reducing ovarian cancer mortality through early detection ...cancer prevention research | minireview...

13
CANCER PREVENTION RESEARCH | MINIREVIEW Reducing Ovarian Cancer Mortality Through Early Detection: Approaches Using Circulating Biomarkers Camille V. Trinidad 1 , Ashley L. Tetlow 1 , Leonidas E. Bantis 2 , and Andrew K. Godwin 1,3 ABSTRACT More than two-thirds of all women diagnosed with epithelial ovarian cancer (EOC) will die from the disease (>14,000 deaths annually), a fact that has not changed considerably in the last three decades. Although the 5-year survival rates for most other solid tumors have improved steadily, ovarian cancer remains an exception, making it the deadliest of all gynecologic cancers and ve times deadlier than breast cancer. When diagnosed early, treat- ment is more effective, with a 5-year survival rate of up to 90%. Unfortunately, most cases are not detected until after the cancer has spread, resulting in a dismal 5-year survival rate of less than 30%. Current screening methods for ovarian cancer typically use a combination of a pelvic examination, transvaginal ultrasonography, and serum cancer antigen 125 (CA125), but these have made minimal impact on improving mortality. Thus, there is a compel- ling unmet need to develop new molecular tools that can be used to diagnose early-stage EOC and/or assist in the clinical management of the disease after a diagnosis, given that more than 220,000 women are living with ovarian cancer in the United States and are at risk of recurrence. Here, we discuss the state of advancing liquid-based approaches for improving the early detection of ovarian cancer. See all articles in this Special Collection Honoring Paul F. Engstrom, MD, Champion of Cancer Prevention Introduction Ovarian cancers are a heterogeneous group of malignancies arising from or involving the ovary and/or fallopian tubes (1). Ovarian cancers are broadly classied into nonepithelial ovar- ian cancer and epithelial ovarian cancer (EOC) and that the latter is attributed to the majority of ovarian cancerrelated deaths (2). Overall, EOC is the fth leading cause of cancer- related death among American women, making it the deadliest cancer of the reproductive system (3). EOC is a heterogeneous disease, comprising several histotypes (serous, 70%; endo- metrioid, 10%; clear cell, 10%; and mucinous, 3%10%) with distinct epidemiologic, molecular, and clinical features. High-grade serious carcinomas (HGSC) are estimated to be 50% to 60% of all ovarian malignancies and account for half of all EOCs. EOCs are diagnosed predominantly at an advanced stage with widespread metastases throughout the peritoneal cavity (46). Manifestations of the disease are typically vague and do not become apparent until the disease is advanced and difcult to treat. Thus, ovarian cancer is caught at an early stage only in about one-fth of all cases according to the National Ovarian Cancer Coalition. Furthermore, no reliable screening tests are currently available for ovarian cancer, and current diagnostic tools for early detection remain inadequate. Early detection of ovarian cancer at a localized stage (stages IA and IB) results in far better disease prognosis: according to the American Cancer Society (2019), the projected 5-year survival rate for these patients is about 92%. However, only 15% of all ovarian cancers are successfully diagnosed at this stage. Patients with advanced-stage HGSC have a worst prognosis (32% and 15% for 5- and 10-year survival, respectively) compared with patients diagnosed with early-stage HGSC [71% and 53% (5- and 10-year survival, respectively); ref. 7]. Moreover, 30 years of statistics gathered by the NIH Surveillance, Epide- miology, and End Results (SEER) Program indicate that approximately 70% of patients diagnosed with ovarian cancer will experience disease recurrence after initial benet from chemotherapy (8). These statistics clearly emphasize the need to develop tools to detect ovarian cancer at an earlier stage. The two most commonly used screening tests in the clinic are transvaginal ultrasound (TVUS) and blood tests for elevated cancer antigen-125 (CA125) protein levels. It is difcult to distinguish benign from malignant tumors by TVUS and, hence, the need for more invasive biopsies. On the other hand, CA125 protein levels are typically not elevated in the serum of up to 50% of patients with stage I ovarian cancer; thereby, it is not as reliable for early detection (9). Also, blood tests quan- tifying CA125 protein level are not specic to ovarian cancer, as CA125 elevation can be potentiated by many factors (10). Unlike breast cancer in which a biopsy can be performed for diagnosis, EOC screening requires invasive surgery to make 1 Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas. 2 Department of Biostatistics, University of Kansas Medical Center, Kansas City, Kansas. 3 The University of Kansas Cancer Center, Kansas City, Kansas. C.V. Trinidad and A.L. Tetlow contributed equally to this article. Corresponding Author: Andrew K. Godwin, University of Kansas Medical Center, 3901 Rainbow Boulevard, MS 3040, Kansas City, KS 66160. Phone: 913- 945-6373; Fax: 913-945-6327; E-mail: [email protected] Cancer Prev Res 2020;13:24152 doi: 10.1158/1940-6207.CAPR-19-0184 Ó2020 American Association for Cancer Research. AACRJournals.org | 241 Research. on May 26, 2020. © 2020 American Association for Cancer cancerpreventionresearch.aacrjournals.org Downloaded from

Upload: others

Post on 24-May-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Reducing Ovarian Cancer Mortality Through Early Detection ...CANCER PREVENTION RESEARCH | MINIREVIEW Reducing Ovarian Cancer Mortality Through Early ... standard in the initial diagnosis

CANCER PREVENTION RESEARCH | MINIREVIEW

Reducing Ovarian Cancer Mortality Through EarlyDetection: Approaches Using Circulating BiomarkersCamille V. Trinidad1, Ashley L. Tetlow1, Leonidas E. Bantis2, and Andrew K. Godwin1,3

ABSTRACT◥

More than two-thirds of all women diagnosed withepithelial ovarian cancer (EOC) will die from the disease(>14,000 deaths annually), a fact that has not changedconsiderably in the last three decades. Although the 5-yearsurvival rates for most other solid tumors have improvedsteadily, ovarian cancer remains an exception, making itthe deadliest of all gynecologic cancers and five timesdeadlier than breast cancer. When diagnosed early, treat-ment is more effective, with a 5-year survival rate of up to90%. Unfortunately, most cases are not detected until afterthe cancer has spread, resulting in a dismal 5-year survivalrate of less than 30%. Current screening methods forovarian cancer typically use a combination of a pelvic

examination, transvaginal ultrasonography, and serumcancer antigen 125 (CA125), but these have made minimalimpact on improving mortality. Thus, there is a compel-ling unmet need to develop new molecular tools that canbe used to diagnose early-stage EOC and/or assist in theclinical management of the disease after a diagnosis, giventhat more than 220,000 women are living with ovariancancer in the United States and are at risk of recurrence.Here, we discuss the state of advancing liquid-basedapproaches for improving the early detection of ovariancancer.See all articles in this Special CollectionHonoring Paul

F. Engstrom, MD, Champion of Cancer Prevention

IntroductionOvarian cancers are a heterogeneous group of malignancies

arising from or involving the ovary and/or fallopian tubes (1).Ovarian cancers are broadly classified into nonepithelial ovar-ian cancer and epithelial ovarian cancer (EOC) and that thelatter is attributed to the majority of ovarian cancer–relateddeaths (2). Overall, EOC is the fifth leading cause of cancer-related death among American women, making it the deadliestcancer of the reproductive system (3). EOC is a heterogeneousdisease, comprising several histotypes (serous, �70%; endo-metrioid, �10%; clear cell, �10%; and mucinous, �3%–10%)with distinct epidemiologic, molecular, and clinical features.High-grade serious carcinomas (HGSC) are estimated to be50% to 60% of all ovarian malignancies and account for half ofall EOCs.EOCs are diagnosed predominantly at an advanced stage

with widespread metastases throughout the peritonealcavity (4–6). Manifestations of the disease are typically vagueand do not become apparent until the disease is advanced and

difficult to treat. Thus, ovarian cancer is caught at an early stageonly in about one-fifth of all cases according to the NationalOvarian Cancer Coalition. Furthermore, no reliable screeningtests are currently available for ovarian cancer, and currentdiagnostic tools for early detection remain inadequate. Earlydetection of ovarian cancer at a localized stage (stages IA andIB) results in far better disease prognosis: according to theAmerican Cancer Society (2019), the projected 5-year survivalrate for these patients is about 92%. However, only 15% of allovarian cancers are successfully diagnosed at this stage. Patientswith advanced-stage HGSC have a worst prognosis (�32% and�15% for 5- and 10-year survival, respectively) compared withpatients diagnosed with early-stage HGSC [�71% and �53%(5- and 10-year survival, respectively); ref. 7]. Moreover,30 years of statistics gathered by the NIH Surveillance, Epide-miology, and End Results (SEER) Program indicate thatapproximately 70% of patients diagnosed with ovarian cancerwill experience disease recurrence after initial benefit fromchemotherapy (8). These statistics clearly emphasize the needto develop tools to detect ovarian cancer at an earlier stage.The twomost commonly used screening tests in the clinic are

transvaginal ultrasound (TVUS) and blood tests for elevatedcancer antigen-125 (CA125) protein levels. It is difficult todistinguish benign from malignant tumors by TVUS and,hence, the need for more invasive biopsies. On the other hand,CA125 protein levels are typically not elevated in the serum ofup to 50% of patients with stage I ovarian cancer; thereby, it isnot as reliable for early detection (9). Also, blood tests quan-tifying CA125 protein level are not specific to ovarian cancer, asCA125 elevation can be potentiated by many factors (10).Unlike breast cancer in which a biopsy can be performedfor diagnosis, EOC screening requires invasive surgery tomake

