Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

The Clinicopathologic and Prognostic Value of Altered Chromosome 17 Centromere Copy Number in HER2 Fish Equivocal Breast Carcinomas

  • Hongfei Ji ,

    Contributed equally to this work with: Hongfei Ji, Qijia Xuan

    Affiliation Department of Cancer Molecular and Biology, Cancer Institute of Harbin Medical University, Harbin, China

  • Qijia Xuan ,

    Contributed equally to this work with: Hongfei Ji, Qijia Xuan

    Affiliation Department of Medical Oncology, Tumor Hospital of Harbin Medical University, Harbin, China

  • Abiyasi Nanding,

    Affiliation Department of Pathology, Tumor Hospital of Harbin Medical University, Harbin, China

  • Haiyu Zhang,

    Affiliation Department of Biostatistics, Harbin Medical University, Harbin, China

  • Qingyuan Zhang

    zqyxsci@126.com

    Affiliations Department of Cancer Molecular and Biology, Cancer Institute of Harbin Medical University, Harbin, China, Department of Medical Oncology, Tumor Hospital of Harbin Medical University, Harbin, China

Abstract

Chromosome 17 centromere (CEP17) gain is frequently observed in breast cancer by fluorescence in situ hybridization (FISH). To address the biologic characteristics and clinical significance of CEP17 gain in a large population of breast cancer patients, we performed FISH on a series of 770 breast cancer tissues to evaluate the status of human epidermal growth factor receptor 2 (HER2) gene and CEP17 by immunohistochemistry (IHC) and FISH. Among the 770 specimens, 184 cases showed CEP17 gain (23.9%). Histological grade, nodal status, HER2 by IHC, Ki 67 index, and p53 expression were significantly different between CEP17 gain tumors and HER2-positive tumors. In contrast with HER2-negative tumors, CEP17 gain tumors showed higher histological grade, higher HER2 score by IHC, and higher Ki 67 index. The patients with CEP17 gain tumors had an intermediate survival between HER2-negative and HER2-positive patients. By comparison to HER2-negative and HER2-positive patients, survival in luminal B patients with CEP17 gain tumors also fell in between. In conclusion, CEP17 gain tumors show specific differences compared with HER2-negative and HER2-positive tumors in clinical parameters and prognosis.

Introduction

Breast cancer is one of the most common cancers in women and one of the leading causes of death among women [13]. Human epidermal growth factor 2 (HER2), which is located on the long arm of chromosome 17 (17q12-21-21.32), is an important oncogene in breast cancer [45]. Many studies have demonstrated overexpression and amplification of the HER2 transmembrane tyrosine kinase receptor in approximately 20–30% of breast cancer cases, and this is associated with not only poor clinical outcome, but also response to clinical therapies [68]. Therefore, it is crucial to accurately determine HER2 status in breast cancer patients for selecting the appropriate therapeutic regimen.

Fluorescence in situ hybridization (FISH) and immunohistochemistry (IHC) are the most common testing methods and commonly used for the assessment of HER2 status. Although sufficient evidence indicates that FISH and IHC can accurately and efficiently define HER2 positivity in breast cancer, some specimens show ambiguous HER2 results, which are mostly due to CEP17 gain, a common genotypic abnormality [912]. Few studies has specifically examined CEP17 status and compared differences of potential effects, clinical parameters, and prognosis with HER2-negative status and-positive status. The aim of the present study was to analyze the biologic characteristics and clinical significance of CEP17 gain and explore the impact on luminal B molecular subclassification combined with estrogen receptor (ER) and progesterone receptor (PR) expression in a large population of breast cancer patients.

Materials and Methods

Ethics Statement

This study, including the procedures for patient enrollment and recruitment, was approved by the Institutional Review Board of the Affiliated Tumor Hospital of Harbin Medical University, and all patients who participated in the study provided written informed consent.

Patient samples

We evaluated the tissues of patients with invasive breast cancer that showed equivocal or positive HER2 status on IHC. Patient samples were analyzed by FISH from 1/1/2007 to 5/30/2009 in the Tumor Hospital of Harbin Medical University. We excluded the patients who were with metastatic lesions and receiving non-standard treatment. The remaining 670 patients and the 100 patients with negative HER2 status on IHC were enrolled in the study. The date of surgery was used to represent the beginning of the follow-up period and follow-ups were terminated in May 2014. Patients who developed recurrence or metastasis were identified during the follow-ups by tumor marker measurements and adequate diagnostic imaging modalities.

