Ascites analysis by a microfluidic chip allows tumor-cell ...€¦ · Ascites analysis by a...

23
Ascites analysis by a microuidic chip allows tumor-cell proling Vanessa M. Peterson a,b,1 , Cesar M. Castro a,c,1 , Jaehoon Chung a , Nathan C. Miller a,b , Adeeti V. Ullal a,b , Maria D. Castano a , Richard T. Penson c , Hakho Lee a , Michael J. Birrer c , and Ralph Weissleder a,c,d,2 a Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114; d Department of Systems Biology, Harvard Medical School, Boston, MA 02115; c Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114; and b Massachusetts Institute of Technology, Cambridge, MA 02139 Edited* by Patricia K. Donahoe, Massachusetts General Hospital, Boston, MA, and approved October 30, 2013 (received for review August 15, 2013) Ascites tumor cells (ATCs) represent a potentially valuable source of cells for monitoring treatment of ovarian cancer as it would obviate the need for more invasive surgical biopsies. The ability to perform longitudinal testing of ascites in a point-of-care setting could signicantly impact clinical trials, drug development, and clinical care. Here, we developed a microuidic chip platform to enrich ATCs from highly heterogeneous peritoneal uid and then perform molecular analyses on these cells. We evaluated 85 putative ovarian cancer protein markers and found that nearly two-thirds were either nonspecic for malignant disease or had low abundance. Using four of the most promising markers, we prospectively studied 47 patients (33 ovarian cancer and 14 control). We show that a marker set (ATC dx ) can sensitively and specically map ATC numbers and, through its reliable enrichment, facilitate additional treatment-response measurements related to prolifera- tion, protein translation, or pathway inhibition. biomarkers | molecular proling | personalized medicine O varian cancer is the deadliest of gynecologic cancers, with fewer than 50% of women surviving at 5 y following di- agnosis (1). Unfortunately, this statistic has changed little over the years, and most patients are still treated with a one-size-ts- all approach (2). Such a treatment strategy does not account for the broad genomic and proteomic diversity evident within ovarian tumors. Accurate measurement of protein markers will be critical in distinguishing effective from ineffective therapies. Despite the current push for biopsy-driven clinical trials, there are no minimally invasive tests or reliable biomarker panels ca- pable of identifying ovarian cancer treatment failures before radiographic evidence of progression. The reasons are several- fold, including heterogeneity of disease (3), variable expression levels of single biomarkers (4, 5), and markers that fail to dis- tinguish malignant from benign disease (6, 7). However, an expanding pipeline of targeted therapies and increased appre- ciation for the molecular drivers within ovarian cancers have spawned a number of novel approaches for detection and treatment monitoring; these approaches include primarily blood tests for circulating tumor cells (8), tumor-derived exosomes (9), stem/progenitor cells (10), and soluble tumor markers (11, 12), as well as the use of genomic (13, 14) or proteomic information (15). Lacking, however, are practical yet highly effective point- of-care (POC) platforms that can improve currently limited clinical practices (16). We hypothesized that peritoneal uid rather than blood might be a superior source of study material for liquid biopsyanalyses. Excess peritoneal uid accumulation (ascites) in ovarian can- cer is routinely drained (paracentesis) for symptomatic relief. Although often discarded, ascites could provide a source of abundant cellular material and potentially be preferable over blood samples. The precise cellular composition of ascites tends to vary across patients; the fraction of ascites tumor cells (ATCs) is generally believed to be <0.1% of harvested cells, with the remainder being host cells (37% lymphocytes, 29% mesothelial cells, and 32% macrophages) (17). The hurdle lies in reliably identifying and isolating ATCs from their highly heterogeneous environment. To help overcome these challenges, we developed a microuidic chip platform to enrich ATCs directly from ascites and then perform molecular analyses on these cells. We evalu- ated 85 putative ovarian cancer protein markers and found that a reduced marker set (ATC dx ) can map ATC numbers. This approach is poised to expand the utility of analyzing ATCs during cytotoxic and/or molecularly targeted therapy ovarian- cancer trials. Results Experimental Approach. Fig. 1 summarizes the experimental ap- proach. Surveying the literature (10, 18-26) and scientic data- bases (27), we tested putative diagnostic markers of ovarian cancer, mesothelial, and other host cells, as well as mechanistic markers of treatment response (19, 20, 28, 29). Initially, we tested over 100 commercially available antibodies against 85 biomarkers in 12 ovarian cancer cell lines (OV-90, OVCAR-3, SK-OV-3, ES-2, OVCA429, CaOV-3, UCI-101, UCI-107, UWB1.289, TOV-21G, TOV-112D, A2780), two mesothelial cell lines (LP9, LP3), two benign ovarian cell lines (TIOSE4, TIOSE6), and in lymphocytes and neutrophils (SI Appendix, Fig. S1). From these data, 31 markers were identied by ow cytometry and proled in a training set of human ascites col- lected under an Institutional Review Board (IRB)-approved protocol (Fig. 2). Based on these ndings, we then sought to Signicance Serial molecular analyses of tumor cells during treatment- and biopsy-driven clinical trials are emerging norms for many can- cers. Yet surgical and image-guided biopsies are expensive and invasive, explaining why alternative sources for tumor cells are being sought. In ovarian cancer (and other abdominopelvic cancers), abdominal uid buildup (ascites) occurs frequently. We demonstrate that ascites tumor cells (ATCs) present a valuable source of tumor cells, rendering ascites another form of liquid biopsy.We evaluated 85 ovarian cancer-related markers and developed a unique, low cost miniaturized microuidic ATC chip for on-chip enrichment and molecular proling using small amounts of ascites. This approach could expand the utility of ATCs within cytotoxic and/or molecularly targeted ovarian cancer therapeutic trials. Author contributions: V.M.P., C.M.C., J.C., R.T.P., H.L., M.J.B., and R.W. designed research; V.M.P., N.C.M., and M.D.C. performed research; J.C., A.V.U., and H.L. contributed new reagents/analytic tools; V.M.P., C.M.C., H.L., and R.W. analyzed data; and V.M.P., C.M.C., M.J.B., and R.W. wrote the paper. The authors declare no conict of interest. *This Direct Submission article had a prearranged editor. 1 V.M.P. and C.M.C. contributed equally to this work. 2 To whom correspondence should be addressed. E-mail: [email protected]. This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. 1073/pnas.1315370110/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1315370110 PNAS Early Edition | 1 of 9 MEDICAL SCIENCES PNAS PLUS

Transcript of Ascites analysis by a microfluidic chip allows tumor-cell ...€¦ · Ascites analysis by a...

Page 1: Ascites analysis by a microfluidic chip allows tumor-cell ...€¦ · Ascites analysis by a microfluidic chip allows tumor-cell profiling Vanessa M. Petersona,b,1, Cesar M. Castroa,c,1,

Ascites analysis by a microfluidic chip allowstumor-cell profilingVanessa M. Petersona,b,1, Cesar M. Castroa,c,1, Jaehoon Chunga, Nathan C. Millera,b, Adeeti V. Ullala,b,Maria D. Castanoa, Richard T. Pensonc, Hakho Leea, Michael J. Birrerc, and Ralph Weissledera,c,d,2

aCenter for Systems Biology, Massachusetts General Hospital, Boston, MA 02114; dDepartment of Systems Biology, Harvard Medical School, Boston, MA 02115;cMassachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114; and bMassachusetts Institute of Technology, Cambridge,MA 02139

Edited* by Patricia K. Donahoe, Massachusetts General Hospital, Boston, MA, and approved October 30, 2013 (received for review August 15, 2013)

Ascites tumor cells (ATCs) represent a potentially valuable sourceof cells for monitoring treatment of ovarian cancer as it wouldobviate the need for more invasive surgical biopsies. The ability toperform longitudinal testing of ascites in a point-of-care settingcould significantly impact clinical trials, drug development, andclinical care. Here, we developed a microfluidic chip platform toenrich ATCs from highly heterogeneous peritoneal fluid and thenperform molecular analyses on these cells. We evaluated 85putative ovarian cancer protein markers and found that nearlytwo-thirds were either nonspecific for malignant disease or hadlow abundance. Using four of the most promising markers, weprospectively studied 47 patients (33 ovarian cancer and 14 control).We show that a marker set (ATCdx) can sensitively and specificallymap ATC numbers and, through its reliable enrichment, facilitateadditional treatment-response measurements related to prolifera-tion, protein translation, or pathway inhibition.

biomarkers | molecular profiling | personalized medicine

Ovarian cancer is the deadliest of gynecologic cancers, withfewer than 50% of women surviving at 5 y following di-

agnosis (1). Unfortunately, this statistic has changed little overthe years, and most patients are still treated with a one-size-fits-all approach (2). Such a treatment strategy does not account forthe broad genomic and proteomic diversity evident withinovarian tumors. Accurate measurement of protein markers willbe critical in distinguishing effective from ineffective therapies.Despite the current push for biopsy-driven clinical trials, thereare no minimally invasive tests or reliable biomarker panels ca-pable of identifying ovarian cancer treatment failures beforeradiographic evidence of progression. The reasons are several-fold, including heterogeneity of disease (3), variable expressionlevels of single biomarkers (4, 5), and markers that fail to dis-tinguish malignant from benign disease (6, 7). However, anexpanding pipeline of targeted therapies and increased appre-ciation for the molecular drivers within ovarian cancers havespawned a number of novel approaches for detection andtreatment monitoring; these approaches include primarily bloodtests for circulating tumor cells (8), tumor-derived exosomes (9),stem/progenitor cells (10), and soluble tumor markers (11, 12),as well as the use of genomic (13, 14) or proteomic information(15). Lacking, however, are practical yet highly effective point-of-care (POC) platforms that can improve currently limitedclinical practices (16). We hypothesized that peritoneal fluidrather than blood might be a superior source of study materialfor “liquid biopsy” analyses.Excess peritoneal fluid accumulation (ascites) in ovarian can-

cer is routinely drained (paracentesis) for symptomatic relief.Although often discarded, ascites could provide a source ofabundant cellular material and potentially be preferable overblood samples. The precise cellular composition of ascites tendsto vary across patients; the fraction of ascites tumor cells (ATCs)is generally believed to be <0.1% of harvested cells, with theremainder being host cells (37% lymphocytes, 29% mesothelial

cells, and 32% macrophages) (17). The hurdle lies in reliablyidentifying and isolating ATCs from their highly heterogeneousenvironment. To help overcome these challenges, we developeda microfluidic chip platform to enrich ATCs directly from ascitesand then perform molecular analyses on these cells. We evalu-ated 85 putative ovarian cancer protein markers and found thata reduced marker set (ATCdx) can map ATC numbers. Thisapproach is poised to expand the utility of analyzing ATCsduring cytotoxic and/or molecularly targeted therapy ovarian-cancer trials.