1Department of Pathology and Laboratory Medicine, University of KansasMedical Center, Kansas City, Kansas. 2Department of Biostatistics, Universityof KansasMedical Center, Kansas City, Kansas. 3The University of Kansas CancerCenter, Kansas City, Kansas.

C.V. Trinidad and A.L. Tetlow contributed equally to this article.

Corresponding Author: Andrew K. Godwin, University of Kansas MedicalCenter, 3901 Rainbow Boulevard, MS 3040, Kansas City, KS 66160. Phone: 913-945-6373; Fax: 913-945-6327; E-mail: [email protected]

Cancer Prev Res 2020;13:241–52

doi: 10.1158/1940-6207.CAPR-19-0184

�2020 American Association for Cancer Research.

AACRJournals.org | 241

Research. on May 26, 2020. © 2020 American Association for Cancercancerpreventionresearch.aacrjournals.org Downloaded from

Page 2: Reducing Ovarian Cancer Mortality Through Early Detection ...CANCER PREVENTION RESEARCH | MINIREVIEW Reducing Ovarian Cancer Mortality Through Early ... standard in the initial diagnosis

a definitive diagnosis (11, 12) and therefore false-positivesurgeries must be limited to at most 10 per screen-detectedcancer, that is, a positive predictive value (PPV) exceeding 10%,which for the patient is quite unacceptable. Thus, the lowprevalence of this cancer in the general postmenopausal pop-ulation (1 in 2,500) requires an effective screening strategy tohave a high sensitivity for early-stage disease (>75%) and a veryhigh combined specificity (99.6%) to achieve a PPV exceeding10% (11–19). When CA125 is interpreted longitudinally withthe risk of ovarian cancer algorithm (ROCA) referring 2%annually at highest risk to transvaginal ultrasound (TVUS), this“2-stage screening strategy” achieved a combined specificity of99.8%, a 22% PPV—greater than the 10% lower limit, anddetected an increased but still modest proportion of EOCs inearly stage (11, 20, 21). In a 2017 landmark study, the UnitedKingdom Collaborative Trial of Ovarian Cancer Screeningcompared several screening approaches in more than200,000 postmenopausal women (median follow-up of 11.1years): (i) serum CA125 level evaluated using ROCA (50,640women), (ii) annual TVUS screening (50,639 women), or (iii)no screening (101,359 women). Unfortunately, like otherovarian cancer screening studies primarily focused on mea-suring serum CA125 levels (22–27), this study did not show asignificant decrease in ovarian cancer mortality (28). Thisclinical problem highlights the urgent need to expand the classof liquid-based biomarkers to detect a substantially greaterproportion of cancers in early stage, while maintaining a 98%annual specificity for referral to imaging (e.g., TVUS).

Liquid-Based Biopsies: Can TheseDevelopingMolecular ToolsBeUsedto Improve Early Detection ofCancer?Pathologic analysis of tumor tissue biopsies has been the gold

standard in the initial diagnosis of cancer, but liquid biopsies,which analyze tumor-derivedmaterial circulating in the blood-stream and other bodily fluids, are rapidly gaining traction inthe clinic. Liquid biopsy is a minimal or noninvasive technol-ogy that detects molecular biomarkers using liquid samplewithout the need for costly or invasive procedures. These testshave considerable potential in oncology, e.g., for early detectionof cancer, treatment and recurrence monitoring, and as surro-gates for traditional biopsies with the purpose of predictingresponse to treatments and the development of acquiredresistance. A liquid biopsy can provide information aboutthe genetic landscape of all cancerous lesions (primary andmetastases) as well as offer the opportunity to systematicallytrack genomic evolution. The liquid biopsy biomarker types areprimarily segmented into circulating tumor cells (CTCs), cell-free DNA (cfDNA), and extracellular vesicles (EV; Fig. 1). Thediscovery of CTCs dates back to the 1860s, when cells that weremorphologically identical to the tumor were identified in theblood of a patient with metastatic cancer (29). Their potentialsignificance was not fully realized until the late 2000s, when the

number of CTCs in the bloodstream were shown to haveprognostic significance in various different tumor types, forexample, metastatic breast, metastatic castration-resistantprostate, and metastatic colorectal cancer (30). Another waythat tumors release biomarker information is through tumorcell necrosis and the release of dead cells or cell fragments.These cells are engulfed by phagocytes that process the tumorcell DNA into small fragments (160–180 base pairs in length) ofnucleic acids, which is then released into the blood-stream (31, 32). Although levels of tumor DNA have beenshown to mirror tumor burden, interpretation is often com-plicated by the presence of other cfDNA derived from non-tumor cells. Therefore, a number of highly sensitive methodshave been developed to detect aberrations found in circulatingtumor DNA (ctDNA), including mutation, amplification,chromosomal rearrangement, and hypermethylation. A thirdbiomarker, which has been known for years, has reemerged andis showing great promise. This member of the EV family,known as exosomes or small EVs, are nano-sized vesicles(30–150 nm) of endocytic origin, which are produced andreleased by most cell types under normal physiologic and indiseased states (33, 34). Once considered little more thangarbage cans whose job was to discard unwanted cellularcomponents, recent discoveries have sparked considerableinterest in exosomes as circulating biomarkers. Exosomes areinformative molecules that carry cargo representative of theiroriginating cell including nucleic acids, cytokines, membrane-bound receptors, and a wide assortment of other, biologicallyactive lipids and proteins (35–38). This cargo remains func-tional upon entry or fusion with a recipient cell, thus exosomaltransfer is now considered an important form of cell–cellcommunication in normal and pathologic states, such ascancer. Because exosomes can travel systemically throughoutthe body, efforts are underway to exploit them as potentialbiomarkers to detect and monitor disease states. In this review,we will focus on the current methods and advancements usingliquid-based biopsies that have shown promising potential inthe early detection of ovarian cancer.

Circulating Tumor CellsFirstobserved in1869byThomasAshworth in thebloodof an

individualwithmetastaticdisease,CTCshave longbeenthoughtto be an important component of tumor dissemina-tion (29, 39, 40). CTCs are cancer cells that have detached fromthe tumor and are found at extremely low levels in the blood-stream. Modern research on CTCs has focused on CTC enu-meration and characterization. Some speculate that tumor cellshedding is an early event in tumorigenesis andmight be usefulfor early detection, but few studies have shown utility in thisclinical setting. As a blood-based, FDA-approved biomarker,CTCs were thought useful for determining the prognosis ofpatients with metastatic breast, colorectal, and prostatecancers (41–45). As the only FDA-cleared device for the enu-meration of CTCs inwhole blood, CellSearch (Menarini SiliconBiosystems, Inc.) was originally used for the evaluation of CTC

Trinidad et al.

