose should not exceed 40 to 45 Gy when doses of approximately 40 Gy are delivered to point A with LDR intracavitary insertions.
Kuske et al.,719 reported results of therapy in 99 patients with carcinoma of the cervix on whom minicolpostats were used, noted a 15% incidence of grade 2 and 3 complications, which was higher than the 8% incidence noted in a similar group of patients treated with regular colpostats during the same period (p = .08).
Perez et al.816 reported that the incidence and type of complications with interstitial therapy were approximately the same as in patients treated with intracavitary technique only. In contrast, Kasibhatla et al.826 noted 6% small-bowel obstruction in 36 women with gynecologic cancer treated with EBRT and interstitial brachytherapy, which was aggravated by previous abdominopelvic surgery. The 3-year risk of rectovaginal fistula was 18%, and it was significantly higher in patients who received total doses of >76 Gy (100% vs. 7%; p = .009).
Irradiation of the para-aortic lymph nodes has been reported to cause increased morbidity, particularly if it is done after transperitoneal staging para-aortic lymphadenectomy. In a randomized study reported by Rotman et al.,827 a somewhat higher incidence of grade 2 and 3 complications was reported in 170 patients (10 complications) given 45 Gy to the para-aortic area in addition to standard pelvic irradiation, compared with 5 complications in 167 patients treated by pelvic irradiation only. The incidence of fatal (grade 5) complications was 4 and 1, respectively. In a similar randomized study by Haie et al.541 for the EORTC, the incidence of grade 3 small-bowel injury was 2.3% in the para-aortic irradiation group and 0.9% in the pelvic irradiation–only group. The overall incidences of severe complications were 9% and 4.8%, respectively.
Willett et al.828 reported on 28 patients with inflammatory bowel disease (10 with Crohn’s disease, 18 with ulcerative colitis) who underwent external-beam abdominal or pelvic irradiation. Patients were treated either by specialized techniques (16 patients) to minimize small- and large-bowel irradiation or by conventional approaches (12 patients). The overall incidence of severe toxicity was 46% (13 of 28 patients), and 6 patients (21%) experienced severe acute toxicity necessitating cessation of radiation therapy. Late toxicity requiring hospitalization or surgical intervention was observed in 8 of 28 patients (29%). For patients treated with conventional approaches, the 5-year actuarial rate of late toxicity was 73% versus 23% for patients treated by specialized techniques (p = .02). In patients with inflammatory bowel disease abdominal or pelvic irradiation, must be used judiciously.
In contrast, Song et al.,411 in a review of 24 patients with a history of inflammatory bowel disease who received RT (median dose of 45 Gy in 1.8- to 3-Gy fractions) to fields encompassing some portion of the gastrointestinal tract, noted that 5 patients (21%) experienced acute intestinal toxicity of grade 3 or greater and 2 (8%) had grade 3 or greater late intestinal toxicity. Fifteen patients also received concurrent chemotherapy. The authors believed that the gastrointestinal toxicity in these patients was more modest than generally perceived. Tiersten and Saltz829 noted that five patients with inflammatory bowel disease and gastrointestinal malignancy completed planned radiation therapy (30 to 54 Gy), usually with concurrent 5-FU, without difficulty.
Salama et al.443 reported preliminary observations on acute toxicity with extended-field IMRT in 13 patients with gynecologic cancer. With median follow-up of 11 months, 2 patients treated with chemoradiation experienced grade 3 or higher morbidity and 1 (with a history of previous surgeries) developed small-bowel obstruction.
Levenback et al.830 identified 116 of 1,784 patients (6.5%) with stage IB carcinoma of the cervix treated with irradiation in whom hemorrhagic cystitis developed, 23% grade 2 (repeated minor bleeding) and 18% grade 3 (hospitalization required for medical management). The median interval to onset of hematuria was 35.5 months. The risk of severe hematuria requiring surgical intervention was 1.4% at 10 years and 2.3% at 20 years. Minor episodes of hematuria are managed by antibiotic therapy. Cystoscopic, laser, or cautery treatment of bleeding points is indicated. Clot evacuation and continuous bladder irrigation are important elements in the acute management of patients with heavy bleeding. Occasionally, a urinary diversion is required for intractable severe hematuria.
Genitourinary Toxicity
Ureteral stricture at 20 years was observed in 2.5% of 1,784 patients with stage IB carcinoma of the cervix treated with irradiation (274 followed for up to 20 years or longer).831 The most common presenting symptoms were flank pain and urinary tract infection. In 5 patients, ureteral stricture was complicated by a vesicovaginal fistula. Seven of 43 patients who had no evidence of cancer and had hydroureter or hydronephrosis died of radiation complications. Treatment of ureteral stenosis may consist of stenting or resection of the fibrotic segment and reimplantation of the ureter with either ureteroneocystostomy or ureteroileocystostomy. In approximately half of the patients, diversion of urinary stream and ileal conduits are necessary. Occasionally, a nephrectomy is performed for removal of a nonfunctional kidney. Buglione et al.832 reported a 10% incidence of late urinary morbidity and 1% ureteral fibrosis, grade III or IV, in 191 patients. They postulated the role of TGF-β1 in the activation of fibroblasts and remodeling of extracellular matrix, which may be important in the induction of these sequelae.
Patients with gynecologic malignancies, including those receiving radiation therapy, are prone to development of urinary tract infections. Prasad et al.833 collected 216 urine samples from 36 patients receiving pelvic irradiation, 12 of whom had urinary tract infection. The most common organism isolated was Escherichia coli, followed by Enterococcus species. Appropriate urine bacterial studies and cultures are indicated in patients suspected of having superimposed urinary tract infection during the course of radiation therapy.
Parkin et al.834 reported a 26% incidence of severe urinary symptoms (urgency, incontinence, and frequency) in patients treated with irradiation alone for cervical carcinoma. They carried out urodynamic studies in 42 women and compared them with 28 women having urodynamic evaluations before and after treatment. There was no difference in the mean maximum flow rate or mean residual volume in the two groups. However, mean volume of full bladder sensation was significantly lower in the postirradiation group than in the pretreatment group, as was the mean maximum cystometric capacity. This same dysfunction may be noted in approximately 10% of the general female population, and the incidence increases in older women.302
Ureteroarterial fistula is a rare occurrence, and it is associated with a high mortality rate. When profuse urinary tract bleeding occurs in patients previously diagnosed with a gynecologic malignancy and treated with radiation therapy and extensive surgery, ureteroarterial fistula should be considered in the differential diagnoses.835
Neurologic Toxicity
Although extremely rare, lumbosacral plexopathy has been occasionally reported in patients treated for pelvic tumors with doses of 60 to 67.5 Gy. This syndrome was observed in 4 of 2,410 patients with cervical or endometrial carcinoma receiving 45 Gy to the para-aortic lymph nodes (without spinal cord shielding) or external pelvic irradiation (60 Gy to the parametria) and brachytherapy, with the lumbosacral plexus receiving total doses of 70 to 79 Gy.836 Lower-extremity paralysis secondary to lumbosacral plexopathy was reported in one patient after standard radiation therapy for cervical cancer.837
Patients previously reported as having radiation myelopathy to the lumbar spine may have suffered a lumbar and sacral nerve plexopathy instead of or in addition to the spinal cord injury. The differential diagnosis of plexopathy with recurrent tumors is sometimes difficult. In a comparison of 20 patients with lumbosacral plexopathy after irradiation and 30 patients with plexus damage from pelvic malignancy, Thomas et al.838 noted that indolent leg weakness occurred early in radiation-induced plexopathy (pain occurred initially in 10% of patients, although ultimately it was present in 50%), whereas pain was most frequently associated with tumor plexopathy. Muscular weakness, numbness, and paresthesia are common in both groups. Electromyography showed abnormal myokymic discharges in 57% of patients, whereas this finding was very unusual in tumor-induced plexopathy. CT is extremely helpful in the detection of pelvic masses or bone destruction caused by tumor. The authors also reported extensive retroperitoneal fibrosis of the lumbosacral plexus in 2 patients and femoral nerve fibrosis with plexopathy in 1 patient. Although cystometrograms have demonstrated bladder atonicity in some cases, several authors have failed to observe bladder or rectal sphincter disturbances. Unfortunately, as in radiation myelopathy, the neurologic deficit is irreversible, and no effective therapy other supportive care has been found.
Sexual Function
Other types of clinically significant sequelae have been described. Bruner et al.,839 in 90 patients treated with intracavitary irradiation for either carcinoma of the cervix (42 patients) or endometrial carcinoma (48 patients), 78 of whom also received external pelvic irradiation (44.5-Gy mean dose), noted that vaginal length decreased in most patients (at 24 months, in endometrial carcinoma from 8.8 to 7.8 cm, and in cervical carcinoma from 7.6 to 6.2 cm). Pretreatment sexual activity was reported by 31% of women in comparison with 43% after treatment. However, 22% of women reported a decrease in sexual frequency and 37% a decrease in sexual satisfaction. This was correlated with increased dyspareunia, which was noted in 31% of women treated for carcinoma of the cervix and 44% of those treated for endometrial carcinoma. Grigsby et al.840 described complex problems with sexual adjustment in women with gynecologic tumors treated with radiation therapy, with decreased frequency of sexual intercourse, desire, orgasm, and enjoyment of intercourse in 16% to 47% of patients.
Regular vaginal dilation is widely recommended to maintain vaginal health and sexual functioning; however, the compliance rate with this recommendation is not consistent. In a study to test the effectiveness of an “information-motivation-behavior skills” model, the intervention improved the use of vaginal dilation after radiotherapy, and decreased fear about sex after treatment841 There was no evidence that the experimental intervention improved global sexual health. Jensen et al.842 described persistent sexual dysfunction throughout 2 years after RT in 118 women; 85% had low or no sexual interest, 35% had lack of vaginal lubrication, and 55% had mild to severe dyspareunia. However, 63% of the sexually active patients before RT remained active, although with decreased frequency.
Radiation causes ovarian failure with a cessation of menses over a 6-month to 1-year period after treatment. Radiation also causes uterine fibrosis in a dose-dependent fashion. The dose required for radical cervical cancer treatment causes sufficient uterine fibrosis that even if a woman were to become pregnant through embryo donation, the pregnancy terminates as a stillbirth due to insufficient uterine distensibility.586,843
Bone Toxicity
Grigsby et al.,844 in 1,313 patients with gynecologic tumors treated with radiation therapy, identified 207 who received pelvic irradiation to the inguinal areas, including the hips. Femoral neck fractures developed in 10 patients (4.8%); 4 were bilateral. The cumulative actuarial incidence of fracture was 11% at 5 years and 15% at 10 years. Most of the fractures occurred in patients receiving 45 to 63 Gy, and although radiation dose could not be correlated with the occurrence of fracture, no fractures were noted in patients receiving <42 Gy. Cigarette smoking and osteoporosis were significant prognostic factors for increased risk of fracture.
A retrospective cohort study using SEER cancer registry data linked to Medicare claims data analyzed 6,428 women of age 65 years and older diagnosed with pelvic malignancies from 1986 through 1999, and compared results for women who did (n = 2,855) with those who did not (n = 3,573) undergo radiation therapy. Results demonstrated that women who underwent radiation therapy were more likely to have a pelvic fracture than women who did not undergo radiation therapy. The cumulative 5-year fracture rate was 8.2% versus 5.9% in women with cervical cancer; the difference was statistically significant, and most fractures (90%) were hip fractures.845 Concurrent chemoradiation may result in a higher risk than for patients treated with RT alone because the highest fracture rates were seen in patients with anal carcinoma treated with concurrent chemoradiation.
Blomlie et al.846 reported radiation-induced insufficiency fractures of the pelvis on MRI (characterized by edema on T1-weighted images) in 16 of 18 women (9 premenopausal and 9 postmenopausal) with advanced cervical carcinoma. During the study, the fractures associated with edema subsided without treatment in 41 of 52 (79%) lesions in 15 of 16 (94%) patients. Moreno et al.847 described eight patients with pelvic cancer who developed insufficiency fractures after pelvic irradiation, with an average onset 13.7 months after treatment. The bone and CT scan showed abnormalities in the sacroiliac joint in all cases and in the pubis in three cases. In five patients, the initial diagnosis was incorrectly labeled as bone metastases.
Huh et al.848 reported on 463 patients treated for cervical cancer with RT alone, 1.7% of whom developed insufficiency fractures between 7 and 19 months (median, 12 months) after treatment. All had resolution of symptoms within <1 year with conservative therapy, including nonsteroidal anti-inflammatory medication and rest.
The most common complaint is persistent low back pain. Insufficiency fractures may be falsely diagnosed as metastases on PET/CT. The most common form of treatment is conservative management, followed by sacroplasty with polymethylmethacrylate. Bye et al.849 assessed health-related quality of life (HRQOL) at 3 to 4 years after pelvic radiation therapy for carcinoma of the endometrium and cervix in 94 survivors, 79 (84%) of whom answered a survey. The treated women scored lower than the general population on role functioning (81.5 vs. 90.6; p < .01) and higher on diarrhea (23.8 vs. 9.5; p < .01). Compared with pretreatment conditions, an increase in cases with pain in the lower back, hips, and thighs was seen and was associated with deterioration in HRQOL.
Toxicities Related to Brachytherapy
Descriptions of sequelae vary among institutions because toxicity-grading scales are not uniform and the scoring system for complications is not clearly stated in all reports. It is helpful to institute preventive measures when initiating radiation; for example, Dusenbery et al.850 reported 21 (6.4%) life-threatening complications in 327 of 462 patient implants. Lanciano et al.,851 in 95 tandem and ovoid insertions for cervical cancer in 91 patients and for endometrial cancer in 4, observed 2 uterine perforations and a vaginal laceration in 2 patients. Twenty-four percent of implants in 16 patients were associated with temperatures >100.5°F. Five implants (5%) were removed because of presumed sepsis, pulmonary disease, arterial hypotension, change in mental status, and myocardial infarction.
Jhingran and Eifel,852 in 4,043 patients with carcinoma of the cervix who had undergone 7,662 intracavitary procedures, observed 11 (0.3%) documented or suspected thromboembolisms, resulting in 4 deaths; the incidence of postimplant thromboembolism did not decrease significantly with the routine use of minidose heparin prophylaxis. Other life-threatening perioperative complications included myocardial infarction (1 death in 5 patients), cerebrovascular accident (2 patients), congestive heart failure (3 patients), and halothane liver toxicity (2 deaths). Intraoperative complications included uterine perforation (2.8%) and vaginal laceration (0.3%), which occurred more frequently in patients 60 years of age or older (p < .01).
Wollschlaeger et al.853 reported morbidity during hospitalization in 128 patients with cervical carcinoma undergoing 110 LDR intracavitary brachytherapy insertions. Forty-two implants (24.7%) were associated with acute problems; the most common were fever/infection (14.1%) or gastrointestinal problems (5.9%).
Acute gastrointestinal side effects of pelvic irradiation include diarrhea, abdominal cramping, rectal discomfort, and occasionally rectal bleeding, which may be caused by transient enteroproctitis. Patients with hemorrhoids may experience discomfort earlier than other patients. Diarrhea and abdominal cramping can be controlled with the oral administration of diphenoxylate hydrochloride, with loperamide, atropine sulfate, or opium preparations or emollients such as kaolin and pectin. Proctitis and rectal discomfort can be alleviated by small enemas with hydrocortisone and anti-inflammatory suppositories containing bismuth, benzyl benzoate, zinc oxide, or Peruvian balsam. Some suppositories contain cortisone. Small enemas with cod liver oil are also effective. A low-residue diet with no grease or spices and increased fiber in the stool (psyllium, polycarbophil) usually helps to decrease gastrointestinal symptoms.
Genitourinary symptoms secondary to cystourethritis are dysuria, frequency, and nocturia. The urine is usually clear, although there may be microscopic or even gross hematuria. Methenamine mandelate and antispasmodics such as phenazopyridine hydrochloride or a smooth muscle antispasmodic such as flavoxate hydrochloride, hyoscyamine sulfate, oxybutynin chloride, or tolterodine tartrate can relieve symptoms. Fluid intake should be at least 2,000 to 2,500 mL daily. Urinary tract infections may occur; diagnosis should be established with appropriate urine culture studies, including sensitivity to sulfonamides and antibiotics. Therapy should be promptly instituted.
Erythema and dry or moist desquamation may develop in the perineum or intergluteal fold. Proper skin hygiene and topical application of petroleum jelly, petrolatum, or lanolin should relieve these symptoms. U.S.P. zinc oxide ointment and intensive skin care may be needed for severe cases.
Management of acute radiation vaginitis includes douching every day or at least three times weekly with a 1:5 mixture of hydrogen peroxide and water. Douching should be continued on a weekly basis until the mucositis has resolved or for 2 or 3 months as necessary. Superficial ulceration of the vagina responds to topical (intravaginal) estrogen creams, which stimulate epithelial regeneration within 3 months after irradiation. Use of vaginal dilators several times daily, started during the course of treatment, prevents vaginal stenosis. Psychoeducational intervention and motivation improve the compliance in use of dilators.854 More-severe necrosis may require debridement on a weekly basis until healing takes place. Judicious use of biopsies is recommended to rule out persistent or recurrent cancer.
Petereit et al.855 reported 16 acute events (9.5%) in 169 patients treated with HDR brachytherapy (128 with cervical cancer also receiving external irradiation, and 41 medically inoperable endometrial carcinomas). The overall 30-day morbidity rate for the patients with cervical cancer was 5.5%, and the 30-day mortality rate was 1.6% (2 patients; 1 died of pulmonary edema 12 days after first HDR insertion and the other had enteritis and died in a nursing home).
The complication rates for HDR and LDR techniques are usually equivalent.608,610 Petereit et al.606 observed a 12% 3-year actuarial overall toxicity (2.6% genitourinary and 5.6% rectum) with LDR, compared to 15% overall (3% genitourinary and 4.6% rectum) with HDR brachytherapy. However, in the series by Cikaric,711 the rectal complication rate was significantly higher in the LDR group. Bladder complication rates reported, in general, are lower than rectal complication rates; again, except for the series by Cikaric711 showing a higher complication rate with the LDR technique, there were no significant differences with the two techniques.
Ogino et al.,856 in 253 patients with invasive carcinoma of the cervix treated with HDR brachytherapy, noted that grade 4 rectal complications were not observed in patients with a time–dose factor of <130 or biologic equivalent dose of <147, assuming an α/βratio of 3 Gy for late reactions.
Spontaneous intraperitoneal rupture of the urinary bladder, an extremely rare event, was reported by Fujikawa et al.857 after radiation therapy for cervical cancer in 6 of 148 patients treated with HDR intracavitary brachytherapy combined with EBRT. All 6 patients underwent laparotomy and repair of the perforation; however, rerupture of the bladder occurred in 3 of these patients.
Clark et al.858 reported on 43 patients treated with pelvic EBRT (46 Gy) and three HDR intracavitary treatments given weekly combined with concomitant chemotherapy (cisplatin, 30 mg/m2 weekly) for advanced carcinoma of the cervix. At 40 months after treatment, 9 of 13 patients who received a dose to the rectal reference point greater than the prescribed point A dose had a 46% actuarial rate of serious (grade 3 and 4) rectal complications, compared with 14% in the remainder. A strong dose response was observed, with a threshold for complications at a brachytherapy dose of 8 Gy per fraction.
Hyperbaric Oxygen
In 13 patients with hemorrhagic cystitis treated with hyperbaric oxygen, all but 1 experienced durable cessation of hematuria.859 Lee et al.860 also noted that, in 16 of 20 patients (80%) with hemorrhagic radiation cystitis, significant improvement was observed after treatment with hyperbaric oxygen at 2.5 atm (44 sessions).
Several reports evaluated the efficacy of hyperbaric oxygen combined with irradiation in the treatment of a variety of human tumors, including carcinoma of the uterine cervix. Watson et al.,861 in a randomized clinical trial of 320 patients (stages III to IVA), reported a 5-year survival rate of 33% in the oxygen-treated group in contrast to 27% in the control group treated in normal air (p = .08). The local recurrence rate was 33% in the 161 patients treated with oxygen and 53% in 159 patients treated in normal air (p< .001). Morbidity in the patients treated with oxygen was greater (20 severe and 13 moderate complications) than in those treated in normal air (6 severe and 8 moderate complications, respectively). The difference was particularly striking in the bowel (13 and 2 severe complications, respectively).
Dische et al.862 reviewed the data in a randomized study of patients with advanced carcinoma of the cervix treated with radiation therapy and hyperbaric oxygen or air and noted that the patients treated with oxygen had improved survival at Mount Vernon and Glasgow but not at Cape Town. Data from the three centers were merged, and analysis showed that local tumor control was significantly worse in patients treated in normal air who had a prior blood transfusion, but in the oxygen group this effect was reversed. The same interaction was noted in the survival results (p = .042).
A trial reported by Fletcher165 in which 233 patients with stages IIB, III, and IV carcinoma of the cervix were randomized to be treated with irradiation in normal air or with hyperbaric oxygen demonstrated no significant benefit in survival or tumor control (20 of 109 patients treated with oxygen failed in the pelvis, in contrast to 29 of 124 treated in normal air). Furthermore, morbidity was greater (26 complications) in patients treated with hyperbaric oxygen compared with the control group (15 complications).
Dische et al.862 published results of a four-arm randomized trial of hyperbaric oxygen and radiation therapy of stages IIB and III carcinoma of the cervix in which 335 patients were randomized to treatment in 10 or 28 fractions in hyperbaric oxygen or in normal air. Data from 327 cases were analyzed. There was no advantage in tumor control with the use of hyperbaric oxygen. There was an increase in late radiation morbidity when treatment was given in hyperbaric oxygen rather than in normal air, and when using 10 fractions, a total dose of 45 Gy rather than 40 Gy was administered.
No definite conclusions can be drawn concerning the use of hyperbaric oxygen in carcinoma of the cervix. It is possible that hyperbaric oxygen administered with fewer high-dose fractions may be more efficacious than when combined with conventional dose and fractionation schemes. The trials reported have not shown an increased incidence of distant metastasis, which has been observed in a clinical study and in some animal experiments.863
Hormonal Replacement After Treatment of Cervical Cancer
After pelvic irradiation or bilateral salpingo-oophorectomy, usually carried out with a radical hysterectomy in patients treated for carcinoma of the uterine cervix, symptoms of menopause may occur. They can be treated with replacement hormones, although some gynecologists have expressed reservations. During the past 25 years, hormonal replacement therapy has been shown to reduce the risk of cardiovascular diseases, osteoporotic fractures, and colon carcinoma. On the other hand, there is a significant increase of the risk in breast cancer with prolonged use of estrogen plus progesterone for >5 years. Consideration may be given to progesterone alone, which does not increase the risk of endometrial cancer but has potential thromboembolic risks.864
Burger et al.865 concluded that squamous cell cancers of the cervix, vulva, and vagina are unlikely to be influenced by hormonal replacement therapy. In a study of women 50 years of age or younger with ovarian cancer, estrogen replacement therapy did not have a negative influence on disease-free survival. Long-term hormonal replacement therapy in women treated for a gynecologic cancer must be based on the medical history of and discussion of risk with the individual patient (and her family when warranted). Usually, 0.625 to 1.25 mg of coagulated estrogen daily is sufficient.866
Second Malignancy
The risk for induction of secondary primary cancers by pelvic irradiation is low, and many potential confounders are either unknown or may not be fully accounted for, given available information.867 Using the population-based cancer registries of Denmark, Finland, Norway, Sweden, and the United States, Chaturvedi et al.868 found a significantly increased cancer risk in both SCC and AC survivors, with standardized incidence ratio of 1.31 (95% CI = 1.29 to 1.34) and 1.29 (95% CI = 1.22 to 1.38), respectively. The risk of smoking-related lung cancer was higher in the SCC than in the AC population, whereas second malignancies of the colon, soft tissue, melanoma, and non-Hodgkin lymphoma were higher in the AC population. Similarly, 1-year survivors of cervical cancer had an increased risk of HPV-related cancers, including pharynx, genital, and anal cancers. Higher hazard ratios for second cancer of the rectum, anus, bladder, and genital sites was seen for younger patients, with a 40-year cumulative risk of any second cancer of 22% for women diagnosed with cervical cancer before age 50 years versus 16% for those diagnosed at age >50 years.869 In contrast, Lee et al.870 observed no significant increase in the incidence of second malignancies in patients irradiated for carcinoma of the cervix in comparison with the Connecticut Tumor Registry prevailing rates.
Boice et al.,339 in a review of 68,730 women with carcinoma of the cervix treated with radiation therapy, observed a second malignant tumor in 3,324, compared with 3,063 expected (4.8% increase; p < .001). The excess was concentrated in the lung, other genital organs, bladder, and rectum. In addition, in 10,817 women with invasive cervical cancer not treated with irradiation, 479 secondary malignant tumors were observed versus 435 expected (4.4%; p = .02). Thus, the incidence of secondary tumors in women treated for carcinoma of the cervix with or without irradiation is only slightly greater than in the general population. Pelvic organs receiving a high dose of irradiation appear to have a somewhat greater incidence of a second primary.
Storm,871 in a comprehensive analysis of the Danish Cancer Registry data of 24,970 women with invasive cervical cancer and 19,470 with carcinoma in situ of the cervix treated between 1943 and 1982, noted a small overall excess of secondary primary cancers in the lung, stomach, pancreas, rectum, and bladder and connective tissue sarcomas, although there was a decreased incidence of breast cancer in the irradiated patients compared with nonirradiated patients (attributable to ovarian ablation by radiation therapy). In the patients irradiated for invasive carcinoma, there was an excess of 64 cases per 10,000 women per year of tumors in organs close to or at an intermediate distance from the cervix, reaching a maximum after 30 years or longer of follow-up. A high risk for development of acute nonlymphatic leukemia was observed in irradiated patients with carcinoma in situ but not in those with invasive lesions. This could be explained by the lower doses of irradiation delivered to the bone marrow in the in situ tumors treated with brachytherapy alone, with greater induction of mutations and less cell killing, which may be responsible for the leukemogenic effect. Decreased risk was noted for tumors of the brain, myeloma of the skin, and tumors of the colon other than rectal.
In a study of 117,830 women diagnosed with cervical carcinoma in situ and 17,556 with invasive cervical carcinoma in Sweden, treatment not specified and in situ lesions traditionally treated with surgery alone, there was an increased incidence (RR = 2.3 to 3) of second primary tumors in the anus, rectum, urinary bladder, pancreas, esophagus, and lung compared with the standardized incidence rate for all women.872 The data showed consistent increases in suggested targets for HPV at tobacco-related sites. A contributing role for a depressed immune response was considered.
Werner-Wasik et al.,873 in an analysis of 125 women with stages I and II carcinoma of the cervix treated with radiation therapy, observed 11 secondary primary tumors in 10 patients (4 breast, 2 lung, and 1 each of myeloma, non-Hodgkin lymphoma, bladder, thyroid, and vulva). All secondary primary tumors were located outside the irradiation fields. The increased relative risk of breast cancer in these patients was 2.64, higher than reported by Boice et al.339
In an analysis of 199,268 individuals by Wright et al.,874 the risk of secondary leukemia increased 72% in patients who received pelvic radiotherapy, with a peak at 5 to 10 years after treatment; there was no increased risk of multiple myeloma. Mark et al.875 identified 13 of 114 patients diagnosed with uterine sarcoma who had a prior history of pelvic irradiation (doses of 40 to 80 Gy). Criteria for radiation-induced sarcomas included a prior history of pelvic irradiation, a latent period of several years, development of sarcoma within previously irradiated field, and histologic confirmation of malignancy. Histologic types of tumor were mixed Müllerian in 6, leiomyosarcoma in 4 patients, endometrial stroma sarcoma in 1, fibrosarcoma in 1, and angiosarcoma in 1. Sarcoma developed in the uterus in 12 patients and at the vaginal cuff in 1 patient. Ten patients were treated with surgery and 2 with radiation therapy. The 5-year disease-free survival rate after salvage therapy was 17%.
In a theoretical analysis of IMRT risk in postoperative cases relative to three-dimensional conformal radiotherapy, the estimated increase in second cancer risk was 6% for 6-MV IMRT and 26% for 18 MV IMRT, with large increases in organs away from the primary beam and skin because with IMRT a much larger volume of skin was exposed.876
Seidman et al.877 reviewed 15 cases of second malignancies after pelvic radiation; 5 were HPV-related vaginal primary tumors. The average latency period for development was approximately 20 years.