Methods

IHC.

All breast cancer tissues were formalin-fixed and paraffin-embedded. For each specimen, a 4-micrometers-thick tissue section was provided on a slide. The immunohistochemical analysis was executed as depicted previously [13]. In brief, slides were incubated with HER2 polyclonal antibody (Dako, California, USA), P53 (1:500, DO-7; Dako), Ki-67 (1:300, MIB-1; Dako). The stained tissues were evaluated according to the densities of staining and the number of stained cells. Tissues were considered positive for Ki-67 with more than 20% stained of the cells examined. Any staining of cells was deemed as P53 positive. The staining of cell membrane was considered HER2 protein expression. The slides have been reevaluated by two pathologists to assess HER2 status following the 2013 ASCO/CAP recommendations (Fig 1) [10]. The positive controls were the sections with strong membrane staining. Each section was scanned at ×100 and×400 magnification by microscope (Olympus BX51).

thumbnail
Fig 1. Classification of evaluation of HER2 protein expression by IHC of the invasive component of a breast cancer specimen.

https://doi.org/10.1371/journal.pone.0132824.g001

FISH.

FISH analysis was performed on paraffinized 5-micrometers-thick sections using the Vysis LSI HER2 SpectrumOrange and CEP17 SpectrumGreen Dual Color DNA probe kit (VysisPath Vysion, Abbot Laboratories, Illinois, USA) in line with the manufacturer’s instructions. In short, pretreated procedure comprised the following steps: sections baking, deparaffin, dehydration, deproteinization and refixation. Then, according to the protocol, the sections were denatured and hybridized hybridization oven. Finally the sections were washed, counterstained with 4',6-Diamidino-2-Phenylindole (DAPI, invitrogen, NY, USA) for signal calculation. Scoring of HER2 and CEP17 probe signals were achieved by a fluorescence microscope (Olympus BX51). The slides have been reevaluated by two independent and certified pathologists and with identified the invasive area. The pathologists assessed HER2 status following the 2013 ASCO/CAP recommendations [10]. FISH results were interpreted depending on two different scoring methods: (1) based on absolute HER2 gene copy number or (2) based on the ratio of HER2 gene/CEP17 copy number (HER2/CEP17 ratio). As recommended by the ASCO/CAP guidelines, a HER2/CEP17 ratio of less than 2.0 with an average HER2 copy number less than 4.0 signals/cell was considered HER2 negative, and a HER2/CEP17 ratio of higher than or equal to 2.0 or an HER2/CEP17 ratio of less than 2.0 with an average HER2 copy number higher than 6.0 signals/cell was considered HER2 positive. CEP17 gain was defined as an average CEP17 copy number between 3.76 and 6.

Statistical methods

The Chi-square test were used for comparing clinicopathologicfeatures between tumor groups. The time of disease-free survival was defined as the time between diagnosis and local recurrence, distant metastasis, or death from breast cancer. The survival curves were analyzed by the Kaplan–Meier method and the comparison between curves was evaluated by the log-rank test. All the statistical analyses were accomplished by the IBM SPSS statistics version 20.0 (SPSS Inc. Illinois, USA). Statistical significance was determined as p-values <0.05.

Results

Clinicopathologic characteristics of CEP17 gain tumors

Representative images of evaluations of HER2 expression by IHC and FISH, respectively, are presented in Figs 2 and 3. Among the 770 specimens studied by IHC and FISH, 184 cases showed CEP17 gain (23.9%). The patients were divided into 3 groups by HER2 results on FISH (HER2-negative, HER2-positive and CEP17 gain). Table 1 shows the clinicopathologic parameters of the patients in the 3 groups. In contrast with HER2-negative tumors, CEP17 gain tumors presented higher histological grade (P = 0.042), higher HER2 score by IHC (P<0.001), and higher Ki67 index (P = 0.030). Histological grade (P<0.001), nodal status (P<0.001), HER2 score by IHC (P<0.001), Ki 67 index (P<0.001), and P53 expression (P = 0.003) were strikingly different between HER2-positive tumors and CEP17 gain tumors. By comparison with HER2-negative tumors, the HER2-positive tumors expressed higher histological grade (P = 0.026), more lymph mode metastasis (P<0.001), higher HER2 score by IHC (P<0.001), higher Ki 67 status (P<0.001) and higher frequency of P53 positive status (P<0.001). Taken together, these data indicate that CEP17 gain tumors are significantly different from HER2-negative and HER2-positive tumors.

thumbnail
Fig 2. HER2 protein expression detected by IHC.

