Assessing Pelvic Epithelial Cancer Risk and Intercepting Early Malignancy




Abstract


This chapter addresses several important aspects of ovarian cancer prevention. One pertains to the risk factors and efforts at early detection, critically reviewing the data on screening tests for ovarian cancer. Another addresses the role of the fallopian tube, a topic that has been at the center of efforts to reduce the risk of high-grade serous cancer via both risk reduction salpingo-oophorectomy and opportunistic salpingectomy. The role of the pathologist is first and foremost by more careful scrutinizing of the fallopian tube using the Sectioning and Extensively Examining the FIMbraited End (SEE-FIM) protocol, which has allowed all pathologists to participate in this paradigm shift to the oviduct as a potential source and target of prevention. Both supporting data and caveats are presented on this compelling issue that continues to evolve and the various entities that deserve attention as either potential premalignant lesions or their mimics are illustrated.




Keywords

BRCA , BOCS, hereditary ovarian cancer, serous tubal intraepithelial carcinoma (STIC), risk-reducing salpingo-oophorectomy

 





Introduction


This chapter addresses the risk factors for pelvic (ovarian and fallopian tube) epithelial cancer, detection, and the role of the pathologist in risk reduction. The 5-year survival for stages IA and IV ovarian cancer are 88% and 18%, respectively, indicating that early detection may improve survival. However, the opportunity to detect the tumors when they are limited to the ovary or fallopian tube may be difficult, because the period of time when these tumors are so localized is brief. Thus, with the exception of discovering novel therapies, reducing the death rate from pelvic epithelial cancer will involve (1) identification of women at high risk for pelvic cancers, (2) risk reduction, and (3) early detection of patients with cancer at lower and more curable stages.




Risk Identification


Genetic Ovarian Cancer Syndromes


There are three well-recognized genetic syndromes that account for the vast majority of familial ovarian cancer and approximately 10% of all ovarian cancers. These are breast ovarian cancer syndrome (BOCS), site-specific ovarian cancer syndrome (SSOCS), and hereditary nonpolyposis colorectal cancer (HNPCC) syndrome (or Lynch syndrome). Both BOCS and SSOCS are caused by inherited mutations in the BRCA-1 and BRCA-2 genes. In fact, although often described as separate entities, these two syndromes are most likely phenotypic variants of the same genetic mutations. BRCA-1 and BRCA-2 function as classic tumor suppressor genes and are inherited in an autosomal dominant fashion. Lynch syndrome is caused by mutations in DNA mismatch repair genes responsible for repairing errors in DNA replication. Inactivation of these genes result in a high incidence of right-sided colon cancer, endometrial cancer, and ovarian cancer. Hereditary ovarian cancers associated with BRCA-1 and BRCA-2 mutations will be discussed first, followed by Lynch syndrome–related ovarian cancer.


Breast Ovarian Cancer Syndrome and Site-Specific Ovarian Cancer Syndrome


Germline mutations in the BRCA-1 and BRCA-2 tumor suppressor genes account for approximately 90% of cases of hereditary ovarian epithelial cancers and confer a risk of ovarian carcinoma by age 70 of 40% to 50% and 10% to 20%, respectively. BRCA-1 and BRCA-2 are located on chromosomes 17q21 and 13q12-13, respectively, and are inherited in an autosomal dominant fashion. They encode nuclear proteins that are functionally similar. Both proteins participate in the repair of double-stranded DNA damage, as well as the regulation of gene expression at a transcriptional level. Loss of BRCA protein function leads to failure to repair DNA damage, resulting in the activation of p53, with subsequent initiation of cell cycle arrest or apoptosis. In the absence of functional p53, however, the cell continues to proliferate, DNA damage accumulates, and the likelihood of ensuing malignancy increases.


The lifetime risk of developing ovarian cancer in the United States is about 1.4%, but among women with BRCA-1 and BRCA-2 mutations, the risk has been estimated to be about 40% and 20%, respectively. These genes also impart a significant lifetime risk of breast cancer in women and, in the case of BRCA-2, in men as well. Less than 0.3% of those in the general population are carriers of BRCA-1 or BRCA-2 mutations; however, the carrier rate is dependent on ethnic background.


Founder mutations have been identified among multiple unrelated families in Iceland, the Netherlands, Sweden, and among Jews of Central or Eastern European (Ashkenazi) descent. The best-described founder mutations are the 185delAG and 5382insC mutations in BRCA-1 and the 6174delT mutation in BRCA-2, occurring in Ashkenazi Jews at a carrier rate of 2%. Although women of Ashkenazi Jewish descent do not have an overall increased rate of ovarian cancer, if an Ashkenazi Jewish woman develops ovarian cancer, it is far more likely to be genetic rather than sporadic. Consequently, if a woman of Ashkenazi Jewish descent develops ovarian cancer, there is a 40% chance she carries a mutation in one of these two genes. The implications for her first-degree relatives (mother, sisters, daughters) are that they have a 20% risk for being gene carriers (given autosomal dominant transmission). Therefore, a woman of Ashkenazi Jewish heritage needs only one first-degree relative with ovarian cancer to be considered for further genetic counseling. Routine screening of Ashkenazi women has been proposed given the high likelihood missing carriers based on family history alone.