REFERENCES
1. Anson BJ, McVay CB. Surgical anatomy, 5th ed. Philadelphia: WB Saunders, 1971:800–35.
2. World Health Organization. Age-standardized incidence rates of cervical cancer, 2009. Available at: http://www.who.int/mediacentre/factsheets/fs297/en/. Accessed May 22, 2010.
3. American Cancer Society. Cancer facts and figures. Atlanta, GA: 2012.
4. Barakat R, Markman M, Randall M, eds. Principles and practice of gynecologic oncology, 5th ed. Philadelphia: Lippincott Williams & Wilkins, 2009.
5. Hochman A, Ratzkowski E, Schreiber H. Incidence of carcinoma of the cervix in Jewish women in Israel. Br J Cancer 1955;9:358–364.
6. Bosch FX, Lorincz A, Munoz N, et al. The causal relation between human papillomavirus and cervical cancer. J Clin Pathol 2002;55:244–265.
7. World Health Organization. Human papillomavirus and related cancers, 2010. Available at: http://apps.who.int/hpvcentre/statistics/dynamic/ico/country_pdf/XWX.pdf?CFID=5541015. Accessed March 1, 2012.
8. zur Hausen H. Papillomaviruses causing cancer: evasion from host-cell control in early events in carcinogenesis. J Natl Cancer Inst 2000;92:690–698.
9. Franco EL, Villa LL, Sobrinho JP, et al. Epidemiology of acquisition and clearance of cervical human papillomavirus infection in women from a high-risk area for cervical cancer. J Infect Dis 1999;180:1415–1423.
10. Ho GY, Bierman R, Beardsley L, et al. Natural history of cervicovaginal papillomavirus infection in young women. N Engl J Med 1998;338:423–428.
11. Christopherson WM, Parker JE. Relation of cervical cancer to early marriage and childbearing. N Engl J Med 1965;273:235–239.
12. Keighley E. Carcinoma of the cervix among prostitutes in a women’s prison. Br J Vener Dis 1968;44:254–5.
13. Vriend HJ, Boot HJ, van der Sande MA. Type-specific human papillomavirus infections among young heterosexual male and female STI clinic attendees. Sex Transm Dis 2012;39:72–78.
14. Nahmias AJ, Naib ZM, Josey WE. Epidemiological studies relating genital herpetic infection to cervical carcinoma. Cancer Res 1974;34:1111–1117.
15. Agarwal SS, Sehgal A, Sardana S, et al. Role of male behavior in cervical carcinogenesis among women with one lifetime sexual partner. Cancer 1993;72:1666–1669.
16. Iversen T, Tretli S, Johansen A, et al. Squamous cell carcinoma of the penis and of the cervix, vulva and vagina in spouses: is there any relationship? An epidemiological study from Norway, 1960–92. Br J Cancer 1997;76:658–6560.
17. Boon ME, Susanti I, Tasche MJ, et al. Human papillomavirus (HPV)-associated male and female genital carcinomas in a Hindu population. The male as vector and victim. Cancer 1989;64:559–565.
18. Terris M, Wilson F, Nelson JH Jr. Relation of circumcision to cancer of the cervix. Am J Obstet Gynecol 1973;117:1056–1066.
19. Lynch HT, Rubinstein WS, Locker GY. Cancer in Jews: introduction and overview. Fam Cancer 2004;3:177–192.
20. Joneja MG, Coulson DB. Histopathology and cytogenetics of tumors induced by the application of 7,12-dimethybenz(a)anthracene (DMBA) in mouse cervix. Eur J Cancer 1973;9:367–374.
21. Drill VA. Oral contraceptives: relations to mammary cancer, benign breast lesions, and cervical cancer. Annu Rev Pharmacol 1975;15:367–685.
22. Green J, Berrington de Gonzalez A, et al. Human papillomavirus infection and use of oral contraceptives. Br J Cancer 2003;88:1713–1720.
23. Kaminski PF, Maier RC. Clear cell adenocarcinoma of the cervix unrelated to diethylstilbestrol exposure. Obstet Gynecol 1983;62:720–727.
24. Herbst AL, Cole PL. Epidemiologic and clinical aspects of clear cell adenocarcinoma in young women. In: Herbst AL, ed. Intrauterine exposure to diethylstilbestral in the human. Washington, DC: American College of Obstetricians and Gynecologists, 1978:2–7.
25. Becker TM, Wheeler CM, McGough NS, et al. Sexually transmitted diseases and other risk factors for cervical dysplasia among southwestern Hispanic and non-Hispanic white women. JAMA 1994;271:1181–1188.
26. Gram IT, Austin H, Stalsberg H. Cigarette smoking and the incidence of cervical intraepithelial neoplasia, grade III, and cancer of the cervix uteri. Am J Epidemiol 1992;135:341–346.
27. Louie KS, Castellsague X, de Sanjose S, et al. Smoking and passive smoking in cervical cancer risk: pooled analysis of couples from the IARC multicentric case-control studies. Cancer Epidemiol Biomarkers Prev2011;20:1379–1390.
28. Szarewski A. Smoking and cervical neoplasia: a review of the evidence. J Epidemiol Biostat 1998;3:229–256.
29. Palefsky JM, Minkoff H, Kalish LA, et al. Cervicovaginal human papillomavirus infection in human immunodeficiency virus–1 (HIV)-positive and high-risk HIV-negative women. J Natl Cancer Inst 1999;91:226–236.
30. Castellsague X, Diaz M, Vaccarella S, et al. Intrauterine device use, cervical infection with human papillomavirus, and risk of cervical cancer: a pooled analysis of 26 epidemiological studies. Lancet Oncol 2011;12:1023–31.
31. Barron BA, Richart RM. Statistical model of the natural history of cervical carcinoma. II. Estimates of the transition time from dysplasia to carcinoma in situ. J Natl Cancer Inst 1970;45:1025–1030.
32. Christopherson WM, Gray LA, Parker JE. Microinvasive carcinoma of the uterine cervix: A long-term followup study of eighty cases. Cancer 1976;38:629–632.
33. Bohm JW, Krupp PJ, Lee FY, et al. Lymph node metastasis in microinvasive epidermoid cancer of the cervix. Obstet Gynecol 1976;48:65–67.
34. Inoue T, Okumura M. Prognostic significance of parametrial extension in patients with cervical carcinoma Stages IB, IIA, and IIB. A study of 628 cases treated by radical hysterectomy and lymphadenectomy with or without postoperative irradiation. Cancer 1984;54:1714–1719.
35. Landoni F, Maneo A, Colombo A, et al. Randomised study of radical surgery versus radiotherapy for stage Ib-IIa cervical cancer. Lancet 1997;350:535–540.
36. Perez CA, Camel HM, Askin F, et al. Endometrial extension of carcinoma of the uterine cervix: a prognostic factor that may modify staging. Cancer 1981;48:170–180.
37. Chao KSC, Grigsby PW, Mutch D, et al. Prognostic factors for distant metastasis in carcinoma of the cervix with endometrial extension. J Brachyther Int 2000;16:181–186.
38. Girardi F, Lichtenegger W, Tamussino K, et al. The importance of parametrial lymph nodes in the treatment of cervical cancer. Gynecol Oncol 1989;34:206–211.
39. Henriksen E. The lymphatic spread of carcinoma of the cervix and of the body of the uterus; a study of 420 necropsies. Am J Obstet Gynecol 1949;58:924–942.
40. Benedetti-Panici P, Maneschi F, Scambia G, et al. Lymphatic spread of cervical cancer: an anatomical and pathological study based on 225 radical hysterectomies with systematic pelvic and aortic lymphadenectomy. Gynecol Oncol 1996;62:19–24.
41. Fagundes H, Perez CA, Grigsby PW, et al. Distant metastases after irradiation alone in carcinoma of the uterine cervix. Int J Radiat Oncol Biol Phys 1992;24:197–204.
42. Ampil FL, Apple S, Bell MC. Spinal epidural compression complicating cancer of the cervix: review of seven cases. Eur J Gynaecol Oncol 1998;19:105–107.
43. Lemus JF, Abdulhay G, Sobolewski C, et al. Cardiac metastasis from carcinoma of the cervix: report of two cases. Gynecol Oncol 1998;69:264–268.
44. Mahmoud-Ahmed AS, Suh JH, Barnett GH, et al. Tumor distribution and survival in six patients with brain metastases from cervical carcinoma. Gynecol Oncol 2001;81:196–200.
45. Viswanathan AN, Deavers MT, Jhingran A, et al. Small cell neuroendocrine carcinoma of the cervix: outcome and patterns of recurrence. Gynecol Oncol 2004;93:27–33.
46. American College of Obstetricians and Gynecologists. Guidelines for women’s health care: a resource manual, Washington, DC: Author, 2009.
47. Kern W, Zivolich MR. The accuracy and consistency of the cytologic classification of squamous lesions of the uterine cervix. Acta Cytol 1977;21:519–23.
48. Solomon D, Davey D, Kurman R, et al. The 2001 Bethesda System: terminology for reporting results of cervical cytology. JAMA 2002;287:2114–9.
49. Novak ER, Jones GS, Jones HW 3rd, eds. Novak’s textbook of gynecology. Philadelphia: Williams & Wilkins, 1970.
50. Kim SH, Choi BI, Han JK, et al. Preoperative staging of uterine cervical carcinoma: comparison of CT and MRI in 99 patients. J Comput Assist Tomogr 1993;17:633–640.
51. Lagasse LD, Ballon SC, Berman MS, et al. Pretreatment lymphangiography and operative evaluation in carcinoma of the cervix. Am J Obstet Gynecol 1979;134:219–224.
52. Kim SH, Han MC. Invasion of the urinary bladder by uterine cervical carcinoma: evaluation with MR imaging. AJR Am J Roentgenol 1997;168:393–397.
53. Mitchell DG, Snyder B, Coakley F, et al. Early invasive cervical cancer: MRI and CT predictors of lymphatic metastases in the ACRIN 6651/GOG 183 intergroup study. Gynecol Oncol 2009;112:95–103.
54. Camilien L, Gordon D, Fruchter RG, et al. Predictive value of computerized tomography in the presurgical evaluation of primary carcinoma of the cervix. Gynecol Oncol 1988;30:209–15.
55. Heller PB, Malfetano JH, Bundy BN, Barnhill DR, Okagaki T. Clinical-pathologic study of stage IIB, III, and IVA carcinoma of the cervix: Extended diagnostic evaluation for paraaortic node metastasis—a Gynecologic Oncology Group study. Gynecol Oncol 1990;38:425–430.
56. Wong F, Bhimji S. The usefulness of ultrasonography in intracavitary radiotherapy using selectron applicators. Int J Radiat Oncol Biol Phys 1990;19:477–82.
57. Viswanathan AN, Moughan J, Small W Jr, et al. The quality of cervical cancer brachytherapy implantation and the impact on local recurrence and disease-free survival in radiation therapy oncology group prospective trials 0116 and 0128. Int J Gynecol Cancer 2012;22:123–131.
58. Liu FY, Yen TC, Chen MY, et al. Detection of hematogenous bone metastasis in cervical cancer: 18F-fluorodeoxyglucose-positron emission tomography versus computed tomography and magnetic resonance imaging. Cancer2009;115:5470–5480.
59. Hricak H, Powell CB, Yu KK, et al. Invasive cervical carcinoma: role of MR imaging in pretreatment work-up—cost minimization and diagnostic efficacy analysis. Radiology 1996;198:403–409.
60. Ascher SM, Cooper C, Scoutt L, et al. Diagnostic imaging techniques in gynecologic oncology. In: Hoskins WJ, Perez CA, Young RC, eds. Principles and practice of gynecologic oncology, 4th ed. Philadelphia: Lippincott Williams & Wilkins, 2005:223–266.
61. Sironi S, Belloni C, Taccagni GL et al. Carcinoma of the cervix: value of MR in detecting parametrial involvement. AJR Am J Roentgenol 1991;156:753–756.
62. Hawnaur JM, Johnson RJ, Carrington BM, et al. Predictive value of clinical examination, transrectal ultrasound and magnetic resonance imaging prior to radiotherapy in carcinoma of the cervix. Br J Radiol 1998;71:819–827.
63. Postema S, Pattynama PM, van den Berg-Huysmans A, et al. Effect of MRI on therapeutic decisions in invasive cervical carcinoma. Direct comparison with the pelvic examination as a preperative test. Gynecol Oncol2000;79:485–489.
64. Hansen MA, Pedersen PH, Andreasson B, et al. Staging uterine cervical carcinoma with low-field MR imaging. Acta Radiol 2000;41:647–652.
65. Mitchell DG, Snyder B, Coakley F, et al. Early invasive cervical cancer: tumor delineation by magnetic resonance imaging, computed tomography, and clinical examination, verified by pathologic results, in the ACRIN 6651/GOG 183 Intergroup Study. J Clin Oncol 2006;24:5687–5694.
66. Hricak H, Gatsonis C, Coakley FV, et al. Early invasive cervical cancer: CT and MR imaging in preoperative evaluation—ACRIN/GOG comparative study of diagnostic performance and interobserver variability. Radiology2007;245:491–498.
67. Haider MA, Patlas M, Jhaveri K, et al. Adenocarcinoma involving the uterine cervix: magnetic resonance imaging findings in tumours of endometrial, compared with cervical, origin. Can Assoc Radiol J 2006;57:43–48.
68. Kodaira T, Fuwa N, Kamata M, et al. Clinical assessment by MRI for patients with stage II cervical carcinoma treated by radiation alone in multicenter analysis: are all patients with stage II disease suitable candidates for chemoradiotherapy? Int J Radiat Oncol Biol Phys 2002;52:627–636.
69. Ebner F, Kressel HY, Mintz MC, et al. Tumor recurrence versus fibrosis in the female pelvis: differentiation with MR imaging at 1.5 T. Radiology 1988;166:333–340.
70. Corn BW, Schnall MD, Milestone B, et al. Signal characteristics of tumors shown by high-resolution endorectal coil magnetic resonance imaging may predict outcome among patients with cervical carcinoma treated with irradiation. A preliminary study. Cancer 1996;78:2535–2542.
71. Mayr NA, Yuh WT, Magnotta VA, et al. Tumor perfusion studies using fast magnetic resonance imaging technique in advanced cervical cancer: a new noninvasive predictive assay. Int J Radiat Oncol Biol Phys 1996;36:623–633.
72. Hatano K, Sekiya Y, Araki H, et al. Evaluation of the therapeutic effect of radiotherapy on cervical cancer using magnetic resonance imaging. Int J Radiat Oncol Biol Phys 1999;45:639–644.
73. Gong QY, Brunt JN, Romaniuk CS, et al. Contrast enhanced dynamic MRI of cervical carcinoma during radiotherapy: early prediction of tumour regression rate. Br J Radiol 1999;72:1177–1184.
74. van de Bunt L, van der Heide UA, Ketelaars M, et al. Conventional, conformal, and intensity-modulated radiation therapy treatment planning of external beam radiotherapy for cervical cancer: the impact of tumor regression. Int J Radiat Oncol Biol Phys2006;64:189–196.
75. Dimopoulos JC, Schard G, Berger D, et al. Systematic evaluation of MRI findings in different stages of treatment of cervical cancer: potential of MRI on delineation of target, pathoanatomic structures, and organs at risk. Int J Radiat Oncol Biol Phys2006;64:1380–1388.
76. Rose PG, Adler LP, Rodriguez M, et al. Positron emission tomography for evaluating para-aortic nodal metastasis in locally advanced cervical cancer before surgical staging: a surgicopathologic study. J Clin Oncol 1999;17:41–45.
77. Grigsby PW, Siegel BA, Dehdashti F. Lymph node staging by positron emission tomography in patients with carcinoma of the cervix. J Clin Oncol 2001;19:3745–3749.
78. Grigsby PW, Siegel BA, Dehdashti F, et al. Posttherapy surveillance monitoring of cervical cancer by FDG-PET. Int J Radiat Oncol Biol Phys 2003;55:907–913.
79. Grigsby PW, Siegel BA, Dehdashti F, et al. Posttherapy [18F] fluorodeoxyglucose positron emission tomography in carcinoma of the cervix: response and outcome. J Clin Oncol 2004;22:2167–2171.
80. Grigsby PW, Singh AK, Siegel BA, et al. Lymph node control in cervical cancer. Int J Radiat Oncol Biol Phys 2004;59:706–712.
81. Hope AJ, Saha P, Grigsby PW. FDG-PET in carcinoma of the uterine cervix with endometrial extension. Cancer 2006;106:196–200.
82. Lin LL, Yang Z, Mutic S, et al. FDG-PET imaging for the assessment of physiologic volume response during radiotherapy in cervix cancer. Int J Radiat Oncol Biol Phys 2006;65:177–181.
83. Kidd EA, Grigsby PW. Intratumoral metabolic heterogeneity of cervical cancer. Clin Cancer Res 2008;14:5236–5241.
84. Kidd EA, Siegel BA, Dehdashti F, et al. The standardized uptake value for F-18 fluorodeoxyglucose is a sensitive predictive biomarker for cervical cancer treatment response and survival. Cancer 2007;110:1738–44.
85. Kidd EA, Siegel BA, Dehdashti F, et al. Pelvic lymph node F-18 fluorodeoxyglucose uptake as a prognostic biomarker in newly diagnosed patients with locally advanced cervical cancer. Cancer 2010;116:1469–1475.
86. Kidd EA, Siegel BA, Dehdashti F, et al. Lymph node staging by positron emission tomography in cervical cancer: relationship to prognosis. J Clin Oncol 2010;28:2108–2113.
87. Kidd EA, Spencer CR, Huettner PC, et al. Cervical cancer histology and tumor differentiation affect 18F-fluorodeoxyglucose uptake. Cancer 2009;115:3548–3554.
88. Tran BN, Grigsby PW, Dehdashti F, et al. Occult supraclavicular lymph node metastasis identified by FDG-PET in patients with carcinoma of the uterine cervix. Gynecol Oncol 2003;90:572–576.
89. Schwarz JK, Grigsby PW, Dehdashti F, et al. The role of 18F-FDG PET in assessing therapy response in cancer of the cervix and ovaries. J Nucl Med 2009;50(Suppl 1):64S–73S.
90. Schwarz JK, Lin LL, Siegel BA, et al. 18-F-fluorodeoxyglucose-positron emission tomography evaluation of early metabolic response during radiation therapy for cervical cancer. Int J Radiat Oncol Biol Phys 2008;72:1502–1507.
91. Schwarz JK, Siegel BA, Dehdashti F, et al. Association of posttherapy positron emission tomography with tumor response and survival in cervical carcinoma. JAMA 2007;298:2289–2295.
92. Schwarz JK, Siegel BA, Dehdashti F, et al. Metabolic response on posttherapy FDG-PET predicts patterns of failure after radiotherapy for cervical cancer. Int J Radiat Oncol Biol Phys 2011;83:185–190.
93. Horn LC, Fischer U, Raptis G, et al. Tumor size is of prognostic value in surgically treated FIGO stage II cervical cancer. Gynecol Oncol 2007;107:310–315.
94. American Joint Committee on Cancer. AJCC cancer staging handbook, 7th ed. New York: Springer, 2010.
95. Kraus FT, Perezmesa C. Verrucous carcinoma. Clinical and pathologic study of 105 cases involving oral cavity, larynx and genitalia. Cancer 1966;19:26–38.
96. Look KY, Brunetto VL, Clarke-Pearson DL, et al. An analysis of cell type in patients with surgically staged stage IB carcinoma of the cervix: a Gynecologic Oncology Group study. Gynecol Oncol 1996;63:304–311.
97. Drescher CW, Hopkins MP, Roberts JA. Comparison of the pattern of metastatic spread of squamous cell cancer and adenocarcinoma of the uterine cervix. Gynecol Oncol 1989;33:340–343.
98. Chao A, Wang TH, Lee YS, et al. Molecular characterization of adenocarcinoma and squamous carcinoma of the uterine cervix using microarray analysis of gene expression. Int J Cancer 2006;119:91–98.
99. Contag SA, Gostout BS, Clayton AC, et al. Comparison of gene expression in squamous cell carcinoma and adenocarcinoma of the uterine cervix. Gynecol Oncol 2004;95:610–617.
100. An HJ, Kim KR, Kim IS, et al. Prevalence of human papillomavirus DNA in various histological subtypes of cervical adenocarcinoma: a population-based study. Mod Pathol 2005;18:528–534.
101. Silverberg SG, Hurt WG. Minimal deviation adenocarcinoma (“adenoma malignum”) of the cervix: a reappraisal. Am J Obstet Gynecol 1975;121:971–975.
102. Srivatsa PJ, Keeney GL, Podratz KC. Disseminated cervical adenoma malignum and bilateral ovarian sex cord tumors with annular tubules associated with Peutz-Jeghers syndrome. Gynecol Oncol 1994;53:256–264.
103. Ulbright TM, Gersell DJ. Glassy cell carcinoma of the uterine cervix. A light and electron microscopic study of five cases. Cancer 1983;51:2255–2263.
104. Littman P, Clement PB, Henriksen B, et al. Glassy cell carcinoma of the cervix. Cancer 1976;37:2238–2246.
105. Piura B, Rabinovich A, Meirovitz M, et al. Glassy cell carcinoma of the uterine cervix. J Surg Oncol 1999;72:206–210.
106. Gordon HW, McMahon NJ, Agliozzo CM, et al. Adenoid cystic (cylindromatous) carcinoma of the uterine cervix: report of two cases. Am J Clin Pathol 1972;58:51–57.
107. Nishida M, Nasu K, Takai N, et al. Adenoid cystic carcinoma of the uterine cervix. Int J Clin Oncol 2005;10:198–200.
108. Clement PB, Zubovits JT, Young RH, et al. Malignant mullerian mixed tumors of the uterine cervix: a report of nine cases of a neoplasm with morphology often different from its counterpart in the corpus. Int J Gynecol Pathol1998;17:211–222.
109. Abeler VM, Holm R, Nesland JM, et al. Small cell carcinoma of the cervix. A clinicopathologic study of 26 patients. Cancer 1994;73:672–677.
110. Gersell DJ, Mazoujian G, Mutch DG, et al. Small-cell undifferentiated carcinoma of the cervix. A clinicopathologic, ultrastructural, and immunocytochemical study of 15 cases. Am J Surg Pathol 1988;12:684–698.
111. Ishida GM, Kato N, Hayasaka T, et al. Small cell neuroendocrine carcinomas of the uterine cervix: a histological, immunohistochemical, and molecular genetic study. Int J Gynecol Pathol 2004;23:366–372.
112. van Nagell JR Jr, Powell DE, Gallion HH, et al. Small cell carcinoma of the uterine cervix. Cancer 1988;62:1586–1593.
113. Daroca PJ Jr, Dhurandhar HN. Basaloid carcinoma of uterine cervix. Am J Surg Pathol 1980;4:235–239.
114. Layton-Henry J, Scurry J, Planner R, et al. Cervical adenoid basal carcinoma: five cases and literature review. Int J Gynecol Cancer 1996;6:193–199.
115. Toyoshima M, Okamura C, Niikura H, et al. Epithelioid leiomyosarcoma of the uterine cervix: a case report and review of the literature. Gynecol Oncol 2005;97:957–960.
116. el Omari-Alaoui H, Kebdani T, Benjaafar N, et al. Non-Hodgkin’s lymphoma of the uterus: apropos of 4 cases and review of the literature [in French]. Cancer Radiother 2002;6:39–45.
117. Bozaci EA, Atabekoglu C, Sertcelik A, et al. Metachronous metastases from renal cell carcinoma to uterine cervix and vagina: case report and review of literature. Gynecol Oncol 2005;99:232–235.
118. Pauer HU, Viereck V, Burfeind P, et al. Uterine cervical metastasis of breast cancer: a rare complication that may be overlooked. Onkologie 2003;26:58–60.
119. Shimada M, Kigawa J, Nishimura R, et al. Ovarian metastasis in carcinoma of the uterine cervix. Gynecol Oncol 2006;101:234–237.
120. Delaloye JF, Pampallona S, Coucke PA, et al. Younger age as a bad prognostic factor in patients with carcinoma of the cervix. Eur J Obstet Gynecol Reprod Biol 1996;64:201–205.
121. Dattoli MJ, Gretz HF 3rd, Beller U, et al. Analysis of multiple prognostic factors in patients with stage IB cervical cancer: age as a major determinant. Int J Radiat Oncol Biol Phys 1989;17:41–47.
122. Meanwell CA, Kelly KA, Wilson S, et al. Young age as a prognostic factor in cervical cancer: analysis of population based data from 10,022 cases. Br Med J (Clin Res Ed) 1988;296:386–391.
123. Rutledge FN, Mitchell MF, Munsell M, et al. Youth as a prognostic factor in carcinoma of the cervix: a matched analysis. Gynecol Oncol 1992;44:123–130.
124. Mitchell PA, Waggoner S, Rotmensch J, et al. Cervical cancer in the elderly treated with radiation therapy. Gynecol Oncol 1998;71:291–298.
125. Mundt AJ, Connell PP, Campbell T, et al. Race and clinical outcome in patients with carcinoma of the uterine cervix treated with radiation therapy. Gynecol Oncol 1998;71:151–158.
126. Grigsby PW, Hall-Daniels L, Baker S, et al. Comparison of clinical outcome in black and white women treated with radiotherapy for cervical carcinoma. Gynecol Oncol 2000;79:357–361.
127. Brooks SE, Chen TT, Ghosh A, et al. Cervical cancer outcomes analysis: impact of age, race, and comorbid illness on hospitalizations for invasive carcinoma of the cervix. Gynecol Oncol 2000;79:107–115.
128. Katz A, Eifel PJ, Moughan J, et al. Socioeconomic characteristics of patients with squamous cell carcinoma of the uterine cervix treated with radiotherapy in the 1992 to 1994 patterns of care study. Int J Radiat Oncol Biol Phys2000;47:443–450.
129. Hirst D. Anemia: a problem or an opportunity in radiotherapy? Int J Radiat Oncol Biol Phys 1986;12:2009–2017.
130. Kavanagh BD, Secomb TW, Hsu R, et al. A theoretical model for the effects of reduced hemoglobin-oxygen affinity on tumor oxygenation. Int J Radiat Oncol Biol Phys 2002;53:172–179.
131. Sundfor K, Lyng H, Kongsgard UL, et al. Polarographic measurement of pO2 in cervix carcinoma. Gynecol Oncol 1997;64:230–236.
132. Hockel M, Schlenger K, Mitze M, et al. Hypoxia and radiation response in human tumors. Semin Radiat Oncol 1996;6:3–9.
133. Knocke TH, Weitmann HD, Feldmann HJ, et al. Intratumoral pO2-measurements as predictive assay in the treatment of carcinoma of the uterine cervix. Radiother Oncol 1999;53:99–104.
134. Rofstad EK, Sundfor K, Lyng H, et al. Hypoxia-induced treatment failure in advanced squamous cell carcinoma of the uterine cervix is primarily due to hypoxia-induced radiation resistance rather than hypoxia-induced metastasis. Br J Cancer2000;83:354–359.
135. Cooper RA, West CM, Logue JP, et al. Changes in oxygenation during radiotherapy in carcinoma of the cervix. Int J Radiat Oncol Biol Phys 1999;45:119–126.
136. Dunst J, Hansgen G, Lautenschlager C, et al. Oxygenation of cervical cancers during radiotherapy and radiotherapy + cis-retinoic acid/interferon. Int J Radiat Oncol Biol Phys 1999;43:367–373.
137. Fyles AW, Pintilie M, Kirkbride P, et al. Prognostic factors in patients with cervix cancer treated by radiation therapy: results of a multiple regression analysis. Radiother Oncol 1995;35:107–117.
138. Haensgen G, Krause U, Becker A, et al. Tumor hypoxia, p53, and prognosis in cervical cancers. Int J Radiat Oncol Biol Phys 2001;50:865–872.
139. Bush R. The significance of anemia in clinical radiation therapy. Int J Radiat Oncol Biol Phys 1986;12:2047–2050.
140. Fyles AW, Milosevic M, Pintilie M, et al. Anemia, hypoxia and transfusion in patients with cervix cancer: a review. Radiother Oncol 2000;57:13–19.
