Development and initial clinical testing of a multiplexed circulating tumor cell assay in patients with clear cell renal cell carcinoma

Rory M Bade, Jennifer L Schehr, Hamid Emamekhoo, Benjamin K Gibbs, Tamara S Rodems, Matthew C Mannino, Joshua A Desotelle, Erika Heninger, Charlotte N Stahlfeld, Jamie M Sperger, Anupama Singh, Serena K Wolfe, David J Niles, Waddah Arafat, John A Steinharter, E Jason Abel, David J Beebe, Xiao X Wei, Rana R McKay, Toni K Choueri, Joshua M Lang, Rory M Bade, Jennifer L Schehr, Hamid Emamekhoo, Benjamin K Gibbs, Tamara S Rodems, Matthew C Mannino, Joshua A Desotelle, Erika Heninger, Charlotte N Stahlfeld, Jamie M Sperger, Anupama Singh, Serena K Wolfe, David J Niles, Waddah Arafat, John A Steinharter, E Jason Abel, David J Beebe, Xiao X Wei, Rana R McKay, Toni K Choueri, Joshua M Lang

Abstract

Although therapeutic options for patients with advanced renal cell carcinoma (RCC) have increased in the past decade, no biomarkers are yet available for patient stratification or evaluation of therapy resistance. Given the dynamic and heterogeneous nature of clear cell RCC (ccRCC), tumor biopsies provide limited clinical utility, but liquid biopsies could overcome these limitations. Prior liquid biopsy approaches have lacked clinically relevant detection rates for patients with ccRCC. This study employed ccRCC-specific markers, CAIX and CAXII, to identify circulating tumor cells (CTC) from patients with metastatic ccRCC. Distinct subtypes of ccRCC CTCs were evaluated for PD-L1 and HLA-I expression and correlated with patient response to therapy. CTC enumeration and expression of PD-L1 and HLA-I correlated with disease progression and treatment response, respectively. Longitudinal evaluation of a subset of patients demonstrated potential for CTC enumeration to serve as a pharmacodynamic biomarker. Further evaluation of phenotypic heterogeneity among CTCs is needed to better understand the clinical utility of this new biomarker.

Trial registration: ClinicalTrials.gov NCT03203473 NCT04071223.

Keywords: biomarkers; circulating tumor cells; clear cell renal cell carcinoma; exclusion-based sample preparation; pharmacodynamic; prognostic.

Conflict of interest statement

David J. Beebe and Joshua M. Lang hold equity in Salus Discovery LLC, which has licensed technology utilized in the manuscript. D. J. Beebe holds equity in Tasso, Inc., Stacks for the Future, LLC, and Bellbrook Labs, LLC. Rana R. McKay is a consultant/Advisory Board Member for Bristol‐Myers Squibb, Dendreon, Exelixis, Janssen, Pfizer, Novartis, and Tempus; receives institutional research funding from Bayer and Pfizer. Xiao X. Wei receives institutional research funding from Bristol‐Myers Squibb.

© 2021 The Authors. Published by FEBS Press and John Wiley & Sons Ltd.

Figures

Fig. 1
Fig. 1
Heterogeneity of RCC biomarkers on cells in circulation. Flow cytometric evaluation of the frequency of expression of different biomarkers on CTCs from n = 3 different patients (A, B, or C). The first row of pie graphs represents the distribution of CK‐positive CTCs (gray) vs. CK‐negative / exclusion channel negative cells (purple). The second row of Euler diagrams portrays the frequency of expression of other markers of renal cancer origin (CAIX, CAXII, and EpCAM) within the CK‐positive vs. CK‐negative / exclusion negative cell fractions. Overlapping circles indicate the co‐expression of different biomarkers on the same cells. Subpopulation frequencies are rounded to the nearest whole percent, with percentages 

Fig. 2

Representative Images and Clinical Correlations…

Fig. 2

Representative Images and Clinical Correlations of CTC Enumeration. (A) Schematic overview of the…