In contrast to hereditary breast cancers, in which BRCA-1 and BRCA-2 mutations are found with equal frequency, BRCA-1 mutations are found more commonly than BRCA-2 mutations in patients with familial ovarian carcinoma. The mean age of developing ovarian cancer in the setting of a BRCA-1 mutation is younger than in the women without a mutation (53 vs. 63 in the latter group). Although BRCA-1 carriers have a 2% to 3% risk of ovarian carcinoma by age 40, for BRCA-2 carriers, this risk is not until age 50. Mutations in BRCA genes are rare in the setting of sporadic ovarian tumors, but loss of function of their encoded proteins may play a role in tumor development. Inactivation of BRCA-1 in sporadic ovarian tumors has been attributed to promoter hypermethylation.


The Society of Gynecologic Oncologists (SGO) has issued recommended criteria for referral of women to genetic counselors and consideration for genetic testing for BRCA-1 and BRCA-2 genes. These risk variables are summarized in Box 24.1 . Because women with high-grade serous carcinoma probably have a higher rate of underlying germline mutations in BRCA (16% to 20%) than other ovarian tumors, testing should be considered for all women with this type of tumor. Some have argued for population-based testing, independent of history, given the feasibility of such testing, risk associated with missing carriers, and variability in assessment of family history and referral to genetic counselors. In addition, it is currently far more common to obtain expanded panel testing to include other lower penetrance genes.



Box 24.1

Society of Gynecologic Oncologists Guidelines for Identifying Women at Increased Risk for Having an Inherited Genetic Predisposition to Breast and Ovarian/Tubal/Peritoneal Cancer Who Should Receive Genetic Counseling and Be Offered Genetic Testing


Women affected with:




  • High grade epithelial ovarian/tubal/peritoneal cancer



  • Breast cancer ≤45 years old



  • Breast cancer with a close relative a


    a Close relative is defined as first-degree relative (parent, sibling, offspring), second-degree relative (grandparent, grandchild, uncle, aunt, nephew, niece, half-sibling), or third-degree relative (first cousin, great-grandparent, or great-grandchild).

    with breast cancer ≤50 years old or close relative a with epithelial ovarian/tubal/peritoneal cancer



  • Breast cancer ≤50 years old with a limited family history b


    b Limited family history includes less than two first- or second-degree female relative of female relatives surviving beyond 45 years old.




  • Breast cancer with two or more close relatives a with breast cancer at any age, with pancreatic cancer, or with aggressive prostate cancer (Gleason score ≥7)



  • Two breast primaries, with the first diagnosed prior to age 50



  • Triple negative breast cancer ≤60 years old



  • With breast cancer and Ashkenazi Jewish ancestry



  • Pancreatic cancer with two or more close relatives a with breast, ovarian/tubal/peritoneal, pancreatic, or aggressive prostate cancer (Gleason score ≥7)



Women unaffected with cancer, but with:




  • A first degree or several close relatives a that meet one of the above criteria



  • A close relative a carrying a known BRCA-1 or BRCA-2 mutation



  • A close relative a with male breast cancer



From Lancaster JM, Powell CB, Chen LM, et al: Society of Gynecologic Oncology statement on risk assessment for inherited gynecologic cancer predispositions. Gynecol Oncol 136(1):3-7, 2015.


Most ovarian malignancies diagnosed in women with BRCA mutations are high-grade serous carcinomas of advanced stage. These tumors tend to have morphologic features known as SET, referring to more than 50% solid, pseudoendometrioid, or transitional. Other histologic types have less frequently been described in patients with BRCA-1 mutations, including endometrioid, clear cell, transitional, and undifferentiated carcinomas; carcinosarcoma; and dysgerminoma. Borderline tumors and mucinous tumors do not appear to be associated with BRCA mutations. The overwhelming majority of malignancies identified in the fallopian tube in women with BRCA mutations are also high-grade serous carcinomas.


Lynch Syndrome


Mutations in mismatch repair genes (Lynch syndrome) account for only a small percentage of hereditary ovarian cancer; these women have a lifetime risk of up to 12% for ovarian cancer. Mutations in DNA mismatch repair genes (MLH1, MSH2, MSH6, and PMS2) most frequently lead to loss of function and, therefore, to microsatellite instability (MSI). Mismatch repair proteins function to DNA base-pair mismatches. Microsatellites are repetitive DNA sequences that are prone to replication errors, such as base-pair mismatches. Loss of mismatch repair protein function results in an inability to repair these mismatches with resultant MSI. MSI is characterized by these repetitive DNA areas retracting or expanding and causing frameshift mutations. Tumors presumably form when the frameshift mutations occur within the coding region of genes involved in tumor development. Not all mismatch repair gene mutations bear an equivalent risk for ovarian cancer; mutations in PMS2 are associated with the lowest overall risk.