ResultsExperimental Approach. Fig. 1 summarizes the experimental ap-proach. Surveying the literature (10, 18-26) and scientific data-bases (27), we tested putative diagnostic markers of ovariancancer, mesothelial, and other host cells, as well as mechanisticmarkers of treatment response (19, 20, 28, 29). Initially, wetested over 100 commercially available antibodies against 85biomarkers in 12 ovarian cancer cell lines (OV-90, OVCAR-3,SK-OV-3, ES-2, OVCA429, CaOV-3, UCI-101, UCI-107,UWB1.289, TOV-21G, TOV-112D, A2780), two mesothelial celllines (LP9, LP3), two benign ovarian cell lines (TIOSE4,TIOSE6), and in lymphocytes and neutrophils (SI Appendix, Fig.S1). From these data, 31 markers were identified by flowcytometry and profiled in a training set of human ascites col-lected under an Institutional Review Board (IRB)-approvedprotocol (Fig. 2). Based on these findings, we then sought to

Significance

Serial molecular analyses of tumor cells during treatment- andbiopsy-driven clinical trials are emerging norms for many can-cers. Yet surgical and image-guided biopsies are expensive andinvasive, explaining why alternative sources for tumor cells arebeing sought. In ovarian cancer (and other abdominopelviccancers), abdominal fluid buildup (ascites) occurs frequently.We demonstrate that ascites tumor cells (ATCs) present avaluable source of tumor cells, rendering ascites another formof “liquid biopsy.” We evaluated 85 ovarian cancer-relatedmarkers and developed a unique, low cost miniaturizedmicrofluidic ATC chip for on-chip enrichment and molecularprofiling using small amounts of ascites. This approach couldexpand the utility of ATCs within cytotoxic and/or molecularlytargeted ovarian cancer therapeutic trials.

Author contributions: V.M.P., C.M.C., J.C., R.T.P., H.L., M.J.B., and R.W. designed research;V.M.P., N.C.M., and M.D.C. performed research; J.C., A.V.U., and H.L. contributed newreagents/analytic tools; V.M.P., C.M.C., H.L., and R.W. analyzed data; and V.M.P., C.M.C.,M.J.B., and R.W. wrote the paper.

The authors declare no conflict of interest.

*This Direct Submission article had a prearranged editor.1V.M.P. and C.M.C. contributed equally to this work.2To whom correspondence should be addressed. E-mail: [email protected].

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1315370110/-/DCSupplemental.

www.pnas.org/cgi/doi/10.1073/pnas.1315370110 PNAS Early Edition | 1 of 9

MED

ICALSC

IENCE

SPN

ASPL

US

Page 2: Ascites analysis by a microfluidic chip allows tumor-cell ...€¦ · Ascites analysis by a microfluidic chip allows tumor-cell profiling Vanessa M. Petersona,b,1, Cesar M. Castroa,c,1,

establish a reliable and manageable protein-marker panel thatcould be adapted to microfluidic point-of-care testing. Alto-gether, we accrued a patient cohort involving 65 patients [n = 46OvCA (Table 1), n= 19 benign] across training (n= 18 samples)and testing (n = 47 samples) sets (Table 2 and Fig. 3). Controlsamples included ascites collected from patients with end-stageliver disease or advanced heart failure without known malig-nancy. Based on these profiling studies, we tested aliquots ofthese patient samples in the ATC chip (Fig. 4) via on-chipstaining (Fig. 5) or chip-based harvesting for subsequent mRNAanalysis (Fig. 6). In addition, serial samples (Fig. 7) wereobtained in a subset of patients during therapy (n = 7); thesetemporal samples were not included in the training or test por-tions of the study.

Protein Expression in a Training Set of Human Ascites Samples. Theclinical training set comprised 18 patients (13 ovarian cancer, 5nonovarian cancer). First, ascites samples were purified fromcluster of differentiation 45 (CD45)-positive cells using magneticseparation. Then, multicolor flow cytometry was performed todetermine marker expression levels in CD45-positive leukocytes,calretinin-positive mesothelial cells(18), and calretinin/CD45-negative cells (Fig. 2A). The latter were tested for multiple bio-markers (Fig. 2B) (see Methods for details). The clinical perfor-mance of each marker was determined by receiver operatingcharacteristic (ROC) analyses adapted to flow cytometry (30, 31)(SI Appendix, Fig. S4). The ROC curves were used to calculate

optimal cutoff values for individual markers and subsequently thesensitivity, specificity, and accuracy for each marker (30, 31).The markers unique to cancer cells with the highest sensitivity

were epithelial cell adhesion molecule (EpCAM), cluster ofdifferentiation 24 (CD24), and tumor-associated glycoprotein 72(TAG-72) (Table 3). Markers that were nonspecific and expressedin both cancer and mesothelial cells included vimentin, mucin 1(MUC1), CD44, cancer antigen 125 (CA-125), folate receptor 1(FOLR1), Wilms tumor protein (WT1), and epidermal growthfactor receptor (EGFR). Markers unique to mesothelial cells in-cluded D2-40 and thrombomodulin. Noteworthy was the lowsensitivity of FOLR1 (69.2%) and CA-125 (53.8%). FOLR1 hasbeen often cited in literature as a promising therapeutic target (32,33), and CA-125 is the most frequently used biomarker for ovar-ian-cancer monitoring (34). Also of note, the three ovarian-cancerpatients with the lowest EpCAM expression level had the highestvimentin levels, suggestive of epithelial-to-mesenchymal transition(EMT); their median survival was 5 mo (range 1–8 mo). It isthought that, during EMT, epithelial cancer cells undergo bio-chemical changes resulting in a mesenchymal cell phenotype thatenhances migratory capacity, invasiveness, and resistance to apo-ptosis (35, 36). We next identified an ascites-derived tumor sig-nature termed ATCdx referring to cells that are either EpCAM+

and/or V3-positive (Vimentin+/Calretinin−/CD45−). This ATCdxpanel had higher sensitivity, specificity, and accuracy than anyindividual marker alone and was able to correctly identify all13 ovarian-cancer samples in the training set (Table 3).

Fig. 1. Schematic approach. A total of 85 putative ovarian cancer protein markers were identified through literature, database, and other screens (UpperLeft). Markers were tested in 12 ovarian cell lines (SI Appendix, Fig. S1), and a subset were examined in ascites from human patients (Lower Left; n = 65) (Figs.2 and 3). A microfluidic chip (Fig. 4) was developed for point-of-care analysis (Lower Right).

2 of 9 | www.pnas.org/cgi/doi/10.1073/pnas.1315370110 Peterson et al.

Page 3: Ascites analysis by a microfluidic chip allows tumor-cell ...€¦ · Ascites analysis by a microfluidic chip allows tumor-cell profiling Vanessa M. Petersona,b,1, Cesar M. Castroa,c,1,

Prospective ATC Profiling in a Test Set of Human Ascites Samples.Wenext investigated the diagnostic performance of ATCdx in a pro-spective study (Fig. 3). Using a test set of 47 patients (n = 33OvCA, n = 14 benign), we were able to demonstrate high sen-sitivity and specificity. Namely, the presence or absence of ATCdxcorrectly identified 33 ovarian-cancer patients and 14 benignascites samples (Fig. 3A). In patients with sufficient cell numbersfor flow analyses, the four markers with the highest sensitivity inthe training set were evaluated in the test set. The cutoff valuesdetermined from ROC analyses (SI Appendix, Fig. S4) in thetraining set were applied to the test set (Fig. 3B). The sensitivity,specificity, and accuracy of these individual markers were thencalculated (SI Appendix, Table S1). EpCAM had the highestsensitivity (93.9%) and accuracy (95.7%), followed by CD24(sensitivity, 85.7%; accuracy, 89.7%). Adding V3 (Vimentin+/Calretinin−/CD45−) to EpCAM (ATCdx panel) increased sensi-tivity and accuracy.

Ascites Specimen Cellular Composition. The total cell and ATCcount in ascites specimens was determined for each of the 65patients (n = 46 OvCA, n = 19 Ctrl) (SI Appendix, Fig. S5). Themean total cell number (host cells and ATCs) for the 65 patientswas 1.2 × 105 cells per mL (median, 4.1 × 104 ; range, 3 × 103 to1.5 × 106; SEM, 2.7 × 104) and similar (P value > 0.05) in boththe 46 OvCA (mean, 1.5 × 105; median, 6.8 × 104; range, 1.6 ×103 to 1.5 × 106; SEM, 3.5 × 104) and 19 control samples (mean,6.7 × 104; median, 3.2 × 104; range, 3.1 × 103 to 5 × 105; SEM,2.6 × 104). ATCs were identified in all 46 ovarian-cancer patients(mean, 2.7 × 104; median, 2 × 103; range, 1.5 × 101 to 6 × 105;SEM, 1.4 × 104).