Fig. 2
Representative Images and Clinical Correlations of CTC Enumeration. (A) Schematic overview of the method used to isolate and identify CTCs from whole blood. (B) Three representative CTCs and one WBC from a patient blood sample. Examples of CTCs with intact nuclei that were CAXII S+ (top), Double+ (middle), CK S+ (bottom), all Exclusion. Scale bars represent 5 microns. (C) Enumeration of all CTCs for n = 20 patients and n = 2 healthy donors where each symbol represents the number of CTCs from one patient. Significance was determined by a one tailed Mann‐Whitney test p < 0.05 (*). Error bars represent standard error. ROC curve showing sensitivity and specificity of total CTC number to differentiate between patients whose disease was progressing vs responding. (D) CTC enumeration separated by phenotype for the same n = 20 patients: CAXII single+ (green), double+ (blue), and CK single+ (gray). Percentages of different CTC subpopulations are listed in supplementary table 2. (E) two‐tailed Mann‐Whitney tests and (F) ROC evaluation of the ability of the different populations of CTCs to differentiate between therapeutic progression or response for the same n = 20 patients. ROC curve parameters are tabulated in table below ROC curves.

Fig. 3

Quantification of Immunotherapy Biomarkers on…

Fig. 3

Quantification of Immunotherapy Biomarkers on CTCs. (A) Positive and negative expression of PD‐L1…

Fig. 3
Quantification of Immunotherapy Biomarkers on CTCs. (A) Positive and negative expression of PD‐L1 and HLA‐I expression on CTCs is shown with scale bars representing 5 microns. (B) Biomarker evaluation was performed as a single ‘All CTCs’ (black) population as well as by each subpopulation, CK single+ (gray), double+ (blue), and CAXII single+ (green), where each dot represents a single CTC. Average CTC expression is represented by a black bar through each population, and % positive is defined as the frequency of CTCs with biomarker expression above the red dotted line cutoff for positivity. Representative dataset shown from one patient sample. C) The % positive and average expression of CTCs from each patient were determined for PD‐L1 and HLA‐I for cohort = 20 patients. Samples without CTCs are indicated with an ‘X’.

Fig. 4

Clinical Utility of Biomarker Evaluation…

Fig. 4

Clinical Utility of Biomarker Evaluation with Different CTC Populations. Scatter plots showing the…

Fig. 4
Clinical Utility of Biomarker Evaluation with Different CTC Populations. Scatter plots showing the average expression of either PD‐L1 or HLA‐I on either CK + or CAXII S + CTCs in patients either responding or stable (Res) vs. progressing (Prog) on either ICIs or TKIs. AUC values annotated on graphs are a measure of the ability of the assay to identify patients who are progressing. Data represent 42 total samples from 24 unique patients.

Fig. 5

Longitudinal Sampling of CTCs. CTC…

Fig. 5

Longitudinal Sampling of CTCs. CTC biomarker evaluation of 4 patients (#21‐24) over time…