Most ovarian cancers in Lynch syndrome are non-serous histology, specifically endometrioid, clear cell, or undifferentiated carcinomas. Up to 10% of ovarian carcinomas in patients less than or equal to 50 years old are associated with Lynch syndrome. There is a strong association between Lynch syndrome and clear cell carcinoma of the ovary, with mismatch repair protein mutations in 14% or 17% of clear cell carcinomas.


Guidelines for Lynch syndrome in ovarian cancer are not well defined. The Amsterdam and Bethesda criteria primarily focus on colorectal carcinoma, but the SGO has also outlined guidelines for gynecologic tumors. In general, testing for Lynch syndrome should be considered in woman with non-serous ovarian epithelial carcinoma. Because most women with Lynch syndrome who present with ovarian cancer are younger than 50 years old, many age-based algorithms must take this into account. Molecular analysis of the DNA mismatch repair genes is the gold standard for definitive diagnosis of Lynch syndrome, but it is not routinely employed as a screening method due to cost and is used mostly for confirmation. Various algorithms for using mismatch repair protein immunohistochemistry, MSI analysis, and MLH1 methylation studies have been employed. In our institution, we screen all non-serous ovarian epithelial carcinomas by mismatch repair protein immunohistochemistry, with reflex to MLH1 methylation studies in cases where MLH1 expression is lost. This information is directly forwarded to our genetic counseling department to determine the need for further assessment and potential germline testing. Parenthetically, all women with ovarian cancers at the Dana Farber Cancer Institute/Brigham and Women’s Hospital are referred for genetic counseling and undergo expanded panel testing if consenting. We no longer use family history to guide us.


Hereditary Predisposition to Ovarian Cancer: Beyond BRCA and Mismatch Repair Genes


Given that there are still families with strong histories of ovarian cancer with no identifiable mutations in BRCA-1, BRCA-2, or mismatch repair genes, current efforts are focused on identification of other inherited genetic mutations, which might account for the increased risk. Analysis of the large genomic library of single nucleotide repeats in large populations of cases and controls has been performed with the intent to identify loci that segregate with malignancy. This has been successful to a degree, identifying potentially predictive genomic markers for ovarian cancer. The downside has been the relatively low overall risk imposed by individual changes (less than twofold), which are too small to justify their use in a clinical setting, but combined risk models are emerging. Multigene panel testing for cancer risk has become available to patients complicating management recommendations and further stressing the invaluable role of genetic counselors in the process. The risk associated with the genes is highly variable with about 20% of mutations considered to be variants of uncertain significance.


Although BRCA-1, BRCA-2, and mismatch repair genes are certainly the most well-known and studied genes associated with a hereditary disposition to ovarian cancer, recent advances have identified other genes associated with potential increased risk. Mutations in BRIP1, RAD51C, and RAD51D confer a lifetime risk of 10% to 15%. A multi-gene profile (BROCA) is also in use. PALB2 gene mutations have been identified in some families with strong breast and ovarian cancer histories, but the actual risk of ovarian cancer with this gene is currently unknown. Some recently identified mutations are associated with particular types of ovarian tumors. For example, DICER1 mutations are associated with increased risk for the development of Sertoli-Leydig cell tumors, and SMARCA4 gene mutations confer risk for ovarian small cell carcinoma.


Demographic Risk Factors for Ovarian Cancer


Dietary Factors


Obesity has been reported to be associated with an increased risk of ovarian cancer mortality. There may also be an increased risk in women eating a diet high in saturated fat and low in vegetable fiber. In 1989, the observation that Swedes had both a high risk of ovarian cancer and the highest per capita dairy consumption in the world led some investigators to postulate a relationship between lactose consumption and ovarian cancer risk. Specifically, ovarian cancer cases were more likely to have high levels of galactose, a component sugar of the disaccharide lactose and a known oocyte toxin, than matched controls. This observation, however, has been inconsistent. Overall, observational studies of diet and ovarian cancer risk have not shown a consistent pattern of association; therefore, no specific dietary strategy for ovarian cancer risk reduction can be recommended.