141. Thomas G. The effect of hemoglobin level on radiotherapy outcomes: the Canadian experience. Semin Oncol 2001;28:60–5.
142. Dunst J, Kuhnt T, Strauss HG, et al. Anemia in cervical cancers: impact on survival, patterns of relapse, and association with hypoxia and angiogenesis. Int J Radiat Oncol Biol Phys 2003;56:778–87.
143. Munstedt K, Johnson P, Bohlmann MK, et al. Adjuvant radiotherapy in carcinomas of the uterine cervix: the prognostic value of hemoglobin levels. Int J Gynecol Cancer 2005;15:285–291.
144. Grogan M, Thomas GM, Melamed I, et al. The importance of hemoglobin levels during radiotherapy for carcinoma of the cervix. Cancer 1999;86:1528–1536.
145. Kapp KS, Poschauko J, Geyer E, et al. Evaluation of the effect of routine packed red blood cell transfusion in anemic cervix cancer patients treated with radical radiotherapy. Int J Radiat Oncol Biol Phys 2002;54:58–66.
146. Vaupel P, Mayer A, Hockel M. Impact of hemoglobin levels on tumor oxygenation: the higher, the better? Strahlenther Onkol 2006;182:63–71.
147. Thomas G, Ali S, Hoebers FJ, et al. Phase III trial to evaluate the efficacy of maintaining hemoglobin levels above 12.0 g/dL with erythropoietin vs above 10.0 g/dL without erythropoietin in anemic patients receiving concurrent radiation and cisplatin for cervical cancer. Gynecol Oncol 2008;108:317–325.
148. Blohmer JU, Paepke S, Sehouli J, et al. Randomized phase III trial of sequential adjuvant chemoradiotherapy with or without erythropoietin Alfa in patients with high-risk cervical cancer: results of the NOGGO-AGO intergroup study. J Clin Oncol2011;29:3791–3797.
149. Jenkin RD, Stryker JA. The influence of the blood pressure on survival in cancer of the cervix. Br J Radiol 1968;41:913–920.
150. Kapp DS, Lawrence R. Temperature elevation during brachytherapy for carcinoma of the uterine cervix: adverse effect on survival and enhancement of distant metastasis. Int J Radiat Oncol Biol Phys 1984;10:2281–2292.
151. Campbell OB, Arowojolu AO, Adu FD, et al. Human immunodeficiency virus antibody in patients with cancer of the uterine cervix undergoing radiotherapy: clinical stages, histological grade and outcome of radiotherapy. J Obstet Gynaecol 1999;19:403–405.
152. Klevens RM, Fleming PL, Mays MA, et al. Characteristics of women with AIDS and invasive cervical cancer. Obstet Gynecol 1996;88:269–273.
153. Wright TC Jr, Ellerbrock TV, Chiasson MA, et al. Cervical intraepithelial neoplasia in women infected with human immunodeficiency virus: prevalence, risk factors, and validity of Papanicolaou smears. New York Cervical Disease Study. Obstet Gynecol1994;84:591–597.
154. Tomita LY, Longatto Filho A, Costa MC, et al. Diet and serum micronutrients in relation to cervical neoplasia and cancer among low-income Brazilian women. Int J Cancer 2010;126:703–714.
155. Cho H, Kim MK, Lee JK, et al. Relationship of serum antioxidant micronutrients and sociodemographic factors to cervical neoplasia: a case-control study. Clin Chem Lab Med 2009;47:1005–1012.
156. Goodman MT, Kiviat N, McDuffie K, et al. The association of plasma micronutrients with the risk of cervical dysplasia in Hawaii. Cancer Epidemiol Biomarkers Prev 1998;7:537–544.
157. Lai CH, Chang CJ, Huang HJ, et al. Role of human papillomavirus genotype in prognosis of early-stage cervical cancer undergoing primary surgery. J Clin Oncol 2007;25:3628–3634.
158. Lai CH, Huang HJ, Hsueh S, et al. Human papillomavirus genotype in cervical cancer: a population-based study. Int J Cancer 2007;120:1999–2006.
159. Wang CC, Lai CH, Huang HJ, et al. Clinical effect of human papillomavirus genotypes in patients with cervical cancer undergoing primary radiotherapy. Int J Radiat Oncol Biol Phys 2010;78:1111–20.
160. Alvarez RD, Soong SJ, Kinney WK, et al. Identification of prognostic factors and risk groups in patients found to have nodal metastasis at the time of radical hysterectomy for early-stage squamous carcinoma of the cervix. Gynecol Oncol 1989;35:130–135.
161. Delgado G, Bundy BN, Fowler WC Jr, et al. A prospective surgical pathological study of stage I squamous carcinoma of the cervix: a Gynecologic Oncology Group Study. Gynecol Oncol 1989;35:314–320.
162. Finan MA, DeCesare S, Fiorica JV, et al. Radical hysterectomy for stage IB1 vs IB2 carcinoma of the cervix: does the new staging system predict morbidity and survival? Gynecol Oncol 1996;62:139–147.
163. Toita T, Nakano M, Higashi M, et al. Prognostic value of cervical size and pelvic lymph node status assessed by computed tomography for patients with uterine cervical cancer treated by radical radiation therapy. Int J Radiat Oncol Biol Phys 1995;33:843–849.
164. Piver MS, Chung WS. Prognostic significance of cervical lesion size and pelvic node metastases in cervical carcinoma. Obstet Gynecol 1975;46:507–510.
165. Fletcher GH. Cancer of the uterine cervix. Janeway Lecture, 1970. Am J Roengenol Radium Ther Nucl Med 1971;3:225–242.
166. Eifel PJ, Morris M, Wharton JT, et al. The influence of tumor size and morphology on the outcome of patients with FIGO stage IB squamous cell carcinoma of the uterine cervix. Int J Radiat Oncol Biol Phys 1994;29:9–16.
167. Perez CA, Grigsby PW, Nene SM, et al. Effect of tumor size on the prognosis of carcinoma of the uterine cervix treated with irradiation alone. Cancer 1992;69:2796–2806.
168. Eifel PJ, Thoms WW, Smith TL, et al. The relationship between brachytherapy dose and outcome in patients with bulky endocervical tumors treated with radiation alone. Int J Radiat Oncol Biol Phys 1994;28:113–118.
169. Perez CA, Grigsby PW, Chao KS, et al. Tumor size, irradiation dose, and long-term outcome of carcinoma of uterine cervix. Int J Radiat Oncol Biol Phys 1998;41:307–317.
170. Leveque J, Laurent JF, Burtin F, et al. Prognostic factors of the uterine cervix adenocarcinoma. Eur J Obstet Gynecol Reprod Biol 1998;80:209–214.
171. Silver DF, Hempling RE, Piver MS, et al. Stage I adenocarcinoma of the cervix: does lesion size affect treatment options and prognosis? Am J Clin Oncol 1998;21:431–435.
172. Grigsby PW, Perez CA, Chao KS, et al. Lack of effect of tumor size on the prognosis of carcinoma of the uterine cervix Stage IB and IIA treated with preoperative irradiation and surgery. Int J Radiat Oncol Biol Phys1999;45:645–651.
173. Grimard L, Genest P, Girard A, et al. Prognostic significance of endometrial extension in carcinoma of the cervix. Gynecol Oncol 1986;31:301–309.
174. Noguchi H, Shiozawa I, Kitahara T, et al. Uterine body invasion of carcinoma of the uterine cervix as seen from surgical specimens. Gynecol Oncol 1988;30:173–182.
175. Estape RE, Angioli R, Madrigal M, et al. Close vaginal margins as a prognostic factor after radical hysterectomy. Gynecol Oncol 1998;68:229–232.
176. Hong JH, Tsai CS, Lai CH, et al. Postoperative low-pelvic irradiation for stage I-IIA cervical cancer patients with risk factors other than pelvic lymph node metastasis. Int J Radiat Oncol Biol Phys 2002;53:1284–1290.
177. Viswanathan AN, Lee H, Hanson E, et al. Influence of margin status and radiation on recurrence after radical hysterectomy in Stage IB cervical cancer. Int J Radiat Oncol Biol Phys 2006;65:1501–1507.
178. Alfsen GC, Kristensen GB, Skovlund E, et al. Histologic subtype has minor importance for overall survival in patients with adenocarcinoma of the uterine cervix: a population-based study of prognostic factors in 505 patients with nonsquamous cell carcinomas of the cervix. Cancer 2001;92:2471–2483.
179. Chen F, Trapido EJ, Davis K. Differences in stage at presentation of breast and gynecologic cancers among whites, blacks, and Hispanics. Cancer 1994;73:2838–2842.
180. Waldenstrom AC, Horvath G. Survival of patients with adenocarcinoma of the uterine cervix in western Sweden. Int J Gynecol Cancer 1999;9:18–23.
181. Reagan JW, Fu YS. Histologic types and prognosis of cancers of the uterine cervix. Int J Radiat Oncol Biol Phys 1979;5:1015–1020.
182. Crissman JD, Budhraja M, Aron BS, et al. Histopathologic prognostic factors in stage II and III squamous cell carcinoma of the uterine cervix. An evaluation of 91 patients treated primarily with radiation therapy. Int J Gynecol Pathol1987;6:97–103.
183. Monk BJ, Wang J, Im S, et al. Rethinking the use of radiation and chemotherapy after radical hysterectomy: a clinical-pathologic analysis of a Gynecologic Oncology Group/Southwest Oncology Group/Radiation Therapy Oncology Group trial. Gynecol Oncol 2005;96:721–728.
184. Petereit DG, Sarkaria JN, Chappell R, et al. The adverse effect of treatment prolongation in cervical carcinoma. Int J Radiat Oncol Biol Phys 1995;32:1301–1307.
185. Fyles A, Keane TJ, Barton M, et al. The effect of treatment duration in the local control of cervix cancer. Radiother Oncol 1992;25:273–279.
186. Girinsky T, Rey A, Roche B, et al. Overall treatment time in advanced cervical carcinomas: a critical parameter in treatment outcome. Int J Radiat Oncol Biol Phys 1993;27:1051–1056.
187. Lanciano RM, Pajak TF, Martz K, et al. The influence of treatment time on outcome for squamous cell cancer of the uterine cervix treated with radiation: a patterns-of-care study. Int J Radiat Oncol Biol Phys 1993;25:391–397.
188. Chatani M, Matayoshi Y, Masaki N, et al. High-dose rate intracavitary irradiation for carcinoma of the uterine cervix. The adverse effect of treatment prolongation. Strahlenther Onkol 1997;173:379–384.
189. Perez CA, Grigsby PW, Castro-Vita H, et al. Carcinoma of the uterine cervix. I. Impact of prolongation of overall treatment time and timing of brachytherapy on outcome of radiation therapy. Int J Radiat Oncol Biol Phys1995;32:1275–1288.
190. Noordhuis MG, Eijsink JJ, Roossink F, et al. Prognostic cell biological markers in cervical cancer patients primarily treated with (chemo)radiation: a systematic review. Int J Radiat Oncol Biol Phys 2011;79:325–334.
191. Bachtiary B, Schindl M, Potter R, et al. Overexpression of hypoxia-inducible factor 1alpha indicates diminished response to radiotherapy and unfavorable prognosis in patients receiving radical radiotherapy for cervical cancer. Clin Cancer Res2003;9:2234–2240.
192. Burri P, Djonov V, Aebersold DM, et al. Significant correlation of hypoxia-inducible factor–1alpha with treatment outcome in cervical cancer treated with radical radiotherapy. Int J Radiat Oncol Biol Phys 2003;56:494–501.
193. Hutchison GJ, Valentine HR, Loncaster JA, et al. Hypoxia-inducible factor 1alpha expression as an intrinsic marker of hypoxia: correlation with tumor oxygen, pimonidazole measurements, and outcome in locally advanced carcinoma of the cervix. Clin Cancer Res 2004;10:8405–8412.
194. Kawanaka T, Kubo A, Ikushima H, Sano T, Takegawa Y, Nishitani H. Prognostic significance of HIF-2alpha expression on tumor infiltrating macrophages in patients with uterine cervical cancer undergoing radiotherapy. J Med Invest 2008;55:78–86.
195. Loncaster JA, Cooper RA, Logue JP, et al. Vascular endothelial growth factor (VEGF) expression is a prognostic factor for radiotherapy outcome in advanced carcinoma of the cervix. Br J Cancer 2000;83:620–625.
196. Kabuubi P, Loncaster JA, Davidson SE, et al. No relationship between thymidine phosphorylase (TP, PD-ECGF) expression and hypoxia in carcinoma of the cervix. Br J Cancer 2006;94:115–120.
197. Chen HH, Su WC, Chou CY, et al. Increased expression of nitric oxide synthase and cyclooxygenase-2 is associated with poor survival in cervical cancer treated with radiotherapy. Int J Radiat Oncol Biol Phys 2005;63:1093–1100.
198. Lee S, Shin HJ, Han IO, et al. Tumor carbonic anhydrase 9 expression is associated with the presence of lymph node metastases in uterine cervical cancer. Cancer Sci 2007;98:329–333.
199. Loncaster JA, Harris AL, Davidson SE, et al. Carbonic anhydrase (CA IX) expression, a potential new intrinsic marker of hypoxia: correlations with tumor oxygen measurements and prognosis in locally advanced carcinoma of the cervix. Cancer Res2001;61:6394–6399.
200. Kim JY, Shin HJ, Kim TH, et al. Tumor-associated carbonic anhydrases are linked to metastases in primary cervical cancer. J Cancer Res Clin Oncol 2006;132:302–308.
201. Obermair A, Wanner C, Bilgi S, et al. Tumor angiogenesis in stage IB cervical cancer: correlation of microvessel density with survival. Am J Obstet Gynecol 1998;178:314–319.
202. Cooper RA, Wilks DP, Logue JP, et al. High tumor angiogenesis is associated with poorer survival in carcinoma of the cervix treated with radiotherapy. Clin Cancer Res 1998;4:2795–800.
203. Kristensen GB, Kaern J, Abeler VM, et al. No prognostic impact of flow-cytometric measured DNA ploidy and S-phase fraction in cancer of the uterine cervix: a prospective study of 465 patients. Gynecol Oncol 1995;57:79–85.
204. Strang P, Eklund G, Stendahl U, et al. S-phase rate as a predictor of early recurrences in carcinoma of the uterine cervix. Anticancer Res 1987;7:807–810.
205. Wootipoom V, Lekhyananda N, Phungrassami T, et al. Prognostic significance of Bax, Bcl–2, and p53 expressions in cervical squamous cell carcinoma treated by radiotherapy. Gynecol Oncol 2004;94:636–642.
206. Cho NH, Kim YB, Park TK, et al. P63 and EGFR as prognostic predictors in stage IIB radiation-treated cervical squamous cell carcinoma. Gynecol Oncol 2003;91:346–353.
207. Ohno T, Nakano T, Niibe Y, et al. Bax protein expression correlates with radiation-induced apoptosis in radiation therapy for cervical carcinoma. Cancer 1998;83:103–110.
208. Jain D, Srinivasan R, Patel FD, et al. Evaluation of p53 and Bcl–2 expression as prognostic markers in invasive cervical carcinoma stage IIb/III patients treated by radiotherapy. Gynecol Oncol 2003;88:22–28.
209. Mukherjee G, Freeman A, Moore R, et al. Biologic factors and response to radiotherapy in carcinoma of the cervix. Int J Gynecol Cancer 2001;11:187–193.
210. Kainz C, Kohlberger P, Gitsch G, et al. Mutant p53 in patients with invasive cervical cancer stages IB to IIB. Gynecol Oncol 1995;57:212–214.
211. Ebara T, Mitsuhashi N, Saito Y, et al. Prognostic significance of immunohistochemically detected p53 protein expression in stage IIIB squamous cell carcinoma of the uterine cervix treated with radiation therapy alone. Gynecol Oncol 1996;63:216–218.
212. Cerciello F, Hofstetter B, Fatah SA, et al. G2/M cell cycle checkpoint is functional in cervical cancer patients after initiation of external beam radiotherapy. Int J Radiat Oncol Biol Phys 2005;62:1390–1398.
213. Tsang RW, Wong CS, Fyles AW, et al. Tumour proliferation and apoptosis in human uterine cervix carcinoma II: correlations with clinical outcome. Radiother Oncol 1999;50:93–101.
214. West CM, Davidson SE, Burt PA, et al. The intrinsic radiosensitivity of cervical carcinoma: correlations with clinical data. Int J Radiat Oncol Biol Phys 1995;31:841–846.
215. Oka K, Suzuki Y, Nakano T. Expression of p27 and p53 in cervical squamous cell carcinoma patients treated with radiotherapy alone: radiotherapeutic effect and prognosis. Cancer 2000;88:2766–2773.
216. Hayashi Y, Hachisuga T, Iwasaka T, et al. Expression of ras oncogene product and EGF receptor in cervical squamous cell carcinomas and its relationship to lymph node involvement. Gynecol Oncol 1991;40:147–151.
217. Koulos JP, Wright TC, Mitchell MF, et al. Relationships between c-Ki-ras mutations, HPV types, and prognostic indicators in invasive endocervical adenocarcinomas. Gynecol Oncol 1993;48:364–369.
218. Lee JH, Lee SK, Yang MH, et al. Expression and mutation of H-ras in uterine cervical cancer. Gynecol Oncol 1996;62:49–54.
219. Iwasaka T, Yokoyama M, Oh-uchida M, et al. Detection of human papillomavirus genome and analysis of expression of c-myc and Ha-ras oncogenes in invasive cervical carcinomas. Gynecol Oncol 1992;46:298–303.
220. Riou G, Barrois M, Le MG, et al. C-myc proto-oncogene expression and prognosis in early carcinoma of the uterine cervix. Lancet 1987;1:761–763.
221. Santucci MA, Barbieri E, Frezza G, et al. Radiation-induced gadd45 expression correlates with clinical response to radiotherapy of cervical carcinoma. Int J Radiat Oncol Biol Phys 2000;46:411–416.
222. Vieira SC, Silva BB, Pinto GA, et al. CD34 as a marker for evaluating angiogenesis in cervical cancer. Pathol Res Pract 2005;201:313–318.
223. Zhang JM, Hashimoto M, Kawai K, et al. CD109 expression in squamous cell carcinoma of the uterine cervix. Pathol Int 2005;55:165–169.
224. Mandai M, Konishi I, Koshiyama M, et al. Altered expression of nm23-H1 and c-erbB–2 proteins have prognostic significance in adenocarcinoma but not in squamous cell carcinoma of the uterine cervix. Cancer1995;75:2523–2529.
225. Perez-Regadera J, Sanchez-Munoz A, De-la-Cruz J, et al. Negative prognostic impact of the coexpression of epidermal growth factor receptor and c-erbB-2 in locally advanced cervical cancer. Oncology 2009;76:133–141.
226. Harima Y, Sawada S, Nagata K, et al. Mutation of the PTEN gene in advanced cervical cancer correlated with tumor progression and poor outcome after radiotherapy. Int J Oncol 2001;18:493–497.
227. van Bommel PF, Kenemans P, Helmerhorst TJ, et al. Expression of cytokeratin 10, 13, and involucrin as prognostic factors in low stage squamous cell carcinoma of the uterine cervix. Cancer 1994;74:2314–2320.
228. Tsai CC, Lin H, Huang EY, et al. The role of the preoperative serum carcinoembryonic antigen level in early-stage adenocarcinoma of the uterine cervix. Gynecol Oncol 2004;94:363–367.
229. Borras G, Molina R, Xercavins J, et al. Tumor antigens CA 19.9, CA 125, and CEA in carcinoma of the uterine cervix. Gynecol Oncol 1995;57:205–211.
230. Bolli JA, Doering DL, Bosscher JR, et al. Squamous cell carcinoma antigen: clinical utility in squamous cell carcinoma of the uterine cervix. Gynecol Oncol 1994;55:169–173.
231. Ngan HY, Cheng GTS, Cheng D, et al. Post-treatment serial serum squamous cell carcinoma antigen (SCC) in the monitoring of squamous cell carcinoma of the cervix. Int J Gynecol Cancer 1996;6:115–119.
232. Hong JH, Tsai CS, Chang JT, et al. The prognostic significance of pre- and posttreatment SCC levels in patients with squamous cell carcinoma of the cervix treated by radiotherapy. Int J Radiat Oncol Biol Phys 1998;41:823–830.
233. Micke O, Bruns F, Schafer U, et al. The impact of squamous cell carcinoma (SCC) antigen in patients with advanced cancer of uterine cervix treated with (chemo-)radiotherapy. Anticancer Res 2005;25:1663–1666.
234. Hong JH, Tsai CS, Lai CH, et al. Risk stratification of patients with advanced squamous cell carcinoma of cervix treated by radiotherapy alone. Int J Radiat Oncol Biol Phys 2005;63:492–499.
235. Huang EY, Wang CJ, Chen HC, et al. Multivariate analysis of para-aortic lymph node recurrence after definitive radiotherapy for stage IB-IVA squamous cell carcinoma of uterine cervix. Int J Radiat Oncol Biol Phys2008;72:834–842.
236. Huang EY, Huang YJ, Chanchien CC, et al. Pretreatment carcinoembryonic antigen level is a risk factor for para-aortic lymph node recurrence in addition to squamous cell carcinoma antigen following definitive concurrent chemoradiotherapy for squamous cell carcinoma of the uterine cervix. Radiat Oncol 2012;7:13.
237. Kitano Y, Fujisaki S, Nakamura N, et al. Immunological disorder against the Epstein-Barr virus infection and prognosis in patients with cervical carcinoma. Gynecol Oncol 1995;57:150–157.
238. Dickson J, Davidson SE, Hunter RD, et al. Pretreatment plasma TGF beta 1 levels are prognostic for survival but not morbidity following radiation therapy of carcinoma of the cervix. Int J Radiat Oncol Biol Phys 2000;48:991–995.
239. Hazelbag S, Kenter GG, Gorter A, et al. Prognostic relevance of TGF-beta1 and PAI–1 in cervical cancer. Int J Cancer 2004;112:1020–1028.
240. Gruber G, Hess J, Stiefel C, et al. Correlation between the tumoral expression of beta3-integrin and outcome in cervical cancer patients who had undergone radiotherapy. Br J Cancer 2005;92:41–46.
241. Gaffney DK, Holden J, Davis M, et al. Elevated cyclooxygenase-2 expression correlates with diminished survival in carcinoma of the cervix treated with radiotherapy. Int J Radiat Oncol Biol Phys 2001;49:1213–1217.
242. Kim YB, Kim GE, Pyo HR, et al. Differential cyclooxygenase-2 expression in squamous cell carcinoma and adenocarcinoma of the uterine cervix. Int J Radiat Oncol Biol Phys 2004;60:822–829.
243. Pyo H, Kim YB, Cho NH, et al. Coexpression of cyclooxygenase-2 and thymidine phosphorylase as a prognostic indicator in patients with FIGO stage IIB squamous cell carcinoma of uterine cervix treated with radiotherapy and concurrent chemotherapy. Int J Radiat Oncol Biol Phys 2005;62:725–732.
244. Kang S, Kim MH, Park IA, et al. Elevation of cyclooxygenase-2 is related to lymph node metastasis in adenocarcinoma of uterine cervix. Cancer Lett 2006;237:305–311.
245. Suzuki Y, Nakano T, Arai T, et al. Progesterone receptor is a favorable prognostic factor of radiation therapy for adenocarcinoma of the uterine cervix. Int J Radiat Oncol Biol Phys 2000;47:1229–1234.
246. Nasiell K, Roger V, Nasiell M. Behavior of mild cervical dysplasia during long-term follow-up. Obstet Gynecol 1986;67:665–669.
247. Ferenczy A. Management of patients with high grade squamous intraepithelial lesions. Cancer 1995;76:1928–1933.
248. Michelin MA, Merino LM, Franco CA, et al. Pregnancy outcome after treatment of cervical intraepithelial neoplasia by the loop electrosurgical excision procedure and cold knife conization. Clin Exp Obstet Gynecol2009;36:17–19.
249. Kyrgiou M, Koliopoulos G, Martin-Hirsch P, et al. Obstetric outcomes after conservative treatment for intraepithelial or early invasive cervical lesions: systematic review and meta-analysis. Lancet 2006;367:489–498.
250. Arbyn M, Kyrgiou M, Simoens C, et al. Perinatal mortality and other severe adverse pregnancy outcomes associated with treatment of cervical intraepithelial neoplasia: meta-analysis. BMJ 2008;337:a1284.
251. Maneo A, Sideri M, Scambia G, et al. Simple conization and lymphadenectomy for the conservative treatment of stage IB1 cervical cancer. An Italian experience. Gynecol Oncol 2011;123:557–560.
252. Riva HL, Hefner JD, Marchetti AA, et al. Prophylactic trachelectomy of cervical stump: two hundred and twelve cases. South Med J 1961;54:1082–1084.
253. Dargent D, Mathevet P. Schauta’s vaginal hysterectomy combined with laparoscopic lymphadenectomy. Baillieres Clin Obstet Gynaecol 1995;9:691–705.
254. Peppercorn PD, Jeyarajah AR, Woolas R, et al. Role of MR imaging in the selection of patients with early cervical carcinoma for fertility-preserving surgery: initial experience. Radiology 1999;212:395–399.
255. Smith JR, Boyle DC, Corless DJ, et al. Abdominal radical trachelectomy: a new surgical technique for the conservative management of cervical carcinoma. Br J Obstet Gynaecol 1997;104:1196–1200.
256. Piver MS, Rutledge F, Smith JP. Five classes of extended hysterectomy for women with cervical cancer. Obstet Gynecol 1974;44:265–272.
257. Wertheim E. The extended abdominal operation for carcinoma uteri (based on 500 operative cases). Am J Obstet 1912:169–174.
258. Meigs J. Carcinoma of the cervix: the Wertheim operation. Gynecol Obstet 1944:195–199.
259. Querleu D, Morrow CP. Classification of radical hysterectomy. Lancet Oncol 2008;9:297–303.
260. Kramer C, Peschel RE, Goldberg N, et al. Radiation treatment of FIGO stage IVA carcinoma of the cervix. Gynecol Oncol 1989;32:323–326.
261. Million RR, Rutledge F, Fletcher GH. Stage IV carcinoma of the cervix with bladder invasion. Am J Obstet Gynecol 1972;113:239–246.
262. Upadhyay SK, Symonds RP, Haelterman M, et al. The treatment of stage IV carcinoma of cervix by radical dose radiotherapy. Radiother Oncol 1988;11:15–19.
263. Van Dyke AH, Van Nagell JR Jr. The prognostic significance of ureteral obstruction in patients with recurrent carcinoma of the cervix uteri. Surg Gynecol Obstet 1975;141:371–373.
264. Lagasse LD, Creasman WT, Shingleton HM, et al. Results and complications of operative staging in cervical cancer: experience of the Gynecologic Oncology Group. Gynecol Oncol 1980;9:90–98.
265. Stehman FB, Bundy BN, DiSaia PJ, et al. Carcinoma of the cervix treated with radiation therapy. I. A multi-variate analysis of prognostic variables in the Gynecologic Oncology Group. Cancer 1991;67:2776–2785.
266. Berman ML, Keys H, Creasman W, et al. Survival and patterns of recurrence in cervical cancer metastatic to periaortic lymph nodes (a Gynecologic Oncology Group study). Gynecol Oncol 1984;19:8–16.
267. Cosin JA, Fowler JM, Chen MD, et al. Pretreatment surgical staging of patients with cervical carcinoma: the case for lymph node debulking. Cancer 1998;82:2241–2248.
268. Potish RA. Surgical staging, extended field radiation, and enteric morbidity in the treatment of cervix cancer. Int J Radiat Oncol Biol Phys 1995;31:1009–1010.
269. Potish RA, Downey GO, Adcock LL, et al. The role of surgical debulking in cancer of the uterine cervix. Int J Radiat Oncol Biol Phys 1989;17:979–984.
270. Inoue T, Morita K. Long-term observation of patients treated by postoperative extended-field irradiation for nodal metastases from cervical carcinoma stages IB, IIA, and IIB. Gynecol Oncol 1995;58:4–10.