Cancer Prev Res; 13(3) March 2020 CANCER PREVENTION RESEARCH242

Research. on May 26, 2020. © 2020 American Association for Cancercancerpreventionresearch.aacrjournals.org Downloaded from

Page 3: Reducing Ovarian Cancer Mortality Through Early Detection ...CANCER PREVENTION RESEARCH | MINIREVIEW Reducing Ovarian Cancer Mortality Through Early ... standard in the initial diagnosis

numbers in patients with cancer to help predict prognosis andoverall survival (30). However, clinical enumeration of thesecirculating cells has become less useful and costly, due to the factCTC tests are rarely reimbursed by insurance. Regardless, theinterest in CTCs continues and many different platforms forCTC isolation have been developed for research as well asclinical use in recent years (46). Nevertheless, CTC isolationand characterization remain technically challenging and prog-ress has been hampered by the difficulty in rapidly and effec-tively isolating pure populations of CTCs using liquid biopsy.The three main categories of isolation techniques are

immune-affinity–based methods, size-/density-based meth-ods, and microfluidic device–based methods (ref. 47; Fig. 2).The earliest approaches utilized positive and negative selectionfor commonCTC surface antigens or organ-specificmarkers todifferentiate CTCs from other cell types. In these studies,researchers used antibody-coated beads to positively selectCTCs from the cellular milieu or deplete the milieu of othercell types with a negative selection marker. EpCAM, a celladhesion protein found on epithelial cells, is often used as apositive selection marker for CTCs (48). The most commonnegative selection marker is CD45 because it is found only on

cells of hematopoietic origin. First reported in 2000, the ISET(isolation by size of epithelial tumor cells) system is based oncell size filtration and has been shown to effectively recoverCTCs for diagnostic cytopathologic analysis (49). However,smaller CTCs less than 8 mm in diameter can be lost using thismethod, due to the selective pore size.Microfluidic devices offerseveral attractive advantages for CTC-based studies, such ascontinuous sample processing using a small volume to reduceloss and integration of downstream analyses on the chip itself.These devices can also reduce hands-on time and increaseconsistency of heterogeneous CTC profiles (50). Some micro-fluidic devices integrate existing CTC isolation techniqueswithin their design, such as the size-dictated immunocapturechip (SDI-Chip; ref. 51). The SDI-chip is composed of twomirrored anti-EpCAMantibody-coatedmicropillar arrays thatselectively enhance the interaction of CTCs with the selectionantibodies, resulting in efficient capture of CTCs based on theprinciple of deterministic lateral displacement (52). The dis-advantage to the EpCAM-based capture is that not all CTCsexpress EpCAM (53). In an effort to move away from the biasintroduced by affinity-based isolation, researchers have begunto focus on label-free surface expression–independent

Figure 1.

A graphical representation of a variety of approaches ovarian cancer researchers can take to evaluate blood-based liquid biopsies for cancer-associated biomarkersto aid in early detection of malignancy.

Reducing Ovarian Cancer Mortality Through Early Detection

AACRJournals.org Cancer Prev Res; 13(3) March 2020 243

Research. on May 26, 2020. © 2020 American Association for Cancercancerpreventionresearch.aacrjournals.org Downloaded from

Page 4: Reducing Ovarian Cancer Mortality Through Early Detection ...CANCER PREVENTION RESEARCH | MINIREVIEW Reducing Ovarian Cancer Mortality Through Early ... standard in the initial diagnosis

microfluidic devices to isolate CTCs. A label-free approach isoptimal because CTCs are very heterogeneous and fluid in theirgene expression, making any affinity-based approach biasedtoward cells expressing a certainmarker. Several label-freeCTCseparation techniques have arisen in recent years includinga microfluidic flow fractionation approach, acoustic-basedseparation, and deterministic lateral displacement separa-tion (52, 54, 55). The Labyrinth device developed by Lin andcolleagues in 2017 is a label-free circular microfluidic chamberthat uses a hydrodynamic maze to isolate CTCs by size (56).The device functions by flowing blood through a labyrinth(hence the name) of 56 sharp turns, which results in size-based

enrichment of CTCs depleted of hematopoietic cells. Improve-ments can be made on the purity of resultant CTCs by doubleLabyrinth separation. Labyrinth output can even be used withvarious downstream single-cell analysis tools including Bio-Mark Real-Time PCR System (Fluidigm), DEPArray (SiliconBiosystems), and droplet digital PCR (Rain-dance).CTC enumeration data can provide only limited insight into

the underlying biology of malignant disease. More recentresearch efforts have been focused on the molecular character-ization and functional analysis of CTCs. Using less than 1mL ofblood, Racila and colleagues demonstrated that 12 of 14 patientswith clinically organ-confined breast cancers and 3 of 3 patientswith organ-confined prostate cancers had excess epithelial cellsdetected in their blood (57). However, the diagnostic utility ofCTCs in ovarian cancer has been decidedly underwhelming. Inthe first studies led by us, CTCs (as circulating endothelial cells/circulating endothelial progenitors) were found to be rare inpatients with ovarian cancer and were not overly informativewhen evaluated along with activity of molecular targeted ther-apies (58, 59). In a 2018 study, CTCs were detected in only 5 outof 29 patients (17.2%) with proven primary ovarian cancer(Table 1). CTCs were shown to be more prevalent in patientswith metastases to the ovary, rather than in primary ovariancancer (43). Although CTCsmay not be useful as early detectionbiomarkers, CTC status in ovarian cancer is associated withoverall survival and progression-free survival according to arecent meta-analysis of 11 publications and 1,129 patients (60).

Circulating Tumor DNAIt was reported for the first time in 1977 by Leon and

colleagues that patients with cancer have increased levels of

Figure 2.

Examples of methods to capture circulating tumor cell from whole blood.

Table 1. Examples of liquid-based biopsy studies to detect ovarian cancer using ctDNA and CTCs.

Platformused

Selectionparameters Case size

Controlsize Sensitivitya Specificitya CI (95%) References

CTCs CellSearch� EpCAM Histologicdiagnosis ofovarian cancer

n ¼ 29 n ¼ 17a

Benigndisease

25.7% 100% Se ¼ 12.5% � 43.3%Sp ¼ 76.8% � 100%

43

Cancer withmetastasesto the ovary

n ¼ 5

CirculatingtumorDNA(ctDNA)

Microarray(MethDet56)

Promoters:RASSF1A,CALCA,EP300

Benign disease n ¼ 30 n ¼ 30 90% 86.7% Se ¼ 80% � 100%Sp ¼ 66.7% � 96.7%

75Ovarian cancer n ¼ 30

Microfluidicdigital PCR

TP53-mutantallelefraction

Ovarian n ¼ 32 n ¼ 5 71% 88% Se ¼ 42% � 92%Sp ¼ 64% � 99%

71Primary peritoneal n ¼ 5Fallopian tube n ¼ 3

Ultra-highcoveragebi-sulfitesequencing

Unspecified3 DNAmemarkerpanel

Ovarian cancer n ¼ 41 n ¼ 21 41.4% 90.7% Se ¼ 24.1% � 60.9%Sp ¼ 84.3% � 94.8%

76Other cancer n ¼ 37Nonepithelial tumors n ¼ 5Borderline n ¼ 27Benign tumors n ¼ 119

Note: Pairs of sensitivity and specificity in this table are not directly comparable with each other because they are calculated on the basis of different cutoffsthroughout.aSensitivity (Se) and specificity (Sp) pair values listed correspond to a specific decision cutoff utilized in each study.

Trinidad et al.

Cancer Prev Res; 13(3) March 2020 CANCER PREVENTION RESEARCH244

Research. on May 26, 2020. © 2020 American Association for Cancercancerpreventionresearch.aacrjournals.org Downloaded from

Page 5: Reducing Ovarian Cancer Mortality Through Early Detection ...CANCER PREVENTION RESEARCH | MINIREVIEW Reducing Ovarian Cancer Mortality Through Early ... standard in the initial diagnosis

free DNA in their blood serum (61). Cell-free DNA broadlyrefers to all circulating DNA, whereas ctDNA is specificallytumor-derived as the name suggests. Unfortunately, typicalDNA isolation methods do not distinguish DNA by its origin,so scientists have developed several strategies to increase theproportion of ctDNA isolated from serum or plasma. Enrich-ment for ctDNA is optimal because it increases the likelihood ofuncovering biomarkers specific to the cellular heterogeneity ofthe tumor. Apoptotic DNA fragmentation is thought to be themain source of ctDNA due to its average size of 70 to 200 basepairs, which corresponds to the length of DNA around anucleosome (62). The amount of ctDNA can be highly variable.ctDNA can comprise approximately 0.01% � 90% of totalcirculating DNA (63–65). Even so, ctDNA has been used todetect tumor-specific mutations, loss of heterozygosity, DNAintegrity, microsatellite alterations, and epigenetic altera-tions (62, 66). Importantly, ctDNA can be used to quantifythe level of disease burden and reveal the genomic landscape ofthe tumor. Researchers have estimated that the half-life ofctDNA ranges from 16 minutes to 2.5 hours, based on thedegradation kinetics of DNA in circulation (67–69). Thismakes ctDNA a temporal snapshot of tumor status. Even withthis short half-life, promising results have been shown incolorectal cancer, in which >60% of early-stage tumors canbe detected through sensitive mutational analysis of DNAfragments in plasma (64). Figure 3 depicts typical analysismethods used to evaluate ctDNA and its required sensitivity aswell as various applications.As we move toward the future, more and more advance-