Talc Exposure


Talc placed on the perineum may enter the vagina and ascend to the upper genital tract. Structurally similar to asbestos, there is theoretic concern that talc may potentially increase ovarian cancer risk. In addition, women who undergo tubal sterilization procedures or hysterectomy have a lower risk of ovarian cancer, supporting the ascending carcinogen hypothesis. Multiple case-control studies have shown a small but consistent increased risk with talc exposure, and a recent pooled analysis demonstrates a 24% increase in epithelial ovarian cancer (odds ratio [OR] = 1.24, 95% confidence interval [CI] = 1.15–1.33). However, two prospective cohort studies found no overall increased risk for ovarian cancer associated with talc use. Of note, one of those studies did show that there was a modest risk of the development of invasive serous carcinoma (relative risk [RR] = 1.4, 95% CI = 1.02–1.09). Overall, the evidence is mixed and inconclusive, with a possible risk associated with talc usage. Given the widespread availability and quality of cornstarch-based dusting powders and potential risk of talc-based powders, the practice of applying genuine talc to the perineum should be discouraged.


Infertility and Infertility Drugs


One of the most difficult issues to study is the association of infertility drugs and the risk of ovarian cancer. It is known, for example, that unexplained infertility is an independent risk factor for the development of ovarian cancer. One retrospective study claimed an association between prolonged clomiphene exposure and an increased risk of ovarian cancer. This study, however, was not restricted to invasive epithelial ovarian cancers but also included granulosa cell tumors. These estrogen-secreting neoplasms of stromal origin may contribute to infertility directly by disrupting normal follicular maturation and the menstrual cycle. There are, however, a number of studies, including a large collaborative analysis of 12 case-control studies, that have reported an association between fertility drugs and invasive epithelial ovarian cancer. In addition, many of the theoretic models of epithelial ovarian cancer pathogenesis implicate both incessant ovulation and high gonadotropin levels as important steps in malignant transformation of ovarian epithelium. Oral contraceptives that reduce ovulatory events and moderate gonadotropin levels are associated with a consistent and significant protective effect. It therefore seems prudent, in the absence of convincing data, to use fertility medication only when absolutely indicated, at the lowest effective dose, and for the shortest duration possible without compromising successful fertility treatment. Prior exposure to these agents should not be considered an indication for increased surveillance or prophylactic surgery.


Hormone Replacement Therapy


There appears to be an increased risk of ovarian cancer among women on estrogen replacement therapy (ERT). When compared with nonusers, users of ERT had a RR of ovarian cancer of 2.2 (95% CI = 1.53–3.17). This risk increased with the duration of use. Long-term users, defined as at least 20 years of ERT use, had a RR of 3.2 (95% CI = 1.7–5.7). Although some studies suggest a protective effect of combination replacement regimens including both estrogen and progesterone, this observation has not been confirmed. Based on these observations, long-term users of ERT should consider an increased risk of developing ovarian cancer as a factor in whether or not to initiate or continue ERT. Two recent meta-analyses suggested that women who use hormone therapy for 5 years from around 50 years old have about one extra ovarian cancer per 1000 users and, if its prognosis if typical, about one extra ovarian cancer death per 1700 users.


Endometriosis


Endometriosis increased ovarian cancer risk with a RR of 1.3 to 1.8. Such cancers are more often low stage and of low-grade endometrioid and clear cell histology. The risk is higher with increasing age and cyst complexity on ultrasound.




Reducing Risk


Oral Contraceptives


Oral contraceptive pills (OCPs) significantly reduce the risk of developing ovarian cancer. A number of studies have demonstrated a 10% per year risk reduction up to 5 to 7 years of use. This effect seems to persist for at least 10 years after OCPs are discontinued. This protective effect has also been observed in patients known to be carriers of the BRCA-1 and BRCA-2 genes and is the basis for recommending OCPs as a chemoprophylactic agent in known carriers who wish to retain their fertility. There has recently been some controversy about the protective effects of OCPs in BRCA patients. An Israeli population-based study of OCPs and ovarian cancer demonstrated a protective effect of pregnancy but not of OCPs. It is unclear why the Israeli data are inconsistent with prior published reports.


Tubal Ligation


Tubal ligation reduces risk by more than half and may be effective in subsets of women with BRCA mutations and family history of ovarian cancer. The mechanism by which this procedure reduces risk is unknown, but the popular theory is that the transfer of growth factors or carcinogens is interrupted.


Risk Reduction Surgery


Women with inherited mutations in BRCA-1 or BRCA-2 genes carry a sizable risk of “pelvic” (ovarian/fallopian tube/peritoneal) cancer. This risk is attenuated as much as 80% to 90% in those who elect to undergo risk reduction surgery. Following the increased attention to the fallopian tube as a source of high-grade serous cancer over the past 10 years, opportunistic salpingectomy has emerged as a recommended approach to reducing cancer risk in women undergoing sterilization or hysterectomy for benign disease. This involves the removal of fallopian tubes for primary prevention of epithelial carcinoma of the fallopian tube, ovary, or peritoneum in a woman undergoing pelvic surgery for another indication. Based on retrospective population studies, the reduction in ovarian cancer risk is estimated at about 50% with a hazard ratio of 0.65. Although not universal, this practice is being employed in many centers and more precise estimates as to the effectiveness of this technique should be forthcoming over the next one or two decades. Professional societies in several countries now recommend considering salpingectomy at the time of hysterectomy when convenient. Opportunistic salpingectomy with temporary preservation of the ovaries is also being explored in women with BRCA mutations who refuse oophorectomy but there are no outcome data at present. Table 24.1 provides a summary of the risk reduction for ovarian cancer provided by these potential preventative measures.