271. Esthappan J, Mutic S, Malyapa RS, et al. Treatment planning guidelines regarding the use of CT/PET-guided IMRT for cervical carcinoma with positive paraaortic lymph nodes. Int J Radiat Oncol Biol Phys 2004;58:1289–1297.
272. Portelance L, Chao KS, Grigsby PW, et al. Intensity-modulated radiation therapy (IMRT) reduces small bowel, rectum, and bladder doses in patients with cervical cancer receiving pelvic and para-aortic irradiation. Int J Radiat Oncol Biol Phys2001;51:261–266.
273. Mutic S, Malyapa RS, Grigsby PW, et al. PET-guided IMRT for cervical carcinoma with positive para-aortic lymph nodes-a dose-escalation treatment planning study. Int J Radiat Oncol Biol Phys 2003;55:28–35.
274. Esthappan J, Chaudhari S, Santanam L, et al. Prospective clinical trial of positron emission tomography/computed tomography image-guided intensity-modulated radiation therapy for cervical carcinoma with positive para-aortic lymph nodes. Int J Radiat Oncol Biol Phys 2008;72:1134–1139.
275. Poorvu PD, Sadow CA, Townamchai K et al. Duodenal and other gastrointestinal toxicity in cervical and endometrial cancer treated with extended-field intensity modulated radiation therapy to paraaortic lymph nodes. Int J Radiat Oncol Biol Phys 2012; doi: 10.1016/j.ijrobp.2012.10.004. [epub ahead of print].
276. Weiser EB, Bundy BN, Hoskins WJ, et al. Extraperitoneal versus transperitoneal selective paraaortic lymphadenectomy in the pretreatment surgical staging of advanced cervical carcinoma (a Gynecologic Oncology Group study). Gynecol Oncol1989;33:283–289.
277. Fine BA, Hempling RE, Piver MS, et al. Severe radiation morbidity in carcinoma of the cervix: impact of pretherapy surgical staging and previous surgery. Int J Radiat Oncol Biol Phys 1995;31:717–723.
278. Berman ML, Lagasse LD, Watring WG, et al. The operative evaluation of patients with cervical carcinoma by an extraperitoneal approach. Obstet Gynecol 1977;50:658–664.
279. Wharton JT, Jones HW 3rd, Day TG Jr, et al. Preirradiation celiotomy and extended field irradiation for invasive carcinoma of the cervix. Obstet Gynecol 1977;49:333–338.
280. Leblanc E, Gauthier H, Querleu D, et al. Accuracy of 18-fluoro-2-deoxy-D-glucose positron emission tomography in the pretherapeutic detection of occult para-aortic node involvement in patients with a locally advanced cervical carcinoma. Ann Surg Oncol 2011;18:2302–2309.
281. Brockbank E, Kokka F, Bryant A, et al. Pre-treatment surgical para-aortic lymph node assessment in locally advanced cervical cancer. Cochrane Database Syst Rev 2011:CD008217.
282. Lai CH, Huang KG, Hong JH, et al. Randomized trial of surgical staging (extraperitoneal or laparoscopic) versus clinical staging in locally advanced cervical cancer. Gynecol Oncol 2003;89:160–167.
283. Fowler JM, Carter JR, Carlson JW, et al. Lymph node yield from laparoscopic lymphadenectomy in cervical cancer: a comparative study. Gynecol Oncol 1993;51:187–192.
284. Ramirez PT, Jhingran A, Macapinlac HA, et al. Laparoscopic extraperitoneal para-aortic lymphadenectomy in locally advanced cervical cancer: a prospective correlation of surgical findings with positron emission tomography/computed tomography findings. Cancer 2011;117:1928–1934.
285. Perez-Mesa C, Spratt JS. Scalene node biopsy in the pretreatment staging of carcinoma of the cervix uteri. Am J Obstet Gynecol 1976;125:93–95.
286. Manetta A, Podczaski ES, Larson JE, et al. Scalene lymph node biopsy in the preoperative evaluation of patients with recurrent cervical cancer. Gynecol Oncol 1989;33:332–334.
287. Chereau E, Feron JG, Ballester M, et al. Contribution of pelvic and para-aortic lymphadenectomy with sentinel node biopsy in patients with IB2-IIB cervical cancer. Br J Cancer 2012;106:39–44.
288. Di Stefano AB, Acquaviva G, Garozzo G, et al. Lymph node mapping and sentinel node detection in patients with cervical carcinoma: a 2-year experience. Gynecol Oncol 2005;99:671–679.
289. Bats AS, Buenerd A, Querleu D, et al. Diagnostic value of intraoperative examination of sentinel lymph node in early cervical cancer: a prospective, multicenter study. Gynecol Oncol 2011;123:230–235.
290. Lecuru F, Mathevet P, Querleu D, et al. Bilateral negative sentinel nodes accurately predict absence of lymph node metastasis in early cervical cancer: results of the SENTICOL study. J Clin Oncol 2011;29:1686–1691.
291. Yamamoto R, Okamoto K, Yukiharu T, et al. A study of risk factors for ovarian metastases in stage Ib-IIIb cervical carcinoma and analysis of ovarian function after a transposition. Gynecol Oncol 2001;82:312–316.
292. Parker M, Bosscher J, Barnhill D, et al. Ovarian management during radical hysterectomy in the premenopausal patient. Obstet Gynecol 1993;82:187–190.
293. Flay LD, Matthews JH. The effects of radiotherapy and surgery on the sexual function of women treated for cervical cancer. Int J Radiat Oncol Biol Phys 1995;31:399–404.
294. Anderson B, LaPolla J, Turner D, et al. Ovarian transposition in cervical cancer. Gynecol Oncol 1993;49:206–214.
295. Feeney DD, Moore DH, Look KY, et al. The fate of the ovaries after radical hysterectomy and ovarian transposition. Gynecol Oncol 1995;56:3–7.
296. Buekers TE, Anderson B, Sorosky JI, et al. Ovarian function after surgical treatment for cervical cancer. Gynecol Oncol 2001;80:85–88.
297. Morice P, Juncker L, Rey A, et al. Ovarian transposition for patients with cervical carcinoma treated by radiosurgical combination. Fertil Steril 2000;74:743–748.
298. Le Bouedec G, Rabishong B, Canis M, et al. Ovarian transposition by laparoscopy in young women before curietherapy for cervical cancer [in French]. J Gynecol Obstet Biol Reprod (Paris) 2000;29:564–570.
299. Stockle E, Verdier G, Thomas L, et al. Functional outcome of laparoscopically transposed ovaries in the multidisciplinary treatment of cervical cancers. Analysis of risk factors [in French]. J Gynecol Obstet Biol Reprod (Paris)1996;25:244–252.
300. Pahisa J, Martinez-Roman S, Martinez-Zamora MA, et al. Laparoscopic ovarian transposition in patients with early cervical cancer. Int J Gynecol Cancer 2008;18:584–589.
301. Greer BE, Koh WJ, Figge DC, et al. Gynecologic radiotherapy fields defined by intraoperative measurements. Gynecol Oncol 1990;38:421–424.
302. Perez CA, Kavanagh BD. Uterine cervix. In: Perez CA, Brady LW, Halperin EC, et al., eds. Principles and practice of radiation oncology. Philadelphia: Lippincott Williams & Wilkins, 2004:1800–915.
303. Russell AH, Walter JP, Anderson MW, et al. Sagittal magnetic resonance imaging in the design of lateral radiation treatment portals for patients with locally advanced squamous cancer of the cervix. Int J Radiat Oncol Biol Phys1992;23:449–455.
304. Bonin SR, Lanciano RM, Corn BW, et al. Bony landmarks are not an adequate substitute for lymphangiography in defining pelvic lymph node location for the treatment of cervical cancer with radiotherapy. Int J Radiat Oncol Biol Phys 1996;34:167–172.
305. Zunino S, Rosato O, Lucino S, et al. Anatomic study of the pelvis in carcinoma of the uterine cervix as related to the box technique. Int J Radiat Oncol Biol Phys 1999;44:53–59.
306. Knocke TH, Pokrajac B, Fellner C, et al. A comparison of CT-supported 3D planning with simulator planning in the pelvic irradiation of primary cervical carcinoma [in German]. Strahlenther Onkol 1999;175:68–73.
307. Finlay MH, Ackerman I, Tirona RG, et al. Use of CT simulation for treatment of cervical cancer to assess the adequacy of lymph node coverage of conventional pelvic fields based on bony landmarks. Int J Radiat Oncol Biol Phys 2006;64:205–209.
308. Taylor A, Rockall AG, Reznek RH, Powell ME. Mapping pelvic lymph nodes: guidelines for delineation in intensity-modulated radiotherapy. Int J Radiat Oncol Biol Phys 2005;63:1604–1612.
309. van den Berg HA, Olofsen-van Acht MJ, van Santvoort JP, et al. Definition and validation of a reference target volume in early stage node-positive cervical carcinoma, based on lymphangiograms and CT-scans. Radiother Oncol2000;54:163–170.
310. Yamazaki A, Shirato H, Nishioka T, et al. Reduction of late complications after irregularly shaped four-field whole pelvic radiotherapy using computed tomographic simulation compared with parallel-opposed whole pelvic radiotherapy. Jpn J Clin Oncol2000;30:180–184.
311. Burnett AF, Coe FL, Klement V, et al. The use of a pelvic displacement prosthesis to exclude the small intestine from the radiation field following radical hysterectomy. Gynecol Oncol 2000;79:438–443.
312. Bouchard M, Nadeau S, Germain I, et al. Anatomy-based MLC field optimization for the treatment of gynecologic malignancies. Int J Radiat Oncol Biol Phys 2005;63:S344–S345.
313. Han Y, Shin EH, Huh SJ, et al. Interfractional dose variation during intensity-modulated radiation therapy for cervical cancer assessed by weekly CT evaluation. Int J Radiat Oncol Biol Phys 2006;65:617–623.
314. Beadle BM, Jhingran A, Salehpour M, et al. Cervix regression and motion during the course of external beam chemoradiation for cervical cancer. Int J Radiat Oncol Biol Phys 2009;73:235–241.
315. Haripotepornkul NH, Nath SK, Scanderbeg D, et al. Evaluation of intra- and inter-fraction movement of the cervix during intensity modulated radiation therapy. Radiother Oncol 2011;98:347–351.
316. Tyagi N, Lewis JH, Yashar CM, et al. Daily online cone beam computed tomography to assess interfractional motion in patients with intact cervical cancer. Int J Radiat Oncol Biol Phys 2011;80:273–280.
317. Lim K, Chan P, Dinniwell R, et al. Cervical cancer regression measured using weekly magnetic resonance imaging during fractionated radiotherapy: radiobiologic modeling and correlation with tumor hypoxia. Int J Radiat Oncol Biol Phys 2008;70:126–133.
318. Lee CM, Shrieve DC, Gaffney DK. Rapid involution and mobility of carcinoma of the cervix. Int J Radiat Oncol Biol Phys 2004;58:625–230.
319. van de Bunt L, Jurgenliemk-Schulz IM, de Kort GA, et al. Motion and deformation of the target volumes during IMRT for cervical cancer: what margins do we need? Radiother Oncol 2008;88:233–240.
320. Lim K, Small W Jr, Portelance L, et al. Consensus guidelines for delineation of clinical target volume for intensity-modulated pelvic radiotherapy for the definitive treatment of cervix cancer. Int J Radiat Oncol Biol Phys2011;79:348–355.
321. Small W Jr, Mell LK, Anderson P, et al. Consensus guidelines for delineation of clinical target volume for intensity-modulated pelvic radiotherapy in postoperative treatment of endometrial and cervical cancer. Int J Radiat Oncol Biol Phys 2008;71:428–434.
322. Ma DJ, Michaletz-Lorenz M, Goddu SM, et al. Magnitude of interfractional vaginal cuff movement: implications for external irradiation. Int J Radiat Oncol Biol Phys 2012;82:1439–1444.
323. Jhingran A, Salehpour M, Sam M, et al. Vaginal motion and bladder and rectal volumes during pelvic intensity-modulated radiation therapy after hysterectomy. Int J Radiat Oncol Biol Phys 2012;82:256–262.
324. A phase ii study of postoperative intensity modulated radiation therapy (IMRT) with concurrent cisplatin and bevacizumab followed by carboplatin and paclitaxel for patients with endometrial cancer. Radiation Therapy Oncology Group, 2010. Available at: http://www.rtog.org/ClinicalTrials/ProtocolTable/StudyDetails.aspx?study=0921. Accessed February 1, 2012.
325. Miller TR, Grigsby PW. Measurement of tumor volume by PET to evaluate prognosis in patients with advanced cervical cancer treated by radiation therapy. Int J Radiat Oncol Biol Phys 2002;53:353–359.
326. Kavanagh BD, Pan CC, Dawson LA, et al. Radiation dose-volume effects in the stomach and small bowel. Int J Radiat Oncol Biol Phys 2010;76:S101–S107.
327. Michalski JM, Gay H, Jackson A, et al. Radiation dose-volume effects in radiation-induced rectal injury. Int J Radiat Oncol Biol Phys 2010;76:S123–S129.
328. Viswanathan AN, Yorke ED, Marks LB, et al. Radiation dose-volume effects of the urinary bladder. Int J Radiat Oncol Biol Phys 2010;76:S116–S122.
329. Ahmed RS, Kim RY, Duan J, et al. IMRT dose escalation for positive para-aortic lymph nodes in patients with locally advanced cervical cancer while reducing dose to bone marrow and other organs at risk. Int J Radiat Oncol Biol Phys 2004;60:505–512.
330. Heron DE, Gerszten K, Selvaraj RN, et al. Conventional 3D conformal versus intensity-modulated radiotherapy for the adjuvant treatment of gynecologic malignancies: a comparative dosimetric study of dose-volume histograms small star, filled. Gynecol Oncol 2003;91:39–45.
331. D’Souza WD, Ahamad AA, Iyer RB, et al. Feasibility of dose escalation using intensity-modulated radiotherapy in posthysterectomy cervical carcinoma. Int J Radiat Oncol Biol Phys 2005;61:1062–1070.
332. Adli M, Mayr NA, Kaiser HS, et al. Does prone positioning reduce small bowel dose in pelvic radiation with intensity-modulated radiotherapy for gynecologic cancer? Int J Radiat Oncol Biol Phys 2003;57:230–238.
333. Brixey CJ, Roeske JC, Lujan AE, et al. Impact of intensity-modulated radiotherapy on acute hematologic toxicity in women with gynecologic malignancies. Int J Radiat Oncol Biol Phys 2002;54:1388–1396.
334. Lujan AE, Mundt AJ, Yamada SD, et al. Intensity-modulated radiotherapy as a means of reducing dose to bone marrow in gynecologic patients receiving whole pelvic radiotherapy. Int J Radiat Oncol Biol Phys 2003;57:516–521.
335. Hasselle MD, Rose BS, Kochanski JD, et al. Clinical outcomes of intensity-modulated pelvic radiation therapy for carcinoma of the cervix. Int J Radiat Oncol Biol Phys 2011;80:1436–1445.
336. Guerrero M, Li XA, Ma L, Linder J, et al. Simultaneous integrated intensity-modulated radiotherapy boost for locally advanced gynecological cancer: radiobiological and dosimetric considerations. Int J Radiat Oncol Biol Phys2005;62:933–939.
337. Kavanagh BD, Gieschen HL, Schmidt-Ullrich RK, et al. A pilot study of concomitant boost accelerated superfractionated radiotherapy for stage III cancer of the uterine cervix. Int J Radiat Oncol Biol Phys 1997;38:561–568.
338. Hall EJ. Intensity-modulated radiation therapy, protons, and the risk of second cancers. Int J Radiat Oncol Biol Phys 2006;65:1–7.
339. Boice JD Jr, Day NE, Andersen A, et al. Second cancers following radiation treatment for cervical cancer. An international collaboration among cancer registries. J Natl Cancer Inst 1985;74:955–975.
340. Kamrava M, Mell LK, Yashar C. In-room image-guided radiation therapy for cervical cancers. In: Mundt A, Yashar C, Mell LK, eds. Gyncologic cancer. New York: Demos Medical, 2012:357–370.
341. Higginson DS, Morris DE, Jones EL, et al. Stereotactic body radiotherapy (SBRT): Technological innovation and application in gynecologic oncology. Gynecol Oncol 2011;120:404–412.
342. Viswanathan A. Image-guided brachytherapy in cervical cancer. In: Mundt A, Yashar C, Mell LK, eds. Gynecologic cancer. New York: Demos Medical, 2012:371–382.
343. Wolfson AH, Abdel-Wahab M, Markoe AM, et al. A quantitative assessment of standard vs. customized midline shield construction for invasive cervical carcinoma. Int J Radiat Oncol Biol Phys 1997;37:237–242.
344. Lee LJ, Sadow CA, Russell A, et al. Correlation of point B and lymph node dose in 3D-planned high-dose-rate cervical cancer brachytherapy. Int J Radiat Oncol Biol Phys 2009;75:803–809.
345. Fenkell L, Assenholt M, Nielsen SK, et al. Parametrial boost using midline shielding results in an unpredictable dose to tumor and organs at risk in combined external beam radiotherapy and brachytherapy for locally advanced cervical cancer. Int J Radiat Oncol Biol Phys 2011;79:1572–1579.
346. Townamchai K, Lee LJ, Poorvu PD, et al. Clinical outcomes with dose escalation using intensity modulated radiotherapy for node-positive endometrial and cervical cancer. Int J Radiat Oncol Biol Phys 2012;84:S454.
347. Podczaski E, Stryker JA, Kaminski P, et al. Extended-field radiation therapy for carcinoma of the cervix. Cancer 1990;66:251–258.
348. Russell AH, Jones DC, Russell KJ, et al. High dose para-aortic lymph node irradiation for gynecologic cancer: technique, toxicity, and results. Int J Radiat Oncol Biol Phys 1987;13:267–271.
349. Kodaira T, Karasawa K, Shimizu T, et al. Clinical efficacy of applying four-field portals to paraaortic irradiation in the treatment of cervical carcinoma. Radiat Oncol Invest 1999;7:170–177.
350. Holcomb K, Gabbur N, Tucker T, et al. 60Cobalt vs. linear accelerator in the treatment of locally advanced cervix carcinoma: a comparison of survival and recurrence patterns. Eur J Gynaecol Oncol 2001;22:16–19.
351. Biggs PJ, Russell MD. Effect of a femoral head prosthesis on megavoltage beam radiotherapy. Int J Radiat Oncol Biol Phys 1988;14:581–586.
352. Allt WE. Supervoltage radiation treatment in advanced cancer of the uterine cervix. A preliminary report. Can Med Assoc J 1969;100:792–797.
353. Johns HE. Optimization of energy and equipment. In: Kramer S, Suntharalingam N, Zinniger GF, eds. High-energy photons and electrons: clinical applications in cancer management. New York: Wiley; 1976:333–345.
354. Maor MH, Gillespie BW, Peters LJ, et al. Neutron therapy in cervical cancer: results of a phase III RTOG Study. Int J Radiat Oncol Biol Phys 1988;14:885–891.
355. Kato S, Ohno T, Tsujii H, et al. Dose escalation study of carbon ion radiotherapy for locally advanced carcinoma of the uterine cervix. Int J Radiat Oncol Biol Phys 2006;65:388–397.
356. MacLeod C, Bernshaw D, Leung S, et al. Accelerated hyperfractionated radiotherapy for locally advanced cervix cancer. Int J Radiat Oncol Biol Phys 1999;44:519–524.
357. Viswanathan FR, Varghese C, Peedicayil A, et al. Hyperfractionation in carcinoma of the cervix: tumor control and late bowel complications. Int J Radiat Oncol Biol Phys 1999;45:653–656.
358. Grigsby PW, Heydon K, Mutch DG, et al. Long term follow up of Radiation Therapy Oncology Group 92–10: cervical cancer with positive para-aortic lymph nodes. Int J Radiat Oncol Biol Phys 2001;51:982–987.
359. Calkins AR, Harrison CR, Fowler WC Jr, et al. Hyperfractionated radiation therapy plus chemotherapy in locally advanced cervical cancer: results of two phase I dose-escalation Gynecologic Oncology Group trials. Gynecol Oncol 1999;75:349–355.
360. Thomas G, Dembo A, Ackerman I, et al. A randomized trial of standard versus partially hyperfractionated radiation with or without concurrent 5-fluouracil in locally advanced cervical cancer. Gynecol Oncol 1998;69:137–145.
361. Bjerre B, Eliasson G, Linell F, et al. Conization as only treatment of carcinoma in situ of the uterine cervix. Am J Obstet Gynecol 1976;125:143–152.
362. Randall ME, Michael H, Ver Morken J, et al. Uterine cervix. In: Hoskins WJ, Perez CA, Young RC, eds. Principles and practice of gynecologic oncology, 4th ed. Philadelphia: Lippincott: Williams & Wilkins; 2005:743–822.
363. Delregato JA, Cox JD. Transvaginal Roentgen therapy in the conservative management of carcinoma in situ of the uterine cervix. Radiology 1965;84:1090–1095.
364. Grigsby PW, Perez CA. Radiotherapy alone for medically inoperable carcinoma of the cervix: Stage IA and carcinoma in situ. Int J Radiat Oncol Biol Phys 1991;21:375–378.
365. Ogino I, Kitamura T, Okajima H, et al. High-dose-rate intracavitary brachytherapy in the management of cervical and vaginal intraepithelial neoplasia. Int J Radiat Oncol Biol Phys 1998;40:881–887.
366. Brunschwig A. The surgical treatment of cancer of the cervix: stage I and II. Am J Roentgenol Radium Ther Nucl Med 1968;102:147–151.
367. Liu W, Meigs JV. Radical hysterectomy and pelvic lymphadenectomy; a review of 473 cases including 244 for primary invasive carcinoma of the cervix. Am J Obstet Gynecol 1955;69:1–32.
368. Eifel PJ, Moughan J, Erickson B, et al. Patterns of radiotherapy practice for patients with carcinoma of the uterine cervix: a patterns of care study. Int J Radiat Oncol Biol Phys 2004;60:1144–1153.
369. Eifel PJ, Khalid N, Erickson B, et al. Patterns of radiotherapy practice for patients treated for intact cervical cancer in 2005–2007: a QRRO Study. Int J Radiat Oncol Biol Phys 2010;78:S119.
370. Barbera L, Paszat L, Thomas G, et al. The rapid uptake of concurrent chemotherapy for cervix cancer patients treated with curative radiation. Int J Radiat Oncol Biol Phys 2006;64:1389–1394.
371. National Cancer Institute. Concurrent chemoradiation for cervical cancer (Clinical Announcement). Bethesda, MD: Author, 1999.
372. Andras EJ, Fletcher G, Rutledge F. Radiotherapy of carcinoma of the cervix following simple hysterectomy. Am J Obstet Gynecol 1973;115:647–655.
373. Ampil F, Datta R, Datta S. Elective postoperative external radiotherapy after hysterectomy in early-stage carcinoma of the cervix. Is additional vaginal cuff irradiation necessary? Cancer 1987;60:280–288.
374. Green TH Jr, Morse WJ Jr. Management of invasive cervical cancer following inadvertent simple hysterectomy. Obstet Gynecol 1969;33:763–769.
375. Crane CH, Schneider BF. Occult carcinoma discovered after simple hysterectomy treated with postoperative radiotherapy. Int J Radiat Oncol Biol Phys 1999;43:1049–1053.
376. Huerta Bahena J, Labastida Almendaro S, Cortez Arroyo H, et al. Postoperative radiotherapy in patients with invasive uterine cervix cancer treated previously with simple hysterectomy. Results from the Hospital de Oncologia, Centro Medico Nacional SXXI [in Spanish]. Ginecol Obstet Mex 2003;71:304–311.
377. Munstedt K, Johnson P, von Georgi R, et al. Consequences of inadvertent, suboptimal primary surgery in carcinoma of the uterine cervix. Gynecol Oncol 2004;94:515–520.
378. Saibishkumar EP, Patel FD, Ghoshal S, et al. Results of salvage radiotherapy after inadequate surgery in invasive cervical carcinoma patients: a retrospective analysis. Int J Radiat Oncol Biol Phys 2005;63:828–833.
379. Park JY, Kim DY, Kim JH, et al. Management of occult invasive cervical cancer found after simple hysterectomy. Ann Oncol 2010;21:994–1000.
380. Smith KB, Amdur RJ, Yeung AR, et al. Postoperative radiotherapy for cervix cancer incidentally discovered after a simple hysterectomy for either benign conditions or noninvasive pathology. Am J Clin Oncol 2010;33:229–232.
381. Ramirez PT, Schmeler KM, Wolf JK, et al. Robotic radical parametrectomy and pelvic lymphadenectomy in patients with invasive cervical cancer. Gynecol Oncol 2008;111:18–21.
382. Kolstad P. Follow-up study of 232 patients with stage Ia1 and 411 patients with stage Ia2 squamous cell carcinoma of the cervix (microinvasive carcinoma). Gynecol Oncol 1989;33:265–272.
383. Raspagliesi F, Ditto A, Quattrone P, et al. Prognostic factors in microinvasive cervical squamous cell cancer: long-term results. Int J Gynecol Cancer 2005;15:88–93.
384. Benson WL, Norris HJ. A critical review of the frequency of lymph node metastasis and death from microinvasive carcinoma of the cervix. Obstet Gynecol 1977;49:632–638.
385. Abu-Rustum NR, Sonoda Y. Fertility-sparing surgery in early-stage cervical cancer: indications and applications. J Natl Compr Canc Netw 2010;8:1435–1438.
386. Gadducci A, Sartori E, Maggino T, et al. The clinical outcome of patients with stage Ia1 and Ia2 squamous cell carcinoma of the uterine cervix: a Cooperation Task Force (CTF) study. Eur J Gynaecol Oncol 2003;24:513–516.
387. Balega J, Michael H, Hurteau J, et al. The risk of nodal metastasis in early adenocarcinoma of the uterine cervix. Int J Gynecol Cancer 2004;14:104–109.
388. Webb JC, Key CR, Qualls CR, et al. Population-based study of microinvasive adenocarcinoma of the uterine cervix. Obstet Gynecol 2001;97:701–706.
389. Zola P, Volpe T, Castelli G, et al. Is the published literature a reliable guide for deciding between alternative treatments for patients with early cervical cancer? Int J Radiat Oncol Biol Phys 1989;16:785–797.
390. Nelson JH Jr, Boyce J, Macasaet M, et al. Incidence, significance, and follow-up of para-aortic lymph node metastases in late invasive carcinoma of the cervix. Am J Obstet Gynecol 1977;128:336–340.
391. Kupets R, Thomas GM, Covens A. Is there a role for pelvic lymph node debulking in advanced cervical cancer? Gynecol Oncol 2002;87:163–170.
392. Whitney CW, Stehman FB. The abandoned radical hysterectomy: a Gynecologic Oncology Group Study. Gynecol Oncol 2000;79:350–635.
393. Akine Y, Arimoto H, Ogino T, et al. High-dose-rate intracavitary irradiation in the treatment of carcinoma of the uterine cervix: Early experience with 84 patients. Int J Radiat Oncol Biol Phys 1988;14:893–898.
394. Hamberger AD, Fletcher GH, Wharton JT. Results of treatment of early stage I carcinoma of the uterine cervix with intracavitary radium alone. Cancer 1978;41:980–985.
395. Volterrani F, Lombardi F. Long term results of radium therapy in cervical cancer. Int J Radiat Oncol Biol Phys 1980;6:565–570.
396. Saibishkumar EP, Patel FD, Sharma SC. Results of radiotherapy alone in the treatment of carcinoma of uterine cervix: a retrospective analysis of 1069 patients. Int J Gynecol Cancer 2005;15:890–897.
397. Eifel PJ, Winter K, Morris M, et al. Pelvic irradiation with concurrent chemotherapy versus pelvic and para-aortic irradiation for high-risk cervical cancer: an update of radiation therapy oncology group trial (RTOG) 90–01. J Clin Oncol 2004;22:872–880.
398. Newton M. Radical hysterectomy or radiotherapy for stage I cervical cancer. A prospective comparison with 5 and 10 years follow-up. Am J Obstet Gynecol 1975;123:535–542.