ments are taking place to increase the sensitivity of currentDNA-based analysis methods such as real-time PCR, micro-

fluidic digital-drop PCR, BEAMing, Safe-SeqS, CAPP-Seq, andother next-generation sequencing techniques (70). In a recentretrospective study, researchers determined that detection ofmutations in TP53 using high-grade serous ovarian carcinomapatient plasma has the potential to assess clinical prognosis andresponse. This study also demonstrated that the amount ofctDNA correlated with tumor volume as determined by three-dimensional (3D) volume reconstruction fromCT images (71).Overall, all patients with cancer evaluated in this study with atumor volume greater than 20 cm3 had reliably detectableTP53mutations; however, the role of ctDNAmutation screening forearly detection remains unproven. Using a mathematical mod-el, researchers postulate that tumors in themillimeter diameterrange can only be detected using secreted blood biomarkersunder ideal conditions of extremely high rates of biomarkersecretion and essentially zero background from healthycells (72, 73). Thus, very sensitive technology is necessary todetect ctDNA from early-stage ovarian cancer.Epigenetic changes can also be reliably detected in ctDNA.

Aberrant methylation in circulating DNA has been shown inmany cancer types (74). Cancer cells use promotermethylationto deregulate gene expression and can be informative as anoninvasive biomarker. The hypermethylation of importantDNA repair genes is often an early step in carcinogenesis. In2011, Liggett and colleagues determined that themethylation ofthree promoters differentiated patients with ovarian cancerfrom healthy controls with a sensitivity of 90% and a specificityof 86.7% (75). These methylated genes, RASSF1A, CALCA,and EP300, are associated with tumor suppression, calciumregulation, and histone acetylation, respectively. In addition,promoter methylation of RASSF1A and PGR-PROX was

Figure 3.

Sensitivity, methods, and applications for circulatingtumor DNA analysis. Overview of the most commonapplications and techniques for ctDNA analysis. ctDNArepresented by red DNA strands in blood collectiontubes (adapted from ref. 102).

Reducing Ovarian Cancer Mortality Through Early Detection

AACRJournals.org Cancer Prev Res; 13(3) March 2020 245

Research. on May 26, 2020. © 2020 American Association for Cancercancerpreventionresearch.aacrjournals.org Downloaded from

Page 6: Reducing Ovarian Cancer Mortality Through Early Detection ...CANCER PREVENTION RESEARCH | MINIREVIEW Reducing Ovarian Cancer Mortality Through Early ... standard in the initial diagnosis

informative for distinguishing ovarian cancer from benignovary disease (sensitivity 80%, specificity 73.3%; ref. 75). Meth-ylation patterns found in ctDNAhave even shown the potentialto diagnose a subset of ovarian cancers up to 2 years in advanceof elevated CA-125 levels (Table 1; ref. 76).It is thought that solid malignant neoplasms shed DNA into

the circulatory system most often by necrosis, rather thanapoptosis as in normal tissue (77). Necrosis typically resultsin larger and less uniform DNA fragments than apoptosis.Because of this, DNA integrity is increased in cancer and can beanalyzed using ctDNA. Using the most common mobile ele-ment in the human genome, Alu repeats, researchers can derivea DNA integrity index that can differentiate patients withcancer from healthy individuals by isolating circulating DNAfrom plasma. This approach was used byWang and colleaguesto establish a cutoff index value of 0.59, meaning all samplesabove this index are likely malignant (77). This study dem-onstrated inadequacies in sensitivity of the assay (62%);however, specificity was 100% (Table 1). DNA integrityanalysis represents a less-expensive and simpler alternativeto DNA sequencing.

EVs in Ovarian CancerExtracellular vesicles compared with CTCs and cfDNA are

considered a relatively new class of cancer biomarker. EVs aresecreted by cells as a form of intercellular communication.These EVs can shuttle nucleic acids, lipids, and proteins fromtheir cell of origin to surrounding cells to regulate the functionof other cells (78). EVs are classified according to size (from afew nanometers to a few micrometers) and subcellular ori-gin (79). A subtype of EVs, termed exosomes, is endocytic inorigin and includes 60- to 80-nm small exosomes (Exo-S), aswell as 90- to 120-nm large exosomes (Exo-L; ref. 80). Ingeneral, the term “exosomes” is broadly used to refer to aheterogenous mixture of small EVs (sEVs) that are less than200 nm in size; this is due to the fact that widely usedpurification methods (such as differential ultracentrifugation)cannot definitively isolate EV class based on subcellularorigin (81).The role of exosomes in cancer metastasis has been dem-

onstrated in various cancer types (82–85), establishing a ratio-nale for exploring exosome-based diagnostics. In the ovariancancer context, our laboratory has shown that tumor-derivedexosomes from CP30 and CP70, which are both platinum-resistant ovarian cancer cell lines, imparted platinum resistanceto a platinum-sensitive cell line (A2780; ref. 86). In anotherin vitro study, EOC cells were shown to secrete exosomes thataffected the phenotype of mesothelial cells of the peritoneumvia transfer of CD44, a cell surface glycoprotein that plays a rolein cell adhesion and migration. The mesothelial cells, in turn,increased CD44 surface and MMP-9 secretion, resulting in thedegradation of the extracellular matrix and promoting ovariancell invasion (87). Collectively, these studies suggest that exo-somes represent a general mechanism by which one cell type

canmodulate the phenotype and characteristics of neighboringcells.Ovarian tumor–derived EVs are not only rich in proteins

but these contain miRNAs. miRNAs are small noncodingRNAs that can target messenger RNA thereby altering geneexpression. miRNA profiling of EpCAM-captured exosomesfrom ovarian cancer patients’ serum identified miR-21,miR-141, miR200a, miR200b, miR-200c, miR-203, miR-205, and miR-214 to be elevated compared with controls.A high expression of let-7 has been correlated to ovariancancer cell invasiveness in SKOV3 (88). It has been dem-onstrated that miR-1246 expression in ovarian tumor–derived exosomes can aid tumor growth by downregulatingCav1 expression, thereby increasing P-glycoprotein expres-sion in infiltrating pro-tumorigenic immune cells (M2-typemacrophages; ref. 89). This makes the tumor microenviron-ment favorable because this confers drug resistance to pac-litaxel (89). In addition, other studies have shown thatmiRNAs from ovarian cancer tumor–derived EVs can conferdrug resistance through other pathways (82, 90–92).Because there are various evidence of tumor-derived EVs

contributing to the progression of ovarian cancer (87, 93, 94),it is logical to assess the potential of EVs for diagnostics.Recently, several technologies have been developed to cap-ture exosomes from minute volumes of starting material tosurpass the problem of requiring larger sample volumeswhen using conventional methods such as ELISA and immu-noblotting (Table 2). Technologies such as microfluidicchips and the nano-plasmonic exosome (nPLEX) platformcan be conjugated to antibodies to allow for capture andprofiling of exosomes. One study has assessed exosomesderived from ascites of patients with ovarian cancer usingthe nPLEX platform (95). Initially, molecular profiling ofantibodies was used to coat the surface of nanoholes usingselected ovarian cancer markers based from literature. Ovar-ian cell lines and exosomes derived from the same cell lineswere then profiled. The authors found that the combinationof EpCAM and CD24 can distinguish ovarian tumor–derivedexosomes with a detection accuracy of 97%. Furthermore, itwas found that these two markers are highly expressed in theascites of patients with ovarian cancer (n ¼ 20) comparedwith the control population (n ¼ 10), which consisted ofascites from nonmalignant conditions (95). These findingscorrespond with a previous report that identified thatEpCAM and CD24 are found in exosomes of cultured celllines and malignant ascites (96). Our laboratory was the firstto demonstrate the capture of exosomes directly from theplasma of patients with ovarian cancer using a microfluidicchip (97). This was followed by the development of amicrofluidic chip called ExoSearch (Fig. 4) by Zhao andcolleagues (2016), wherein exosomes from plasma of healthycontrols and patients with ovarian cancer were capturedusing anti-CA-125, anti-EpCAM (Epithelial Cell AdhesionMolecule), and anti-CD24 (cluster of differentiation 24 orheat stable antigen CD24; ref. 98). In this study, it was

Trinidad et al.