(×400) A: Negative (score 0). B: Negative (score 1+). C: Equivocal (score 2+). D: Positive (score 3+).

https://doi.org/10.1371/journal.pone.0132824.g002

thumbnail
Fig 3. HER2 gene status identified by FISH.

Red signals represent HER2 gene, green signals represent CEP17 (×600). A: HER2 gene negative case; B: HER2 gene positive case, HER2/CEP17 ratio of higher than 2.0; C: HER2 gene positive case, HER2 signals are clustered; D: HER2 gene with CEP17 gain, average CEP17 copy number ≥3.76 to 6.

https://doi.org/10.1371/journal.pone.0132824.g003

thumbnail
Table 1. Distribution of clinicopathologic features in CEP17 gain tumors compared with HER2–negative and HER2–positive tumors defined by FISH.

https://doi.org/10.1371/journal.pone.0132824.t001

Survival curves characteristics of the CEP17 gain patients

We preformed Kaplan–Meier survival analysis in the three groups (CEP17 gain, HER2- negative and HER2-positive patients). And the result demonstrated that disease-free survival in patients with CEP17 gain tumors is shorter than HER2-negative patients, and longer compared with HER2-positive patients. The patients with CEP17 gain tumors had an intermediate survival (P = 0.014) (Fig 4).

thumbnail
Fig 4. Kaplan–Meier analysis of disease-free survival time in the enrolled patients.

https://doi.org/10.1371/journal.pone.0132824.g004

Survival curves characteristics of CEP17 gain in the luminal B patients

The characteristics of luminal B tumors present ER/PR positive, HER2 negative and Ki67 index ≥14% or ER/PR positive and HER2 positive on IHC. Despite expressing ER, the luminal B subtype confers increased risk of early relapse with endocrine therapy compared with the luminal A subtype [1416]. We divided the luminal B patients into three subgroups by HER-2 signal number. HER2-positive, HER2-negative and patients with CEP17 gain tumors. Among the luminal B patients, Kaplan–Meier survival curves (Fig 5) illustrated different disease-free survival in three subgroups patients., the survival time in patients with CEP17 gain tumors was between (P = 0.025) HER2-negative and HER2-positive patients.

thumbnail
Fig 5. Kaplan–Meier analysis of disease-free survival time in the luminal B patients.

https://doi.org/10.1371/journal.pone.0132824.g005

Discussion

Breast cancer, one of heterogeneous diseases, is categorized into four major subtypes based on the breast cancer-related receptors ER, PR, and HER2: luminal A, luminal B, HER2+/ER−, and basal-like [1516]. These subtypes exhibit distinct differences in a variety of aspects, including genetic alterations, clinical features, responses to specific chemotherapy, and prognosis [1618]. Therefore, accurate assessment of ER, PR, and HER2 status is a key point for the classification of breast cancer. Several limitations and disadvantages in the identification of HER2 status compared with ER and PR still exist. As a consequence, a portion of breast cancer patients have been determined as equivocal status instead of specifically positive or negative status.

A growing body of recent studies suggested that CEP17 gain is the major mechanism underlying HER2 equivocal status [1824]. The CEP17 probe was used to measure the copy number of CEP17. Several different thresholds (between two and four copies per cell) have been used regarding CEP17 gain because of a lack of official guidelines [2526]. Based on these varying thresholds, the frequency of CEP17 gain in breast cancer specimens varies from 13% to 46% [2729]. In this study, the threshold for CEP17 gain was used 3.76 to 6 copies, and 23.9% specimens (184 in 770) were observed to harbor CEP17 gain.