Fig. 5
Longitudinal Sampling of CTCs. CTC biomarker evaluation of 4 patients (#21‐24) over time were compared to therapeutic history (colored bars) and radiographic assessment of response (numbers on graphs and corresponding radiographic scan images) for each patient. Graphs represent enumeration for each CTC population. Dashed line represents the optimal cutoff for CK + CTC number (2.6) identified in Figure 2.
Fig. 2
Fig. 2
Representative Images and Clinical Correlations of CTC Enumeration. (A) Schematic overview of the method used to isolate and identify CTCs from whole blood. (B) Three representative CTCs and one WBC from a patient blood sample. Examples of CTCs with intact nuclei that were CAXII S+ (top), Double+ (middle), CK S+ (bottom), all Exclusion. Scale bars represent 5 microns. (C) Enumeration of all CTCs for n = 20 patients and n = 2 healthy donors where each symbol represents the number of CTCs from one patient. Significance was determined by a one tailed Mann‐Whitney test p < 0.05 (*). Error bars represent standard error. ROC curve showing sensitivity and specificity of total CTC number to differentiate between patients whose disease was progressing vs responding. (D) CTC enumeration separated by phenotype for the same n = 20 patients: CAXII single+ (green), double+ (blue), and CK single+ (gray). Percentages of different CTC subpopulations are listed in supplementary table 2. (E) two‐tailed Mann‐Whitney tests and (F) ROC evaluation of the ability of the different populations of CTCs to differentiate between therapeutic progression or response for the same n = 20 patients. ROC curve parameters are tabulated in table below ROC curves.
Fig. 3
Fig. 3
Quantification of Immunotherapy Biomarkers on CTCs. (A) Positive and negative expression of PD‐L1 and HLA‐I expression on CTCs is shown with scale bars representing 5 microns. (B) Biomarker evaluation was performed as a single ‘All CTCs’ (black) population as well as by each subpopulation, CK single+ (gray), double+ (blue), and CAXII single+ (green), where each dot represents a single CTC. Average CTC expression is represented by a black bar through each population, and % positive is defined as the frequency of CTCs with biomarker expression above the red dotted line cutoff for positivity. Representative dataset shown from one patient sample. C) The % positive and average expression of CTCs from each patient were determined for PD‐L1 and HLA‐I for cohort = 20 patients. Samples without CTCs are indicated with an ‘X’.
Fig. 4
Fig. 4
Clinical Utility of Biomarker Evaluation with Different CTC Populations. Scatter plots showing the average expression of either PD‐L1 or HLA‐I on either CK + or CAXII S + CTCs in patients either responding or stable (Res) vs. progressing (Prog) on either ICIs or TKIs. AUC values annotated on graphs are a measure of the ability of the assay to identify patients who are progressing. Data represent 42 total samples from 24 unique patients.
Fig. 5
Fig. 5
Longitudinal Sampling of CTCs. CTC biomarker evaluation of 4 patients (#21‐24) over time were compared to therapeutic history (colored bars) and radiographic assessment of response (numbers on graphs and corresponding radiographic scan images) for each patient. Graphs represent enumeration for each CTC population. Dashed line represents the optimal cutoff for CK + CTC number (2.6) identified in Figure 2.