Cancer Prev Res; 13(3) March 2020 CANCER PREVENTION RESEARCH246

Research. on May 26, 2020. © 2020 American Association for Cancercancerpreventionresearch.aacrjournals.org Downloaded from

Page 7: Reducing Ovarian Cancer Mortality Through Early Detection ...CANCER PREVENTION RESEARCH | MINIREVIEW Reducing Ovarian Cancer Mortality Through Early ... standard in the initial diagnosis

observed that exosomal CA125 and EpCAM may be used todiscriminate patients with ovarian cancer from the healthycontrols (98). Most recently, our group together with Zhangand colleagues (2019) has reported the development of anultrasensitive analysis of exosomes using a 3D nanostructuredherringbone chip (nano-HB; ref. 99). In this small case–controlstudy, we report that exo-folate receptor alpha as well as exo-EpCAM and exo-CD24 could be detected in small volumes ofplasma samples (as little as 2 mL) and could significantlydistinguish patients with ovarian cancer from cancer-free con-trols (area under the curve¼ 1; Fig. 5; ref. 99).While these typesof devices have yet to be assessed under the rigor of clinical trialsor in longitudinal samples from asymptotic patients who laterdevelop cancer, these devices clearly hold promise.

ConclusionLiquid-based approaches to detect cancer, although gaining

traction, are not new. Complete blood cell counts, includingwhite blood cells, red blood cells, and platelets, are used to helpdiagnose blood disorders, including leukemia and lymphoma.Prostate-specific antigen is used to help diagnosis prostatecancer, whereas calcitonin, a-fetoprotein, and human chori-onic gonadotropin are used to diagnosis medullary thyroidcancer, liver cancer, and germ cell tumors, such as testicularcancer and ovarian cancer, respectively. Liquid-based biopsiesare likely here to stay and as technologies improve so will theirclinical utility. These types of assays, which are considerablyless invasive when compared with the tissue biopsy procedure,will help further advance personalized targeted therapy and

Table 2. Comparison of studies using circulating EVs to detect ovarian cancer.

Platform used Selection parameters Case size Control size AUC Sensitivitya Specificitya References

nPLEX EpCAM 20 10 0.968 90%b 100%b 95CD24 0.900 80%b 100%b

EpCAM þ CD24 0.995 95%b 100%b

CD63 0.670 75%b 60%b

Microfluidic chip (ExoSearch) EpCAM 15 5 1.00 93%a 100%a 98CA-125 1.00 93%a 100%a

CD24 0.91 93%a 80%a

Microfluidic chip (nanoHB) EpCAM 20 10 1.00 100%a 100%a 99CD24 1.00 100%a 100%a

FR-a 0.995 95%a 100%a

NTA 1.00 69%a 86%a

Note: Pairs of sensitivity and specificity in this table are not directly comparable with each other because they are calculated on the basis of different cutoffsthroughout.aSensitivity and specificity pair values listed correspond to a specific decision cutoff utilized in each study. The sensitivity and specificity correspond to themaximizedYouden index.bAs reported by each study.

Figure 4.

A, Schematic of the ExoSearch chip. Bright-field microscopy images of Y-shaped injector (B), mixer (C), immunomagnetic bead (D), and TEM of exosomes (E)captured on the surface of the immunomagnetic bead (reprinted with permission from ref. 98).

Reducing Ovarian Cancer Mortality Through Early Detection

AACRJournals.org Cancer Prev Res; 13(3) March 2020 247

Research. on May 26, 2020. © 2020 American Association for Cancercancerpreventionresearch.aacrjournals.org Downloaded from

Page 8: Reducing Ovarian Cancer Mortality Through Early Detection ...CANCER PREVENTION RESEARCH | MINIREVIEW Reducing Ovarian Cancer Mortality Through Early ... standard in the initial diagnosis

immunotherapy by offering a source of easily obtainablematerial for mutation analysis. Liquid biopsies also have thepotential to be useful in efficacy assessment, especially ifimaging cannot be used or the interpretation is problematic,

and in real-time monitoring of molecular profiles and clonalevolution in patients undergoing cancer therapy to detectmetastatic relapse or metastatic progression as well as mechan-isms of resistance.

Figure 5.

A, Schematic of the 3D nanostructured herringbone chip (nano-HB), method of nano-HB chip fabrication (B), a nano-HB chip with magnified SEM images in themiddle (C), and testing clinical samples on the nano-HB chip (D, left). CD24, EpCAM, and FRa on exosomes derived from the plasma of patients with ovarian cancer(n¼ 20) and control population (n¼ 10) were quantified. Error bars, SD (n¼ 3). E, Plot of the exosomal protein concentration captured on the nano-HB chip versusovarian cancer staging, with stage I/II considered as early and stage III/IV as advanced (reprintedwith permission from ref. 99). BKG, Background; CSA, colloidal self-assembly; SEM, scanning electron microscopy.

Trinidad et al.

Cancer Prev Res; 13(3) March 2020 CANCER PREVENTION RESEARCH248

Research. on May 26, 2020. © 2020 American Association for Cancercancerpreventionresearch.aacrjournals.org Downloaded from

Page 9: Reducing Ovarian Cancer Mortality Through Early Detection ...CANCER PREVENTION RESEARCH | MINIREVIEW Reducing Ovarian Cancer Mortality Through Early ... standard in the initial diagnosis

Even with the growing excitement surrounding this area oftranslational research, significant improvements in liquidbiopsy platforms and techniques will be required to advancethe promises of precision cancer medicine. At present, aliquid biopsy test in itself cannot simply replace the goldstandard tissue biopsy–based test; they primarily serve as acomplimentary test. As such, there are several remainingchallenges impeding the wider adoption of liquid biopsy–based clinical tests. First, it is important to determine thelevel of sensitivity of a given liquid-based assay. It is wellestablished that CTCs, ctDNA, and tumor-derived EVs arerelatively rare compared with the number of “other mole-cules” found in a blood or bodily fluid sample; therefore,major hurdles remain to improve the test's detection ability.Furthermore, it is not clear if a given test can accuratelydissect the heterogeneity of a tumor and identify the “badactors” among the other tumor subclones. Second, themajority of liquid biopsy assays lack extensive clinical val-idation resulting in limited reimbursements and utilizationof these next-generation tests within the medical commu-nity (100). It is, therefore, essential to rigorously validate anddemonstrate to both providers and payers the value of liquidbiopsies in the clinical setting before they impact clinicalpractice and are routinely reimbursed. As we move towardthe future, the technologies supporting the development ofliquid biopsy assays will continue to improve and in turn thesensitivity and specificity of the test. Currently, strides arebeing made in improving single CTC capture approachesand platforms. With the adoption of digital drop PCR,researchers are able to increase the sensitivity of detectionof low-abundance mutations in the ctDNA (101). Micro-fluidic technologies are also proving suitable for isolation oflow-abundance cancer-associated EVs/exosomes (99).In summary, developing effective screening tests for early

detection of ovarian cancer remains one of the most significant

unmet needs in the diagnosis and treatment of this disease. It isvital to identify new liquid-based tests and to develop aneffective strategy to identify ovarian cancer at its earliest stagesso that women stand the best chance for successful treatmentand improved survival. To implement a population-wideovarian cancer screening strategy, there must be evidence thatthe test is both sensitive enough to detect the cancer early andspecific enough not to cause harm to healthy people. It is clearthat when cancer, any cancer, is detected late, that is, after it hasmetastasized to other parts of the body, the outcomes aregenerally grim. These new liquid-based platforms representpotentially valuable clinical tools to evaluate various circulatingbiomarkers at a specificity and sensitivity necessary to detectovarian cancer while it is still localized. In the next decade,liquid-based biopsies will become important clinical tools forcancer screening, diagnosis, prognosis evaluation, drugresponse predictions, and disease monitoring. However, theanalytical and clinical utility of such tools must be rigorouslyestablished before this can be applied. Successful “bench tobedside” transition will require extensive clinical validation,which demonstrates a significant decrease in the mortality ofwomen diagnosed with ovarian cancer.

Disclosure of Potential Conflicts of InterestNo potential conflicts of interest were disclosed.