ATC Enrichment and Detection Using a Point-Of-Care MicrofluidicChip. Many of the ascites samples we procured containedclumps and extracellular debris that pose a challenge for con-ventional microfluidic approaches (SI Appendix, Fig. S6). Wethus developed a unique ATC chip equipped with (i) a miniature

filter (70 μm) at the inlet to prevent downstream clogging (Fig.4), (ii) negative magnetic selection of benign cells captured by

Fig. 2. Profiling of primary human samples in a training set. (A) Multicolor flow cytometry was used for gating of mesothelial (calretinin+), leukocytes(CD45+), and CD45/Calretinin− cells. (B) A subgroup of 31 markers identified in the cell-line screen (SI Appendix, Fig. S1) were subsequently tested in a trainingset (n = 18). The markers were placed into four different categories: unique malignant, overlapping markers (ubiquitous), benign, and absent, using cutoffsdescribed in Methods. The three OvCA patients with lowest EpCAM expression levels had the highest vimentin expression levels, suggestive of epithelial-to-mesenchymal transition (EMT). This training set led to identification of the ATCdx panel where malignancy is defined by either having an EpCAM+ and/orV3+ (Vimentin+/Calretinin−/CD45−) signature. Heatmap values are the log ratio of the fluorescent signal over the ctrl [λ = (Sig-Ctrl)/Ctrl] where the ctrl is thesecondary antibody without the primary antibody (see Methods for more details) (yellow, lowest; red, highest).

Table 1. Characteristics of ovarian-cancer patients (n = 46)

Characteristic No. Percentage

Ovarian-cancer patients 46Age

Median 60Range 36–85

StageIC 1 2IIIC 27 59IV 18 39

Surgical debulkingOptimal 25 55Suboptimal 6 13Interval 14 30None 1 2

Survival (avg mo from collection)Alive 14 (26) 30Deceased 32 (9) 70

ChemotherapyActive 21 45Not yet initiated 25 55

Platinum responseSensitive 18 39Resistant 18 39Refractory 4 9Not applicable 6 13

Disease courseResponse 19 41Stable 1 2Progression 24 52.5Mixed 2 4.5

Peterson et al. PNAS Early Edition | 3 of 9

MED

ICALSC

IENCE

SPN

ASPL

US

Page 4: Ascites analysis by a microfluidic chip allows tumor-cell ...€¦ · Ascites analysis by a microfluidic chip allows tumor-cell profiling Vanessa M. Petersona,b,1, Cesar M. Castroa,c,1,

a magnet located under the device inlet, and (iii) multiple, se-rially smaller microwells (40–15 μm) in which purified andenriched cells are captured (SI Appendix, Fig. S7). The multistep,on-chip purification approach yielded an approximate 1,000-foldATC enrichment (SI Appendix, Fig. S8). Perhaps more impor-tant, the microwell-captured cells facilitated visualization of thecell populations and simplified multicolor image analysis.The ATC chip was designed to be optically transparent so that

cells captured in microwells, can be stained on-chip and visual-ized with a charge-coupled device (CCD). Fig. 5 illustrates arepresentative example of ATCdx-based staining of ATCs (red),and CD45/Calretinin staining of mesothelial and host cellstaining (green). Compared with conventional flow-cytometryneeds (∼10,000 cells), the ATC chip affords reduced sample-sizerequirements due to its single cell detection capabilities. Forexample, an ovarian-cancer patient harboring 15 ATCs per mLwould require >500 mL of ascites processing for flow cytometrycompared with only 0.1 mL for on-chip processing. Moreover,this microfluidic chip can be fabricated using soft lithographictechniques (37, 38) with inexpensive materials such as PDMS,

providing a practical, affordable (<$1.00 per chip), and scalablealternative to flow cytometry. We also demonstrated that chip-enriched cells (Fig. 6A) can be further analyzed such as formRNA expression levels. Measurements of mRNA on cells fromthe ATC chip showed excellent correlation to bulk measure-ments (R2 = 0.995) (Fig. 6B). Ascites samples from five ovarian-cancer patients were enriched with ATC chip and subsequentlyprofiled for gene-expression levels relative to benign cell lines(Fig. 6C). EpCAM and CD24 had the highest expression levelsconcurrent with our earlier findings in the training and test sets(Table 3 and SI Appendix, Table S1).

Serial Testing to Measure Treatment Response in Individual Patients.A major application of a point-of-care approach would be toleverage the use of readily accessible (but otherwise discarded)ascites as a source of ATCs for treatment monitoring. Fig. 7exemplifies this potential using ascites collected and profiledfrom a single patient over a 14 wk treatment period. The patientwas initially treated with cytotoxic agents (Carboplatin andPaclitaxel; weeks 2 and 5) but was transitioned to antiangiogenic

Fig. 3. Prospective testing of ATC marker panels in 47 patients. (A) Ascites samples were tested for the presence of six individual markers in 33 ovarian-cancerpatients (Left) and 14 controls (right). EpCAM alone was positive in 31 samples. By using the V3 marker set (Vimentin+/Calretinin−/CD45−) and EpCAM to-gether (ATCdx), all 33 samples were correctly identified (green heat map). ATCs were identified in malignant samples, but not in benign samples. Gray squaresrepresent data not measured due to insufficient number of cells for flow cytometry. Color scale is same as Fig. 2B. (B) Waterfall plots of the individual markersprofiled in the test set. Dotted red lines represent the optimal threshold values determined from ROC analyses performed on the training set (SI Appendix,Fig. S4).

Table 2. Sample numbers of different data sets

Dataset Definition Malignant (n) Benign (n) Total (n)

Profiling Profiling of cell lines; no primary patient samples 12 6 18Training Validation of markers identified in cell screens 13 5 18Test Prospective analysis in patient cohort 33 14 47Complete All above patient samples combined 46 19 65Serial Tx Patients with repeat samples 7 NA 7

4 of 9 | www.pnas.org/cgi/doi/10.1073/pnas.1315370110 Peterson et al.

Page 5: Ascites analysis by a microfluidic chip allows tumor-cell ...€¦ · Ascites analysis by a microfluidic chip allows tumor-cell profiling Vanessa M. Petersona,b,1, Cesar M. Castroa,c,1,

therapy (Bevazucimab/Avastin; week 7) due to disease progres-sion. ATC numbers were initially observed to decrease steadilyduring treatment response, only to increase when disease pro-gression occurred. Avastin attenuated the increase in ATC burdenconcurrent with the patient’s improved clinical symptoms.ATC analysis could also be used for early detection of treatment

response through the profiling of protein markers related to bi-ological processes such as proliferation [Ki67, phospho histoneH3 (pH3), pCyclin D], mRNA translation [phospho 4E-binding

protein 1 (p4E-BP1)], and/or pathway inhibition [phospho S6 ri-bosomal protein (pS6RP), phospho extra-signal-regulated kinase(pERK)]. As expected, the proliferation markers Ki67, pH3, andpCyclinD had a decreasing trend mirroring ATC counts. Aftereach Carboplatin/Taxol administration, the levels of the growthpathway markers p4E-BP1, pERK, and pS6RP were reducedand stable, but, upon switching to single agent Avastin, the levelsof these markers increased. In this clinical example, the datasuggest that resuming cytotoxic therapy (which had maintainedpathway inhibition) alongside Avastin treatment might haveslowed this patient’s rapid clinical decline. Indeed, recent late-phase clinical-trial data have reported progression-free survivaladvantages when cytotoxic and antiangiogenic strategies are com-bined in ovarian cancer (39).Using an expanded set of mechanistic markers (SI Appendix,

Fig. S9), we also compared the profiles of treatment responderswith nonresponders (based on tumor burden, as determined byimaging or clinical course) (Table 1). The panels indicate thatthe two groups could be distinguished based on ATC molecularprofiles. Moreover, these profiles could be potentially useful forproviding additional biological insight into the drivers of treatmentresponse or disease progression. For example, unlike in treatmentresponders, levels of pS6RP and p4E-BP1 (readouts of the phos-phatidylinositol 3/PI3-kinase pathway) remained elevated aftertherapy in the nonresponders. These trends are consistent withprevious findings that have associated activated PI3-kinase signalingwith chemoresistance in advanced ovarian cancer (40).

DiscussionIn the current study, we developed and tested a bedside-com-patible “ATC chip” simplifying enrichment and molecularanalysis of shed cancer cells in peritoneal fluid. Unlike in blood,cells harvested from the peritoneum often form clumps and cancontain extracellular debris that challenges conventional micro-fluidic devices. We circumvented this challenge by microfiltration,negative magnetic separation, and positioning of individual cells in

Fig. 4. Schematic of on-chip purification and labeling. First, ascites fluid is collected from the patient that contains malignant cells among an inflammatorymilieu of host cells. Ascites cells are added to the chip followed by an antibody mixture (EpCAM-FITC, Vimentin-PE/Cy7 Calretinin-Biotin/AF647, CD45-Biotin/AF647, Marker-AF555). Streptavidin-coated magnetic particles then bind to the benign mesothelial cells (Calret+) and leukocytes (CD45+). A magnet under theinlet captures the benign cells whereas the malignant cells pass freely through the microchip. The four different size microwells (15, 20, 30, and 40 μm) allowfor capture of the malignant cells while allowing for the typically smaller leukocytes to pass through the device. The ATCdx signature (EpCAM+ and/orVimentin+/Calretinin−/CD45−) can then be imaged to determine number of ATCs.

Fig. 5. On-chip ATCdx and profiling. Multiple stains performed on-chip al-low diagnosis of cancer cells (red) and evaluation of specific biomarkers(Ki67 in this example). The Lower Right image is the merge of the pro-liferation marker Ki67 with the ATC markers EpCAM and Vimentin. Red,EpCAM; green, CD45/Calretinin (benign host cells); blue, Vimentin; cyan,Ki67; yellow, nuclei. Note alignment of cells of chip and simple imageanalysis.