References

    1. Jemal A, Bray F, Center MM, Ferlay J, Ward E & Forman D (2011) Global cancer statistics. CA Cancer J Clin 61, 69–90.
    1. Howlader N, Ries LA, Mariotto AB, Reichman ME, Ruhl J & Cronin KA (2010) Improved estimates of cancer‐specific survival rates from population‐based data. J Natl Cancer Inst 102, 1584–1598.
    1. Dorai T, Sawczuk I, Pastorek J, Wiernik PH & Dutcher JP (2006) Role of carbonic anhydrases in the progression of renal cell carcinoma subtypes: proposal of a unified hypothesis. Cancer Invest 24, 754–779.
    1. Motzer R, Jonasch E, Michaelson M, Nandagopal L, Gore J, George S, Alva A, Haas N, Harrison M, Plimack Eet al., (2019) NCCN guidelines insights: kidney cancer, version 2.2020. Journal of the National Comprehensive Cancer Network: JNCCN 17.
    1. Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, Tykodi SS, Sosman JA, Procopio G, Plimack ERet al., (2015) Nivolumab versus Everolimus in advanced renal‐cell carcinoma. N Engl J Med 373, 1803–1813.
    1. Iacovelli R, Nole F, Verri E, Renne G, Paglino C, Santoni M, Cossu Rocca M, Giglione P, Aurilio G, Cullura Det al., (2016) Prognostic role of PD‐L1 expression in renal cell carcinoma. A systematic review and meta‐analysis. Target Oncol 11, 143–148.
    1. Rodriguez‐Vida A, Strijbos M & Hutson T (2016) Predictive and prognostic biomarkers of targeted agents and modern immunotherapy in renal cell carcinoma. ESMO Open 1.
    1. Kitamura H, Honma I, Torigoe T, Asanuma H, Sato N & Tsukamoto T (2007) Down‐regulation of HLA class I antigen is an independent prognostic factor for clear cell renal cell carcinoma. J Urol 177, 1269–1272.discussion 1272.
    1. Wang J, Liu L, Qu Y, Xi W, Xia Y, Bai Q, Xiong Y, Long Q, Xu J & Guo J (2018) HLA class I expression predicts prognosis and therapeutic benefits from tyrosine kinase inhibitors in metastatic renal‐cell carcinoma patients. Cancer Immunol Immunother 67, 79–87.
    1. Gerlinger M, Horswell S, Larkin J, Rowan AJ, Salm MP, Varela I, Fisher R, McGranahan N, Matthews N, Santos CRet al., (2014) Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nat Genet 46, 225–233.
    1. Kalbasi A & Ribas A (2020) Tumour‐intrinsic resistance to immune checkpoint blockade. Nat Rev Immunol 20, 25–39.
    1. Yuan J, Liu S, Yu Q, Lin Y, Bi Y, Wang Y & An R (2013) Down‐regulation of human leukocyte antigen class I (HLA‐I) is associated with poor prognosis in patients with clear cell renal cell carcinoma. Acta Histochem 115, 470–474.
    1. Eichelberg C, Chun FK, Bedke J, Heuer R, Adam M, Moch H, Terracciano L, Hinrichs K, Dahlem R, Fisch Met al., (2013) Epithelial cell adhesion molecule is an independent prognostic marker in clear cell renal carcinoma. Int J Cancer 132, 2948–2955.
    1. Zimpfer A, Maruschke M, Rehn S, Kundt G, Litzenberger A, Dammert F, Zettl H, Stephan C, Hakenberg OW & Erbersdobler A (2014) Prognostic and diagnostic implications of epithelial cell adhesion/activating molecule (EpCAM) expression in renal tumours: a retrospective clinicopathological study of 948 cases using tissue microarrays. BJU Int 114, 296–302.
    1. Bluemke K, Bilkenroth U, Meye A, Fuessel S, Lautenschlaeger C, Goebel S, Melchior A, Heynemann H, Fornara P & Taubert H (2009) Detection of circulating tumor cells in peripheral blood of patients with renal cell carcinoma correlates with prognosis. Cancer Epidemiol Biomarkers Prev 18, 2190–2194.
    1. Lee C, Park JW, Suh JH, Nam KH & Moon KC (2013) Histologic variations and immunohistochemical features of metastatic clear cell renal cell carcinoma. Korean J Pathol 47, 426–432.