AcknowledgmentsThis report was supported in part by The Kansas Institute

for Precision Medicine (GM130423, to A.K. Godwin), a graduatestudent award from the OVERRUN Ovarian Cancer Foundation (toC. Trinidad) and the Honorable Tina Brozman Foundation, Inc. (Tina'swish to A.K. Godwin). A.K. Godwin is the Chancellors DistinguishedChair in Biomedical Sciences.

Received April 4, 2019; revised June 20, 2019; accepted August 15, 2019;published first March 4, 2020.

References1. Cho KR, Shih Ie M. Ovarian cancer.Annu Rev Pathol 2009;4:

287–313.2. KurmanRJ, Shih IeM.The dualisticmodel of ovarian carcinogenesis:

revisited, revised, and expanded. Am J Pathol 2016;186:733–47.3. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J

Clin 2018;68:7–30.4. Sahdev A, Hughes JH, Barwick T, Rockall AG, Gallagher CJ, Reznek

RH. Computed tomography features of recurrent ovarian carcinomaaccording to time to relapse. Acta Radiol 2007;48:1038–44.

5. Hirose S, Tanabe H, Nagayoshi Y, Hirata Y, Narui C, Ochiai K, et al.Retrospective analysis of sites of recurrence in stage I epithelialovarian cancer. J Gynecol Oncol 2018;29:e37.

6. GuthU, ArndtV, Stadlmann S,HuangDJ, SingerG. Epidemiology inovarian carcinoma: lessons from autopsy. Gynecol Oncol 2015;138:417–20.

7. Kim J, Park EY, Kim O, Schilder JM, Coffey DM, Cho CH, et al. Cellorigins of high-grade serous ovarian cancer. Cancers 2018;10(11):pii:E433.

8. Giornelli GH. Management of relapsed ovarian cancer: a review.Springerplus 2016;5:1197.

9. Scholler N, UrbanN. CA125 in ovarian cancer. BiomarkMed 2007;1:513–23.

10. Sevinc A, Adli M, Kalender ME, Camci C. Benign causes ofincreased serum CA-125 concentration. Lancet Oncol 2007;8:1054–5.

11. Lu KH, Skates S, Hernandez MA, Bedi D, Bevers T, Leeds L, et al. A2-stage ovarian cancer screening strategy using the Risk of OvarianCancer Algorithm (ROCA) identifies early-stage incident cancersand demonstrates high positive predictive value. Cancer 2013;119:3454–61.

12. Jelovac D, Armstrong DK. Recent progress in the diagnosis andtreatment of ovarian cancer. CA Cancer J Clin 2011;61:183–203.

13. Rein BJ, Gupta S, Dada R, Safi J, Michener C, Agarwal A. Potentialmarkers for detection and monitoring of ovarian cancer. J Oncol2011;2011:475983.

14. Urban N, Drescher C. Potential and limitations in early diagnosis ofovarian cancer. Adv Exp Med Biol 2008;6222008:3–14.

15. Bast RC Jr, Brewer M, Zou C, Hernandez MA, Daley M, Ozols R,et al. Prevention and early detection of ovarian cancer: missionimpossible? Recent Results Cancer Res 2007;174:91–100.

Reducing Ovarian Cancer Mortality Through Early Detection

AACRJournals.org Cancer Prev Res; 13(3) March 2020 249

Research. on May 26, 2020. © 2020 American Association for Cancercancerpreventionresearch.aacrjournals.org Downloaded from

Page 10: Reducing Ovarian Cancer Mortality Through Early Detection ...CANCER PREVENTION RESEARCH | MINIREVIEW Reducing Ovarian Cancer Mortality Through Early ... standard in the initial diagnosis

16. Cohen JG, White M, Cruz A, Farias-Eisner R. In 2014, can we dobetter than CA125 in the early detection of ovarian cancer? World JBiol Chem 2014;5:286–300.

17. Jacobs I, Bast RC Jr. The CA 125 tumour-associated antigen: a reviewof the literature. Hum Reprod 1989;4:1–12.

18. Jacobs I, Oram D. Prevention of ovarian cancer: a survey of thepractice of prophylactic oophorectomy by fellows and members ofthe Royal College of Obstetricians and Gynaecologists. Br J ObstetGynaecol 1989;96:510–5.

19. Skates SJ, Xu FJ, YuYH, Sjovall K, EinhornN, ChangY, et al. Towardan optimal algorithm for ovarian cancer screening with longitudinaltumor markers. Cancer 1995;76:2004–10.

20. Pinsky PF, Zhu C, Skates SJ, Black A, Partridge E, Buys SS, et al.Potential effect of the risk of ovarian cancer algorithm (ROCA) on themortality outcome of the Prostate, Lung, Colorectal and Ovarian(PLCO) trial. Int J Cancer 2013;132:2127–33.

21. Menon U, Ryan A, Kalsi J, Gentry-Maharaj A, Dawnay A, Habib M,et al. Risk algorithm using serial biomarker measurements doublesthe number of screen-detected cancers compared with a single-threshold rule in the United KingdomCollaborative Trial of OvarianCancer Screening. J Clin Oncol 2015;33:2062–71.

22. Hogg R, FriedlanderM. Biology of epithelial ovarian cancer: implica-tions for screening women at high genetic risk. J Clin Oncol 2004;22:1315–27.

23. Chan A, Gilks B, Kwon J, Tinker AV. New insights into thepathogenesis of ovarian carcinoma: time to rethink ovarian cancerscreening. Obstet Gynecol 2012;120:935–40.

24. Buys SS, Partridge E, BlackA, JohnsonCC, Lamerato L, IsaacsC, et al.Effect of screening on ovarian cancer mortality: the Prostate, Lung,Colorectal and Ovarian (PLCO) Cancer Screening RandomizedControlled Trial. JAMA 2011;305:2295–303.

25. Menon U. Ovarian cancer screening has no effect on disease-specificmortality. Evid Based Med 2012;17:47–8.

26. Henderson JT, Webber EM, Sawaya GF. Screening for ovariancancer: updated evidence report and systematic review for the USPreventive Services Task Force. JAMA 2018;319:595–606.

27. Kobayashi H, Yamada Y, Sado T, SakataM, Yoshida S, Kawaguchi R,et al. A randomized study of screening for ovarian cancer: amulticenter study in Japan. Int J Gynecol Cancer 2008;18:414–20.

28. Jacobs IJ, Menon U, Ryan A, Gentry-Maharaj A, Burnell M, Kalsi JK,et al. Ovarian cancer screening andmortality in theUKCollaborativeTrial of Ovarian Cancer Screening (UKCTOCS): a randomisedcontrolled trial. Lancet 2016;387:945–56.

29. Ashworth T. A case of cancer in which cells similar to those in thetumours were seen in the blood after death. AustralasianMed J 1869;14:146–7.

30. Miller MC, Doyle GV, Terstappen LW. Significance of circulatingtumor cells detected by the cell search system in patients withmetastatic breast colorectal and prostate cancer. J Oncol 2010;2010:617421.

31. Thierry AR, El Messaoudi S, Gahan PB, Anker P, StrounM. Origins,structures, and functions of circulating DNA in oncology. CancerMetastasis Rev 2016;35:347–76.

32. Jahr S, Hentze H, Englisch S, Hardt D, Fackelmayer FO, Hesch RD,et al. DNA fragments in the blood plasma of cancer patients:quantitations and evidence for their origin from apoptotic andnecrotic cells. Cancer Res 2001;61:1659–65.

33. Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, micro-vesicles, and friends. J Cell Biol 2013;200:373–83.

34. Simons M, Raposo G. Exosomes–vesicular carriers for intercellularcommunication. Curr Opin Cell Biol 2009;21:575–81.

35. Wubbolts R, Leckie RS, Veenhuizen PT, SchwarzmannG,MobiusW,Hoernschemeyer J, et al. Proteomic and biochemical analyses ofhuman B cell-derived exosomes. Potential implications for their

function and multivesicular body formation. J Biol Chem 2003;278:10963–72.

36. Hemler ME. Targeting of tetraspanin proteins—potential benefitsand strategies. Nat Rev Drug Discov 2008;7:747–58.

37. Hemler ME. Tetraspanin proteins mediate cellular penetration,invasion, and fusion events and define a novel type of membranemicrodomain. Annu Rev Cell Dev Biol 2003;19:397–422.

38. Th�ery C, Regnault A, Garin J, Wolfers J, Zitvogel L, Ricciardi-Castagnoli P, et al. Molecular characterization of dendritic cell-derived exosomes. J Cell Biol 1999;147:599–610.