There are several reasons for choosing the threshold of 3.76 to 6 copies per cell. Firstly, abnormality of CEP17 number is generally can be divided into, CEP17 low polysomy (2.26 to 3.75 signals per cell), and CEP17 high polysomy (3.76 to 6 signals per cell) [3031]. According to published reports, specimens with less than 3.75 copies (hypodisomy and low polysomy) were close to disomy. And in the high polysomy and low polysomy patients, HER2 protein expression was significantly different [3234]. Furthermore, this threshold will confer a precise definition CEP17 gain to the specimens identified as HER2 equivocal status [10]. Using the threshold, these equivocal specimens would be stratified into the CEP17 gain status and HER2-negative status. Most importantly, our results suggest that the CEP17 gain tumors are significantly different from HER2-negative and HER2-positive tumors in severity-related parameters. For example, histological grade, Ki67 index, nodal status, and p53 expression. In addition, survival analysis indicated that the survival rates in CEP17 gain and HER2-negative and HER2-positive subgroups were significantly different. When the luminal B patients were stratified into three subgroups by FISH results, the significance differences were also observed between the different subgroups. Previous studies have shown controversial results regarding the association between CEP17 gain and clinical parameters, and only a few studies could provide evidence about clinicopathologic value of chromosome gain 17 polysomy, specifically about prognosis [3539]. We speculate that the lower threshold, along with genetic heterogeneity, may be responsible for such equivocal results.

In summary, we have investigated the frequency and clinical significance of CEP17 gain in 770 breast cancer specimens. Based on the threshold, CEP17 gain tumors are significantly different from HER2-negative and HER2-positive tumors. This study provides applicable implications for the definition of CEP17 gain that argue against the previous classification. Patients with CEP17 gain should be subtyped and can be distinguished from patients with HER2 gene amplification or no amplification. This study will help to determine the best therapeutic response to HER2-targeted therapy in breast cancer patients with CEP17 gain.

Author Contributions

Conceived and designed the experiments: QZ HJ. Performed the experiments: QX HJ. Analyzed the data: HZ. Contributed reagents/materials/analysis tools: AN. Wrote the paper: HJ QX.