399. Roddick JW Jr, Greenelaw RH. Treatment of cervical cancer. A randomized study of operation and radiation. Am J Obstet Gynecol 1971;109:754–764.
400. Rosa DD, Medeiros LR, Edelweiss MI, et al. Adjuvant platinum-based chemotherapy for early stage cervical cancer. Cochrane Database Syst Rev 2009:CD005342.
401. Rydzewska L, Tierney J, Vale CL, et al. Neoadjuvant chemotherapy plus surgery versus surgery for cervical cancer. Cochrane Database Syst Rev 2012;doi: 10.1002/14651858.CD007406.
402 Neoadjuvant Chemotherapy for Locally Advanced Cervical Cancer Meta-analysis Collaboration. Neoadjuvant chemotherapy for locally advanced cervical cancer: a systematic review and meta-analysis of individual patient data from 21 randomised trials. Eur J Cancer 2003;39(17):2470–2486.
403. Keilbinska S, Ludwika, Tarlowska, et al. Studies of mortality and health status in women cured of cancer of the cervix uteri. Comparison of long-term results of radiotherapy and combined surgery and radiotherapy. Cancer1973;32:245–252.
404. Piver MS, Marchetti DL, Patton T, et al. Radical hysterectomy and pelvic lymphadenectomy versus radiation therapy for small (less than or equal to 3 cm) stage IB cervical carcinoma. Am J Clin Oncol 1988;11:21–24.
405. Einhorn N, Patek E, Sjöberg B. Outcome of different treatment modalities in cervix carcinoma stage IB and IIA. Cancer 1985;55:949–955.
406. Perez CA, Camel HM, Kao MS, et al. Randomized study of preoperative radiation and surgery or irradiation alone in the treatment of stage IB and IIA carcinoma of the uterine cervix: final report. Gynecol Oncol 1987;27:129–140.
407. Perez CA, Grigsby PW, Camel HM, et al. Irradiation alone or combined with surgery in stage IB, IIA, and IIB carcinoma of uterine cervix: update of a nonrandomized comparison. Int J Radiat Oncol Biol Phys 1995;31:703–716.
408. Jobson VW, Girtanner RE, Averette HE. Therapy and survival of early invasive carcinoma of the cervix uteri with metastases to the pelvic nodes. Surg Gynecol Obstet 1980;151:27–9.
409. Peters WA 3rd, Liu PY, Barrett RJ 2nd, et al. Concurrent chemotherapy and pelvic radiation therapy compared with pelvic radiation therapy alone as adjuvant therapy after radical surgery in high-risk early-stage cancer of the cervix. J Clin Oncol2000;18:1606–1613.
410. Sedlis A, Bundy BN, Rotman MZ, et al. A randomized trial of pelvic radiation therapy versus no further therapy in selected patients with stage IB carcinoma of the cervix after radical hysterectomy and pelvic lymphadenectomy: a Gynecologic Group Study. Gynecol Oncol 1999;73:177–183.
411. Song S, Song C, Kim HJ, et al. 20 year experience of postoperative radiotherapy in IB-IIA cervical cancer patients with intermediate risk factors: impact of treatment period and concurrent chemotherapy. Gynecol Oncol2012;124:63–67.
412. Rotman M, Sedlis A, Piedmonte MR, et al. A phase III randomized trial of postoperative pelvic irradiation in stage IB cervical carcinoma with poor prognostic features: follow-up of a gynecologic oncology group study. Int J Radiat Oncol Biol Phys2006;65:169–176.
413. Rogers L, Siu SS, Luesley D, et al. Adjuvant radiotherapy and chemoradiation after surgery for cervical cancer. Cochrane Database Syst Rev 2009:CD007583.
414. Sundfør K, Trope CG, Kjørstad KE. Radical radiotherapy versus brachytherapy plus surgery in carcinoma of the cervix 2A and 2B—long-term results from a randomized study 1968–1980. Acta Oncol 1996;35(Suppl 8):99–107.
415. Snijders-Keilholz A, Hellebrekers BW, Zwinderman AH, et al. Adjuvant radiotherapy following radical hysterectomy for patients with early-stage cervical carcinoma (1984–1996). Radiother Oncol 1999;51:161–167.
416. Garipagaoglu M, Tulunay G, Kose MF, et al. Prognostic factors in stage IB-IIA cervical carcinomas treated with postoperative radiotherapy. Eur J Gynaecol Oncol 1999;20:131–135.
417. Lahousen M, Haas J, Pickel H, et al. Chemotherapy versus radiotherapy versus observation for high-risk cervical carcinoma after radical hysterectomy: A randomized, prospective, multicenter trial. Gynecol Oncol 1999;73:196–201.
418. González González D, Ketting BW, et al. Carcinoma of the uterine cervix stage IB and IIA: results of postoperative irradiation in patients with microscopic infiltration in the parametrium and/or lymph node metastasis. Int J Radiat Oncol Biol Phys1989;16:389–395.
419. Bianchi UA, Sartori E, Pecorelli S, et al. Treatment of primary invasive cervical cancer. Considerations on 997 consecutive cases. Eur J Gynaecol Oncol 1988;9:47–53.
420. Chatani M, Nose T, Masaki N, et al. Adjuvant radiotherapy after radical hysterectomy of the cervical cancer. Prognostic factors and complications. Strahlenther Onkol 1998;174:504–509.
421. Uno T, Ito H, Itami J, et al. Postoperative radiation therapy for stage IB-IIB carcinoma of the cervix with poor prognostic factors. Anticancer Res 2000;20:2235–2239.
422. Kinney WK, Alvarez RD, Reid GC, et al. Value of adjuvant whole-pelvis irradiation after Wertheim hysterectomy for early-stage squamous carcinoma of the cervix with pelvic nodal metastasis: a matched-control study. Gynecol Oncol 1989;34:258–262.
423. Kinney WK, Hodge DO, Egorshin EV, et al. Surgical treatment of patients with stages IB and IIA carcinoma of the cervix and palpably positive pelvic lymph nodes. Gynecol Oncol 1995;57:145–149.
424. Mitsuhashi N, Takahashi M, Yamakawa M, et al. Results of postoperative radiation therapy for patients with carcinoma of the uterine cervix: evaluation of intravaginal cone boost with an electron beam. Gynecol Oncol1995;57:321–326.
425. Mabuchi S, Okazawa M, Isohashi F, et al. Radical hysterectomy with adjuvant radiotherapy versus definitive radiotherapy alone for FIGO stage IIB cervical cancer. Gynecol Oncol 2011;123:241–247.
426. Mabuchi S, Okazawa M, Isohashi F, et al. Postoperative whole pelvic radiotherapy plus concurrent chemotherapy versus extended-field irradiation for early-stage cervical cancer patients with multiple pelvic lymph node metastases. Gynecol Oncol2011;120:94–100.
427. Lee HN, Lee KH, Lee DW, et al. Weekly cisplatin therapy compared with triweekly combination chemotherapy as concurrent adjuvant chemoradiation therapy after radical hysterectomy for cervical cancer. Int J Gynecol Cancer 2011;21:128–136.
428. Small W Jr, Beriwal S, Demanes DJ, et al. American Brachytherapy Society consensus guidelines for adjuvant vaginal cuff brachytherapy after hysterectomy. Brachytherapy 2012;11:58–67.
429. Hart K, Han I, Deppe G, et al. Postoperative radiation for cervical cancer with pathologic risk factors. Int J Radiat Oncol Biol Phys 1997;37:833–838.
430. Busch M, Rath W, Schaffer M, et al. Results of postoperative radiotherapy of cervix carcinoma after radical versus non radical hysterectomy. Radiol Med 1997;93:110–114.
431. Atkovar G, Uzel O, Ozsahin M, et al. Postoperative radiotherapy in carcinoma of the cervix: treatment results and prognostic factors. Radiother Oncol 1995;35:198–205.
432. Christie DR, Bull CA, Gebski V, et al. Concurrent 5-fluorouracil, mitomycin C and irradiation in locally advanced cervix cancer. Radiother Oncol 1995;37:181–189.
433. Coughlin CT, Richmond RC. Biologic and clinical developments of cisplatin combined with radiation: concepts, utility, projections for new trials, and the emergence of carboplatin. Semin Oncol 1989;16:31–43.
434. Park TK, Choi DH, Kim SN, et al. Role of induction chemotherapy in invasive cervical cancer. Gynecol Oncol 1991;41:107–112.
435. Rotman MZ. Chemoirradiation: a new initiative in cancer treatment. 1991 RSNA annual oration in radiation oncology. Radiology 1992;184:319–327.
436. Douple EB. Platinum-radiation interactions. NCI Monogr 1988:315–319.
437. Durand RE, Aquino-Parsons C. Predicting response to treatment in human cancers of the uterine cervix: sequential biopsies during external beam radiotherapy. Int J Radiat Oncol Biol Phys 2004;58:555–560.
438. Green JA, Kirwan JM, Tierney JF, et al. Survival and recurrence after concomitant chemotherapy and radiotherapy for cancer of the uterine cervix: a systematic review and meta-analysis. Lancet 2001;358:781–786.
439. Deckers PJ, Ketcham AS, Sugerbaker EV, et al. Pelvic exenteration for primary carcinoma of the uterine cervix. Obstet Gynecol 1971;37:647–659.
440. Niibe Y, Hayakawa K, Kanai T, et al. Optimal dose for stage IIIB adenocarcinoma of the uterine cervix on the basis of biological effective dose. Eur J Gynaecol Oncol 2006;27:47–49.
441. Small W Jr, Winter K, Levenback C, et al. Extended-field irradiation and intracavitary brachytherapy combined with cisplatin chemotherapy for cervical cancer with positive para-aortic or high common iliac lymph nodes: results of ARM 1 of RTOG 0116. Int J Radiat Oncol Biol Phys 2007;68:1081–1087.
442. Small W Jr, Winter K, Levenback C, et al. Extended-field irradiation and intracavitary brachytherapy combined with cisplatin and amifostine for cervical cancer with positive para-aortic or high common iliac lymph nodes: results of arm II of Radiation Therapy Oncology Group (RTOG) 0116. Int J Gynecol Cancer 2011;21:1266–1275.
443. Salama JK, Mundt AJ, Roeske J, et al. Preliminary outcome and toxicity report of extended-field, intensity-modulated radiation therapy for gynecologic malignancies. Int J Radiat Oncol Biol Phys 2006;65:1170–1176.
444. Yin M, Zhang H, Li H, et al. The toxicity and long-term efficacy of nedaplatin and paclitaxel treatment as neoadjuvant chemotherapy for locally advanced cervical cancer. J Surg Oncol 2012;105:206–211.
445. Lehman M, Thomas G. Is concurrent chemotherapy and radiotherapy the new standard of care for locally advanced cervical cancer? Int J Gynecol Cancer 2001;11:87–99.
446. Whitney CW, Sause W, Bundy BN, et al. Randomized comparison of fluorouracil plus cisplatin versus hydroxyurea as an adjunct to radiation therapy in stage IIB-IVA carcinoma of the cervix with negative para-aortic lymph nodes: a Gynecologic Oncology Group and Southwest Oncology Group study. J Clin Oncol 1999;17:1339–1348.
447. Rose PG, Bundy BN, Watkins EB, et al. Concurrent cisplatin-based radiotherapy and chemotherapy for locally advanced cervical cancer. N Engl J Med 1999;340:1144–1153.
448. Rose PG, Ali S, Watkins E, et al. Long-term follow-up of a randomized trial comparing concurrent single agent cisplatin, cisplatin-based combination chemotherapy, or hydroxyurea during pelvic irradiation for locally advanced cervical cancer: a Gynecologic Oncology Group Study. J Clin Oncol 2007;25:2804–2810.
449. Morris M, Eifel PJ, Lu J, Grigsby PW, et al Pelvic radiation with concurrent chemotherapy compared with pelvic and para-aortic radiation for high-risk cervical cancer N Engl J Med 1999 ;340(15):1137–1143.
450. Keys HM, Bundy BN, Stehman FB, et al. Cisplatin, radiation, and adjuvant hysterectomy compared with radiation and adjuvant hysterectomy for bulky stage IB cervical carcinoma. N Engl J Med 1999;340:1154–1161.
451. Stehman FB, Ali S, Keys HM, et al. Radiation therapy with or without weekly cisplatin for bulky stage 1B cervical carcinoma: follow-up of a Gynecologic Oncology Group trial. Am J Obstet Gynecol 2007;197:503e1–6.
452. Rose PG, Eifel PJ. Combined radiation therapy and chemotherapy for carcinoma of the cervix. Cancer J 2001;7:86–94.
453. Curtin JP, Hoskins WJ, Venkatraman ES, et al. Adjuvant chemotherapy versus chemotherapy plus pelvic irradiation for high-risk cervical cancer patients after radical hysterectomy and pelvic lymphadenectomy (RH-PLND): a randomized phase III trial. Gynecol Oncol 1996;61:3–10.
454. Pearcey R, Brundage M, Drouin P, et al. Phase III trial comparing radical radiotherapy with and without cisplatin chemotherapy in patients with advanced squamous cell cancer of the cervix. J Clin Oncol 2002;20:966–972.
455. Green J, Kirwan J, Tierney J, et al. Concomitant chemotherapy and radiation therapy for cancer of the uterine cervix. Cochrane Database Syst Rev 2005:CD002225.
456. Reducing uncertainties about the effects of chemoradiotherapy for cervical cancer: a systematic review and meta-analysis of individual patient data from 18 randomized trials. J Clin Oncol 2008;26:5802–5812.
457. Nugent EK, Case AS, Hoff JT, et al. Chemoradiation in locally advanced cervical carcinoma: an analysis of cisplatin dosing and other clinical prognostic factors. Gynecol Oncol 2010;116:438–441.
458. Einstein MH, Novetsky AP, Garg M, et al. Survival and toxicity differences between 5-day and weekly cisplatin in patients with locally advanced cervical cancer. Cancer 2007;109:48–53.
459. Duenas-Gonzalez A, Zarba JJ, Patel F, et al. Phase III, open-label, randomized study comparing concurrent gemcitabine plus cisplatin and radiation followed by adjuvant gemcitabine and cisplatin versus concurrent cisplatin and radiation in patients with stage IIB to IVA carcinoma of the cervix. J Clin Oncol 2011;29:1678–1685.
460. Lanciano R, Calkins A, Bundy BN, et al. Randomized comparison of weekly cisplatin or protracted venous infusion of fluorouracil in combination with pelvic radiation in advanced cervix cancer: a gynecologic oncology group study. J Clin Oncol2005;23:8289–8295.
461. Lorvidhaya V, Tonusin A, Sukthomya W, et al. Induction chemotherapy and irradiation in advanced carcinoma of the cervix. Gan To Kagaku Ryoho 1995;22(Suppl 3):244–251.
462. Kim YS, Shin SS, Nam JH, et al. Prospective randomized comparison of monthly fluorouracil and cisplatin versus weekly cisplatin concurrent with pelvic radiotherapy and high-dose rate brachytherapy for locally advanced cervical cancer. Gynecol Oncol2008;108:195–200.
463. Tseng CJ, Chang CT, Lai CH, et al. A randomized trial of concurrent chemoradiotherapy versus radiotherapy in advanced carcinoma of the uterine cervix. Gynecol Oncol 1997;66:52–58.
464. Geara FB, Shamseddine A, Khalil A, et al. A phase II randomized trial comparing radiotherapy with concurrent weekly cisplatin or weekly paclitaxel in patients with advanced cervical cancer. Radiat Oncol 2010;5:84.
465. Martinez-Monge R, Gaztanaga M, Aramendia JM, et al. A phase II trial of less than 7 weeks of concomitant cisplatin-paclitaxel chemoradiation in locally advanced cervical cancer. Int J Gynecol Cancer 2010;20:133–140.
466. Katanyoo K, Tangjitgamol S, Chongthanakorn M, et al. Treatment outcomes of concurrent weekly carboplatin with radiation therapy in locally advanced cervical cancer patients. Gynecol Oncol 2011;123:571–576.
467. Takekida S, Fujiwara K, Nagao S, et al. Phase II study of combination chemotherapy with docetaxel and carboplatin for locally advanced or recurrent cervical cancer. Int J Gynecol Cancer 2010;20:1563–1568.
468. Hreshchyshyn MM, Aron BS, Boronow RC, et al. Hydroxyurea or placebo combined with radiation to treat stages IIIB and IV cervical cancer confined to the pelvis. Int J Radiat Oncol Biol Phys 1979;5:317–322.
469. Piver MS, Barlow JJ, Vongtama V, et al. Hydroxyurea as a radiation sensitizer in women with carcinoma of the uterine cervix. Am J Obstet Gynecol 1977;129:379–383.
470. Symonds RP, Collingwood M, Kirwan J, et al. Concomitant hydroxyurea plus radiotherapy versus radiotherapy for carcinoma of the uterine cervix: a systematic review. Cancer Treat Rev 2004;30:405–414.
471. Stehman FB, Bundy BN, Thomas G, et al. Hydroxyurea versus misonidazole with radiation in cervical carcinoma: long-term follow-up of a Gynecologic Oncology Group trial. J Clin Oncol 1993;11:1523–1528.
472. Leibel S, Bauer M, Wasserman T, et al. Radiotherapy with or without misonidazole for patients with stage IIIB or stage IVA squamous cell carcinoma of the uterine cervix: preliminary report of a Radiation Therapy Oncology Group randomized trial. Int J Radiat Oncol Biol Phys 1987;13:541–549.
473. Overgaard J, Bentzen SM, Kolstad P, et al. Misonidazole combined with radiotherapy in the treatment of carcinoma of the uterine cervix. Int J Radiat Oncol Biol Phys 1989;16:1069–1072.
474. Grigsby PW, Winter K, Wasserman TH, et al. Irradiation with or without misonidazole for patients with stages IIIB and IVA carcinoma of the cervix: final results of RTOG 80–05. Radiation Therapy Oncology Group. Int J Radiat Oncol Biol Phys1999;44:513–517.
475. Bonomi P, Blessing J, Ball H, et al. A phase II evaluation of cisplatin and 5-fluorouracil in patients with advanced squamous cell carcinoma of the cervix: a Gynecologic Oncology Group study. Gynecol Oncol 1989;37:354–358.
476. Perez CA, Grigsby PW. Adjuvant chemotherapy and irradiation in locally advanced squamous cell carcinoma of the uterine cervix. PPGO Updates 1993;1:1–20.
477. Sardi JE, Giaroli A, Sananes C, et al. Long-term follow-up of the first randomized trial using neoadjuvant chemotherapy in stage Ib squamous carcinoma of the cervix: the final results. Gynecol Oncol 1997;67:61–69.
478. Souhami L, Seymour R, Roman TN, et al. Weekly cisplatin plus external beam radiotherapy and high dose rate brachytherapy in patients with locally advanced carcinoma of the cervix. Int J Radiat Oncol Biol Phys1993;27:871–8.
479. Park TK, Lee SK, Kim SN, et al. Combined chemotherapy and radiation for bulky stages I-II cervical cancer: comparison of concurrent and sequential regimens. Gynecol Oncol 1993;50:196–201.
480. Lee JW, Kim BG, Lee SJ, et al. Preliminary results of consolidation chemotherapy following concurrent chemoradiation after radical surgery in high-risk early-stage carcinoma of the uterine cervix. Clin Oncol (R Coll Radiol)2005;17:412–417.
481. Grigsby PW, Mutch DG, Rader J, et al. Lack of benefit of concurrent chemotherapy in patients with cervical cancer and negative lymph nodes by FDG-PET. Int J Radiat Oncol Biol Phys 2005;61:444–449.
482. Cetina L, Garcia-Arias A, Uribe Mde J, et al. Concurrent chemoradiation with carboplatin for elderly, diabetic and hypertensive patients with locally advanced cervical cancer. Eur J Gynaecol Oncol 2008;29:608–612.
483. Roberts KB, Urdaneta N, Vera R, et al. Interim results of a randomized trial of mitomycin C as an adjunct to radical radiotherapy in the treatment of locally advanced squamous-cell carcinoma of the cervix. Int J Cancer2000;90:206–223.
484. Smith HO, Jiang CS, Weiss GR, et al. Tirapazamine plus cisplatin in advanced or recurrent carcinoma of the uterine cervix: a Southwest Oncology Group study. Int J Gynecol Cancer 2006;16:298–305.
485. Wun T, Law L, Harvey D, et al. Increased incidence of symptomatic venous thrombosis in patients with cervical carcinoma treated with concurrent chemotherapy, radiation, and erythropoietin. Cancer 2003;98:1514–1520.
486. Anders JC, Grigsby PW, Singh AK. Cisplatin chemotherapy (without erythropoietin) and risk of life-threatening thromboembolic events in carcinoma of the uterine cervix: the tip of the iceberg? A review of the literature. Radiat Oncol 2006;1:14.
487. Schefter TE, Winter K, Kwon JS, et al. A phase II study of bevacizumab in combination with definitive radiotherapy and cisplatin chemotherapy in untreated patients with locally advanced cervical carcinoma: preliminary results of RTOG 0417. Int J Radiat Oncol Biol Phys 2012;83:1179–1184.
488. Nogueira-Rodrigues A, do Carmo CC, Viegas C, et al. Phase I trial of erlotinib combined with cisplatin and radiotherapy for patients with locally advanced cervical squamous cell cancer. Clin Cancer Res 2008;14:6324–6329.
489. Morris M, Eifel PJ, Burke TW, et al. Treatment of locally advanced cervical cancer with concurrent radiation and intra-arterial chemotherapy. Gynecol Oncol 1995;57:72–78.
490. Onishi H, Yamaguchi M, Kuriyama K, et al. Effect of concurrent intra-arterial infusion of platinum drugs for patients with stage III or IV uterine cervical cancer treated with radical radiation therapy. Cancer J Sci Am2000;6:40–45.
491. Thomas GM. Is neoadjuvant chemotherapy a useful strategy for the treatment of stage IB cervix cancer? Gynecol Oncol 1993;49:153–155.
492. Benedetti-Panici P, Greggi S, Colombo A, et al. Neoadjuvant chemotherapy and radical surgery versus exclusive radiotherapy in locally advanced squamous cell cervical cancer: results from the Italian multicenter randomized study. J Clin Oncol2002;20:179–188.
493. Chang TC, Lai CH, Hong JH, et al. Randomized trial of neoadjuvant cisplatin, vincristine, bleomycin, and radical hysterectomy versus radiation therapy for bulky stage IB and IIA cervical cancer. J Clin Oncol 2000;18:1740–177.
494. Chauvergne J, Lhomme C, Rohart J, et al. Neoadjuvant chemotherapy of stage IIb or III cancers of the uterine cervix. Long-term results of a multicenter randomized trial of 151 patients [in French]. Bull Cancer 1993;80:1069–1079.
495. Herod J, Burton A, Buxton J, et al. A randomised, prospective, phase III clinical trial of primary bleomycin, ifosfamide and cisplatin (BIP) chemotherapy followed by radiotherapy versus radiotherapy alone in inoperable cancer of the cervix. Ann Oncol2000;11:1175–1181.
496. Kumar L, Grover R, Pokharel YH, et al. Neoadjuvant chemotherapy in locally advanced cervical cancer: two randomised studies. Aust N Z J Med 1998;28:387–390.
497. Leborgne F, Leborgne JH, Doldan R, et al. Induction chemotherapy and radiotherapy of advanced cancer of the cervix: a pilot study and phase III randomized trial. Int J Radiat Oncol Biol Phys 1997;37:343–350.
498. Sundfor K, Trope CG, Hogberg T, et al. Radiotherapy and neoadjuvant chemotherapy for cervical carcinoma. A randomized multicenter study of sequential cisplatin and 5-fluorouracil and radiotherapy in advanced cervical carcinoma stage 3B and 4A. Cancer 1996;77:2371–2378.
499. Symonds RP, Habeshaw T, Reed NS, et al. The Scottish and Manchester randomised trial of neo-adjuvant chemotherapy for advanced cervical cancer. Eur J Cancer 2000;36:994–1001.
500. Colombo A, Landoni F, Maneo A, et al. Neoadjuvant chemotherapy to radiation and concurrent chemoradiation for locally advanced squamous cell carcinoma of the cervix: a review of the recent literature. Tumori 1998;84:229–237.
501. Souhami L, Gil RA, Allan SE, et al. A randomized trial of chemotherapy followed by pelvic radiation therapy in stage IIIB carcinoma of the cervix. J Clin Oncol 1991;9:970–977.
502. Kumar L, Kaushal R, Nandy M, et al. Chemotherapy followed by radiotherapy versus radiotherapy alone in locally advanced cervical cancer: a randomized study. Gynecol Oncol 1994;54:307–315.
503. Long HJ 3rd, Bundy BN, Grendys EC Jr, et al. Randomized phase III trial of cisplatin with or without topotecan in carcinoma of the uterine cervix: a Gynecologic Oncology Group Study. J Clin Oncol 2005;23:4626–4633.
504. Rose PG, Blessing JA, Gershenson DM, et al. Paclitaxel and cisplatin as first-line therapy in recurrent or advanced squamous cell carcinoma of the cervix: a gynecologic oncology group study. J Clin Oncol 1999;17:2676–80.
505 Gebbia V, Caruso M, Testa A, et al. Vinorelbine and cisplatin for the treatment of recurrent and/or metastatic carcinoma of the uterine cervix. Oncology 2002;63(1):31–37.
506. Lacava JA, Leone BA, Machiavelli M, et al. Vinorelbine as neoadjuvant chemotherapy in advanced cervical carcinoma. J Clin Oncol 1997;15:604–609.
507. Morris M, Brader KR, Levenback C, et al. Phase II study of vinorelbine in advanced and recurrent squamous cell carcinoma of the cervix. J Clin Oncol 1998;16:1094–1098.
508. Umesaki N, Fujii T, Nishimura R, et al. Phase II study of irinotecan combined with mitomycin-C for advanced or recurrent squamous cell carcinoma of the uterine cervix: the JGOG study. Gynecol Oncol 2004;95:127–132.
509. Verschraegen CF, Levy T, Kudelka AP, et al. Phase II study of irinotecan in prior chemotherapy-treated squamous cell carcinoma of the cervix. J Clin Oncol 1997;15:625–631.
510. Schilder RJ, Blessing J, Cohn DE. Evaluation of gemcitabine in previously treated patients with non-squamous cell carcinoma of the cervix: a phase II study of the Gynecologic Oncology Group. Gynecol Oncol 2005;96:103–107.
511. Brewer CA, Blessing JA, Nagourney RA, et al. Cisplatin plus gemcitabine in previously treated squamous cell carcinoma of the cervix: a phase II study of the Gynecologic Oncology Group. Gynecol Oncol 2006;100:385–388.
512. Monk BJ, Sill MW, McMeekin DS, et al. Phase III trial of four cisplatin-containing doublet combinations in stage IVB, recurrent, or persistent cervical carcinoma: a Gynecologic Oncology Group study. J Clin Oncol2009;27:4649–4655.
513. Farley J, Sill MW, Birrer M, et al. Phase II study of cisplatin plus cetuximab in advanced, recurrent, and previously treated cancers of the cervix and evaluation of epidermal growth factor receptor immunohistochemical expression: a Gynecologic Oncology Group study. Gynecol Oncol 2011;121:303–308.
514. Downs LS Jr, Chura JC, Argenta PA, et al. Ifosfamide, paclitaxel, and carboplatin, a novel triplet regimen for advanced, recurrent, or persistent carcinoma of the cervix: a phase II trial. Gynecol Oncol 2011;120:265–269.
515. Monk BJ, Mas Lopez L, Zarba JJ, et al. Phase II, open-label study of pazopanib or lapatinib monotherapy compared with pazopanib plus lapatinib combination therapy in patients with advanced and recurrent cervical cancer. J Clin Oncol 2010;28:3562–3569.
516. Mendenhall WM, McCarty PJ, Morgan LS, et al. Stage IB or IIA-B carcinoma of the intact uterine cervix greater than or equal to 6 cm in diameter: is adjuvant extrafascial hysterectomy beneficial? Int J Radiat Oncol Biol Phys1991;21:899–904.
517. Thoms WW, Eifel PJ, Smith TL, et al. Bulky endocervical carcinomas: a 23-year experience. Int J Radiat Oncol Biol Phys 1992;23:491–499.