39. Meropol NJ. The significance of circulating tumor cells as prognosticmarkers for colon cancer. Clin Adv Hematol Oncol 2009;7:247–8.

40. Dotan E, Cohen SJ, Alpaugh KR, Meropol NJ. Circulating tumorcells: evolving evidence and future challenges. Oncologist 2009;14:1070–82.

41. Cohen SJ, Punt CJ, Iannotti N, Saidman BH, Sabbath KD, GabrailNY, et al. Prognostic significance of circulating tumor cells inpatients with metastatic colorectal cancer. Ann Oncol 2009;20:1223–9.

42. Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Matera J, Miller MC,et al. Circulating tumor cells, disease progression, and survival inmetastatic breast cancer. N Engl J Med 2004;351:781–91.

43. Lou E, Vogel RI, Teoh D, Hoostal S, Grad A, Gerber M, et al.Assessment of circulating tumor cells as a predictive biomarker ofhistology inwomenwith suspected ovarian cancer. LabMed 2018;49:134–9.

44. Cohen SJ, Alpaugh RK, Gross S, O’Hara SM, Smirnov DA, Terstap-pen LW, et al. Isolation and characterization of circulating tumorcells in patients with metastatic colorectal cancer. Clin ColorectalCancer 2006;6:125–32.

45. Cohen SJ, Punt CJ, Iannotti N, Saidman BH, Sabbath KD, GabrailNY, et al. Relationship of circulating tumor cells to tumor response,progression-free survival, and overall survival in patients with met-astatic colorectal cancer. J Clin Oncol 2008;26:3213–21.

46. Zhang J, Chen K, Fan ZH. Circulating tumor cell isolation andanalysis. Adv Clin Chem 2016;75:1–31.

47. Gwak H, Kim J, Kashefi-Kheyrabadi L, Kwak B, Hyun KA, Jung HI.Progress in circulating tumor cell research usingmicrofluidic devices.Micromachines 2018;9:pii: E353.

48. de Wit S, van Dalum G, Lenferink AT, Tibbe AG, Hiltermann TJ,Groen HJ, et al. The detection of EpCAM(þ) and EpCAM(�)circulating tumor cells. Sci Rep 2015;5:12270.

49. Vona G, Sabile A, Louha M, Sitruk V, Romana S, Sch€utze K, et al.Isolation by size of epithelial tumor cells. Am J Pathol 2000;156:57–63.

50. Bulfoni M, Turetta M, Del Ben F, Di Loreto C, Beltrami AP, CesselliD. Dissecting the heterogeneity of circulating tumor cells in meta-static breast cancer: going far beyond the needle in the haystack. Int JMol Sci 2016;17:pii: E1775.

51. Ahmed MG, Abate MF, Song Y, Zhu Z, Yan F, Xu Y, et al. Isolation,detection, and antigen-based profiling of circulating tumor cellsusing a size-dictated immunocapture chip. Angew Chem Int Ed2017;56:10681–5.

52. McGrath J, Jimenez M, Bridle H. Deterministic lateral displacementfor particle separation: a review. Lab Chip 2014;14:4139–58.

53. Pantel K, Speicher MR. The biology of circulating tumor cells.Oncogene 2016;35:1216–24.

54. Li P, Mao Z, Peng Z, Zhou L, Chen Y, Huang PH, et al. Acousticseparation of circulating tumor cells. Proc Natl Acad Sci U S A 2015;112:4970–5.

55. Hyun KA, Kwon K, Han H, Kim SI, Jung HI. Microfluidic flowfractionation device for label-free isolation of circulating tumor cells(CTCs) from breast cancer patients. Biosens Bioelectron 2013;40:206–12.

Trinidad et al.

Cancer Prev Res; 13(3) March 2020 CANCER PREVENTION RESEARCH250

Research. on May 26, 2020. © 2020 American Association for Cancercancerpreventionresearch.aacrjournals.org Downloaded from

Page 11: Reducing Ovarian Cancer Mortality Through Early Detection ...CANCER PREVENTION RESEARCH | MINIREVIEW Reducing Ovarian Cancer Mortality Through Early ... standard in the initial diagnosis

56. Lin E, Rivera-Baez L, Fouladdel S, YoonHJ, Guthrie S,Wieger J, et al.High-throughputmicrofluidic labyrinth for the label-free isolation ofcirculating tumor cells. Cell Syst 2017;5:295–304.

57. Racila E, Euhus D, Weiss AJ, Rao C, McConnell J, Terstappen LW,et al. Detection and characterization of carcinoma cells in the blood.Proc Natl Acad Sci U S A 1998;95:4589–94.

58. BehbakhtK, SillMW,DarcyKM, Rubin SC,Mannel RS,Waggoner S,et al. Phase II trial of the mTOR inhibitor, temsirolimus andevaluation of circulating tumor cells and tumor biomarkers inpersistent and recurrent epithelial ovarian and primary peritonealmalignancies: a Gynecologic Oncology Group study. Gynecol Oncol2011;123:19–26.

59. Schilder RJ, SillMW, LankesHA,GoldMA,Mannel RS,Modesitt SC,et al. A phase II evaluation ofmotesanib (AMG706) in the treatmentof persistent or recurrent ovarian, fallopian tube and primary peri-toneal carcinomas: a Gynecologic Oncology Group study. GynecolOncol 2013;129:86–91.

60. Zhou Y, Bian B, Yuan X, Xie G, Ma Y, Shen L. Prognostic value ofcirculating tumor cells in ovarian cancer: a meta-analysis. PLoS One2015;10:e0130873.

61. Leon SA, Shapiro B, Sklaroff DM, Yaros MJ. Free DNA in theserum of cancer patients and the effect of therapy. Cancer Res1977;37:646–50.

62. Schwarzenbach H, Hoon DS, Pantel K. Cell-free nucleic acids asbiomarkers in cancer patients. Nat Rev Cancer 2011;11:426–37.

63. Diaz LA Jr, Bardelli A. Liquid biopsies: genotyping circulating tumorDNA. J Clin Oncol 2014;32:579–86.

64. Diehl F, Li M, Dressman D, He Y, Shen D, Szabo S, et al.Detection and quantification of mutations in the plasma ofpatients with colorectal tumors. Proc Natl Acad Sci U S A2005;102:16368–73.

65. ForshewT,MurtazaM, ParkinsonC,GaleD, TsuiDW,Kaper F, et al.Noninvasive identification and monitoring of cancer mutations bytargeted deep sequencing of plasma DNA. Sci Transl Med 2012;4:136ra68.

66. Giannopoulou L, Kasimir-Bauer S, Lianidou ES. Liquid biopsy inovarian cancer: recent advances on circulating tumor cells andcirculating tumor DNA. Clin Chem Lab Med 2018;56:186–97.

67. Lo YM, Zhang J, Leung TN, Lau TK, Chang AM, Hjelm NM. Rapidclearance of fetal DNA from maternal plasma. Am J Hum Genet1999;64:218–24.

68. Yao W, Mei C, Nan X, Hui L. Evaluation and comparison of in vitrodegradation kinetics of DNA in serum, urine and saliva: a qualitativestudy. Gene 2016;590:142–8.

69. To EW, Chan KC, Leung SF, Chan LY, To KF, Chan AT, et al. Rapidclearance of plasma Epstein-Barr virus DNA after surgical treatmentof nasopharyngeal carcinoma. Clin Cancer Res 2003;9:3254–9.

70. ElazezyM, Joosse SA. Techniques of using circulating tumorDNA asa liquid biopsy component in cancer management. Comput StructBiotechnol J 2018;16:370–8.

71. Parkinson CA, Gale D, Piskorz AM, Biggs H, Hodgkin C, Addley H,et al. Exploratory analysis of TP53 mutations in circulating tumourDNA as biomarkers of treatment response for patients with relapsedhigh-grade serous ovarian carcinoma: a retrospective study. PLoSMed 2016;13:e1002198.

72. Brown PO, Palmer C. The preclinical natural history of serousovarian cancer: defining the target for early detection. PLoS Med2009;6:e1000114.

73. Lutz AM, Willmann JK, Cochran FV, Ray P, Gambhir SS. Cancerscreening: a mathematical model relating secreted blood biomarkerlevels to tumor sizes. PLoS Med 2008;5:e170.