Table 24.1

Potential Preventive Measures for Ovarian Cancer



















Method Risk Reduction
Oral contraceptives OR 0.11-0.80
Tubal ligation OR 0.33-0.72
Prophylactic oophorectomy HR 0.15
Salpingectomy HR 0.65 (estimated 50% reduction)

HR, Hazard ratio; OR, odds ratio.

From Falconer H, Yin L, Grönberg H, et al: Ovarian cancer risk after salpingectomy: a nationwide population-based study. J Natl Cancer Inst 107(2), 2015; Yoon SH, Kim SN, Shim SH, et al: Bilateral salpingectomy can reduce the risk of ovarian cancer in the general population: a meta-analysis. Eur J Cancer 55:38-46, 2016.




Early Detection


There are two obvious theoretical reasons for ovarian cancer screening. The first is that tumors arising in the ovary begin as true stage I (A or B) tumors and early detection will predate de-differentiation or extra-ovarian spread. The second is that (at least theoretically) small neoplasms originating within the tubal mucosa or the ovary that are detected when the tumor burden is low would be more likely to be cured. Here it is important to separate early detection from the incidental removal of early malignancies during risk reduction surgery, which is not technically “early detection.” By far, the role of risk reduction surgery, as implied previously, is to remove the tissue at risk rather than interrupt an evolving neoplasm. In one study, 5-year cancer-free rates for BRCA-1 and BRCA-2 mutation carriers were 96% and 69%, respectively, for those with prophylactic oophorectomy versus intensive surveillance. Moreover, even some of these patients with “early” disease have positive peritoneal fluid cytology, indicating early dissemination of malignant cells presumably came from occult tubal or ovarian primary neoplasms. Nonetheless, when the early malignancy in the fallopian tube is confined to the mucosa (serous tubal intraepithelial carcinoma [STIC]) the risk of a subsequent pelvic serous cancer is approximately 5%, supporting the concept that these early lesions can be intercepted and cured. Favorable short-term outcomes in small studies raise the prospect that sufficiently sensitive detection schemes could identify early disease and possibly enhance survival.


Pelvic Examination


Pelvic examination is a central component of gynecologic care and permits the evaluation of potential abnormalities throughout the reproductive tract. However, the sensitivity of pelvic examination for the detection of ovarian cancer is only about 44%. A 15-year study of pelvic examination alone uncovered six ovarian cancers in more than 18,000 examinations of 1319 women.


Biomarker Screening Alone


Since the discovery of CA-125 in the 1980s, there has been little progress in the development of a molecular screening tool that promised to make a measurable impact on the death rate for ovarian epithelial cancer. The reasons for this are several:




  • CA-125 is a high molecular weight glycoprotein that is elevated above 35 IU/mL in 85% of all epithelial carcinomas but in only 50% of women with stage I disease.



  • CA-125 elevations are associated with a range of other intra-abdominal disorders, cutting into the specificity of this marker.



  • A test with 99% specificity will still necessitate as many as 25 invasive procedures to identify a single malignancy. This level of precision has not been attained, even when multiple biomarkers have been used, including osteopontin and HE4. One study proposed six biomarkers: leptin, prolactin, osteopontin, insulin-like growth factor II, macrophage inhibitory factor, and CA-125, claiming both high sensitivity and specificity, but it was challenged as unduly optimistic.



Another highly challenging variable in reducing the ovarian cancer death rate by conventional serologic screening lies in the nature of the disease itself. One-fourth of ovarian cancers are associated with a pathogenesis that would favor screening—that is, tumors arising in the ovary that have a lag period prior to developing and metastasizing, specifically endometrioid, mucinous, and clear cell carcinomas. Clearly, these tumors would benefit from screening. Accordingly, when detected in stage I, they all have a good to excellent prognosis. Unfortunately, two-thirds or more ovarian cancers are high-grade serous (or high-grade endometrioid) and have spread to the pelvic organs by the time of diagnosis. A recent report estimated the lead time that could be expected for serologic detection. Based on a longitudinal study with serologic biomarker analysis, the study concluded that levels of CA-125, HE4, and mesothelin began to rise as early as 3 years before the clinical presentation with malignancy; however, significant elevations were not seen until the last 12 months before presentation. The efforts to develop diagnostic biomarkers the past two decades have come face to face with the stark implications of this reality if not the profound significance of understanding the origins and precursors of ovarian cancer. At present, the failure of biomarker detection to alter the disease has resulted in recommendations by every society and task force that routine screening for ovarian cancer not be performed.