Peterson et al. PNAS Early Edition | 5 of 9

MED

ICALSC

IENCE

SPN

ASPL

US

Page 6: Ascites analysis by a microfluidic chip allows tumor-cell ...€¦ · Ascites analysis by a microfluidic chip allows tumor-cell profiling Vanessa M. Petersona,b,1, Cesar M. Castroa,c,1,

chip microwells that allow convenient analysis. We show that bothprotein analysis through on-chip staining and mRNA analysisusing chip-harvested cells are feasible and result in clinicallyvaluable information. Moreover, the device leverages the ad-

vantages of microfabrication and microfluidics to facilitate point-of-care and ascites testing, respectively. The ATC chip’s low cost(<$1 per unit), ease-of-scalability (plastics), and simplicity of usecould promote widespread testing and, if clinically validated,

Fig. 6. On-chip Processing. (A) Processing ascites samples on-chip enables purification of ATCs (red) from benign cells (green) for easy detection of ATCs. (B)OvCA cells can be removed from chip after enrichment for downstream molecular profiling. Gene counts from CaOV3 cells isolated from chip had excellentcorrelation to unprocessed CaOV3 cells (R2 = 0.995). Measurements from chip were done in triplicate, and error bars represent the SD. (C) Demonstration ofon-chip processing in clinical samples. Ascites samples were enriched for ATCs on-chip, and then ATCs were removed and mRNA gene expression levels weremeasured. The heatmap shows relative expression levels for each marker (yellow, lowest; red, highest).

Fig. 7. Serial analysis of ATCs. ATCs were obtained serially from a single patient over a 14-wk treatment cycle. Carboplatin and Paclitaxel (Taxol) were givenin weeks 2 and 5, and Bevazucimab (Avastin) was given in week 11. The number of ATCs was measured over the course of treatment (ATC burden) usingATCdx. Additionally, protein markers related to biological processes such as proliferation (Ki67, pH3, pCyclinD), mRNA translation (p4E-BP1), and pathwayinhibition (pS6RP, pERK) were also measured. The broader analyses demonstrate molecular profiling of ascites can be used as a tool to monitor treatmentresponse over the course of therapy. All samples were stained with calretinin and CD45 antibodies to exclude mesothelial cells and and leukocytes, respectively.Data are expressed as the average of the mean fluorescent intensity subtracted from background ± SEM. pH3 data are plotted as % of cells up-regulated.

6 of 9 | www.pnas.org/cgi/doi/10.1073/pnas.1315370110 Peterson et al.

Page 7: Ascites analysis by a microfluidic chip allows tumor-cell ...€¦ · Ascites analysis by a microfluidic chip allows tumor-cell profiling Vanessa M. Petersona,b,1, Cesar M. Castroa,c,1,

ready adoption by tertiary academic medical centers or com-munity hospitals.To help address uncertainties in the biomarker literature, we

ourselves screened over 100 commercially available antibodies toinvestigate 85 putative targets of ovarian-cancer cells in perito-neal fluid. Although we were able to identify a relatively simplemarker set for these cells, we found some unexpected results. Forexample, we found low levels of certain markers that have beengaining traction as drug targets (EphA2) (26, 41-43), or that havebeen touted as specific for (CA-125, FOLR1) (32), or over-abundant in (mesothelin) (44), ovarian cancer. We found rela-tively high expression levels of EpCAM, CD24, and TAG-72 incancer patients, consistent with previous reports (10, 21, 45-49).In contrast, levels of calretinin (50, 51) and D2-40 (51) werehigh in mesothelial cells but not in ovarian-cancer cells andthus served as convenient distinction markers. MUC1, EGFR,PAX8, and ESE-1 displayed mixed expression levels that werenonspecific.Despite the number of markers tested, there were some limi-

tations. Firstly, the list of proteins, although lengthy, was certainlynot exhaustive. Secondly, only commercially available antibodieswere tested because our intention was to leverage screeningfindings for eventual and feasible widespread testing using thepoint-of-care device. It is possible that certain research anti-bodies might have resulted in higher and more specific binding,but this conjecture would need to be tested in additional datasetsbeyond the scope of the current study. Thirdly, despite screeninga number of available antibodies (SI Appendix, Table S2), wewere unable to identify reliable antibodies for FSHR (follicle-stimulating hormone receptor), a target implicated in ovariancancer (52-55) (SI Appendix, Fig. S1). We thus abandoned currentefforts due to low specificity, but FSHR nevertheless remains amarker of interest that requires further exploration.In summary, we show that ascites analysis via microfluidic

chips could become a convenient strategy for serially measuringATC numbers and analyzing their molecular profile. Leveragingthis additional tumor source could help expand the clinicalstrategies needed for paradigm shifts in patient-oriented re-search (56).

MethodsPatient Population and Analyses. The study was approved by the Dana Farber/Harvard Cancer Center Institutional Review Board (IRB), and informed con-sent was obtained from all subjects. Sixty-five subjects with accumulation ofascitic fluid, and requiring drainage, were enrolled in this study. Forty-sixsubjects carried a known diagnosis of ovarian cancer (Table 1) wheras 19subjects were included as controls (e.g., their ascites fluid was as a result ofanother disease such as cirrhosis or liver failure). Ascites fluid samples werecollected from patients per routine in the Massachusetts General Hospital(MGH) Abdominal Imaging and Intervention suites. Two clinicians (C.M.Cand R.W.), blinded to the results, reviewed all of the documented clinical,imaging, and pathology data obtained from each cancer patient. Table 2summarizes the different cohorts included in the training set (n = 18),validation set (n = 47), and serial analyses sets (n = 7).

Cell Culture. The cell lines SK-OV-3, OVCAR-3, A2780, CaOV-3, OV-90, ES-2,TOV-112D, TOV-21G, and UWB1.289 were purchased from American TypeCulture Collection and grown in media following their suggested protocol.UCI-101 and UCI-107 cell lines were kindly provided by G. Scott Rose (Universityof California, Irvine, CA) and OVCA429 was kindly provided by DavidSpriggs (Memorial Sloan Kettering, New York). UCI 101, UCI 107, and OVCA429were grown in RPMI (Cellgro) with 10% (vol/vol) FBS, 1% L-glutamine, and1% penicillin/streptomycin. Mesothelial cells, LP3 and LP9, were purchasedfrom the Corriell Institute for Medical Research and grown according toprotocol. NOSE cell lines were derived from ovarian surface epithelium (OSE)brushings cultured in 1:1 Media 199:MCDB 105 (Sigma-Aldrich) with genta-micin (25 μg/mL) and 15% heat-inactivated serum. TIOSE4 and TIOSE6 celllines were obtained from transfection of hTERT into NOSE cells maintainedin 1:1 Media 199:MCDB 105 with gentamicin (25 μg/mL), 15% heat-inacti-vated serum, and G418 (500 μg/mL) (57). Cells were cultured at low passagenumber under standard conditions at 37 °C in a humidified incubator con-taining 95% room air and 5% CO2 atmosphere. When the cells reached∼90% confluence, they were trypsinized to remove the cells from the cultureflask. Medium was then added, the cells were spun down (300 × g for 5 min),and the supernatant was removed. The cells were then fixed following thesame protocol as used for clinical samples; namely, Lysis/Fix buffer (BD PhosflowLyse/Fix Buffer) was added to the cells for 10 min at 37 °C, before being washedtwice with 5 mL of SB+ (PBS with 2% BSA). The cells were aliquoted into tubes(∼1 × 106 cells per mL) and stored at −20 °C until labeling. The cells were thenlabeled following the same protocol used for clinical samples, with the exceptionthat calretinin and CD45 antibodies were not added to each sample.

Bulk Ascites Processing for More Extensive Profiling. Clinically obtained ascitessamples were transferred into 2–4 separate 225-mL conical bottom tubes (BDFalcon) and centrifuged at 300 × g for 5 min (Eppendorf Centrifuge 5810R).The supernatant was then removed, leaving the cell pellet undisturbed. Theremaining ascites fluid was added to the tubes, and the centrifugation andaspiration step was repeated until all of the fluid was processed. The cellpellet in each tube was then resuspended in SB+ and transferred to a smaller50-mL tube. The cells were spun down at 300 × g for 5 min, and the su-pernatant was aspirated. After vortexing, 40 mL of prewarmed Lysis/Fixbuffer (BD Biosciences) was added, and the sample was incubated on ashaker at 37 °C for 10 min. If there were visual clumps present before thefixation step, collagenase (Sigma Aldrich) was added at 0.2 mg/mL in PBS,and the sample was incubated on a shaker for 30–60 min at 37 °C. The cellswere then washed with SB+ before proceeding to the lysis/fix step (describedabove). After the lysis/fix step, the cells were centrifuged at 400 × g for3 min, and the supernatant was removed. Then two washes with 5 mL ofSB+ (PBS with 2% BSA) were conducted. The supernatant was removed afterthe final wash, and the cells were resuspended in 1 mL of SB+.CD45 negative selection. Cells were counted with the Countess Cell Counter(Life Technologies) and adjusted to a concentration of ∼2 × 107 cells per mL.CD45 antibody (Biolegend; H130) was added (0.5 μL per 106 cells) and in-cubated for 1 h, before washing twice with 5 mL of SB+. The supernatantwas removed, and Anti-Mouse IgG1 magnetic beads (80 μL per 107 cells;Miltenyi Biotec) were added along with SB+ (20 μL per 107 cells). The re-action was incubated for 15 min at 4 °C and was followed by two washes(2 mL per 107 cells) with SB+. The remaining sample was then resuspended in1 mL of SB+. Magnetic separation columns (LS; Miltenyi Biotec) and theQuadroMACS separator were used for negative selection following sug-gested protocols. Preseparation filters (Miltenyi Biotec) were used to removeany clumps or debris from the clinical specimens to prevent obstruction ofthe column. The maximum number of cells used in each column was 1 × 108.The sample was added to the column and was washed three times with 3 mLof SB+. The CD45− cells that passed through the column were subsequentlycollected into a 15-mL tube and centrifuged (400 × g, 3 min); the superna-tant was removed, and the cells were resuspended in SB+. Cells were thencounted and aliquoted into tubes at approximately ∼1 × 106 cells per mL,before being stored at −20 °C until labeling.Labeling. CD45-depleted cells stored in the −20 °C freezer were thawed. Thecells were then centrifuged at 400 × g for 3 min, and the supernatant wasremoved. Perm buffer (PW+: BD perm/wash with 2% BSA) was added, andthe cells were aliquoted into cluster tubes (Costar). The appropriate anti-body mixture of calretinin and CD45 was added (SI Appendix, Table S2)(calretinin, Mouse Dako DAK-Calret 1 or Rabbit Invitrogen DC8; CD45, RatAbcam YTH24.5 or Mouse Biolegend H130). The specific antibody for thebiomarker of interest was then added (SI Appendix, Table S2). The finalreaction volume was 150 μL. This primary reaction was vortexed and in-cubated for 1 h before 0.5 mL of PW+ was added to each sample and thecells spun down at 400 × g for 3 min. The supernatant was removed, and the