74. WartonK, SamimiG.Methylation of cell-free circulatingDNA in thediagnosis of cancer. Front Mol Biosci 2015;2:13.

75. Liggett TE, Melnikov A, Yi Q, Replogle C, HuW, Rotmensch J, et al.Distinctive DNA methylation patterns of cell-free plasma DNA inwomen with malignant ovarian tumors. Gynecol Oncol 2011;120:113–20.

76. Widschwendter M, Zikan M, Wahl B, Lempiainen H, Paprotka T,Evans I, et al. The potential of circulating tumor DNA methylationanalysis for the early detection and management of ovarian cancer.Genome Med 2017;9:116.

77. Wang BG, Huang HY, Chen YC, Bristow RE, Kassauei K, Cheng CC,et al. Increased plasma DNA integrity in cancer patients. Cancer Res2003;63:3966–8.

78. Yanez-MoM, Siljander PR,AndreuZ, ZavecAB, Borras FE, Buzas EI,et al. Biological properties of extracellular vesicles and their phys-iological functions. J Extracell Vesicles 2015;4:27066.

79. Colombo M, Raposo G, Thery C. Biogenesis, secretion, and inter-cellular interactions of exosomes and other extracellular vesicles.Annu Rev Cell Dev Biol 2014;30:255–89.

80. Zijlstra A, Di Vizio D. Size matters in nanoscale communication.Nat Cell Biol 2018;20:228–30.

81. Tkach M, Thery C. Communication by extracellular vesicles: wherewe are and where we need to go. Cell 2016;164:1226–32.

82. Atay S, Banskota S, Crow J, Sethi G, Rink L, Godwin AK. OncogenicKIT-containing exosomes increase gastrointestinal stromal tumorcell invasion. Proc Natl Acad Sci U S A 2014;111:711–6.

83. Fu Q, Zhang Q, Lou Y, Yang J, Nie G, Chen Q, et al. Primary tumor-derived exosomes facilitate metastasis by regulating adhesion ofcirculating tumor cells via SMAD3 in liver cancer. Oncogene2018;37:6105–18.

84. HoshinoA,Costa-Silva B, ShenTL, RodriguesG,HashimotoA, TesicMark M, et al. Tumour exosome integrins determine organotropicmetastasis. Nature 2015;527:329–35.

85. Peinado H, Aleckovic M, Lavotshkin S, Matei I, Costa-Silva B,Moreno-Bueno G, et al. Melanoma exosomes educate bone marrowprogenitor cells toward a pro-metastatic phenotype through MET.Nat Med 2012;18:883–91.

86. Crow J, Atay S, Banskota S, Artale B, Schmitt S, Godwin AK.Exosomes as mediators of platinum resistance in ovarian cancer.Oncotarget 2017;8:11917–36.

87. Nakamura K, Sawada K, Kinose Y, Yoshimura A, Toda A, NakatsukaE, et al. Exosomes promote ovarian cancer cell invasion throughtransfer of CD44 to peritoneal mesothelial cells. Mol Cancer Res2017;15:78–92.

88. Kobayashi M, Salomon C, Tapia J, Illanes SE, Mitchell MD, Rice GE.Ovarian cancer cell invasiveness is associated with discordant exo-somal sequestration of Let-7 miRNA and miR-200. J Transl Med2014;12:4.

89. Kanlikilicer P, Bayraktar R, Denizli M, Rashed MH, Ivan C, Aslan B,et al. ExosomalmiRNA confers chemo resistance via targeting Cav1/p-gp/M2-type macrophage axis in ovarian cancer. EBioMedicine2018;38:100–12.

90. Pink RC, Samuel P, Massa D, Caley DP, Brooks SA, Carter DR.The passenger strand, miR-21-3p, plays a role in mediatingcisplatin resistance in ovarian cancer cells. Gynecol Oncol2015;137:143–51.

91. Weiner-Gorzel K, Dempsey E, Milewska M, McGoldrick A, TohV, Walsh A, et al. Overexpression of the microRNA miR-433promotes resistance to paclitaxel through the induction ofcellular senescence in ovarian cancer cells. Cancer Med 2015;4:745–58.

92. Au Yeung CL, Co NN, Tsuruga T, Yeung TL, Kwan SY, Leung CS,et al. Exosomal transfer of stroma-derived miR21 confers paclitaxelresistance in ovarian cancer cells through targeting APAF1.Nat Commun 2016;7:11150.

Reducing Ovarian Cancer Mortality Through Early Detection

AACRJournals.org Cancer Prev Res; 13(3) March 2020 251

Research. on May 26, 2020. © 2020 American Association for Cancercancerpreventionresearch.aacrjournals.org Downloaded from

Page 12: Reducing Ovarian Cancer Mortality Through Early Detection ...CANCER PREVENTION RESEARCH | MINIREVIEW Reducing Ovarian Cancer Mortality Through Early ... standard in the initial diagnosis

93. Cho JA, Park H, Lim EH, Kim KH, Choi JS, Lee JH, et al. Exosomesfrom ovarian cancer cells induce adipose tissue-derived mesenchy-mal stem cells to acquire the physical and functional characteristics oftumor-supporting myofibroblasts. Gynecol Oncol 2011;123:379–86.

94. Ying X, Wu Q, Wu X, Zhu Q, Wang X, Jiang L, et al. Epithelialovarian cancer-secreted exosomal miR-222–3p induces polarizationof tumor-associated macrophages. Oncotarget 2016;7:43076–87.

95. ImH, ShaoH, Park YI, Peterson VM, Castro CM,Weissleder R, et al.Label-free detection and molecular profiling of exosomes with anano-plasmonic sensor. Nat Biotechnol 2014;32:490–5.

96. Runz S, Keller S, Rupp C, Stoeck A, Issa Y, Koensgen D, et al.Malignant ascites-derived exosomes of ovarian carcinoma patientscontain CD24 and EpCAM. Gynecol Oncol 2007;107:563–71.

97. He M, Crow J, Roth M, Zeng Y, Godwin AK. Integrated immunoi-solation and protein analysis of circulating exosomes using micro-fluidic technology. Lab Chip 2014;14:3773–80.

98. Zhao Z, Yang Y, Zeng Y, He M. A microfluidic ExoSearch chip formultiplexed exosome detection towards blood-based ovarian cancerdiagnosis. Lab Chip 2016;16:489–96.

99. Zhang P, Zhou X, He M, Shang Y, Tetlow AL, Godwin AK, et al.Ultrasensitive detection of circulating exosomes with a3D-nanopatterned microfluidic chip. Nat Biomed Eng 2019;3:438–451.

100. Heitzer E, Haque IS, Roberts CES, Speicher MR. Current and futureperspectives of liquid biopsies in genomics-driven oncology. Nat RevGenet 2019;20:71–88.

101. Ramalingam N, Jeffrey SS. Future of liquid biopsies with growingtechnological and bioinformatics studies: opportunities and chal-lenges in discovering tumor heterogeneity with single-cell levelanalysis. Cancer J 2018;24:104–8.

102. Butler TM, Spellman PT, Gray J. Circulating-tumor DNA as an earlydetection and diagnostic tool. Curr Opin Genet Dev 2017;42:14–21.

Cancer Prev Res; 13(3) March 2020 CANCER PREVENTION RESEARCH252

Trinidad et al.

Research. on May 26, 2020. © 2020 American Association for Cancercancerpreventionresearch.aacrjournals.org Downloaded from

Page 13: Reducing Ovarian Cancer Mortality Through Early Detection ...CANCER PREVENTION RESEARCH | MINIREVIEW Reducing Ovarian Cancer Mortality Through Early ... standard in the initial diagnosis

2020;13:241-252. Cancer Prev Res   Camille V. Trinidad, Ashley L. Tetlow, Leonidas E. Bantis, et al.   Approaches Using Circulating BiomarkersReducing Ovarian Cancer Mortality Through Early Detection:

  Updated version

  http://cancerpreventionresearch.aacrjournals.org/content/13/3/241

Access the most recent version of this article at:

   

   

  Cited articles

  http://cancerpreventionresearch.aacrjournals.org/content/13/3/241.full#ref-list-1

This article cites 102 articles, 19 of which you can access for free at:

  Citing articles

  http://cancerpreventionresearch.aacrjournals.org/content/13/3/241.full#related-urls

This article has been cited by 1 HighWire-hosted articles. Access the articles at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected]

To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://cancerpreventionresearch.aacrjournals.org/content/13/3/241To request permission to re-use all or part of this article, use this link

Research. on May 26, 2020. © 2020 American Association for Cancercancerpreventionresearch.aacrjournals.org Downloaded from