Combining Biomarkers With Ultrasound and Other Imaging Techniques


There is evidence that transvaginal sonography (TVS) improves early detection of ovarian cancer and possibly influences mortality in some instances, but at a significant trade-off in specificity ( Fig. 24.1 ). Van Nagell et al. scored ovaries as abnormal if they exceeded 10 and 20 cm 3 from post- and premenopausal women, respectively. In a study of 8500 women who underwent TVS, 121 were abnormal and underwent surgery. A total of 57 had serous cystadenomas, and eight had ovarian cancers, six of which were stage IA. Only one each had an enlarged ovary by palpation or an elevated CA-125. The implication from studies of this type is that TVS will downstage a proportion of tumors and permit improved survival. Despite these results, this group found that TVS had a sensitivity of 98.7% but a positive predictive value (PPV) of only 6.8%. Partridge and Barnes summarized five studies of 11,283 women, noting a PPV of only 3.1% for ovarian cancer. In a study of TVS encompassing 42,113 screening years, van Nagell et al. defined an ovarian volume exceeding 10 cm in postmenopausal women or 20 cm in premenopausal women, or papillary or complex architecture as abnormal. Some 17 of 180 patients with persisting TVS abnormalities who underwent exploratory laparoscopy or laparotomy had cancer, 11 of which were stage I. Eight of the 11 did not have a palpable mass. TVS screening in this setting had a PPV of 9.4% with a negative predictive value of 99.97%. When nonepithelial tumors were excluded, the survival of ovarian cancer patients in the annually screened population was 92.9% at 2 years and 83.6% at 5 years. The authors cautioned that although the improved detection and reduced mortality were associated with screening, this benefit did not apply to women whose cancers occurred in the setting of a normal ovarian volume. Additional studies have echoed the low PPV (1% to 27%) and false positive/true positive ratios of over 30, while reporting a small number of tumors diagnosed at stage I.






Fig. 24.1


Ultrasound evaluation of benign and malignant ovarian cysts. A, Hemorrhagic smooth-walled cyst. B, Ovarian cancer with irregular wall thickness, lining surface, and septation.

(Courtesy of Beryl Bennaceraf, Boston, MA.)


A recent report summarized sequential screening with a baseline and three follow-up CA-125 and TVS assessments. The PPVs were 1.4% to 1.8%, respectively. The ratio of surgical procedures to cancers detected was 31 : 1 at the initial screening and 14 : 1 on the subsequent three testing events. Similar to the studies described, 67% to 83% were stage III or IV, respectively, when diagnosed. Most of the surgeries were prompted by a positive TVS event. An important caveat is that these studies targeted ovarian enlargement, a strategy that ignores the early phases of serous carcinogenesis in the distal fallopian tube, ovarian, and pelvic surfaces. The same limitations apply to the use of ultrasound and biomarkers in combination. The Prostate, Lung, Colorectal and Ovarian (PLCO) trial show that screening with both CA-125 and transvaginal ultrasound compared with standard care did not reduce ovarian cancer mortality.


A recent trial garnering considerable attention was the United Kingdom Collaborative Trial of Ovarian Cancer Screening (UKTOCS) that compared yearly CA-125 (using an algorithm) and transvaginal ultrasound (multimodality screening [MMS]) to ultrasound alone and no screening. Over 200,000 women were enrolled. Although the primary analysis did not appear to show a reduction in mortality for MMS, there was a 20% mortality reduction at 7 to 14 years. The authors concluded that this was due to removal of prevalent cases (years 1 to 7). Caveats include the following:




  • The notion that the differences in mortality between early and later years of the study are due to prevalent versus incident disease is unproven.



  • Estimates of ovarian cancer mortality were based on death certificate data in the compared populations, and the precise reason for reduced mortality estimates is not known.



  • There is no evidence that this screening algorithm will alter mortality in women with BRCA-1 or BRCA-2 mutations, where the ovarian cancer outcome estimation would be far less prone to error.



This management algorithm is currently not recommended by the U.S. Food and Drug Administration (FDA). Various other algorithms which combine imaging, biomarkers, and clinical signs have been proposed but have not yet been widely adopted.


Molecular Screening for Ovarian Cancer


With the emergence of highly sensitive molecular assays for gene mutations, a few studies have explored the possibility that ovarian or tubal cancers could be detected via analysis of lower genital tract fluids. In theory, these “molecular Papanicolaou (Pap) tests” would detect mutations by sequence analysis targeting common mutations associated with gynecologic cancers. Kinde et al. successfully identified mutations in liquid-based Pap tests from 9 of 22 ovarian carcinomas. It remains to be seen whether this approach will detect early ovarian or tubal carcinomas in their curable stages or will encounter problems with “background mutations” in the genital tract mucosa of healthy women.