518. Kim HK, Silver B, Berkowitz R, et al. Bulky, barrel-shaped cervical carcinoma (stages IB, IIA, IIB): the prognostic factors for pelvic control and treatment outcome. Am J Clin Oncol 1999;22:232–236.
519. Paley PJ, Goff BA, Minudri R, et al. The prognostic significance of radiation dose and residual tumor in the treatment of barrel-shaped endophytic cervical carcinoma. Gynecol Oncol 2000;76:373–379.
520. Keys H, Bundy B, Stehman FB, et al. Adjuvant hysterectomy after radiation therapy reduces detection of local recurrence in “bulky” stage IB cervical cancer without improving survival: results of a prospective randomized GOG trial. Cancer J Sci Am1997;3:117.
521. Coia L, Won M, Lanciano R, et al. The Patterns of Care Outcome Study for cancer of the uterine cervix. Results of the second national practice survey. Cancer 1990;66:2451–2456.
522. Russell AH, Koh WJ, Markette K, et al. Radical reirradiation for recurrent or second primary carcinoma of the female reproductive tract. Gynecol Oncol 1987;27:226–232.
523. Kim RY, Trotti A, Wu CJ, et al. Radiation alone in the treatment of cancer of the uterine cervix: analysis of pelvic failure and dose response relationship. Int J Radiat Oncol Biol Phys 1989;17:973–978.
524. Montana GS, Fowler WC, Varia MA, et al. Carcinoma of the cervix, stage III. Results of radiation therapy. Cancer 1986;57:148–154.
525. Hanks GE, Herring DF, Kramer S. Patterns of care outcome studies. Results of the National Practice in Cancer of the Cervix. Cancer 1983;51:959–967.
526. Komaki R, Brickner TJ, Hanlon AL, et al. Long-term results of treatment of cervical carcinoma in the United States in 1973, 1978, and 1983: Patterns of Care Study (PCS). Int J Radiat Oncol Biol Phys 1995;31:973–982.
527. Montana GS, Hanlon AL, Brickner TJ, et al. Carcinoma of the cervix: patterns of care studies: review of 1978, 1983, and 1988–1989 surveys. Int J Radiat Oncol Biol Phys 1995;32:1481–1486.
528. Arthur D, Kaufam N, Schmidt-Ullrich R, et al. Heuristically derived tumor burden score as a prognostic factor for stage IIIB carcinoma of the cervix. Int J Radiat Oncol Biol Phys 1995;34:743–751.
529. Horiot JC, Pigneux J, Pourquier H, et al. Radiotherapy alone in carcinoma of the intact uterine cervix according to G. H. Fletcher guidelines: A French cooperative study of 1383 cases. Int J Radiat Oncol Biol Phys1988;14:605–611.
530. Barillot I, Horiot JC, Maingon P, et al. Impact on treatment outcome and late effects of customized treatment planning in cervix carcinomas: baseline results to compare new strategies. Int J Radiat Oncol Biol Phys 2000;48:189–200.
531. Marcial VA, Amato DA, Marks RD, et al. Split-course versus continuous pelvis irradiation in carcinoma of the uterine cervix: a prospective randomized clinical trial of the Radiation Therapy Oncology Group. Int J Radiat Oncol Biol Phys 1983;9:431–436.
532. Crozier M, Morris M, Levenback C, z. Pelvic exenteration for adenocarcinoma of the uterine cervix. Gynecol Oncol 1995;58:74–78.
533. Fletcher GH. Textbook of radiotherapy, 3rd ed. Philadelphia, PA: Lea & Febiger, 1980.
534. Charra-Brunaud C, Harter V, Delannes M, et al. Impact of 3D image-based PDR brachytherapy on outcome of patients treated for cervix carcinoma in France: results of the national STIC prospective study. Radiother Oncol2012;103:305–313.
535. Dimopoulos JC, Potter R, Lang S, et al. Dose-effect relationship for local control of cervical cancer by magnetic resonance image-guided brachytherapy. Radiother Oncol 2009;93:311–315.
536. Kunos C, Ali S, Abdul-Karim FW, et al. Posttherapy residual disease associates with long-term survival after chemoradiation for bulky stage 1B cervical carcinoma: a Gynecologic Oncology Group study. Am J Obstet Gynecol2010;203:351e1–8.
537. Morice P, Rouanet P, Rey A, et al. Results of the GYNECO 02 Study, an FNCLCC Phase III trial comparing hysterectomy with no hysterectomy in patients with a (clinical and radiological) complete response after chemoradiation therapy for stage IB2 or II cervical cancer. Oncologist 2012;17:64–71.
538. Motton S, Houvenaeghel G, Delannes M, et al. Results of surgery after concurrent chemoradiotherapy in advanced cervical cancer: comparison of extended hysterectomy and extrafascial hysterectomy. Int J Gynecol Cancer2010;20:268–275.
539. Leguevaque P, Motton S, Delannes M, et al. Completion surgery or not after concurrent chemoradiotherapy for locally advanced cervical cancer? Eur J Obstet Gynecol Reprod Biol 2011;155:188–192.
540. Touboul C, Uzan C, Mauguen A, et al. Prognostic factors and morbidities after completion surgery in patients undergoing initial chemoradiation therapy for locally advanced cervical cancer. Oncologist 2010;15:405–415.
541. Haie C, Pejovic MH, Gerbaulet A, et al. Is prophylactic para-aortic irradiation worthwhile in the treatment of advanced cervical carcinoma? Results of a controlled clinical trial of the EORTC radiotherapy group. Radiother Oncol 1988;11:101–112.
542. Sood BM, Gorla GR, Garg M, et al. Extended-field radiotherapy and high-dose-rate brachytherapy in carcinoma of the uterine cervix: clinical experience with and without concomitant chemotherapy. Cancer 2003;97:1781–1788.
543. Varia MA, Bundy BN, Deppe G, et al. Cervical carcinoma metastatic to para-aortic nodes: extended field radiation therapy with concomitant 5-fluorouracil and cisplatin chemotherapy: a Gynecologic Oncology Group study. Int J Radiat Oncol Biol Phys1998;42:1015–1023.
544. Lovecchio JL, Averette HE, Donato D, et al. 5-year survival of patients with periaortic nodal metastases in clinical stage IB and IIA cervical carcinoma. Gynecol Oncol 1989;34:43–45.
545. Stryker JA, Mortel R. Survival following extended field irradiation in carcinoma of cervix metastatic to para-aortic lymph nodes. Gynecol Oncol 2000;79:399–405.
546. Grigsby PW, Perez CA, Chao KS, et al. Radiation therapy for carcinoma of the cervix with biopsy-proven positive para-aortic lymph nodes. Int J Radiat Oncol Biol Phys 2001;49:733–738.
547. Hacker NF, Wain GV, Nicklin JL. Resection of bulky positive lymph nodes in patients with cervical carcinoma. Int J Gynecol Cancer 1995;5:250–256.
548. Grigsby PW, Lu JD, Mutch DG, et al. Twice-daily fractionation of external irradiation with brachytherapy and chemotherapy in carcinoma of the cervix with positive para-aortic lymph nodes: Phase II study of the Radiation Therapy Oncology Group 92–10. Int J Radiat Oncol Biol Phys 1998;41:817–822.
549. Malfetano JH, Keys H, Cunningham MJ, et al. Extended field radiation and cisplatin for stage IIB and IIIB cervical carcinoma. Gynecol Oncol 1997;67:203–207.
550. Chou HH, Wang CC, Lai CH, et al. Isolated paraaortic lymph node recurrence after definitive irradiation for cervical carcinoma. Int J Radiat Oncol Biol Phys 2001;51:442–448.
551. Goodman HM, Niloff JM, Nelson JR, et al. Cervical malignancies. In: Knapp RC, Berkowitz RS, eds. Gynecologic oncology. New York: Macmillan, 1986:225–273.
552. Kovalic JJ, Grigsby PW, Perez CA, z. Cervical stump carcinoma. Int J Radiat Oncol Biol Phys 1991;20:933–938.
553. Wolff JP, Lacour J, Chassagne D, et al. Cancer of the cervical stump. Study of 173 patients. Obstet Gynecol 1972;39:10–16.
554. Miller BE, Copeland LJ, Hamberger AD, et al. Carcinoma of the cervical stump. Gynecol Oncol 1984;18:100–108.
555. Hannoun-Levi JM, Peiffert D, Hoffstetter S, et al. Carcinoma of the cervical stump: retrospective analysis of 77 cases. Radiother Oncol 1997;43:147–153.
556. Hellstrom AC, Sigurjonson T, Pettersson F. Carcinoma of the cervical stump. The radiumhemmet series 1959–1987. Treatment and prognosis. Acta Obstet Gynecol Scand 2001;80:152–157.
557. Miller B, Dockter M, el Torky M, et al. Small cell carcinoma of the cervix: a clinical and flow-cytometric study. Gynecol Oncol 1991;42:27–33.
558. Sevin BU, Method MW, Nadji M, et al. Efficacy of radical hysterectomy as treatment for patients with small cell carcinoma of the cervix. Cancer 1996;77:1489–1493.
559. Morris M, Gershenson DM, Eifel P, et al. Treatment of small cell carcinoma of the cervix with cisplatin, doxorubicin, and etoposide. Gynecol Oncol 1992;47:62–65.
560. Hoskins PJ, Wong F, Swenerton KD, et al. Small cell carcinoma of the cervix treated with concurrent radiotherapy, cisplatin, and etoposide. Gynecol Oncol 1995;56:218–225.
561. Delaloge S, Pautier P, Kerbrat P, et al. Neuroendocrine small cell carcinoma of the uterine cervix: what disease? What treatment? Report of ten cases and a review of the literature. Clin Oncol (R Coll Radiol) 2000;12:357–362.
562. Boruta DM 2nd, Schorge JO, Duska LA, et al. Multimodality therapy in early-stage neuroendocrine carcinoma of the uterine cervix. Gynecol Oncol 2001;81:82–87.
563. Vinh-Hung V, Bourgain C, Vlastos G, et al. Prognostic value of histopathology and trends in cervical cancer: a SEER population study. BMC Cancer 2007;7:164.
564. Grigsby PW, Perez CA, Kuske RR, et al. Adenocarcinoma of the uterine cervix: lack of evidence for a poor prognosis. Radiother Oncol 1988;12:289–296.
565. Kilgore LC, Soong SJ, Gore H, z. Analysis of prognostic features in adenocarcinoma of the cervix. Gynecol Oncol 1988;31:137–153.
566. Eifel PJ, Morris M, Oswald MJ, et al. Adenocarcinoma of the uterine cervix. Prognosis and patterns of failure in 367 cases. Cancer 1990;65:2507–2514.
567. Rutledge FN, Galakatos AE, Wharton JT, et al. Adenocarcinoma of the uterine cervix. Am J Obstet Gynecol 1975;122:236–245.
568. Eifel PJ, Burke TW, Morris M, et al. Adenocarcinoma as an independent risk factor for disease recurrence in patients with stage IB cervical carcinoma. Gynecol Oncol 1995;59:38–44.
569. Nakano T, Arai T, Morita S, Oka K. Radiation therapy alone for adenocarcinoma of the uterine cervix. Int J Radiat Oncol Biol Phys 1995;32:1331–1336.
570. Kjorstad KE, Martimbeau PW, Iversen T. Stage IB carcinoma of the cervix, the Norwegian Radium Hospital: results and complications. III. Urinary and gastrointestinal complications. Gynecol Oncol 1983;15:42–47.
571. Baalbergen A, Veenstra Y, Stalpers LL, Ansink AC. Primary surgery versus primary radiation therapy with or without chemotherapy for early adenocarcinoma of the uterine cervix. Cochrane Database Syst Rev2010:CD006248.
572. Huang YT, Wang CC, Tsai CS, et al. Clinical behaviors and outcomes for adenocarcinoma or adenosquamous carcinoma of cervix treated by radical hysterectomy and adjuvant radiotherapy or chemoradiotherapy. Int J Radiat Oncol Biol Phys 2012;84:420–427.
573. Oguchi M, Ikeda H, Watanabe T, et al. Experiences of 23 patients > or = 90 years of age treated with radiation therapy. Int J Radiat Oncol Biol Phys 1998;41:407–413.
574. Duggan B, Muderspach LI, Roman L, et al. Cervical cancer in pregnancy: reporting on planned delay in therapy. Obstet Gynecol 1993;82:598–602.
575. Norstrom A, Jansson I, Andersson H. Carcinoma of the uterine cervix in pregnancy. A study of the incidence and treatment in the western region of Sweden 1973 to 1992. Acta Obstet Gynecol Scand 1997;76:583–589.
576. Hopkins MP, Morley GW. The prognosis and management of cervical cancer associated with pregnancy. Obstet Gynecol 1992;80:9–13.
577. Monk BJ, Montz FJ. Invasive cervical cancer complicating intrauterine pregnancy: treatment with radical hysterectomy. Obstet Gynecol 1992;80:199–203.
578. Sood AK, Sorosky JI, Krogman S, z. Surgical management of cervical cancer complicating pregnancy: a case-control study. Gynecol Oncol 1996;63:294–298.
579. Ostrom K, Ben-Arie A, Edwards C, Gregg A, et al. Uterine evacuation with misoprostol during radiotherapy for cervical cancer in pregnancy. Int J Gynecol Cancer 2003;13:340–343.
580. Tewari K, Cappuccini F, Gambino A, et al. Neoadjuvant chemotherapy in the treatment of locally advanced cervical carcinoma in pregnancy: a report of two cases and review of issues specific to the management of cervical carcinoma in pregnancy including planned delay of therapy. Cancer 1998;82:1529–1534.
581. Sorosky JI, Squatrito R, Ndubisi BU, et al. Stage I squamous cell cervical carcinoma in pregnancy: planned delay in therapy awaiting fetal maturity. Gynecol Oncol 1995;59:207–210.
582. Carillon MA, Emmanuelli V, Castelain B, et al. Management of pregnant women with advanced cervical cancer: About five cases observed in Lille from 2002 till 2009. Evaluation of practices referring to the new French recommendations of 2008 [in French]. J Gynecol Obstet Biol Reprod (Paris) 2011;40:514–521.
583. Li J, Wang LJ, Zhang BZ, et al. Neoadjuvant chemotherapy with paclitaxel plus platinum for invasive cervical cancer in pregnancy: two case report and literature review. Arch Gynecol Obstet 2011;284:779–783.
584. Greer BE, Easterling TR, McLennan DA, et al. Fetal and maternal considerations in the management of stage I-B cervical cancer during pregnancy. Gynecol Oncol 1989;34:61–65.
585. Creasman WT, Rutledge FN, Fletcher GH. Carcinoma of the cervix associated with pregnancy. Obstet Gynecol 1970;36:495–501.
586. Wo JY, Viswanathan AN. Impact of radiotherapy on fertility, pregnancy, and neonatal outcomes in female cancer patients. Int J Radiat Oncol Biol Phys 2009;73:1304–1312.
587. Sood AK, Sorosky JI, Mayr N, et al. Radiotherapeutic management of cervical carcinoma that complicates pregnancy. Cancer 1997;80:1073–1078.
588. Sood AK, Sorosky JI, Mayr N, et al. Cervical cancer diagnosed shortly after pregnancy: prognostic variables and delivery routes. Obstet Gynecol 2000;95:832–838.
589. Jones WB, Shingleton HM, Russell A, et al. Cervical carcinoma and pregnancy. A national patterns of care study of the American College of Surgeons. Cancer 1996;77:1479–1488.
590. Senekjian EK, Hubby M, Bell DA, et al. Clear cell adenocarcinoma (CCA) of the vagina and cervix in association with pregnancy. Gynecol Oncol 1986;24:207–219.
591. Strauss A. Irradiation of carcinoma of the cervix uteri in pregnancy. Am J Roentgenol Radium Ther Nucl Med 1940;43:552–566.
592. Mould RF. Invited review: the early years of radiotherapy with emphasis on x-ray and radium apparatus. Br J Radiol 1995;68:567–582.
593. Fletcher GH, Shukovsky LJ. The interplay of radiocurability and tolerance in the irradiation of human cancers. J Radiol Electrol Med Nucl 1975;56:383–400.
594. Montana GS, Fowler WC, Varia MA, Walton LA, Mack Y. Analysis of results of radiation therapy for Stage II carcinoma of the cervix. Cancer 1985;55:956–962.
595. Lanciano RM, Martz K, Coia LR, et al. Tumor and treatment factors improving outcome in stage III-B cervix cancer. Int J Radiat Oncol Biol Phys 1991;20:95–100.
596. Lanciano RM, Won M, Coia LR, et al. Pretreatment and treatment factors associated with improved outcome in squamous cell carcinoma of the uterine cervix: a final report of the 1973 and 1978 patterns of care studies. Int J Radiat Oncol Biol Phys1991;20:667–676.
597. Montana GS, Martz KL, Hanks GE. Patterns and sites of failure in cervix cancer treated in the U.S.A. in 1978. Int J Radiat Oncol Biol Phys 1991;20:87–93.
598. Viswanathan AN, Thomadsen B. American Brachytherapy Society consensus guidelines for locally advanced carcinoma of the cervix. Part I: General principles. Brachytherapy 2012;11:33–46.
599. International Commission on Radiation Units and Measurements. Dose and volume specification for reporting intracavitary therapy in gynecology. Bethesda, MD: Author, 1985.
600. Erickson B, Eifel P, Moughan J, et al. Patterns of brachytherapy practice for patients with carcinoma of the cervix (1996–1999): a patterns of care study. Int J Radiat Oncol Biol Phys 2005;63:1083–1092.
601. Viswanathan AN, Erickson BA. Three-dimensional imaging in gynecologic brachytherapy: a survey of the American Brachytherapy Society. Int J Radiat Oncol Biol Phys 2010;76:104–109.
602. Viswanathan AN, Creutzberg CL, Craighead P, et al. International Brachytherapy Practice Patterns: a survey of the Gynecologic Cancer Intergroup (GCIG). Int J Radiat Oncol Biol Phys 2012;82:250–255.
603. Tomita N, Toita T, Kodaira T, et al. Patterns of radiotherapy practice for patients with cervical cancer in Japan, 2003–2005: changing trends in the pattern of care process. Int J Radiat Oncol Biol Phys 2012;83:1506–1513.
604. Pavamani S, D’Souza DP, Portelance L, et al. Image-guided brachytherapy for cervical cancer: a Canadian Brachytherapy Group survey. Brachytherapy 2011;10:345–351.
605. Stewart AJ, Viswanathan AN. Current controversies in high-dose-rate versus low-dose-rate brachytherapy for cervical cancer. Cancer 2006;107:908–915.
606. Petereit DG, Sarkaria JN, Potter DM, et al. High-dose-rate versus low-dose-rate brachytherapy in the treatment of cervical cancer: analysis of tumor recurrence—the University of Wisconsin experience. Int J Radiat Oncol Biol Phys 1999;45:1267–1274.
607. Patel FD, Sharma SC, Negi PS, et al. Low dose rate vs. high dose rate brachytherapy in the treatment of carcinoma of the uterine cervix: a clinical trial. Int J Radiat Oncol Biol Phys 1994;28:335–341.
608. Fu KK, Phillips TL. High-dose-rate versus low-dose-rate intracavitary brachytherapy for carcinoma of the cervix. Int J Radiat Oncol Biol Phys 1990;19:791–796.
609. Hall E, Brenner D. The dose-rate effect revisited: radiobiological considerations of importance in radiotherapy. Int J Radiat Oncol Biol Phys 1991;21:1403–1414.
610. Orton CG, Seyedsadr M, Somnay A. Comparison of high and low dose rate remote afterloading for cervix cancer and the importance of fractionation. Int J Radiat Oncol Biol Phys 1991;21:1425–1434.
611. Dale RG. The application of the linear quadratic dose-effect equation to fractionated and protracted radiotherapy. Br J Radiol 1985;58:515–528.
612. Joiner MC, van der Kogel AJ. The linear quadratic approach to fractionation and calculation of isoeffect relationships. In: Steel GG, ed. Basic clinical radiobiology, 3th ed. London: Arnold, 1997:106–122.
613. Lang S, Nulens A, Briot E, et al. Intercomparison of treatment concepts for MR image assisted brachytherapy of cervical carcinoma based on GYN GEC-ESTRO recommendations. Radiother Oncol 2006;78:185–193.
614. Bentzen SM JM. The linear quadratic approach in clinical practice. In: Joiner MC, Van der Kogel A, eds. Basic clinical radiobiology, 4th ed. London: Hodder Arnold, 2009:120–134.
615. Potter R, Haie-Meder C, Van Limbergen E, et al. Recommendations from gynaecological (GYN) GEC ESTRO working group (II): Concepts and terms in 3D image-based treatment planning in cervix cancer brachytherapy—3D dose volume parameters and aspects of 3D image-based anatomy, radiation physics, radiobiology. Radiother Oncol 2006;78:67–77.
616. Sturdza A, Potter R. Outcomes related to the disease and the use of 3D-based external beam radiation and image-guided brachytherapy. In: Viswanathan AN, Kirisits C, Erickson BE, et al., eds. Gynecologic radiation therapy: novel approaches to image-guidance and management. Berlin: Springer, 2011:263–282.
617. Noda SE, Ohno T, Kato S, et al. Late rectal complications evaluated by computed tomography-based dose calculations in patients with cervical carcinoma undergoing high-dose-rate brachytherapy. Int J Radiat Oncol Biol Phys2007;69:118–124.
618. Clark BG, Souhami L, Roman TN, et al. The prediction of late rectal complications in patients treated with high dose rate brachytherapy for carcinoma of the cervix. Int J Radiat Oncol Biol Phys 1997;38:989–993.
619. Petereit DG, Pearcey R. Literature analysis of high dose rate brachytherapy fractionation schedules in the treatment of cervical cancer: is there an optimal fractionation schedule? Int J Radiat Oncol Biol Phys 1999;43:359–366.
620. Lang S, Kirisits C, Dimopoulos J, et al. Treatment planning for MRI assisted brachytherapy of gynecologic malignancies based on total dose constraints. Int J Radiat Oncol Biol Phys 2007;69:619–627.
621. Newman G. Increased morbidity following the introduction of remote afterloading, with increased dose rate, for cancer of the cervix. Radiother Oncol 1996;39:97–103.
622. Fowler J, Martinez A, Orton C, et al., eds. The radiobiology of radiotherapy. In: Martinez AA, Orton CG, Mould RF, eds. Brachytherapy HDR and LDR. Columbia, MD: Nucletron, 1990:121–137.
623. Viswanathan AN, Beriwal S, De Los Santos JF, et al. American Brachytherapy Society consensus guidelines for locally advanced carcinoma of the cervix. Part II: High-dose-rate brachytherapy. Brachytherapy 2012;11:47–52.
624. Lee LJ, Das IJ, Higgins SA, et al. American Brachytherapy Society consensus guidelines for locally advanced carcinoma of the cervix. Part III: Low-dose-rate and pulsed-dose-rate brachytherapy. Brachytherapy 2012;11:53–57.
625. Haie-Meder C, Potter R, Van Limbergen E, et al. Recommendations from Gynaecological (GYN) GEC-ESTRO Working Group (I): concepts and terms in 3D image based 3D treatment planning in cervix cancer brachytherapy with emphasis on MRI assessment of GTV and CTV. Radiother Oncol 2005;74:235–245.
626. Potter R, Dimopoulos J, Kirisits C, et al. Recommendations for image-based intracavitary brachytherapy of cervix cancer: the GYN GEC ESTRO Working Group point of view: in regard to Nag et al. (Int J Radiat Oncol Biol Phys 2004;60:1160–1172). Int J Radiat Oncol Biol Phys 2005;62:293–295; author reply 5–6.
627. Arai T, Morita S, Iinuma T, et al. Radiation treatment of cervix cancer using the high dose rate remote afterloading intracavitary irradiation: an analysis of the correlation between optimal dose range and fractionation. Jpn J Cancer Clin 1979;25:605–612.
628. Watkins JM, Kearney PL, Opfermann KJ, et al. Ultrasound-guided tandem placement for low-dose-rate brachytherapy in advanced cervical cancer minimizes risk of intraoperative uterine perforation. Ultrasound Obstet Gynecol2011;37:241–244.
629. Van Dyk S, Narayan K, Fisher R, et al. Conformal brachytherapy planning for cervical cancer using transabdominal ultrasound. Int J Radiat Oncol Biol Phys 2009;75:64–70.
630. Viswanathan AN, Cormack R, Holloway CL, et al. Magnetic resonance-guided interstitial therapy for vaginal recurrence of endometrial cancer. Int J Radiat Oncol Biol Phys 2006;66:91–99.
631. Potish RA. The effect of applicator geometry on dose specification in cervical cancer. Int J Radiat Oncol Biol Phys 1990;18:1513–1520.
632. Corn BW, Hanlon AL, Pajak TF, et al. Technically accurate intracavitary insertions improve pelvic control and survival among patients with locally advanced carcinoma of the uterine cervix. Gynecol Oncol 1994;53:294–300.
633. Potish RA, Deibel FC Jr, Khan FM. The relationship between milligram-hours and dose to point A in carcinoma of the cervix. Radiology 1982;145:479–483.
634. Katz A, Eifel PJ. Quantification of intracavitary brachytherapy parameters and correlation with outcome in patients with carcinoma of the cervix. Int J Radiat Oncol Biol Phys 2000;48:1417–1425.
635. Viswanathan AN, Dimopoulos J, Kirisits C, et al. Computed tomography versus magnetic resonance imaging-based contouring in cervical cancer brachytherapy: results of a prospective trial and preliminary guidelines for standardized contours. Int J Radiat Oncol Biol Phys 2007;68:491–498.
636. Eskander RN, Scanderbeg D, Saenz CC, et al. Comparison of computed tomography and magnetic resonance imaging in cervical cancer brachytherapy target and normal tissue contouring. Int J Gynecol Cancer 2010;20:47–53.
637. Eisbruch A, Johnston CM, Martel MK, et al. Customized gynecologic interstitial implants: CT-based planning, dose evaluation, and optimization aided by laparotomy. Int J Radiat Oncol Biol Phys 1998;40:1087–1093.
638. Erickson B, Albano K, Gillin M. CT-guided interstitial implantation of gynecologic malignancies. Int J Radiat Oncol Biol Phys 1996;36:699–709.
639. Holloway C, Racine M, O’Farrell D, et al. Sigmoid dose using 3D imaging in cervical cancer brachytherapy. Radiother Oncol 2009;93:307–310.
640. Kapur T, Egger J, Damato A, et al. 3-T MR-guided brachytherapy for gynecologic malignancies. Magn Reson Imaging 2012;30(9):1279–1290.
641. Viswanathan AN, Kirisits C, Erickson B, et al. Gynecologic radiation therapy: novel approaches to image-guidance and management. Berlin: Springer, 2011.
642. Kim RY, Pareek P. Radiography-based treatment planning compared with computed tomography (CT)-based treatment planning for intracavitary brachytherapy in cancer of the cervix: analysis of dose-volume histograms. Brachytherapy 2003;2:200–206.
643. Kim H, Beriwal S, Houser C, et al. Dosimetric analysis of 3D image-guided HDR brachytherapy planning for the treatment of cervical cancer: is point A-based dose prescription still valid in image-guided brachytherapy? Med Dosim 2011;36:166–170.
644. Datta NR, Srivastava A, Maria Das KJ, et al. Comparative assessment of doses to tumor, rectum, and bladder as evaluated by orthogonal radiographs vs. computer enhanced computed tomography-based intracavitary brachytherapy in cervical cancer. Brachytherapy 2006;5:223–229.
645. Lindegaard JC, Tanderup K, Nielsen SK, et al. MRI-guided 3D optimization significantly improves DVH parameters of pulsed-dose-rate brachytherapy in locally advanced cervical cancer. Int J Radiat Oncol Biol Phys2008;71:756–764.
646. Arai T, Nakano T, Morita S, et al. High-dose-rate remote afterloading intracavitary radiation therapy for cancer of the uterine cervix. A 20-year experience. Cancer 1992;69:175–180.
647. Petric P, Dimopoulos J, Kirisits C, et al. Inter- and intraobserver variation in HR-CTV contouring: intercomparison of transverse and paratransverse image orientation in 3D-MRI assisted cervix cancer brachytherapy. Radiother Oncol 2008;89:164–171.