Table 3. Sensitivity, specificity, and accuracy of differentprotein markers in the training set

MarkersSensitivity,

%Specificity,

%Accuracy,

%

EpCAM 92.3 100.0 96.9CA19-9 46.2 100.0 78.1CD24 92.3 94.7 93.8TAG-72 76.9 100.0 90.6FOLR1 69.2 78.9 75.0CA-125 53.8 94.7 78.1V3 panel (Vim+/Calret-/CD45-) 23.1 100.0 68.8ATCdx (EpCAM+ and/or V3+) 100.0 100.0 100.0

Peterson et al. PNAS Early Edition | 7 of 9

MED

ICALSC

IENCE

SPN

ASPL

US

Page 8: Ascites analysis by a microfluidic chip allows tumor-cell ...€¦ · Ascites analysis by a microfluidic chip allows tumor-cell profiling Vanessa M. Petersona,b,1, Cesar M. Castroa,c,1,

cells were vortexed and washed with 0.5 mL of PW+. After centrifugation(400 × g, 3 min), the supernatant was aspirated, and the appropriate sec-ondary antibodies were added (SI Appendix, Table S3) [Anti-Mouse FITCAbcam (1:300); Anti-Rabbit APC Abcam (1:300); Anti-Rat PeCY7 Biolegend(1:300)]. The final reaction volume was 150 μL. The samples were vortexedand incubated on ice for 1 h. Cells were then washed twice with 0.5 mL ofPW+. Samples analyzed by flow cytometry the same day were kept on iceunder aluminum foil. Samples run the following day were lightly fixed [BDPhosflow Fix Buffer (1:2)] for 10 min at 37 °C and then washed with SB+.After washing, DAPI was added to the samples at a ratio of 1:500 (Fxcyclestain; Invitrogen) 30 min before analysis by flow cytometry. Antibody in-formation (such as clone and company) for all markers is included in SIAppendix, Tables S2 and S3.Flow cytometry. Fluorescently labeled samples were analyzed using an LSRIIflow cytometer (Becton Dickinson). FlowJo software was used to gate on:singlets using DAPI staining, then Calretinin+, CD45+, and CD45−/Calretinin−

cell populations in the clinical samples. The mean fluorescent intensity (MFI)for each marker of interest was then determined for each of these threepopulations. The background was determined by staining with the second-ary antibody only (no primary). The signal-over-background was then cal-culated [λ = (Signal-Background)/Background] and plotted in the heat mapsusing GENE-E software (Broad Institute). For Fig. 2 and SI Appendix, Fig. S1,each marker was placed into four categories on the following criteria: (i)“Unique Malignant” (if an ovarian-cancer sample had λ > 1.5 and all benignsamples had λ < 1.5); (ii) “Ubiquitous” (if an ovarian-cancer sample anda benign sample had λ > 1.5); (iii) “Benign” (if both an ovarian-cancersample and benign sample had λ < 1.5); and (iv) “Absent” (if both ovarian-cancer and benign samples had λ ≤1.5). Figs. 2 and 3 were plotted on a logscale, and gray signifies data that was not determined. A log scale was usedbecause this resulted in a more symmetric distribution. Histograms of a se-lect set of markers from the cell line screening and training set are shown inSI Appendix, Figs. S2 and S3.Statistical analysis. Receiver operating characteristic (ROC) curves were con-structed for cancer markers relative to benignmesothelial cells in the trainingset (SI Appendix, Fig. S4). For each marker sensitivity versus (1 − specificity)was plotted, and the values of area under the curve (AUC) were computedusing the trapezoidal rule. The empirical ROC curves were smoothed byapplying the binormal fitting model. An AUC of 0.5 was used to indicatethat the test shows no difference between the two groups whereas an AUCof 1.0 was used to indicate that the test gives a perfect separation betweenthe groups. We defined the optimal cutoff value for identifying malignantstatus as the point on the ROC curve with the minimal distance between the0% false-negative and the 100% true-positive rate. Statistical analysis wasperformed using the R-package (version 3.0.1). Using standard formulas, wecalculated sensitivity, specificity, and accuracy.

ATC Device Fabrication. The microfluidic device was designed to contain∼5,000 capture sites ranging in size from 15 to 40 μm (SI Appendix, Fig. S7A)in a 2.6 × 1.0 cm area. The capture sites on the device have bowl-likestructures with an underpass gap that allows smaller cells to pass through.As shown in SI Appendix, Fig. S7B, path A through the capture structure hasa considerably shorter length, therefore smaller flow resistance, than path Bthrough the channel structure. Due to the flow-resistance difference, mostof the fluid flows through path A rather than path B. Cells larger than thecapture structures will be trapped, and smaller cells will pass through. Theflow rate of the device was ∼10 mL/h and allowed ∼1,000-fold ATC en-

richment (58, 59). Microfluidic single cell capture arrays were fabricated inpolydimethylsiloxane (PDMS; Dow Corning) using soft lithography(37, 38).Three layers of epoxy-based photoresist were patterned on 4-in siliconwafers as a mold using conventional photolithography. The first SU8 2015(Microchem) layer defined a 15-μm gap height in the capture sites for the 15-and 20-μm cells. Before developing unexposed SU8, the second SU8 2015layer was coated and aligned to the channel structure for the 15- and 20-μmcell capture sites. This layer also defined the gap height (30 μm) for the 30- and40-μm cell capture sites. Finally, another 30-μm-thick SU8 2025 was applied forthe 30- and 40-μm cell capture structures. All unexposed SU8 photoresist layerswere then developed in the SU8 Developer (Microchem). The mold with thechannel and cell-capture patterns was silanized with a vapor of trimethyl-chlorosilane (TMCS; Sigma) and was cast with 3-mm-thick PDMS prepolymer.After curing, the PDMS layer was peeled off, and an inlet and an outlet werepunched out. The prepared PDMS chip was then bonded (not permanently) toa glass slide. The chip thus remained detachable for easy recovery of cells afterpurification and imaging to enable further molecular profiling of cells.

Processing Cells Through the ATC Chip. For on-chip enrichment and staining ofATCs, ∼100 μl of ascites fluid was added to the inlet of the chip. Then, anantibody mixture of the following antibody conjugates was added: Calreti-nin-Biotin (Invitrogen; Rabbit), CD45-Biotin (Abcam; Mouse), CD45-AlexaFluor 647 (Biolegend; Mouse), EpCAM-FITC (Dako; Mouse), Vimentin (R&D;Rat), and Ki67-Alexa Fluor 555 (BD Pharmingen). This mixture was incubatedfor 15 min. Then, streptavidin-coated magnetic particles (R&D), Anti-RabbitAlexa Fluor 647 (Cell Signaling), and Anti-Rat PE/Cy7 (Biolegend) wereadded, followed by a 15-min incubation (Fig. 4). In the first enrichment step,a magnet was placed under the inlet preventing the benign CD45 and Cal-retinin magnetically labeled cells from passing through the chip. The non-magnetically labeled ATCs entered into the microchip when negativepressure was applied to the outlet of the device (via rubber bulb). The sec-ond enrichment step was based on size: (i) the typically larger ATC cells werecaptured on the chip’s capture sites (decreasing in size from 15 to 40 μm) and(ii) the smaller leukocytes (∼<15 μm) (60) passed through the capture sites.This dual enrichment approach using both immunomagnetic labeling andsize separation (58, 59) improved chip performance and enabled an ap-proximate 1,000-fold enrichment to be achieved (SI Appendix, Fig. S8). Threewashes of 100 μl of SB+ with DAPI (4’,6-diamidino-2-phenylindole; Invi-trogen; 1:500) were then passed through the microchip. Captured ATCs wereimaged using the DeltaVision screening system (Applied Precision Instru-ments) and analyzed using ImageJ software (version 10.2). Enriched cellscould then be removed from the detachable chip for further molecularprofiling. Measurements for mRNA expression levels were done usingnCounter gene expression assay (NanoString) on cell lysate (61, 62). Datawere analyzed using nsolver analysis software 1.1 (NanoString), and theheatmap (Fig. 6C) was plotted using GENE-E (Broad Institute).

ACKNOWLEDGMENTS. We thank M. Pectasides and S. McDermott for samplecollection; C. Krasner, L. Sullivan, and S. McIlvenna for patient referral;V. Vathipadiekal and P. Waterman for help growing cell lines; H. Saya (IAMR,Keio University) for CD44v9 antibody, L. Lizotte (REMS media services) formicrochip pictures, A. Zaltsman for imaging, and Y. Fisher-Jeffes for review ofthe manuscript. This work was funded in part by Grants P50CA086355,R01EB004626, R01EB010011, P01-CA139980, U54-CA151884, K12CA087723-11A1, and DODW81XWH-11-1-0706. V.M.P. was supported in part by a MerckEducational Assistance Fellowship.

1. Siegel R, Naishadham D, Jemal A (2013) Cancer statistics, 2013. CA Cancer J Clin 63(1):

11–30.2. Na YJ, et al. (2009) Ovarian cancer: Markers of response. Int J Gynecol Cancer 19(Suppl

2):S21–S29.3. Bast RCJ, Jr., Hennessy B, Mills GB (2009) The biology of ovarian cancer: New op-

portunities for translation. Nat Rev Cancer 9(6):415–428.4. Lutz AM, et al. (2011) Early diagnosis of ovarian carcinoma: Is a solution in sight?