Presenting Signs and Symptoms


Clinical presentations with borderline early and advanced cancer vary. Webb et al. examined patients in these groups and noted 16%, 7%, and 4% of patients with borderline disease, early ovarian cancer, and advanced ovarian cancer, respectively, were symptom free. Abdominal pain (44%) and swelling (39%) were the most common symptoms as opposed to abdominal mass and gynecologic symptoms (12% each). Gastrointestinal problems and malaise were more common in advanced disease.


Overall, ovarian volume is expected to decrease with age. In patients with a suspected pelvic mass, TVS, with or without CA-125 assessments, is of considerable value in segregating low- versus high-risk patients and can be used as a guide to intervention (see Fig. 24.1 ). The presence of a mass with conspicuous calcium supports the diagnosis of teratoma. In contrast, any solid tumor is of concern. Numerous investigators have proposed scoring systems, which depend on specific characteristics of tumors that predict risk. Certain groups can be identified with a strikingly low risk. Bailey et al. showed that none of unilocular cysts less than 10 cm in diameter proved to be malignant. Half resolved spontaneously; many of the rest were cystadenomas. In contrast, complex cysts had a similar resolution rate, but seven of 114 proved to be malignant. In a study of more than 3200 unilocular cysts, nearly 70% resolved spontaneously. Of the remainder, 16.5% (of the total) developed septation, 5.8% a solid area, and 6.8% persisted as a unilocular lesion. Of 27 cancers emerging in the persistent group, 10 were associated with the unilocular lesions, underscoring the need to follow all patients with a persistent ovarian lesion.




The Role of the Pathologist in Risk Reduction and Early Detection


There are two opportunities for the pathologist to participate in the early detection of ovarian cancer:




  • Routine analysis of the distal fallopian tube in women undergoing procedure for benign disorders: The routine analysis of the distal fallopian tube in salpingectomies from benign disorders will occasionally uncover an early (intraepithelial) carcinoma in that site. Several studies of limited populations employing comprehensive examination of the fallopian tube similar to the Sectioning and Extensively Examining the FIMbraited End (SEE-FIM) protocol have estimated the frequency of occult serous carcinoma to be between 0.6% and 1.1% in women with no history of or risk factors for serous carcinomas of the uterus or other pelvic sites. In a recent report from Brigham and Women’s Hospital covering 10 years, the frequency of tubal serous carcinoma was estimated to be between 0.1% and 0.2%. Notably, many of the early carcinomas were associated with concurrent endometrioid carcinomas of the uterus; the significance of this association (which is still uncommon) is under study.




    • The detection of these rare, but early cancers will permit both a thorough workup of the patient for metastatic disease and, in the right setting, a rationale for genetic testing, which if positive could benefit others in the family. It is our practice to submit the fimbriated end of the fallopian tube in addition to representative cross-sections in these cases.




  • Systematic analysis of fallopian tubes from women who are deemed at risk for ovarian cancer (i.e., those with known inherited mutations in the BRCA-1 or BRCA-2 gene): The more conventional exercise is the evaluation of the prophylactic salpingo-oophorectomy from these women for evidence of occult malignancy. However, in recent years, a proportion of these tumors have been found in the fallopian tube mucosa, leading investigators to propose a tubal origin in many instances. Moreover, the frequent location of these early lesions in the fimbriated portion of the tube mandates thorough attention to this portion of the structure. This discussion will address the following points: (1) the beneficial role of prophylactic surgery, (2) the risk of malignancy in women with mutations in BRCA, (3) the procedure for evaluation of the fallopian tube, (4) the criteria for tubal intraepithelial carcinoma, (5) the pitfalls in the diagnosis of early tubal malignancies, and (6) the potential impact on clinical management and perceptions of serous carcinogenesis.



Prophylactic Surgery and Detection of Early Malignancy


Bilateral salpingo-oophorectomy in women with BRCA mutations reduces the risk of developing pelvic serous cancers by greater than 90%. Laparoscopy (whenever possible this is the preferred modality) and laparotomy (almost never) are the surgical modalities of choice to allow inspection of the peritoneal surfaces at the time of prophylactic salpingo-oophorectomy and collect fluid for cytologic evaluation. Performing a hysterectomy in addition to removing the adnexa does not appear to improve the efficacy of the procedure. Paradoxically, women with BRCA -associated cancers have a better clinical outcome following treatment than those without BRCA mutations, even though the former tend to present at a higher stage. Presumably, this is due in part to BRCA -associated tumors being more susceptible to chemotherapeutic agents, such as cisplatin, that induce double-stranded DNA damage.