    1. Gradilone A, Iacovelli R, Cortesi E, Raimondi C, Gianni W, Nicolazzo C, Petracca A, Palazzo A, Longo F, Frati L & et al., (2011) Circulating tumor cells and "suspicious objects" evaluated through Cell Search(R) in metastatic renal cell carcinoma. Anticancer Res 31, 4219–4221.
    1. Allard WJ, Matera J, Miller MC, Repollet M, Connelly MC, Rao C, Tibbe AG, Uhr JW & Terstappen LW (2004) Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseases. Clin Cancer Res 10, 6897–6904.
    1. Liu S, Tian Z, Zhang L, Hou S, Hu S, Wu J, Jing Y, Sun H, Yu F, Zhao Let al., (2016) Combined cell surface carbonic anhydrase 9 and CD147 antigens enable high‐efficiency capture of circulating tumor cells in clear cell renal cell carcinoma patients. Oncotarget 7, 59877–59891.
    1. Schehr JL, Schultz ZD, Warrick JW, Guckenberger DJ, Pezzi HM, Sperger JM, Heninger E, Saeed A, Leal T, Mattox Ket al., (2016) High specificity in circulating tumor cell identification is required for accurate evaluation of programmed death‐ligand 1. PLoS One 11, e0159397.
    1. Wang CH, Rong MY, Wang L, Ren Z, Chen LN, Jia JF, Li XY, Wu ZB, Chen ZN & Zhu P (2014) CD147 up‐regulates calcium‐induced chemotaxis, adhesion ability and invasiveness of human neutrophils via a TRPM‐7‐mediated mechanism. Rheumatology (Oxford) 53, 2288–2296.
    1. El‐Heliebi A, Kroneis T, Zohrer E, Haybaeck J, Fischereder K, Kampel‐Kettner K, Zigeuner R, Pock H, Riedl R, Stauber Ret al., (2013) Are morphological criteria sufficient for the identification of circulating tumor cells in renal cancer? J Transl Med 11, 214.
    1. Garcia‐Donas J, Leon LA, Esteban E, Vidal‐Mendez MJ, Arranz JA, Garcia Del Muro X, Basterretxea L, Gonzalez Del Alba A, Climent MA, Virizuela JAet al., (2017) A prospective observational study for assessment and outcome association of circulating endothelial cells in clear cell renal cell carcinoma patients who show initial benefit from first‐line treatment. The CIRCLES (CIRCuLating Endothelial cellS) Study (SOGUG‐CEC‐2011‐01). Eur Urol Focus 3, 430–436.
    1. Gruenwald V, Beutel G, Schuch‐Jantsch S, Reuter C, Ivanyi P, Ganser A & Haubitz M(2010) Circulating endothelial cells are an early predictor in renal cell carcinoma for tumor response to sunitinib. BMC Cancer 10, 695.
    1. Namdarian B, Tan KV, Fankhauser MJ, Nguyen TT, Corcoran NM, Costello AJ & Hovens CM (2010) Circulating endothelial cells and progenitors: potential biomarkers of renal cell carcinoma. BJU Int 106, 1081–1087.
    1. Vroling L, van der Veldt AA, de Haas RR, Haanen JB, Schuurhuis GJ, Kuik DJ, van Cruijsen H, Verheul HM, van den Eertwegh AJ, Hoekman Ket al., (2009) Increased numbers of small circulating endothelial cells in renal cell cancer patients treated with sunitinib. Angiogenesis 12, 69–79.
    1. Schodel J, Grampp S, Maher ER, Moch H, Ratcliffe PJ, Russo P & Mole DR (2016) Hypoxia, hypoxia‐inducible transcription factors, and renal cancer. Eur Urol 69, 646–657.
    1. Supuran C & Winum J (2015) Carbonic anhydrase IX inhibitors in cancer therapy: an update. Future medicinal chemistry 7, 1407–1414.
    1. McDonald P, Winum J, Supuran C & Dedhar S (2012) Recent developments in targeting carbonic anhydrase IX for cancer therapeutics. Oncotarget 3, 84–97.
    1. Neri D & Supuran C (2011) Interfering with pH regulation in tumours as a therapeutic strategy. Nat Rev Drug Discov 10, 767–777.