Radiology 259(2):329–345.5. Zhang B, Cai FF, Zhong XY (2011) An overview of biomarkers for the ovarian cancer

diagnosis. Eur J Obstet Gynecol Reprod Biol 158(2):119–123.6. Nolen BM, Lokshin AE (2012) Protein biomarkers of ovarian cancer: The forest and the

trees. Future Oncol 8(1):55–71.7. Li AJ, et al. (2004) The prognostic significance of thrombocytosis in epithelial ovarian

carcinoma. Gynecol Oncol 92(1):211–214.8. Obermayr E, et al. (2013) Molecular characterization of circulating tumor cells in

patients with ovarian cancer improves their prognostic significance: A study of the

OVCAD consortium. Gynecol Oncol 128(1):15–21.9. Taylor DD, Gerçel-Taylor C (2005) Tumour-derived exosomes and their role in cancer-

associated T-cell signalling defects. Br J Cancer 92(2):305–311.

10. Meirelles K, et al. (2012) Human ovarian cancer stem/progenitor cells are stimulated

by doxorubicin but inhibited by Mullerian inhibiting substance. Proc Natl Acad Sci

USA 109(7):2358–2363.11. Taylor DD, Gercel-Taylor C, Parker LP (2009) Patient-derived tumor-reactive anti-

bodies as diagnostic markers for ovarian cancer. Gynecol Oncol 115(1):112–120.12. Yip P, et al. (2011) Comprehensive serum profiling for the discovery of epithelial

ovarian cancer biomarkers. PLoS ONE 6(12):e29533.13. Kinde I, et al. (2013) Evaluation of DNA from the Papanicolaou test to detect ovarian

and endometrial cancers. Sci Transl Med 5(167):167ra4.14. Buckanovich RJ, et al. (2007) Tumor vascular proteins as biomarkers in ovarian cancer.

J Clin Oncol 25(7):852–861.15. Emmanuel C, et al.; Australian Ovarian Cancer Study Group (2011) Comparison of

expression profiles in ovarian epithelium in vivo and ovarian cancer identifies novel

candidate genes involved in disease pathogenesis. PLoS ONE 6(3):e17617.16. Oribabor JW, Ambrosio A, Castro CM, Birrer MJ (2012) Biotargets of Cancer

in Current Clinical Practice, ed Bologna M (Springer/Humana Press, New York),

pp 381–402.17. Kipps E, Tan DS, Kaye SB (2013) Meeting the challenge of ascites in ovarian cancer:

New avenues for therapy and research. Nat Rev Cancer 13(4):273–282.

8 of 9 | www.pnas.org/cgi/doi/10.1073/pnas.1315370110 Peterson et al.

Page 9: Ascites analysis by a microfluidic chip allows tumor-cell ...€¦ · Ascites analysis by a microfluidic chip allows tumor-cell profiling Vanessa M. Petersona,b,1, Cesar M. Castroa,c,1,

18. Attanoos RL, Webb R, Dojcinov SD, Gibbs AR (2002) Value of mesothelial and epi-thelial antibodies in distinguishing diffuse peritoneal mesothelioma in females fromserous papillary carcinoma of the ovary and peritoneum. Histopathology 40(3):237–244.

19. Comin CE, Saieva C, Messerini L (2007) h-caldesmon, calretinin, estrogen receptor, andBer-EP4: A useful combination of immunohistochemical markers for differentiatingepithelioid peritoneal mesothelioma from serous papillary carcinoma of the ovary.Am J Surg Pathol 31(8):1139–1148.

20. Laury AR, et al. (2010) PAX8 reliably distinguishes ovarian serous tumors from ma-lignant mesothelioma. Am J Surg Pathol 34(5):627–635.

21. Ordóñez NG (1998) Role of immunohistochemistry in distinguishing epithelial peri-toneal mesotheliomas from peritoneal and ovarian serous carcinomas. Am J SurgPathol 22(10):1203–1214.

22. Ordóñez NG (2006) The diagnostic utility of immunohistochemistry and electronmicroscopy in distinguishing between peritoneal mesotheliomas and serous carcino-mas: A comparative study. Mod Pathol 19(1):34–48.

23. Cancer Genome Atlas Research Network (2011) Integrated genomic analyses ofovarian carcinoma. Nature 474(7353):609–615.

24. Elschenbroich S, et al. (2011) In-depth proteomics of ovarian cancer ascites: Com-bining shotgun proteomics and selected reaction monitoring mass spectrometry.J Proteome Res 10(5):2286–2299.

25. Karst AM, Drapkin R (2011) The new face of ovarian cancer modeling: Better pros-pects for detection and treatment. F1000 Med Rep 3:22.

26. Scarberry KE, Dickerson EB, Zhang ZJ, Benigno BB, McDonald JF (2010) Selective re-moval of ovarian cancer cells from human ascites fluid using magnetic nanoparticles.Nanomedicine 6(3):399–408.

27. Uhlen M, et al. (2010) Towards a knowledge-based Human Protein Atlas. Nat Bio-technol 28(12):1248–1250.

28. LaRocca PJ, Rheinwald JG (1984) Coexpression of simple epithelial keratins and vi-mentin by human mesothelium and mesothelioma in vivo and in culture. Cancer Res44(7):2991–2999.

29. Laury AR, et al. (2011) A comprehensive analysis of PAX8 expression in human epi-thelial tumors. Am J Surg Pathol 35(6):816–826.

30. Stussi G, et al. (2005) Isotype-specific detection of ABO blood group antibodies usinga novel flow cytometric method. Br J Haematol 130(6):954–963.

31. Ng PC, et al. (2004) Neutrophil CD64 is a sensitive diagnostic marker for early-onsetneonatal infection. Pediatr Res 56(5):796–803.

32. Armstrong DK, White AJ, Weil SC, Phillips M, Coleman RL (2013) Farletuzumab (amonoclonal antibody against folate receptor alpha) in relapsed platinum-sensitiveovarian cancer. Gynecol Oncol 129(3):452–458.

33. Teng L, Xie J, Teng L, Lee RJ (2012) Clinical translation of folate receptor-targetedtherapeutics. Expert Opin Drug Deliv 9(8):901–908.

34. Suh KS, et al. (2010) Ovarian cancer biomarkers for molecular biosensors and trans-lational medicine. Expert Rev Mol Diagn 10(8):1069–1083.

35. Latifi A, et al. (2011) Cisplatin treatment of primary and metastatic epithelial ovariancarcinomas generates residual cells with mesenchymal stem cell-like profile. J CellBiochem 112(10):2850–2864.

36. Kalluri R, Weinberg RA (2009) The basics of epithelial-mesenchymal transition. J ClinInvest 119(6):1420–1428.

37. Voldman J, Gray ML, Schmidt MA (1999) Microfabrication in biology and medicine.Annu Rev Biomed Eng 1:401–425.

38. Xia Y, Whitesides GM (1998) Softlithographie. Angew Chem Int Ed 110:568–594.39. Heitz F, et al. (2012) Bevacizumab in the treatment of ovarian cancer. Adv Ther 29(9):

723–735.

40. Carden CP, et al. (2012) The association of PI3 kinase signaling and chemoresistance inadvanced ovarian cancer. Mol Cancer Ther 11(7):1609–1617.

41. Han L, et al. (2005) The clinical significance of EphA2 and Ephrin A-1 in epithelialovarian carcinomas. Gynecol Oncol 99(2):278–286.

42. Landen CN, Kinch MS, Sood AK (2005) EphA2 as a target for ovarian cancer therapy.Expert Opin Ther Targets 9(6):1179–1187.

43. Lee JW, et al. (2009) EphA2 immunoconjugate as molecularly targeted chemotherapyfor ovarian carcinoma. J Natl Cancer Inst 101(17):1193–1205.

44. Huang CY, et al. (2006) Serum mesothelin in epithelial ovarian carcinoma: A newscreening marker and prognostic factor. Anticancer Res 26(6C):4721–4728.

45. Bellone S, et al. (2009) Overexpression of epithelial cell adhesion molecule in primary,metastatic, and recurrent/chemotherapy-resistant epithelial ovarian cancer: Im-plications for epithelial cell adhesion molecule-specific immunotherapy. Int J GynecolCancer 19(5):860–866.

46. Ghazani AA, Castro CM, Gorbatov R, Lee H, Weissleder R (2012) Sensitive and directdetection of circulating tumor cells by multimarker μ-nuclear magnetic resonance.Neoplasia 14(5):388–395.

47. Kristiansen G, et al. (2002) CD24 is expressed in ovarian cancer and is a new in-dependent prognostic marker of patient survival. Am J Pathol 161(4):1215–1221.

48. Richter CE, et al. (2010) High-grade, chemotherapy-resistant ovarian carcinomasoverexpress epithelial cell adhesion molecule (EpCAM) and are highly sensitive toimmunotherapy with MT201, a fully human monoclonal anti-EpCAM antibody. Am JObstet Gynecol 203(6):582.e1–582.e7.

49. Zecchini S, et al. (2011) The adhesion molecule NCAM promotes ovarian cancer pro-gression via FGFR signalling. EMBO Mol Med 3(8):480–494.

50. Kachali C, Eltoum I, Horton D, Chhieng DC (2006) Use of mesothelin as a marker formesothelial cells in cytologic specimens. Semin Diagn Pathol 23(1):20–24.

51. Kim JH, et al. (2009) Immunocytochemical panel for distinguishing between ad-enocarcinomas and reactive mesothelial cells in effusion cell blocks. Diagn Cytopathol37(4):258–261.

52. Choi J-H, Choi K-C, Auersperg N, Leung PCK (2004) Overexpression of follicle-stimu-lating hormone receptor activates oncogenic pathways in preneoplastic ovariansurface epithelial cells. J Clin Endocrinol Metab 89(11):5508–5516.

53. Ji Q, Liu PI, Chen PK, Aoyama C (2004) Follicle stimulating hormone-induced growthpromotion and gene expression profiles on ovarian surface epithelial cells. Int JCancer 112(5):803–814.

54. Radu A, et al. (2010) Expression of follicle-stimulating hormone receptor in tumorblood vessels. N Engl J Med 363(17):1621–1630.