Prophylactic surgery may reduce, but not completely eliminate, the risk of ovarian cancer in high-risk individuals. Bilateral salpingo-oophorectomy in BRCA carriers reduces ovarian cancer risk by over 90% and breast cancer risk by over 50%. The operation should be reserved for women with known mutations in BRCA-1 or BRCA-2 (or other lower penetrance genes with deleterious mutations) or those who have a family history consistent with one of the genetic syndromes associated with ovarian cancer, and it should include an evaluation by a genetic counselor. The addition of hysterectomy does not appear to increase the efficacy of the operation and should be performed only for concurrent gynecologic indications or if the patient has HNPCC. Patients should be informed that prophylactic surgery does not protect them against the subsequent development of high-grade serous carcinoma of the peritoneum. They should also be warned that about 7% of prophylactic operations detect occult ovarian or tubal carcinoma and that these lesions may not be appreciated until final pathology reports are available. Pathologists should be instructed to submit the entire specimen for sectioning to reduce the risk of missing a microscopic occult malignancy. Finally, the patient should be prepared for the consequences of surgical menopause.


An additional risk that has been addressed recently is that of a subsequent uterine serous adenocarcinoma. One study computed an odds ratio of 22 for women with BRCA-1 and 6.4 for those with BRCA-2 germ line mutations. These estimates were based on a very small number of cancer cases across multiple institutions; however, three of three did show loss of wild type BRCA-1 gene. Thus this risk, albeit small, will enter into the discussion when counseling women with these mutations.


In studies of women undergoing prophylactic salpingo-oophorectomy for BRCA-1 or BRCA-2 mutations (BRCA+), the risk of uncovering an early cancer has been estimated to be about 5% to 10%, a figure that may vary both as a function of the age of the population and the thoroughness with which the specimen is examined. Many of these early cancers have been identified in the distal fallopian tube, the earliest malignancy being a tubal intraepithelial carcinoma. In a review of nearly 20 cases in our institution, the frequency of a tubal intraepithelial carcinoma was over 80%. In the largest study to date (GOG-0199), clinically occult cancers were detected in 15 of 326 (4.6%) BRCA-1 carriers and 8 of 231 (3.5%) BRCA-2 carriers undergoing prophylactic salpingo-oophorectomy; about half of these tumors were more than stage II. With these figures in mind, the following points must be stressed:




  • Early malignancies can be both serous (STIC, 80%) and rarely, endometrioid (ETIC, 20%).



  • Twenty percent of these early serous cancers are not associated with an intraepithelial lesion in the fallopian tube; more extensive tubal sampling might uncover some, but most of the time it will not.



  • Advanced pelvic serous cancers in BRCA+ women are virtually identical in tumor distribution to the general population with this disease (i.e., less than half will have evidence of a tubal origin). Although it may be tempting to attribute this to obliteration of the tubal mucosal origin by expanding tumor or some other sampling artifact, the reader is strongly cautioned that salpingectomy alone may not rid the patient of risk, and leaving the ovary might even place her at a substantial risk of an eventual pelvic serous cancer .



  • Prophylactic specimens from women with a family history of cancer but without a documented BRCA mutation are unlikely to contain an early malignancy. Virtually all genetically related pelvic serous cancers are linked to BRCA+.



  • As mentioned earlier, STICs are also rarely reported in fallopian tubes removed as part of surgery for indications other than serous carcinoma.



The SEE-FIM Protocol


Prophylactic salpingo-oophorectomies from women with mutations in BRCA or strong family histories of ovarian carcinoma should be submitted entirely and examined histologically. Most of the early cancers described in the preceding sections occur in grossly unremarkable specimen, so it is imperative that all the tissue be evaluated in order to properly treat the individual and to further understand the pathogenesis of this disease.


The fallopian tubes and ovaries should be submitted entirely and be evaluated in serial sections by a pathologist with expertise in gynecologic malignancies. At Brigham and Women’s Hospital, the goal is to ensure sectioning and extensive examination of the fimbria (SEE-FIM protocol), because the majority of early serous tumors occur in this area, which is as follows ( Fig. 24.2 ):




  • The entire tube is initially fixed for at least 4 hours to minimize loss of epithelium during manipulation.



  • The distal 2 cm of the fimbriated end is amputated.



  • The fimbrial mucosa is sectioned longitudinally into four pieces. The remainder of the tube is sectioned transversely every 2 to 3 mm into cross sections.



  • The tubal segments combined with the fully sectioned fimbriated end are submitted in toto.


Feb 26, 2019 | Posted by in GYNECOLOGY | Comments Off on Assessing Pelvic Epithelial Cancer Risk and Intercepting Early Malignancy

Full access? Get Clinical Tree

Get Clinical Tree app for offline access