648. Potter R, Haie-Meder C, Van Limbergen E, et al. Recommendations from gynaecological (GYN) GEC ESTRO working group (II): concepts and terms in 3D image-based treatment planning in cervix cancer brachytherapy–3D dose volume parameters and aspects of 3D image-based anatomy, radiation physics, radiobiology. Radiother Oncol 2006;78:67–77.
649. Lang S, Nulens A, Briot E, et al. Intercomparison of treatment concepts for MR image assisted brachytherapy of cervical carcinoma based on GYN GEC-ESTRO recommendations. Radiother Oncol 2006;78:185–193.
650. Kirisits C, Potter R, Lang S, et al. Dose and volume parameters for MRI-based treatment planning in intracavitary brachytherapy for cervical cancer. Int J Radiat Oncol Biol Phys 2005;62:901–911.
651. Chargari C, Magne N, Dumas I, et al. Physics contributions and clinical outcome with 3D-MRI-based pulsed-dose-rate intracavitary brachytherapy in cervical cancer patients. Int J Radiat Oncol Biol Phys 2009;74:133–139.
652. Beriwal S, Kannan N, Kim H, et al. Three-dimensional high dose rate intracavitary image-guided brachytherapy for the treatment of cervical cancer using a hybrid magnetic resonance imaging/computed tomography approach: feasibility and early results. Clin Oncol (R Coll Radiol) 2011;23:685–690.
653. Potter R, Georg P, Dimopoulos JC, et al. Clinical outcome of protocol based image (MRI) guided adaptive brachytherapy combined with 3D conformal radiotherapy with or without chemotherapy in patients with locally advanced cervical cancer. Radiother Oncol 2011;100:116–123.
654. De Brabandere M, Mousa AG, Nulens A, et al. Potential of dose optimisation in MRI-based PDR brachytherapy of cervix carcinoma. Radiother Oncol 2008;88:217–226.
655. Haie-Meder C, Chargari C, Rey A, et al. MRI-based low dose-rate brachytherapy experience in locally advanced cervical cancer patients initially treated by concomitant chemoradiotherapy. Radiother Oncol 2010;96:161–165.
656. Jurgenliemk-Schulz IM, Tersteeg RJ, Roesink JM, et al. MRI-guided treatment-planning optimisation in intracavitary or combined intracavitary/interstitial PDR brachytherapy using tandem ovoid applicators in locally advanced cervical cancer. Radiother Oncol 2009;93:322–330.
657. Ling CC, Schell MC, Working KR, et al. CT-assisted assessment of bladder and rectum dose in gynecological implants. Int J Radiat Oncol Biol Phys 1987;13:1577–1582.
658. Schoeppel SL, LaVigne ML, Martel MK, et al. Three-dimensional treatment planning of intracavitary gynecologic implants: analysis of ten cases and implications for dose specification. Int J Radiat Oncol Biol Phys1994;28:277–283.
659. Kim HJ, Kim S, Ha SW, z. Are doses to ICRU reference points valuable for predicting late rectal and bladder morbidity after definitive radiotherapy in uterine cervix cancer? Tumori 2008;94:327–332.
660. Chen SW, Liang JA, Yeh LS, et al. Comparative study of reference points by dosimetric analyses for late complications after uniform external radiotherapy and high-dose-rate brachytherapy for cervical cancer. Int J Radiat Oncol Biol Phys 2004;60:663–671.
661. Georg P, Lang S, Dimopoulos JC, et al. Dose-volume histogram parameters and late side effects in magnetic resonance image-guided adaptive cervical cancer brachytherapy. Int J Radiat Oncol Biol Phys 2011;79:356–362.
662. Kapp KS, Stuecklschweiger GF, Kapp DS, et al. Dosimetry of intracavitary placements for uterine and cervical carcinoma: results of orthogonal film, TLD, and CT-assisted techniques. Radiother Oncol 1992;24:137–146.
663. Eich HT, Haverkamp U, Micke O, et al. Dosimetric analysis at ICRU reference points in HDR-brachytherapy of cervical carcinoma. Rontgenpraxis 2000;53:62–66.
664. Pelloski CE, Palmer M, Chronowski GM, et al. Comparison between CT-based volumetric calculations and ICRU reference-point estimates of radiation doses delivered to bladder and rectum during intracavitary radiotherapy for cervical cancer. Int J Radiat Oncol Biol Phys 2005;62:131–137.
665. Patil VM, Patel FD, Chakraborty S, et al. Can point doses predict volumetric dose to rectum and bladder: a CT-based planning study in high dose rate intracavitary brachytherapy of cervical carcinoma? Br J Radiol2011;84:441–448.
666. Wachter-Gerstner N, Wachter S, Reinstadler E, et al. Bladder and rectum dose defined from MRI based treatment planning for cervix cancer brachytherapy: comparison of dose-volume histograms for organ contours and organ wall, comparison with ICRU rectum and bladder reference point. Radiother Oncol 2003;68:269–276.
667. Olszewska AM, Saarnak AE, de Boer RW, et al. Comparison of dose-volume histograms and dose-wall histograms of the rectum of patients treated with intracavitary brachytherapy. Radiother Oncol 2001;61:83–85.
668. Koom WS, Sohn DK, Kim JY, et al. Computed tomography-based high-dose-rate intracavitary brachytherapy for uterine cervical cancer: preliminary demonstration of correlation between dose-volume parameters and rectal mucosal changes observed by flexible sigmoidoscopy. Int J Radiat Oncol Biol Phys 2007;68:1446–1454.
669. Georg P, Kirisits C, Goldner G, et al. Correlation of dose-volume parameters, endoscopic and clinical rectal side effects in cervix cancer patients treated with definitive radiotherapy including MRI-based brachytherapy. Radiother Oncol 2009;91:173–180.
670. Lee L, Viswanathan A. Predictors of toxicity following image-guided high dose rate interstitial brachytherapy for gynecologic cancer. Int J Radiat Oncol Biol Phys 2012; 84(5):1192–1197.
671. Kirisits C, Lang S, Dimopoulos J, et al. Uncertainties when using only one MRI-based treatment plan for subsequent high-dose-rate tandem and ring applications in brachytherapy of cervix cancer. Radiother Oncol2006;81:269–275.
672. Himmelmann A, Holmberg E, Oden A, et al. Intracavitary irradiation of carcinoma of the cervix stage IB and IIA, a clinical comparison between a remote high dose rate afterloading system and a low dose rate manual system. Acta Radiol Oncol1985;24:139–44.
673. Thomadsen BR, Shahabi S, Stitt JA, et al. High dose rate intracavitary brachytherapy for carcinoma of the cervix: the Madison System: II. Procedural and physical considerations. Int J Radiat Oncol Biol Phys 1992;24:349–357.
674. Fellner C, Potter R, Knocke TH, et al. Comparison of radiography- and computed tomography-based treatment planning in cervix cancer in brachytherapy with specific attention to some quality assurance aspects. Radiother Oncol 2001;58:53–62.
675. Gebara WJ, Weeks KJ, Jones EL, et al. Carcinoma of the uterine cervix: a 3D-CT analysis of dose to the internal, external and common iliac nodes in tandem and ovoid applications. Radiother Oncol 2000;56:43–48.
676. Dewitt KD, Hsu IC, Speight J, et al. 3D inverse treatment planning for the tandem and ovoid applicator in cervical cancer. Int J Radiat Oncol Biol Phys 2005;63:1270–1274.
677. Suyama S, Nakaguchi T, Kawakami K, et al. Computed tomography analysis of causes of local failure in radiotherapy for cervical carcinoma. Cancer 1998;83:1956–1965.
678. Lee KC, Kim TH, Choi JH, et al. Use of the rectal retractor to reduce the rectal dose in high dose rate intracavitary brachytherapy for a carcinoma of the uterine cervix. Yonsei Med J 2004;45:113–122.
679. Wanderas AD, Sundset M, Langdal I, et al. Adaptive brachytherapy of cervical cancer, comparison of conventional point A and CT based individual treatment planning. Acta Oncol 2011;51:345–354.
680. Dimopoulos JC, Petrow P, Tanderup K, et al. Recommendations from Gynaecological (GYN) GEC-ESTRO Working Group (IV): Basic principles and parameters for MR imaging within the frame of image based adaptive cervix cancer brachytherapy. Radiother Oncol 2012;87:1192–1197.
681. Tanderup K, Nielsen SK, Nyvang GB, et al. From point A to the sculpted pear: MR image guidance significantly improves tumour dose and sparing of organs at risk in brachytherapy of cervical cancer. Radiother Oncol2010;94:173–180.
682. Mahantshetty U, Swamidas J, Khanna N, et al. Magnetic resonance image-based dose volume parameters and clinical outcome with high dose rate brachytherapy in cervical cancers—a validation of GYN GEC-ESTRO brachytherapy recommendations. Clin Oncol (R Coll Radiol) 2011;23:376–377.
683. Mahantshetty U, Swamidas J, Khanna N, et al. Reporting and validation of gynaecological Groupe Euopeen de Curietherapie European Society for Therapeutic Radiology and Oncology (ESTRO) brachytherapy recommendations for MR image-based dose volume parameters and clinical outcome with high dose-rate brachytherapy in cervical cancers: a single-institution initial experience. Int J Gynecol Cancer 2011;21:1110–1116.
684. Jurgenliemk-Schulz IM, Lang S, Tanderup K, et al. Variation of treatment planning parameters (D90 HR-CTV, D 2cc for OAR) for cervical cancer tandem ring brachytherapy in a multicentre setting: comparison of standard planning and 3D image guided optimisation based on a joint protocol for dose-volume constraints. Radiother Oncol 2010;94:339–345.
685. Mahantshetty U, Khanna N, Swamidas J, et al. Trans-abdominal ultrasound (US) and magnetic resonance imaging (MRI) correlation for conformal intracavitary brachytherapy in carcinoma of the uterine cervix. Radiother Oncol 2012;102:130–134.
686. Forrest JL, Ackerman I, Barbera L, et al. Patient outcome study of concurrent chemoradiation, external beam radiotherapy, and high-dose rate brachytherapy in locally advanced carcinoma of the cervix. Int J Gynecol Cancer2010;20:1074–1078.
687. Chatani M, Matayoshi Y, Masaki N, et al. A prospective randomized study concerning the point A dose in high-dose rate intracavitary therapy for carcinoma of the uterine cervix. The final results. Strahlenther Onkol1994;170:636–642.
688. Hama Y, Uematsu M, Nagata I, et al. Carcinoma of the uterine cervix: twice- versus once-weekly high-dose-rate brachytherapy. Radiology 2001;219:207–212.
689. Mayer A, Nemeskeri C, Petnehazi C, et al. Primary radiotherapy of stage IIA/B-IIIB cervical carcinoma. A comparison of continuous versus sequential regimens. Strahlenther Onkol 2004;180:209–215.
690. Nam TK, Ahn SJ. A prospective randomized study on two dose fractionation regimens of high-dose-rate brachytherapy for carcinoma of the uterine cervix: comparison of efficacies and toxicities between two regimens. J Korean Med Sci 2004;19:87–94.
691. Liu WS, Yen SH, Chang CH, et al. Determination of the appropriate fraction number and size of the HDR brachytherapy for cervical cancer. Gynecol Oncol 1996;60:295–300.
692. Gupta BD, Ayyagara S, Sharma SC, et al. Carcinoma of the cervix: optimal time-dose-fractionation of HDR brachytherapy and comparison with conventional dose-rate brachytherapy. In: Mould RF, ed. Brachytherapy 2.Leersum, Netherlands: Nucletron International BV; 1989:307–308.
693. Wang X, Liu R, Ma B, et al. High dose rate versus low dose rate intracavity brachytherapy for locally advanced uterine cervix cancer. Cochrane Database Syst Rev 2010:CD007563.
694. Teshima T, Inoue T, Ikeda H, et al. High-dose rate and low-dose rate intracavitary therapy for carcinoma of the uterine cervix. Final results of Osaka University Hospital. Cancer 1993;72:2409–2414.
695. Hareyama M, Sakata K, Oouchi A, et al. High-dose-rate versus low-dose-rate intracavitary therapy for carcinoma of the uterine cervix: a randomized trial. Cancer 2002;94:117–124.
696. Lertsanguansinchai P, Lertbutsayanukul C, Shotelersuk K, et al. Phase III randomized trial comparing LDR and HDR brachytherapy in treatment of cervical carcinoma. Int J Radiat Oncol Biol Phys 2004;59:1424–1431.
697. Narayan K, van Dyk S, Bernshaw D, et al. Comparative study of LDR (Manchester system) and HDR image-guided conformal brachytherapy of cervical cancer: patterns of failure, late complications, and survival. Int J Radiat Oncol Biol Phys2009;74:1529–1535.
698. Haie-Meder C, Kramar A, Lambin P, et al. Analysis of complications in a prospective randomized trial comparing two brachytherapy low dose rates in cervical carcinoma. Int J Radiat Oncol Biol Phys 1994;29:953–960.
699. Toita T, Kato S, Niibe Y, et al. Prospective multi-institutional study of definitive radiotherapy with high-dose-rate intracavitary brachytherapy in patients with nonbulky (<4-cm) stage I and II uterine cervical cancer (JAROG0401/JROSG04–2). Int J Radiat Oncol Biol Phys 2012;82:e49–e56.
700. Fowler JF. Dose reduction factors when increasing dose rate in LDR or MDR brachytherapy of carcinoma of the cervix. Radiother Oncol 1997;45:49–54.
701. Leborgne F, Fowler JF, Leborgne JH, et al. Fractionation in medium dose rate brachytherapy of cancer of the cervix. Int J Radiat Oncol Biol Phys 1996;35:907–914.
702. Rodrigus P, De Winter K, Venselaar JL, et al. Evaluation of late morbidity in patients with carcinoma of the uterine cervix following a dose rate change. Radiother Oncol 1997;42:137–141.
703. Rotmensch J, Connell PP, Yamada D, et al. One versus two intracavitary brachytherapy applications in early-stage cervical cancer patients undergoing definitive radiation therapy. Gynecol Oncol 2000;78:32–38.
704. Ngo C, Alran S, Plancher C, et al. Outcome in early cervical cancer following pre-operative low dose rate brachytherapy: a ten-year follow up of 257 patients treated at a single institution. Gynecol Oncol 2011;123:248–252.
705. Rogers CL, Freel JH, Speiser BL. Pulsed low dose rate brachytherapy for uterine cervix carcinoma. Int J Radiat Oncol Biol Phys 1999;43:95–100.
706. Kaneyasu Y, Kita M, Okawa T, et al. Treatment outcome of medium-dose-rate intracavitary brachytherapy for carcinoma of the uterine cervix: comparison with low-dose-rate intracavitary brachytherapy. Int J Radiat Oncol Biol Phys 2012;84:137–145.
707. el-Baradie M, Inoue T, Murayama S, et al. HDR and MDR intracavitary treatment for carcinoma of the uterine cervix. A prospective randomized study. Strahlenther Onkol1997;173:155–162.
708. Tanaka E, Suzuki O, Oh RJ, et al. Intracavitary brachytherapy for carcinoma of the uterine cervix—comparison of HDR (Ir-192) and MDR (Cs-137). Radiat Med 2006;24:50–57.
709. Bachtiary B, Dewitt A, Pintilie M, et al. Comparison of late toxicity between continuous low-dose-rate and pulsed-dose-rate brachytherapy in cervical cancer patients. Int J Radiat Oncol Biol Phys 2005;63:1077–1082.
710. Rath GK, Sharma DN, Julka PK, et al. Pulsed-dose-rate intracavitary brachytherapy for cervical carcinoma: the AIIMS experience. Am J Clin Oncol 2010;33:238–241.
711. Cikaric S. Radiation therapy of cervical carcinoma using either HDR or LDR afterloading: comparison of 5-year results and complications. Strahlenther Onkol (Suppl) 1988;82:119–122.
712. Glaser F. Comparison of HDR afterloading with 192Ir versus conventional radium therapy in cervix cancer: 5-year results and complications. Strahlenther Onkol (Suppl) 1988;82:106–113.
713. Joslin CAF. High-activity source afterloading in gynecologic cancer and its future prospects. Endocuriether Hypertherm Oncol 1989;5:69–81.
714. Mahmud A, Brydon B, Tonita J, et al. A population-based study of cervix cancer: incidence, management and outcome in the Canadian province of Saskatchewan. Clin Oncol (R Coll Radiol) 2011;23:691–695.
715. Sarkaria JN, Petereit DG, Stitt JA, et al. A comparison of the efficacy and complication rates of low dose-rate versus high dose-rate brachytherapy in the treatment of uterine cervical carcinoma. Int J Radiat Oncol Biol Phys1994;30:75–82; discussion 247.
716. Kapp KS, Stuecklschweiger GF, Kapp DS, et al. Prognostic factors in patients with carcinoma of the uterine cervix treated with external beam irradiation and IR–192 high-dose-rate brachytherapy. Int J Radiat Oncol Biol Phys1998;42:531–540.
717. Ferrigno R, Nishimoto IN, Novaes PE, et al. Comparison of low and high dose rate brachytherapy in the treatment of uterine cervix cancer. Retrospective analysis of two sequential series. Int J Radiat Oncol Biol Phys2005;62:1108–1116.
718. Falkenberg E, Kim RY, Meleth S, et al. Low-dose-rate vs. high-dose-rate intracavitary brachytherapy for carcinoma of the cervix: the University of Alabama at Birmingham (UAB) experience. Brachytherapy 2006;5:49–55.
719. Kuske RR, Perez CA, Jacobs AJ, et al. Mini-colpostats in the treatment of carcinoma of the uterine cervix. Int J Radiat Oncol Biol Phys 1988;14:899–906.
720. Dimopoulos JC, Kirisits C, Petric P, et al. The Vienna applicator for combined intracavitary and interstitial brachytherapy of cervical cancer: clinical feasibility and preliminary results. Int J Radiat Oncol Biol Phys 2006;66:83–90.
721. Nomden CN, de Leeuw AA, Moerland MA, et al. Clinical use of the Utrecht applicator for combined intracavitary/interstitial brachytherapy treatment in locally advanced cervical cancer. Int J Radiat Oncol Biol Phys2011;82:1424–1430.
722. Anker CJ, Cachoeira CV, Boucher KM, et al. Does the entire uterus need to be treated in cancer of the cervix? Role of adaptive brachytherapy. Int J Radiat Oncol Biol Phys 2010;76:704–712.
723. Le Pechoux C, Akine Y, Sumi M, et al. High dose rate brachytherapy for carcinoma of the uterine cervix: comparison of two different fractionation regimens. Int J Radiat Oncol Biol Phys 1995;31:735–741.
724. Hsu WL, Wu CJ, Jen YM, et al. Twice-per-day fractionated high versus continuous low dose rate intracavitary therapy in the radical treatment of cervical cancer: a nonrandomized comparison of treatment results. Int J Radiat Oncol Biol Phys1995;32:1425–1431.
725. Selke P, Roman TN, Souhami L, et al. Treatment results of high dose rate brachytherapy in patients with carcinoma of the cervix. Int J Radiat Oncol Biol Phys 1993;27:803–809.
726. Choi P, Teo P, Foo W, et al. High-dose-rate remote afterloading irradiation of carcinoma of the cervix in Hong Kong: unexpectedly high complication rate. Clin Oncol (R Coll Radiol) 1992;4:186–191.
727. Kagei K, Shirato H, Nishioka T, et al. High-dose-rate intracavitary irradiation using linear source arrangement for stage II and III squamous cell carcinoma of the uterine cervix. Radiother Oncol 1998;47:207–213.
728. Takeshi K, Katsuyuki K, Yoshiaki T, et al. Definitive radiotherapy combined with high-dose-rate brachytherapy for stage III carcinoma of the uterine cervix: retrospective analysis of prognostic factors concerning patient characteristics and treatment parameters. Int J Radiat Oncol Biol Phys 1998;41:319–327.
729. Wang CJ, Leung SW, Chen HC, et al. High-dose-rate intracavitary brachytherapy (HDR-IC) in treatment of cervical carcinoma: 5-year results and implication of increased low-grade rectal complication on initiation of an HDR-IC fractionation scheme. Int J Radiat Oncol Biol Phys 1997;38:391–398.
730. Lorvidhaya V, Tonusin A, Changwiwit W, et al. High-dose-rate afterloading brachytherapy in carcinoma of the cervix: an experience of 1992 patients. Int J Radiat Oncol Biol Phys 2000;46:1185–1191.
731. Leborgne F, Leborgne JH, Zubizarreta E, et al. High-dose-rate brachytherapy at 14 Gy per hour to point A: preliminary results of a prospectively designed schedule for cancer of the cervix based on the linear-quadratic model. Int J Gynecol Cancer2001;11:445–453.
732. Nakano T, Kato S, Cao J, et al. A regional cooperative clinical study of radiotherapy for cervical cancer in east and south-east Asian countries. Radiother Oncol 2007;84:314–319.
733. Novetsky AP, Einstein MH, Goldberg GL, et al. Efficacy and toxicity of concomitant cisplatin with external beam pelvic radiotherapy and two high-dose-rate brachytherapy insertions for the treatment of locally advanced cervical cancer. Gynecol Oncol2007;105:635–640.
734. Patel FD, Kumar P, Karunanidhi G, et al. Optimization of high-dose-rate intracavitary brachytherapy schedule in the treatment of carcinoma of the cervix. Brachytherapy 2011;10:147–153.
735. Wachter-Gerstner N, Wachter S, Reinstadler E, et al. The impact of sectional imaging on dose escalation in endocavitary HDR-brachytherapy of cervical cancer: results of a prospective comparative trial. Radiother Oncol2003;68:51–59.
736. Potter R, Knocke TH, Fellner C, et al. Definitive radiotherapy based on HDR brachytherapy with iridium 192 in uterine cervix carcinoma: report on the Vienna University Hospital findings (1993–1997) compared to the preceding period in the context of ICRU 38 recommendations. Cancer Radiother 2000;4:159–172.
737. Tan LT, Coles CE, Hart C, et al. Clinical impact of computed tomography-based image-guided brachytherapy for cervix cancer using the tandem-ring applicator—the Addenbrooke’s experience. Clin Oncol (R Coll Radiol)2009;21:175–182.
738. Kaplan S, Townamchai K, Newhouse C, et al. Determining dosimetric factors related to local control in CT-based brachytherapy. Brachytherapy 2012;11:S111.
739. Schoeppel SL, Ellis JH, LaVigne ML, et al. Magnetic resonance imaging during intracavitary gynecologic brachytherapy. Int J Radiat Oncol Biol Phys 1992;23:169–174.
740. Tardivon AA, Kinkel K, Lartigau E, et al. MR imaging during intracavitary brachytherapy of vaginal and cervical cancer: preliminary results. Radiographics 1996;16:1363–1370.
741. Haie-Meder C, Chargari C, Rey A, et al. DVH parameters and outcome for patients with early-stage cervical cancer treated with preoperative MRI-based low dose rate brachytherapy followed by surgery. Radiother Oncol2009;93:316–321.
742. Potter R, Dimopoulos J, Georg P, et al. Clinical impact of MRI assisted dose volume adaptation and dose escalation in brachytherapy of locally advanced cervix cancer. Radiother Oncol 2007;83:148–155.
743. Cozzi L, Dinshaw KA, Shrivastava SK, et al. A treatment planning study comparing volumetric arc modulation with RapidArc and fixed field IMRT for cervix uteri radiotherapy. Radiother Oncol 2008;89:180–191.
744. Zwahlen D, Jezioranski J, Chan P, et al. Magnetic resonance imaging-guided intracavitary brachytherapy for cancer of the cervix. Int J Radiat Oncol Biol Phys 2009;74:1157–1164.
745. Syed AM, Feder BH. Technique of after-loading interstitial implants. Radiol Clin (Basel) 1977;46:458–475.
746. Martinez A, Edmundson GK, Cox RS, et al. Combination of external beam irradiation and multiple-site perineal applicator (MUPIT) for treatment of locally advanced or recurrent prostatic, anorectal, and gynecologic malignancies. Int J Radiat Oncol Biol Phys 1985;11:391–398.
747. Viswanathan AN, Erickson B, Rownd J. Image-based approaches to interstitial brachytherapy. In: Viswanathan A, Kirisits C, Erickson B, et al, eds. Gynecologic radiation therapy: novel approaches to image-guidance and management, 1st ed. Berlin: Springer-Verlag, 2011:247–259.
748. Viswanathan AN, Cormack R, Rawal B, et al. Increasing brachytherapy dose predicts survival for interstitial and tandem-based radiation for stage IIIB cervical cancer. Int J Gynecol Cancer 2009;19:1402–1406.
749. Small W Jr, Strauss JB, Hwang CS, et al. Should uterine tandem applicators ever be placed without ultrasound guidance? No: a brief report and review of the literature. Int J Gynecol Cancer 2011;21:941–944.
750. Kirisits C, Lang S, Dimopoulos J, et al. The Vienna applicator for combined intracavitary and interstitial brachytherapy of cervical cancer: design, application, treatment planning, and dosimetric results. Int J Radiat Oncol Biol Phys 2006;65:624–630.
751. Syed AM, Puthawala AA, Abdelaziz NN, et al. Long-term results of low-dose-rate interstitial-intracavitary brachytherapy in the treatment of carcinoma of the cervix. Int J Radiat Oncol Biol Phys 2002;54:67–78.
752. Pierquin B, Marinello G, Mege JP, et al. Intracavitary irradiation of carcinomas of the uterus and cervix: the Creteil method. Int J Radiat Oncol Biol Phys 1988;15:1465–1473.
753. Prempree T. Parametrial implant in stage III B cancer of the cervix. III. A five-year study. Cancer 1983;52:748–750.
754. Nag S, Martinez-Monge R, Selman AE, et al. Interstitial brachytherapy in the management of primary carcinoma of the cervix and vagina. Gynecol Oncol 1998;70:27–32.
755. Recio FO, Piver MS, Hempling RE, et al. Laparoscopic-assisted application of interstitial brachytherapy for locally advanced cervical carcinoma: results of a pilot study. Int J Radiat Oncol Biol Phys 1998;40:411–414.
756. Sharma DN, Rath GK, Thulkar S, et al. High-dose rate interstitial brachytherapy using two weekly sessions of 10 Gy each for patients with locally advanced cervical carcinoma. Brachytherapy 2011;10:242–248.
757. Sharma DN, Rath GK, Thulkar S, et al. Use of transrectal ultrasound for high dose rate interstitial brachytherapy for patients of carcinoma of uterine cervix. J Gynecol Oncol 2010;21:12–17.
758. Beriwal S, Bhatnagar A, Heron DE, et al. High-dose-rate interstitial brachytherapy for gynecologic malignancies. Brachytherapy 2006;5:218–222.
759. Mikami M, Yoshida K, Takenaka T, et al. Daily computed tomography measurement of needle applicator displacement during high-dose-rate interstitial brachytherapy for previously untreated uterine cervical cancer. Brachytherapy 2011;10:318–324.
760. Shukla P, Chopra S, Engineer R, et al. Quality assurance of multifractionated pelvic interstitial brachytherapy for postoperative recurrences of cervical cancers: a prospective study. Int J Radiat Oncol Biol Phys 2012;82(4):e617–e622.
761. Damato AL, Townamchai K, Bhagwat MS, et al. Quality assurance of multi-fraction gynecologic interstitial brachytherapy: Inter-fraction dosimetric variability from catheter shifts and anatomic changes. Submitted.
762. Magné N, Mancy NC, Chajon E, et al. Patterns of care and outcome in elderly cervical cancer patients: a special focus on brachytherapy. Radiother Oncol 2009;91:197–201.
763. Barraclough LH, Swindell R, Livsey JE, et al. External beam boost for cancer of the cervix uteri when intracavitary therapy cannot be performed. Int J Radiat Oncol Biol Phys 2008;71:772–778.
764. Georg D, Kirisits C, Hillbrand M, et al. Image-guided radiotherapy for cervix cancer: high-tech external beam therapy versus high-tech brachytherapy. Int J Radiat Oncol Biol Phys 2008;71:1272–1278.
765. Akine Y, Hashida I, Kajiura Y, et al. Carcinoma of the uterine cervix treated with external irradiation alone. Int J Radiat Oncol Biol Phys 1986;12:1611–1616.