55. Zheng W, et al. (2000) Ovarian epithelial tumor growth promotion by follicle-stim-ulating hormone and inhibition of the effect by luteinizing hormone. Gynecol Oncol76(1):80–88.

56. Haber DA, Gray NS, Baselga J (2011) The evolving war on cancer. Cell 145(1):19–24.57. Zorn KK, et al. (2003) Choice of normal ovarian control influences determination of

differentially expressed genes in ovarian cancer expression profiling studies. ClinCancer Res 9(13):4811–4818.

58. Chung J, et al. (2013) Rare cell isolation and profiling on a hybrid magnetic/size-sorting chip. Biomicrofluidics 7:054107.

59. Chung J, et al. (2012) Microfluidic cell sorter (μFCS) for on-chip capture and analysis ofsingle cells. Adv Healthc Mater 1(4):432–436.

60. Daniels VG, Wheater PR, Burkitt HG (1979) Functional Histology: A Text and ColourAtlas (Churchill Livingstone, Edinburgh).

61. Malkov VA, et al. (2009) Multiplexed measurements of gene signatures in differentanalytes using the Nanostring nCounter Assay System. BMC Res Notes 2:80.

62. Geiss GK, et al. (2008) Direct multiplexed measurement of gene expression with color-coded probe pairs. Nat Biotechnol 26(3):317–325.

Peterson et al. PNAS Early Edition | 9 of 9

MED

ICALSC

IENCE

SPN

ASPL

US

Page 10: Ascites analysis by a microfluidic chip allows tumor-cell ...€¦ · Ascites analysis by a microfluidic chip allows tumor-cell profiling Vanessa M. Petersona,b,1, Cesar M. Castroa,c,1,

SUPPORTING INFORMATION

Ascites analysis by a microfluidic chip allows tumor cell profiling

Vanessa Peterson1,4*, Cesar M. Castro1,3*, Jaehoon Chung1, Nathan Miller1,4, Adeeti Ullal1,4, Maria Castano1, Richard T. Penson3, Hakho Lee1, Michael Birrer3, Ralph Weissleder1,2,3#

1 Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA 02114,2 Department of Systems Biology, Harvard Medical School, 200 Longwood Ave, Boston, MA 021153 Massachusetts General Hospital Cancer Center, Harvard Medical School Boston, MA 021144 Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139

-1-

Page 11: Ascites analysis by a microfluidic chip allows tumor-cell ...€¦ · Ascites analysis by a microfluidic chip allows tumor-cell profiling Vanessa M. Petersona,b,1, Cesar M. Castroa,c,1,

Fig. S1. Profiling of cancer cell lines. Twelve different ovarian cancer (OvCA) cell lines, and six benign cell lines (two mesothelial cell lines (LP9, LP3); two benign ovarian cell lines (TIOSE4,TIOSE6); primary human lymphocytes/neutrophils) were tested for their expression levels (λ=signal/background-1) of putative diagnostic protein markers using flow cytometry. For each marker the frequency of cell lines with λ >1.5 (red) are shown on right hand side of the heat map (grey bar) and are rank-ordered by abundance of positive cell lines. The data is categorized into 4 subgroups: i) markers present in malignant cells (Unique Malignant; top left), ii) markers in malignant and benign cells (Ubiquitous, right), iii) markers in benign cells only (Benign; middle left), and iv) markers absent in both cell types (Absent; bottom left). This dataset was used to identify markers for subsequent analysis of primary human samples (Fig. 2). Parenthesis represents different antibodies used for the same marker (Table S2).

-2-

Page 12: Ascites analysis by a microfluidic chip allows tumor-cell ...€¦ · Ascites analysis by a microfluidic chip allows tumor-cell profiling Vanessa M. Petersona,b,1, Cesar M. Castroa,c,1,

Fig. S2. Shows representative histograms of a reduced set of markers and cell lines from the large scale cell profiling data shown in Fig. S1. The reduced set includes 7 different markers(EpCAM, CD24, TAG-72, CA19-9, Vimentin, Calretinin, and CD45) measured across 12 cell lines (8 OvCA, 4 Ctrl) using flow cytometry. The fluorescent signal of each marker is shown in blue and the control is shown in red. The control in this example refers to the secondary antibody without the primary antibody.

-3-

Page 13: Ascites analysis by a microfluidic chip allows tumor-cell ...€¦ · Ascites analysis by a microfluidic chip allows tumor-cell profiling Vanessa M. Petersona,b,1, Cesar M. Castroa,c,1,

Fig. S3. Flow cytometry data of 8 representative clinical samples from the training set (Fig. 2). Histograms for 6 different markers (EpCAM, CD24, TAG-72, CA19-9, Vimentin, or Calretinin) are shown for four OvCA populations (Calretinin-/CD45-) and four mesothelial cell populations (Calretinin+). The primary antibody cocktail consisted of CD45 rat antibody, calretinin rabbit antibody, and mouse antibody for the marker of interest (e.g. either EpCAM, CD24, TAG-72, CA19-9, Calretinin, or Vimentin). The secondary antibody cocktail consisted of three fluorophores: Anti-mouse FITC, Anti-Rabbit AF647, and Anti-Rat PeCY7. Calretinin-AF647/CD45-PECy7 negative cells or Calretinin-AF647 positive cells were then gated on (see Fig. 2 for details). The signal over noise ratio (SNR) was determined by dividing the geometric mean of the signal (blue) over that of the ctrl (red) (λ=sig/ctrl-1). Flow cytometry data was analyzed using FlowJo 7.6.3.

-4-

Page 14: Ascites analysis by a microfluidic chip allows tumor-cell ...€¦ · Ascites analysis by a microfluidic chip allows tumor-cell profiling Vanessa M. Petersona,b,1, Cesar M. Castroa,c,1,

Fig. S4. ROC analyses of training set. ROC curves were plotted for individual markers using the 18 patient samples in the training set (top). The area under the curve (AUC) and the optimal cutoff level were calculated and are summarized in the bottom table. The cutoff values were then used to determine the sensitivity, specificity, accuracy of each individual marker and the V3 and ATCdx panel (Table 3, Table S1).

-5-

Page 15: Ascites analysis by a microfluidic chip allows tumor-cell ...€¦ · Ascites analysis by a microfluidic chip allows tumor-cell profiling Vanessa M. Petersona,b,1, Cesar M. Castroa,c,1,

Fig. S5. Ascites cellular composition and volume. Ascites samples from 65 patients with (blue; n=46) or without (green; n=19) ovarian cancer were analyzed for total cell number (top left), malignant cell number (bottom left; ATCs), cell volume (top right) and fraction of malignant cells compared to total cells (bottom right). Viable cells were counted using trypan blue staining and the Countess cell counter (Invitrogen). Malignant cell number were determined using ATCdx via flow cytometry (see Methods for more details). Data are plotted as waterfall plots.

-6-

Page 16: Ascites analysis by a microfluidic chip allows tumor-cell ...€¦ · Ascites analysis by a microfluidic chip allows tumor-cell profiling Vanessa M. Petersona,b,1, Cesar M. Castroa,c,1,

Fig. S6. ATC chip design and measurements. A representative example of an ascites sample before purification (A) and after on-chip purification (B). Captured cells (green represents DAPI staining) in the 20 µm capture sites of the ATC chip.

-7-

Page 17: Ascites analysis by a microfluidic chip allows tumor-cell ...€¦ · Ascites analysis by a microfluidic chip allows tumor-cell profiling Vanessa M. Petersona,b,1, Cesar M. Castroa,c,1,

Fig. S7. ATC chip design and measurements. (A) The layout of the ATC chip consists of four different sized capture sites (15, 20, 30 and 40 µm; n = 4,925). Capture site dimensions are µm scale. (B) The capture site in the device has a bowl-like structure with an underpass gap that allows smaller cells to pass through. The length of Path A is considerably shorter than Path B resulting in less flow resistance. Due to the flow resistance difference, most of the fluid flows through path A rather than Path B. Cells that attempt to pass through Path A that are larger than the capture site will be trapped while smaller cells will pass through.

-8-

Page 18: Ascites analysis by a microfluidic chip allows tumor-cell ...€¦ · Ascites analysis by a microfluidic chip allows tumor-cell profiling Vanessa M. Petersona,b,1, Cesar M. Castroa,c,1,

Fig. S8. Schematic of on-chip labeling and purification. First, ascites fluid is collected from the patient which contains malignant cells (red) amongst an anti-inflammatory milieu of host cells (green). Ascites cells are added to the chip followed by an antibody cocktail (EpCAM-FITC, Vimentin-PE/Cy7 Calretinin-Biotin/AF647, CD45-Biotin/AF647, Marker-AF555). Streptavidin-coated magnetic particles then bind to the benign mesothelial cells (Calretinin+) and leukocytes (CD45+). A magnet under the inlet captures the benign cells (red) while the malignant cells (green) pass freely through the microchip. The four different size microwells (40, 30, 20,15 µm) allow for capture of the malignant cells while allowing for the typically smaller leukocytes to pass through the device. The ATCdx signature (EpCAM+ and/or Vimentin+/Calretinin-/CD45-) can then be imaged to determine number of ATCs.

-9-

Page 19: Ascites analysis by a microfluidic chip allows tumor-cell ...€¦ · Ascites analysis by a microfluidic chip allows tumor-cell profiling Vanessa M. Petersona,b,1, Cesar M. Castroa,c,1,

Fig. S9. Predictive ATC markers of treatment response. Key treatment response markers are plotted for 6 patients who were analyzed serially and either responded to treatment (left) or progressed (right). Responders typically have proliferation (Ki67, p-Histone 3, p-CyclinD), mRNA translation (p-4E-BP1) and protein translation (p-S6RP) markers downregulated compared to the non-responders. Each marker was measured in duplicate for each time point and the error bars represent the SEM.

-10-

Page 20: Ascites analysis by a microfluidic chip allows tumor-cell ...€¦ · Ascites analysis by a microfluidic chip allows tumor-cell profiling Vanessa M. Petersona,b,1, Cesar M. Castroa,c,1,

Table S1. Sensitivity, specificity, and accuracy of different protein markers in the test set.