    1. Türeci O, Sahin U, Vollmar E, Siemer S, Göttert E, Seitz G, Parkkila AK, Shah GN, Grubb JH, Pfreundschuh M & et al., (1998) Human carbonic anhydrase XII: cDNA cloning, expression, and chromosomal localization of a carbonic anhydrase gene that is overexpressed in some renal cell cancers. PNAS 95, 7608–7613.
    1. Mboge M, McKenna R & Frost S (2015) Advances in anti‐cancer drug development targeting carbonic anhydrase IX and XII. Topics in anti‐cancer research 5, 3–42.
    1. Waheed A & Sly W (2017) Carbonic anhydrase XII functions in health and disease. Gene 623, 33–40.
    1. BioGPS – your Gene Portal System . (Accessed 30Dec2020)
    1. Berry SM, Chin EN, Jackson SS, Strotman LN, Goel M, Thompson NE, Alexander CM, Miyamoto S, Burgess RR & Beebe DJ (2014) Weak protein‐protein interactions revealed by immiscible filtration assisted by surface tension (IFAST). Anal Biochem 447, 133–140.
    1. Sperger JM, Strotman LN, Welsh A, Casavant BP, Chalmers Z, Horn S, Heninger E, Thiede SM, Tokar J, Gibbs BKet al., (2017) Integrated analysis of multiple biomarkers from circulating tumor cells enabled by exclusion‐based analyte isolation. Clin Cancer Res 23, 746–756.
    1. Pezzi HM, Guckenberger DJ, Schehr JL, Rothbauer J, Stahlfeld C, Singh A, Horn S, Schultz ZD, Bade RM, Sperger JMet al., (2018) Versatile exclusion‐based sample preparation platform for integrated rare cell isolation and analyte extraction. Lab Chip 18, 3446–3458.
    1. Pezzi H, Niles DJ, Schehr JL, Beebe DJ & Lang JM (2018) Integration of magnetic bead‐based cell selection into complex isolations. ACS Omega 3, 3908–3917.
    1. Verschoor CP, Lelic A, Bramson JL & Bowdish DM (2015) An introduction to automated flow cytometry gating tools and their implementation. Front Immunol 6, 380.
    1. Lopez JI & Angulo JC (2018) Pathological bases and clinical impact of intratumor heterogeneity in clear cell renal cell carcinoma. Curr Urol Rep 19, 3.
    1. Eckel‐Passow JE, Ho TH, Serie DJ, Cheville JC, Houston Thompson R, Costello BA, Dong H, Kwon ED, Leibovich BC & Parker AS (2020) Concordance of PD‐1 and PD‐L1 (B7–H1) in paired primary and metastatic clear cell renal cell carcinoma. Cancer Med 9, 1152–1160.
    1. Bersanelli M, Gnetti L, Varotti E, Ampollini L, Carbognani P, Leonardi F, Rusca M, Campanini N, Ziglioli F, Dadomo CIet al., (2019) Immune context characterization and heterogeneity in primary tumors and pulmonary metastases from renal cell carcinoma. Immunotherapy 11, 21–35.
    1. Skinnider BF, Folpe AL, Hennigar RA, Lim SD, Cohen C, Tamboli P, Young A, de Peralta‐Venturina M & Amin MB (2005) Distribution of cytokeratins and vimentin in adult renal neoplasms and normal renal tissue: potential utility of a cytokeratin antibody panel in the differential diagnosis of renal tumors. Am J Surg Pathol 29, 747–754.
    1. Parkkila S, Rajaniemi H, Parkkila A‐K, Kivelä J, Waheed A, Pastoreková S, Pastorek J & Sly WS (2000) Carbonic anhydrase inhibitor suppresses invasion of renal cancer cells in vitro. PNAS 97, 2220–2224.
    1. Chiche J, Ilc K, Laferriere J, Trottier E, Dayan F, Mazure NM, Brahimi‐Horn MC & Pouyssegur J (2009) Hypoxia‐inducible carbonic anhydrase IX and XII promote tumor cell growth by counteracting acidosis through the regulation of the intracellular pH. Cancer Res 69, 358–368.
    1. Wei XX, McKay RR, Gray KP, Stadler WM, McDermott DF, McGregor BA, Agarwal N, Kyriakopoulos C, Carneiro BA, Rose TLet al., (2018) Optimized management of nivolumab (NIVO) and ipilimumab (IPI) in advanced renal cell carcinoma (OMNIVORE): a response‐based phase II study. Poster Presentation. (2018) ASCO Annual Meeting, June 1–5, 2018. Chicago IL.

Source: PubMed

3
Subskrybuj