766. Saibishkumar EP, Patel FD, Sharma SC, et al. Results of external-beam radiotherapy alone in invasive cancer of the uterine cervix: a retrospective analysis. Clin Oncol (R Coll Radiol) 2006;18:46–51.
767. Wright J, Jones G, Whelan T, et al. Patient preference for high or low dose rate brachytherapy in carcinoma of the cervix. Radiother Oncol 1994;33:187–194.
768. Bastin K, Buchler D, Stitt J, et al. Resource utilization. High dose rate versus low dose rate brachytherapy for gynecologic cancer. Am J Clin Oncol1993;16:256–263.
769. Maruyama Y, Muir W. Human cervical cancer clearance after 252Cf neutron brachytherapy versus conventional photon brachytherapy. Am J Clin Oncol 1984;7:347–352.
770. Marcial VA, Blanco MS, De Leon E. Persistent tumor cells in the vaginal smear during the first year after radiation therapy of carcinoma of the uterine cervix. Prognostic significance. Am J Roentgenol Radium Ther Nucl Med1968;102:170–175.
771. Soisson AP, Geszler G, Soper JT, et al. A comparison of symptomatology, physical examination, and vaginal cytology in the detection of recurrent cervical carcinoma after radical hysterectomy. Obstet Gynecol 1990;76:106–109.
772. Davey DD, Zaleski S, Sattich M, et al. Prognostic significance of DNA cytometry of postirradiation cervicovaginal smears. Cancer 1998;84:11–16.
773. Rintala MA, Rantanen VT, Salmi TA, et al. PAP smear after radiation therapy for cervical carcinoma. Anticancer Res 1997;17:3747–3750.
774. Bodurka-Bevers D, Morris M, Eifel PJ, et al. Posttherapy surveillance of women with cervical cancer: an outcomes analysis. Gynecol Oncol 2000;78:187–193.
775. Vernooij CB, Kruitwagen RF, Rodrigus P, et al. Hematometra after radiotherapy for cervical carcinoma. Gynecol Oncol 1997;67:325–327.
776. Hintz BL, Kagan AR, Chan P, et al. Radiation tolerance of the vaginal mucosa. Int J Radiat Oncol 1980;6:711–716.
777. Larson DM, Copeland LJ, Stringer CA, et al. Recurrent cervical carcinoma after radical hysterectomy. Gynecol Oncol 1988;30:381–387.
778. Ijaz T, Eifel PJ, Burke T, et al. Radiation therapy of pelvic recurrence after radical hysterectomy for cervical carcinoma. Gynecol Oncol 1998;70:241–246.
779. Hille A, Weiss E, Hess CF. Therapeutic outcome and prognostic factors in the radiotherapy of recurrences of cervical carcinoma following surgery. Strahlenther Onkol 2003;179:742–747.
780. Sommers GM, Grigsby PW, Perez CA, et al. Outcome of recurrent cervical carcinoma following definitive irradiation. Gynecol Oncol 1989;35:150–155.
781. Coleman RL, Keeney ED, Freedman RA, et al. Radical hysterectomy after radiotherapy for recurrent carcinoma of the uterine cervix. Gynecol Oncol 1994;55:29–35.
782. Lawhead RA Jr, Clark DG, Smith DH, et al. Pelvic exenteration for recurrent or persistent gynecologic malignancies: a 10-year review of the Memorial Sloan-Kettering Cancer Center experience (1972–1981). Gynecol Oncol1989;33:279–282.
783. Kastritis E, Bamias A, Efstathiou E, et al. The outcome of advanced or recurrent non-squamous carcinoma of the uterine cervix after platinum-based combination chemotherapy. Gynecol Oncol 2005;99:376–382.
784. Tinker AV, Bhagat K, Swenerton KD, et al. Carboplatin and paclitaxel for advanced and recurrent cervical carcinoma: the British Columbia Cancer Agency experience. Gynecol Oncol 2005;98:54–58.
785. Kim JS, Kim SY, Kim KH, et al. Hyperfractionated radiotherapy with concurrent chemotherapy for para-aortic lymph node recurrence in carcinoma of the cervix. Int J Radiat Oncol Biol Phys 2003;55:1247–1253.
786. Singh AK, Grigsby PW, Rader JS, et al. Cervix carcinoma, concurrent chemoradiotherapy, and salvage of isolated paraaortic lymph node recurrence. Int J Radiat Oncol Biol Phys 2005;61:450–455.
787. Jhingran A, Yom SS, Zang X, et al Recurrence above the radiotherapy field after definitive treatment of cervix cancer. Int J Radiat Oncol Biol Phys 2005;63:S217–S218.
788. Mahé MA, Gerard JP, Dubois JB, et al. Intraoperative radiation therapy in recurrent carcinoma of the uterine cervix: report of the French intraoperative group on 70 patients. Int J Radiat Oncol Biol Phys 1996;34:21–6.
789. Garton GR, Gunderson LL, Webb MJ, et al. Intraoperative radiation therapy in gynecologic cancer: the Mayo Clinic experience. Gynecol Oncol 1993;48:328–332.
790. Abe M, Shibamoto Y. The usefulness of intraoperative radiation therapy in the treatment of pelvic recurrence of cervical cancer. Int J Radiat Oncol Biol Phys 1996;34:513–514.
791. Mahé MA, Gérard JP, Dubois JB, et al. Intraoperative radiation therapy in recurrent carcinoma of the uterine cervix: report of the French Intraoperative Group on 70 patients. Int J Radiat Oncol Biol Phys 1996;34:21–26.
792. Martinez-Monge R, Jurado M, Azinovic I, et al. Intraoperative radiotherapy in recurrent gynecological cancer. Radiother Oncol 1993;28:127–133.
793. Giorda G, Boz G, Gadducci A, et al. Multimodality approach in extra cervical locally advanced cervical cancer: chemoradiation, surgery and intra-operative radiation therapy. A phase II trial. Eur J Surg Oncol 2011;37:442–447.
794. van Lonkhuijzen L, Thomas G. Palliative radiotherapy for cervical carcinoma, a systematic review. Radiother Oncol 2011;98:287–291.
795. Grigsby PW, Portelance L, Williamson JF. High dose ratio (HDR) cervical ring applicator to control bleeding from cervical carcinoma. Int J Gynecol Cancer 2002;12:18–21.
796. Spanos W Jr, Guse C, Perez C, et al. Phase II study of multiple daily fractionations in the palliation of advanced pelvic malignancies: preliminary report of RTOG 8502. Int J Radiat Oncol Biol Phys 1989;17:659–661.
797. Spanos WJ Jr, Perez CA, Marcus S, et al. Effect of rest interval on tumor and normal tissue response—a report of phase III study of accelerated split course palliative radiation for advanced pelvic malignancies (RTOG–8502). Int J Radiat Oncol Biol Phys1993;25:399–403.
798. Spanos WJ Jr, Clery M, Perez CA, et al. Late effect of multiple daily fraction palliation schedule for advanced pelvic malignancies (RTOG 8502). Int J Radiat Oncol Biol Phys 1994;29:961–967.
799. Hornback NB, Shupe RE, Shidnia H, et al. Advanced stage IIIB cancer of the cervix treatment by hyperthermia and radiation. Gynecol Oncol 1986;23:160–167.
800. Sharma S, Patel F, Sandhu APS, et al. A prospective randomized study of local hyperthermia as a supplement and radiosensitizer in the treatment of carcinoma of the cervix with radiotherapy. Endocuriether Hyperthermia Oncol1989;5:151–159.
801. Dinges S, Harder C, Wurm R, et al. Combined treatment of inoperable carcinomas of the uterine cervix with radiotherapy and regional hyperthermia. Results of a phase II trial. Strahlenther Onkol 1998;174:517–521.
802. Harima Y, Nagata K, Harima K, et al. A randomized clinical trial of radiation therapy versus thermoradiotherapy in stage IIIB cervical carcinoma. Int J Hyperthermia 2001;17:97–105.
803. Vasanthan A, Mitsumori M, Park JH, et al. Regional hyperthermia combined with radiotherapy for uterine cervical cancers: a multi-institutional prospective randomized trial of the international atomic energy agency. Int J Radiat Oncol Biol Phys2005;61:145–153.
804. Lutgens L, van der Zee J, Pijls-Johannesma M, et al. Combined use of hyperthermia and radiation therapy for treating locally advanced cervix carcinoma. Cochrane Database Syst Rev 2010:CD006377.
805. Wootton JH, Prakash P, Hsu IC, et al. Implant strategies for endocervical and interstitial ultrasound hyperthermia adjunct to HDR brachytherapy for the treatment of cervical cancer. Phys Med Biol 2011;56:3967–3984.
806. Magrina JF, Goodrich MA, Weaver AL, et al. Modified radical hysterectomy: morbidity and mortality. Gynecol Oncol 1995;59:277–282.
807. Barnes W, Waggoner S, Delgado G, et al. Manometric characterization of rectal dysfunction following radical hysterectomy. Gynecol Oncol 1991;42:116–119.
808. Fiorica JV, Roberts WS, Greenberg H, et al. Morbidity and survival patterns in patients after radical hysterectomy and postoperative adjuvant pelvic radiotherapy. Gynecol Oncol 1990;36:343–347.
809. Kizer NT, Thaker PH, Gao F, et al. The effects of body mass index on complications and survival outcomes in patients with cervical carcinoma undergoing curative chemoradiation therapy. Cancer 2011;117:948–956.
810. Montz FJ, Holschneider CH, Solh S, et al. Small bowel obstruction following radical hysterectomy: risk factors, incidence, and operative findings. Gynecol Oncol 1994;53:114–120.
811. Eifel PJ, Levenback C, Wharton JT, et al. Time course and incidence of late complications in patients treated with radiation therapy for FIGO stage IB carcinoma of the uterine cervix. Int J Radiat Oncol Biol Phys1995;32:1289–1300.
812. Jacobs AJ, Perez CA, Camel HM, et al. Complications in patients receiving both irradiation and radical hysterectomy for carcinoma of the uterine cervix. Gynecol Oncol 1985;22:273–280.
813. Pedersen D, Bentzen SM, Overgaard J. Early and late radiotherapeutic morbidity in 442 consecutive patients with locally advanced carcinoma of the uterine cervix. Int J Radiat Oncol Biol Phys 1994;29:941–52.
814. Perez CA, Grigsby PW, Lockett MA, et al. Radiation therapy morbidity in carcinoma of the uterine cervix: dosimetric and clinical correlation. Int J Radiat Oncol Biol Phys 1999;44:855–866.
815. Pourquier H, Dubois JB, Delard R. Cancer of the uterine cervix: dosimetric guidelines for prevention of late rectal and rectosigmoid complications as a result of radiotherapeutic treatment. Int J Radiat Oncol Biol Phys1982;8:1887–1895.
816. Perez CA, Breaux S, Bedwinek JM, et al. Radiation therapy alone in the treatment of carcinoma of the uterine cervix. II. Analysis of complications. Cancer 1984;54:235–246.
817. Lanciano RM, Martz K, Montana GS, et al. Influence of age, prior abdominal surgery, fraction size, and dose on complications after radiation therapy for squamous cell cancer of the uterine cervix. A patterns of care study. Cancer 1992;69:2124–2130.
818. Lee SW, Suh CO, Chung EJ, et al. Dose optimization of fractionated external radiation and high-dose-rate intracavitary brachytherapy for FIGO stage IB uterine cervical carcinoma. Int J Radiat Oncol Biol Phys 2002;52:1338–1344.
819. Roche B, Chautems R, Marti MC. Application of formaldehyde for treatment of hemorrhagic radiation-induced proctitis. World J Surg 1996;20:1092–1094; discussion 4–5.
820. Rubinstein E, Ibsen T, Rasmussen RB, et al. Formalin treatment of radiation-induced hemorrhagic proctitis. Am J Gastroenterol 1986;81:44–45.
821. Seow-Choen F, Goh HS, Eu KW, et al. A simple and effective treatment for hemorrhagic radiation proctitis using formalin. Dis Colon Rectum 1993;36:135–138.
822. Nelson R, Norton N, Cautley E, et al. Community-based prevalence of anal incontinence. JAMA 1995;274:559–561.
823. Kim GE, Lim JJ, Park W, et al. Sensory and motor dysfunction assessed by anorectal manometry in uterine cervical carcinoma patients with radiation-induced late rectal complication. Int J Radiat Oncol Biol Phys 1998;41:835–841.
824. Quilty PM. A report of late rectosigmoid morbidity in patients with advanced cancer of the cervix, treated by a six week pelvic brick technique. Clin Radiol 1988;39:297–300.
825. Stryker JA, Bartholomew M, Velkley DE, et al. Bladder and rectal complications following radiotherapy for cervix cancer. Gynecol Oncol 1988;29:1–11.
826 Kasibhatla M, Clough RW, Montana GS, et al. Predictors of severe gastrointestinal toxicity after external beam radiotherapy and interstitial brachytherapy for advanced or recurrent gynecologic malignancies. Int J Radiat Oncol Biol Phys 2006;65:398.
827. Rotman M, Pajak TF, Choi K, et al. Prophylactic extended-field irradiation of para-aortic lymph nodes in stages IIB and bulky IB and IIA cervical carcinomas. Ten-year treatment results of RTOG 79–20. JAMA 1995;274:387–893.
828. Willett CG, Ooi CJ, Zietman AL, et al. Acute and late toxicity of patients with inflammatory bowel disease undergoing irradiation for abdominal and pelvic neoplasms. Int J Radiat Oncol Biol Phys 2000;46:995–998.
829. Tiersten A, Saltz LB. Influence of inflammatory bowel disease on the ability of patients to tolerate systemic fluorouracil-based chemotherapy. J Clin Oncol 1996;14:2043–2046.
830. Levenback C, Eifel PJ, Burke TW, et al. Hemorrhagic cystitis following radiotherapy for stage Ib cancer of the cervix. Gynecol Oncol 1994;55:206–210.
831. McIntyre JF, Eifel PJ, Levenback C, et al. Ureteral stricture as a late complication of radiotherapy for stage IB carcinoma of the uterine cervix. Cancer 1995;75:836–843.
832. Buglione M, Toninelli M, Pietta N, et al. Post-radiation pelvic disease and ureteral stenosis: physiopathology and evolution in the patient treated for cervical carcinoma. Review of the literature and experience of the Radium Institute [in Italian]. Arch Ital Urol Androl 2002;74:6–11.
833. Prasad KN, Pradhan S, Datta NR. Urinary tract infection in patients of gynecological malignancies undergoing external pelvic radiotherapy. Gynecol Oncol 1995;57:380–382.
834. Parkin DE, Davis JA, Symonds RP. Urodynamic findings following radiotherapy for cervical carcinoma. Br J Urol 1988;61:213–217.
835. DePasquale SE, Mylonas I, Falkenberry SS. Fatal recurrent ureteroarterial fistulas after exenteration for cervical cancer. Gynecol Oncol 2001;82:192–196.
836. Georgiou A, Grigsby PW, Perez CA. Radiation induced lumbosacral plexopathy in gynecologic tumors: clinical findings and dosimetric analysis. Int J Radiat Oncol Biol Phys 1993;26:479–482.
837. Abu-Rustum NR, Rajbhandari D, Glusman S, et al. Acute lower extremity paralysis following radiation therapy for cervical cancer. Gynecol Oncol 1999;75:152–154.
838. Thomas JE, Cascino TL, Earle JD. Differential diagnosis between radiation and tumor plexopathy of the pelvis. Neurology 1985;35:1–7.
839. Bruner DW, Lanciano R, Keegan M, et al. Vaginal stenosis and sexual function following intracavitary radiation for the treatment of cervical and endometrial carcinoma. Int J Radiat Oncol Biol Phys 1993;27:825–830.
840. Grigsby PW, Russell A, Bruner D et al. Late injury of cancer therapy on the female reproductive tract. Int J Radiat Oncol Biol Phys 1995;31:1281–1299.
841. Robinson JW, Faris PD, Scott CB. Psychoeducational group increases vaginal dilation for younger women and reduces sexual fears for women of all ages with gynecological carcinoma treated with radiotherapy. Int J Radiat Oncol Biol Phys 1999;44:497–506.
842. Jensen PT, Groenvold M, Klee MC, et al. Longitudinal study of sexual function and vaginal changes after radiotherapy for cervical cancer. Int J Radiat Oncol Biol Phys 2003;56:937–949.
843. Viswanathan AN. Childhood cancer survivors: stillbirth and neonatal death. Lancet 2010;376:570–572.
844. Grigsby PW, Roberts HL, Perez CA. Femoral neck fracture following groin irradiation. Int J Radiat Oncol Biol Phys 1995;32:63–67.
845. Baxter NN, Habermann EB, Tepper JE, et al. Risk of pelvic fractures in older women following pelvic irradiation. JAMA 2005;294:2587–2593.
846. Blomlie V, Rofstad EK, Talle K, et al. Incidence of radiation-induced insufficiency fractures of the female pelvis: evaluation with MR imaging. AJR Am J Roentgenol 1996;167:1205–1210.
847. Moreno A, Clemente J, Crespo C, et al. Pelvic insufficiency fractures in patients with pelvic irradiation. Int J Radiat Oncol Biol Phys 1999;44:61–66.
848. Huh SJ, Kim B, Kang MK, et al. Pelvic insufficiency fracture after pelvic irradiation in uterine cervix cancer. Gynecol Oncol 2002;86:264–268.
849. Bye A, Trope C, Loge JH, et al. Health-related quality of life and occurrence of intestinal side effects after pelvic radiotherapy—evaluation of long-term effects of diagnosis and treatment. Acta Oncol 2000;39:173–180.
850. Dusenbery KE, Carson LF, Potish RA. Perioperative morbidity and mortality of gynecologic brachytherapy. Cancer 1991;67:2786–2790.
851. Lanciano R, Corn B, Martin E, et al. Perioperative morbidity of intracavitary gynecologic brachytherapy. Int J Radiat Oncol Biol Phys 1994;29:969–974.
852. Jhingran A, Eifel PJ. Perioperative and postoperative complications of intracavitary radiation for FIGO stage I-III carcinoma of the cervix. Int J Radiat Oncol Biol Phys 2000;46:1177–1183.
853. Wollschlaeger K, Connell PP, Waggoner S, et al. Acute problems during low-dose-rate intracavitary brachytherapy for cervical carcinoma. Gynecol Oncol 2000;76:67–72.
854. Jeffries SA, Robinson JW, Craighead PS, et al. An effective group psychoeducational intervention for improving compliance with vaginal dilation: a randomized controlled trial. Int J Radiat Oncol Biol Phys 2006;65:404–411.
855. Petereit DG, Sarkaria JN, Chappell RJ. Perioperative morbidity and mortality of high-dose-rate gynecologic brachytherapy. Int J Radiat Oncol Biol Phys 1998;42:1025–1031.
856. Ogino I, Kitamura T, Okamoto N, et al. Late rectal complication following high dose rate intracavitary brachytherapy in cancer of the cervix. Int J Radiat Oncol Biol Phys 1995;31:725–734.
857. Fujikawa K, Yamamichi F, Nonomura M, et al. Spontaneous rupture of the urinary bladder is not a rare complication of radiotherapy for cervical cancer: report of six cases. Gynecol Oncol 1999;73:439–442.
858. Clark BG, Souhami L, Roman TN, et al. Rectal complications in patients with carcinoma of the cervix treated with concomitant cisplatin and external beam irradiation with high-dose-rate brachytherapy: a dosimetric analysis. Int J Radiat Oncol Biol Phys1994;28:1243–1250.
859. Weiss JP, Mattei DM, Neville EC, et al. Primary treatment of radiation-induced hemorrhagic cystitis with hyperbaric oxygen: 10-year experience. J Urol 1994;151:1514–1517.
860. Lee HC, Liu CS, Chiao C, et al. Hyperbaric oxygen therapy in hemorrhagic radiation cystitis: a report of 20 cases. Undersea Hyperb Med 1994;21:321–327.
861. Watson ER, Halnan KE, Dische S, et al. Hyperbaric oxygen and radiotherapy: a Medical Research Council trial in carcinoma of the cervix. Br J Radiol 1978;51:879–587.
862. Dische S, Saunders MI, Sealy R, et al. Carcinoma of the cervix and the use of hyperbaric oxygen with radiotherapy: a report of a randomised controlled trial. Radiother Oncol 1999;53:93–98.
863. Johnson RJ, Walton RJ. Sequential study on the effect of the addition of hyperbaric oxygen on the 5 year survival rates of carcinoma of the cervix treated with conventional fractional irradiations. Am J Roentgenol Radium Ther Nucl Med 1974;120:111–117.
864. Taylor HS, Manson JE. Update in hormone therapy use in menopause. J Clin Endocrinol Metab 2011;96:255–264.
865. Burger CW, van Leeuwen FE, Scheele F, et al. Hormone replacement therapy in women treated for gynaecological malignancy. Maturitas 1999;32:69–76.
866. Sadan O, Frohlich EP, Driscoll JA, et al. Is it safe to prescribe hormonal contraception and replacement therapy to patients with premalignant and malignant uterine cervices? Gynecol Oncol 1989;34:159–163.
867. Tominaga K, Koyama Y, Sasagawa M, et al. A follow-up study of patients with cervical cancer after resection, with special emphasis on the incidence of second primary cancers. Gynecol Oncol 1995;56:71–74.
868. Chaturvedi AK, Kleinerman RA, Hildesheim A, et al. Second cancers after squamous cell carcinoma and adenocarcinoma of the cervix. J Clin Oncol 2009;27:967–973.
869. Chaturvedi AK, Engels EA, Gilbert ES, et al. Second cancers among 104,760 survivors of cervical cancer: evaluation of long-term risk. J Natl Cancer Inst 2007;99:1634–1643.
870. Lee JY, Perez CA, Ettinger N, et al. The risk of second primaries subsequent to irradiation for cervix cancer. Int J Radiat Oncol Biol Phys 1982;8:207–211.
871. Storm HH. Second primary cancer after treatment for cervical cancer. Late effects after radiotherapy. Cancer 1988;61:679–688.
872. Hemminki K, Dong C, Vaittinen P. Second primary cancer after in situ and invasive cervical cancer. Epidemiology 2000;11:457–461.
873. Werner-Wasik M, Schmid CH, Bornstein LE, et al. Increased risk of second malignant neoplasms outside radiation fields in patients with cervical carcinoma. Cancer 1995;75:2281–2285.
874. Wright JD, St Clair CM, Deutsch I, et al. Pelvic radiotherapy and the risk of secondary leukemia and multiple myeloma. Cancer 2010;116:2486–2492.
875. Mark RJ, Poen J, Tran LM, et al. Postirradiation sarcoma of the gynecologic tract. A report of 13 cases and a discussion of the risk of radiation-induced gynecologic malignancies. Am J Clin Oncol 1996;19:59–64.
876. Zwahlen DR, Ruben JD, Jones P, et al. Effect of intensity-modulated pelvic radiotherapy on second cancer risk in the postoperative treatment of endometrial and cervical cancer. Int J Radiat Oncol Biol Phys 2009;74:539–545.
877. Seidman JD, Kumar D, Cosin JA, et al. Carcinomas of the female genital tract occurring after pelvic irradiation: a report of 15 cases. Int J Gynecol Pathol 2006;25:293–297.
878. Huang H, Liu J, Li Y, et al. Metastasis to deep obturator and para-aortic lymph nodes in 649 patients with cervical carcinoma. Eur J Surg Oncol 2011;37:978–83.
879. Canton-Romero JC, Anaya-Prado R, Rodriguez-Garcia HA, et al. Laparoscopic radical hysterectomy with the use of a modified uterine manipulator for the management of stage IB1 cervix cancer. J Obstet Gynaecol2010;30:49–52.
880. Gortzak-Uzan L, Jimenez W, Nofech-Mozes S, et al. Sentinel lymph node biopsy vs. pelvic lymphadenectomy in early stage cervical cancer: is it time to change the gold standard? Gynecol Oncol 2010;116:28–32.
881. Piver MS, Barlow JJ, Krishnamsetty R. Five-year survival (with no evidence of disease) in patients with biopsy-confirmed aortic node metastasis from cervical carcinoma. Am J Obstet Gynecol 1981;139:575–578.
882. Wharton JT, Jones HWI, Day T, et al. Preirradiation celiotomy and extended field irradiation for invasive carcinoma of the cervix. Obstet Gynecol 1977;49:333–338.
883. Rutledge TL, Kamelle SA, Tillmanns TD, et al. A comparison of stages IB1 and IB2 cervical cancers treated with radical hysterectomy. Is size the real difference? Gynecol Oncol 2004;95:70–76.
884. Kilcheski TS, Arger PH, Mulhern CB Jr, et al. Role of computed tomography in the presurgical evaluation of carcinoma of the cervix. J Comput Assist Tomogr 1981;5:378–383.
885. Yildirim Y, Sehirali S, Avci ME, et al. Integrated PET/CT for the evaluation of para-aortic nodal metastasis in locally advanced cervical cancer patients with negative conventional CT findings. Gynecol Oncol 2008;108:154–159.
886. Sharma DN, Rath GK, Kumar S, et al. Postoperative radiotherapy following inadvertent simple hysterectomy versus radical hysterectomy for cervical carcinoma. Asian Pac J Cancer Prev 2011;12:1537–1541.
887. Kim YS, Kim JH, Ahn SD, et al. High-dose extended-field irradiation and high-dose-rate brachytherapy with concurrent chemotherapy for cervical cancer with positive para-aortic lymph nodes. Int J Radiat Oncol Biol Phys2009;74:1522–1528.
888. Rajasooriyar C, Van Dyk S, Bernshaw D, et al. Patterns of failure and treatment-related toxicity in advanced cervical cancer patients treated using extended field radiotherapy with curative intent. Int J Radiat Oncol Biol Phys2011;80:422–428.
889. Kim WC, Kim GE, Suh CO, et al. High versus low dose rate intracavitary irradiation for adenocarcinoma of the uterine cervix. Jpn J Clin Oncol 2001;31:432–437.
890. Kucera H, Potter R, Knocke TH, et al. High dose versus low dose rate brachytherapy in definitive radiotherapy of cervical cancer. Wien Klin Wochenschr 2001;113:58–62.
891. Okkan S, Atkovar G, Sahinler I, et al. Results and complications of high dose rate and low dose rate brachytherapy in carcinoma of the cervix: Cerrahpaşa experience. Radiother Oncol 2003;67:97–105.
892. Sakata KI, Nagakura H, Oouchi A, et al. High dose rate intracavitary brachytherapy: results of analyses of late rectal complications. Int J Radiat Oncol Biol Phys 2002;54:1369–1376.
893. Souhami L, Corns R, Duclos M, et al. Long-term results of high-dose rate brachytherapy in cervix cancer using a small number of fractions. Gynecol Oncol 2005;97:508–513.
894. Wong FCS, Tung SY, Leung TW, et al. Treatment results of high dose rate remote afterloading brachytherapy for cervical cancer and retrospective comparison of two regimes. Int J Radiat Oncol Biol Phys 2003;55:1254–1264.
895. Sood BM, Gorla G, Gupta S, et al. Two fractions of high dose rate brachytherapy in the management of cervix cancer: clinical experience with and without chemotherapy. Int J Radiat Oncol Biol Phys 2002;53:702–706.
896. Saibishkumar EP, Patel FD, Sharma SC. Results of a phase II trial of concurrent chemoradiation in the treatment of locally advanced carinoma of uterine cervix: an experience from India. Bull Cancer 2005;92:E7–E12.
897. Ozsaran Z, Yalman D, Yurut V, et al. Radiochemotherapy for patients with locally advanced cervical cancer: early results. Eur J Gynaecol Oncol 2003;24:191–194.
898. Strauss HG, Kuhnt T, Laban C, et al. Chemoradiation in cervical cancer with cisplatin and high dose rate brachytherapy combined with external beam radiotherapy. Results of a phase II study. Strahlenther Onkol 2002;178:378–385.
899. Logsdon MD, Eifel PJ. Figo IIIB squamous cell carcinoma of the cervix: an analysis of prognostic factors emphasizing the balance between external beam and intracavitary radiation therapy. Int J Radiat Oncol Biol Phys1999;43:763–775.