Test set (n = 47)Test set (n = 47)Test set (n = 47)Test set (n = 47)Test set (n = 47)Test set (n = 47)Test set (n = 47)

Marker EpCAM CA19-9 CD24 TAG-72 V3 ATCdx

Sensitivity 93.9 35.7 85.7 78.6 15.2 100.0

Specificity 100.0 100.0 100.0 100.0 100.0 100.0

Accuracy 95.7 53.8 89.7 84.6 40.4 100.0

-11-

Page 21: Ascites analysis by a microfluidic chip allows tumor-cell ...€¦ · Ascites analysis by a microfluidic chip allows tumor-cell profiling Vanessa M. Petersona,b,1, Cesar M. Castroa,c,1,

Table S2. List of antibodies used in the study.

Number Biomarker Clone Company Species Dilution1 53BP1 Polyclonal Cell Signaling Rabbit 1 to 1002 B7-H3 185504 R&D Mouse IgG1 1 to 1003 B7-H4 MIH43 Pierce Mouse IgG1 1 to 1004 CA-125 X75 Abcam Mouse IgG1 1 to 1005 CA-19-9 SPM110 Abcam Mouse IgG1 1 to 206 Caldesmon h-CD Dako Mouse IgG1 1 to 307 Calretinin DC8 Invitrogen Rabbit 1 to 1008 Calretinin DAK-Calret 1 Dako Mouse IgG1 1 to 509 CD15 28 AbCam Mouse IgM 1 to 10010 CD24 SN3 A5-2H10 eBioscience Mouse IgG1 1 to 5011 CD44 IM7 Biolegend Rat IgG2b 1 to 5012 CD44v6 2F10 R&D Mouse IgG1 1 to 10013 CD44v9 RV3 Dr. Hideyuki Saya

(Keio University)Rat IgG2a 1 to 250

14 CD45 YTH24.5 Abcam Rat IgG2b 1 to 10015 CD45 H130 Biolegend Mouse IgG1 1 to 5016 CD45-Alexa Fluor

647H130 Biolegend Mouse IgG1 1 to 100

17 CD45 Polyclonal Abcam Rabbit 1 to 10018 CEA (a) M111146 Fitzgerald Mouse IgG1 1 to 10019 CEA (b) M85151A Fitzgerald Mouse IgG1 1 to 10020 CEA (c) M111147 Fitzgerald Mouse IgG1 1 to10021 CEA (d) 487618 R&D Mouse IgG1 1 to 10022 CK18 DA-7 EXBIO Mouse IgG1 1 to 10023 CK19 A53-B/A2.26 Neomarkers (Pierce) Mouse IgG2a 1 to 2024 CK20 Q2 Thermo Scientific Mouse IgG1 1 to 2025 CK7 OV-TL 12/30 Neomarker Mouse IgG1 1 to 2026 CK8 C-43 Affinity Bioreagents Mouse IgG1 1 to 10027 Claudin 3 385021 R&D Mouse IgG2a 1 to 10028 Claudin 7 4D4 Abnova Mouse IgG2a 1 to 5029 Cleaved Caspase 3 5A1E Cell Signaling Rabbit 1 to 10030 Cleaved Caspase 8 18C8 Cell Signaling Rabbit 1 to 10031 Cyclin D1 CD1.1 GeneTex Mouse IgG1 1 to 10032 D2-40 D2-40 Abcam Mouse IgG1 1 to 5033 E-Cadherin HECD-1 Life Technologies Mouse IgG1 1 to 3034 EGFR F4 Abcam Mouse IgG1 1 to 10035 EMA E29 Dako Mouse IgG2a 1 to 3036 EMMPRIN 109403 R&D Mouse IgG2b 1 to 10037 EpCAM (a) MOC-31 Dako Mouse IgG1 1 to 3038 EpCAM (b) BerEP4 Dako Mouse IgG1 1 to 10039 EpCAM-FITC BerEP4 Dako Mouse IgG1 1 to 10040 EpHA2 371805 R&D Mouse IgG2a 1 to 10041 ER (Estrogen

Receptor alpha)6F11 Abcam Mouse IgG1 1 to 100

42 ESE-1 Polyclonal Abcam Rabbit 1 to 10043 FGFR-4 4FR6D3 Biolegend Mouse IgG1 1 to 5044 FOLR1 548908 R&D Mouse IgG1 1 to 10045 FSHR (a) Polyclonal GeneTex Rabbit 1 to 10046 FSHR (b) H-190 Santa Cruz Rabbit 1 to 2047 FSHR (c) Polyclonal Novus Biologicals Rabbit 1 to 10048 FSHR (d) Polyclonal Genetex Rabbit 1 to 100

-12-

Page 22: Ascites analysis by a microfluidic chip allows tumor-cell ...€¦ · Ascites analysis by a microfluidic chip allows tumor-cell profiling Vanessa M. Petersona,b,1, Cesar M. Castroa,c,1,

49 HE4 3F9 Abnova Mouse IgG2b 1 to 5050 Her2 191924 R&D Mouse IgG2b 1 to 10051 Her3 RTJ2 Abcam Mouse IgG1 1 to 10052 Ki67 B56 BD Pharmingen Mouse IgG1 1 to 5053 Ki67-Alexa Fluor 555 B56 BD Pharmingen Mouse IgG1 1 to 3054 Ku80 C48E7 Cell Signaling Rabbit 1 to 10055 MAGP2 Polyclonal Abnova Rabbit 1 to 10056 Mesothelin K1 Abcam Mouse IgG1 1 to 10057 Mesothelioma ME1 Thermo Scientific Mouse IgG1 1 to 5058 MUC1 M01102909 Fitzgerald Mouse IgG1 1 to 10059 MUC18 128018 R&D Mouse IgG1 1 to 10060 MUC2 M53 Neomarker ( Pierce) Mouse IgG2a 1 to 2061 p-4E-BP1 17489 Cell Signaling Rabbit 1 to 10062 P-Cadherin 104805 R&D Mouse IgG1 1 to 10063 p-Cyclin D D29B3 Cell Signaling Rabbit 1 to 10064 p-Histone 3 D2C8 Cell Signaling Rabbit 1 to 10065 p-p44/42 MAPK

(p-ERK 1/2)D13.14.4E Cell Signaling Rabbit 1 to 100

66 p53 1C12 Cell Signaling Mouse IgG1 1 to 10067 Pan Cytokeratin C-11 Axxora Mouse IgG1 1 to 10068 PARP 46D11 Cell Signaling Rabbit 1 to 10069 PAX8 Polyclonal Proteintech Rabbit Poly 1 to 5070 p-p53 FP3.2 Pierce Mouse IgG1 1 to 10071 Podoplanin NZ-1.3 eBioscience Rat IgG2a 1 to 10072 PR (progesterone

receptor)1A6 Millipore Mouse IgG1 1 to 30

73 p-S6RP (a) D57.2.2E Cell Signaling Rabbit 1 to 5074 p-S6RP (b) 2F9 Cell Signaling Rabbit 1 to 5075 S100 6G1 Fitzgerald Mouse IgG1 1 to 20076 S100-A1 1D5 Abgent Mouse IgG1 1 to 10077 S100-A11 2F4 Abgent Mouse IgG2a 1 to 5078 S100-A2 M2 Abgent Mouse IgG2a 1 to 5079 S100-A4 1F12-1G7 Abgent Mouse IgG1 1 to 10080 S100-A6 6B5 Abgent Mouse IgG1 1 to 10081 S100-A7 1A4 Abgent Mouse IgG1 1 to 10082 S100-A8 2H2 Abgent Mouse IgG2a 1 to 10083 S100-A9 1C10 Abgent Mouse IgG 1 to 10084 S100B 472806 R&D Mouse IgG2a 1 to 10085 S100P 4E7 Abnova Mouse IgG2b 1 to 10086 S6RP (a) 5G10 Cell Signaling Rabbit 1 to 5087 S6RP (b) 54D2 Cell Signaling Mouse IgG1 1 to 5088 TAG-72 CC49 Abcam Mouse IgG1 1 to 3089 Thrombomodulin 1009 Dako Mouse IgG1 1 to 3090 Transferrin 29806 R&D Mouse IgG1 1 to 10091 TSPAN8 458811 R&D Rat IgG2b 1 to 10092 uPAR 62022 R&D Mouse IgG1 1 to 10093 VEGF VG1 Dako Mouse IgG1 1 to 3094 VEGFR 2 KDR/EIC Abcam Mouse IgG1 1 to 10095 Vimentin Vim 3B4 Abcam Mouse IgG2a 1 to 10096 Vimentin 280618 R&D Rat IgG2a 1 to 10097 WT1 6F-H2 Millipore Mouse IgG1 1 to 100

-13-

Page 23: Ascites analysis by a microfluidic chip allows tumor-cell ...€¦ · Ascites analysis by a microfluidic chip allows tumor-cell profiling Vanessa M. Petersona,b,1, Cesar M. Castroa,c,1,

Table S3. List of secondary antibodies used in the study.

Fluorophore Biomarker Clone Company Species DilutionAlexa 647 Anti Mouse Polyclonal Cell Signalling Goat 1 to 900FITC Anti Mouse Polyclonal Abcam Goat 1 to 300Dylight 650 Anti Rabbit Polyclonal Abcam Goat 1 to 300PE/Cy7 Anti Rat Poly4054 Biolegend Goat 1 to 300Dylight 488 Anti Rabbit Poly4064 Biolegend Donkey Ig 1 to 300FITC Anti Mouse IgM II/41 eBioscience Rat IgG2a 1 to 150FITC Anti Rat IgG2a RG7/1.30 BD Pharmingen Mouse IgG2b 1 to 150FITC Anti Rat IgG2b G15-337 BD Pharmingen Mouse IgG2b 1 to 150FITC Anti Rabbit RG-96 Sigma Mouse IgG1 1 to 150

-14-