References

  1. 1. DeSantis C, Ma J, Bryan L, Jemal A. Breast cancer statistics, 2013. CA. Cancer J Clin. 2014; 64: 52–62. pmid:24114568
  2. 2. De Felice F, Musio D, Bulzonetti N, Raffetto N, Tombolini V. Relationship of clinical and pathologic nodal staging in locally advanced breast cancer: current controversies in daily practice? J Clin Med Res. 2014; 6: 409–413. pmid:25247013
  3. 3. Chopra I, Chopra A. Follow-up care for breast cancer survivors: improving patient outcomes. Patient Relat Outcome Meas. 2014; 5: 71–85. pmid:25210481
  4. 4. Iqbal N, Iqbal N. Human epidermal growth factor receptor 2 (HER2) in cancers: overexpression and therapeutic implications. Mol Biol Int. 2014; 2014: 852748. pmid:25276427
  5. 5. Cuadros M, Cano C, López FJ, Talavera P, García-Peréz I, Blanco A, et al. HER2 status in breast cancer: experience of a Spanish National Reference Centre. Clin Transl Oncol. 2011; 13: 335–340. pmid:21596662
  6. 6. Moelans CB, de Weger RA, van Diest PJ. Chromosome 17 Polysomy without HER2 Amplification Does Not Predict Response to Lapatinib in Metastatic Breast Cancer–Letter. Clin Cancer Res. 2010; 16: 6177. pmid:21169261
  7. 7. Gaiser T, Rüschoff J, Moll R. In situ hybridization in clinical pathology. Significance of polysomy 17 for HER2 determination and genetic tumor heterogeneity in breast cancer. Pathologe. 2012; 2: 307–310.
  8. 8. Hanna WM, Rüschoff J, Bilous M, Coudry RA, Dowsett M, Osamura RY, et al. HER2 in situ hybridization in breast cancer: clinical implications of polysomy 17 and genetic heterogeneity. Mod Pathol.2013; 27: 4–18. pmid:23807776
  9. 9. Hammond ME, Hayes DF, Wolff AC. Clinical notice for American Society of Clinical Oncology-College of American Pathologists guideline recommendations on ER/PgR and HER2 testing in breast cancer. J Clin Oncol. 2011; 29: e458. pmid:21502545
  10. 10. Wolff AC, Hammond ME, Hicks DG, Dowsett M, McShane LM, Allison KH, et al. Recommendations for Human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American pathologists clinical practice guideline update. J Clin Oncol. 2013; 31: 3997–4013. pmid:24101045
  11. 11. Jiang H, Bai X, Meng F, Zhang C, Zhang X. Evaluation of chromosome 17 polysomy in breast cancer by FISH analysis of whole nuclei, and its clinicopathological significance. Oncol Lett. 2014; 7: 1954–1958. pmid:24932267
  12. 12. Wang S, Hossein Saboorian M, Frenkel EP, Haley BB, Siddiqui MT, Gokaslan S, et al. Aneusomy 17 in breast cancer: its role in HER-2/neu proteinexpression and implication for clinical assessment of HER-2/neu status. ModPathol. 2002; 15:137–145.
  13. 13. Liu Y, Ma L, Liu D, Yang Z, Yang C, Hu Z, et al. Impact of polysomy 17 on HER2 testing of invasive breast cancer patients. Int J Clin Exp Pathol. 2013; 7: 163–173. pmid:24427336
  14. 14. Goldhirsch A, Wood WC, Coates AS, Gelber RD, Thürlimann B, Senn HJ, et al. Strategies for subtypes-dealing with the diversity of breast cancer: highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer. Ann Oncol. 2011; 22: 1736–1747. pmid:21709140
  15. 15. Phipps AI, Buist DS, Malone KE, Barlow WE, Porter PL, Kerlikowske K, et al. Reproductive history and risk of three breast cancer subtypes defined by three biomarkers. Cancer Causes Control. 2011; 22:399–405. pmid:21184265
  16. 16. Prat A, Parker JS, Karginova O, Fan C, Livasy C, Herschkowitz JI, et al. Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancer. Breast Cancer Res.2010; 12: R68. pmid:20813035
  17. 17. Parker JS, Mullins M, Cheang MC, Leung S, Voduc D, Vickery T, et al. Supervised risk predictor of breast cancer based on intrinsic subtypes. J Clin Oncol. 2009; 27: 1160–1167. pmid:19204204
  18. 18. Mackay A, Weigelt B, Grigoriadis A, Kreike B, Natrajan R, A'Hern R, et al. Microarray-based class discovery for molecular classification of breast cancer: analysis of inter observer agreement. J Natl Cancer Inst. 2011; 103: 662–673. pmid:21421860
  19. 19. Vranic S, Teruya B, Repertinger S, Ulmer P, Hagenkord J, Gatalica Z. Assessment of HER2 gene status in breast carcinomas with polysomy of chromosome 17. Cancer. 2011; 117: 48–53. pmid:20803611
  20. 20. Hyun CL, Lee HE, Kim KS, Kim SW, Kim JH, Choe G, et al. The effect of chromosome 17 polysomy on HER-2/neu status in breast cancer. J Clin Pathol. 2008; 61: 317–321. pmid:17761736
  21. 21. Egervari K, Kosa C, Szollosi Z. Impact of chromosome 17 centromere region assessment on HER2 status reported in breast cancer. Pathol Res Pract.2011; 207: 468–471. pmid:21741182
  22. 22. Viale G. Be precise! The need to consider the mechanisms for CEP17 copy number changes in breast cancer. J Pathol. 2009; 219: 1–2. pmid:19670216
  23. 23. Ma Y, Lespagnard L, Durbecq V, Paesmans M, Desmedt C, Gomez-Galdon M, et al. Polysomy 17 in HER-2/neu status elaboration in breast cancer: effect on daily practice. Clin Cancer Res. 2005; 11:4393–4399. pmid:15958623
  24. 24. Shah SS, Wang Y, Tull J, Zhang S. Effect of high copy number of HER2 associated with polysomy 17 on HER2 protein expression in invasive breast carcinoma. Diagn Mol Pathol. 2009; 18:30–33. pmid:19214111
  25. 25. Sahoo R, Babu VC, Harini VV, Patil GV, Dhondalay GK, Kulkarni J, et al. Her-2/neu overexpression due to polysomy 17 in breast cancer: molecular testing to guide therapeutic options. Onkologie. 2011; 34: 356–360. pmid:21734421
  26. 26. Yeh IT, Martin MA, Robetorye RS, Bolla AR, McCaskill C, Shah RK, et al. Clinical validation of an array CGH test for HER2 status inbreast cancer reveals that polysomy 17 is a rare event. Mod Pathol. 2009; 22:1169–1175. pmid:19448591
  27. 27. Marchiò C, Lambros MB, Gugliotta P, Di Cantogno LV, Botta C, Pasini B, et al. Does chromosome 17 centromere copy number predict polysomy in breast cancer? A fluorescence in situ hybridization and microarray-based CGH analysis. J Pathol. 2009; 219: 16–24. pmid:19670217
  28. 28. Pathmanathan N, Bilous AM. HER2 testing in breast cancer: an overview of current techniques and recent developments. Pathology. 2012; 44: 587–595. pmid:23111474
  29. 29. Moelans CB, de Weger RA, van Diest PJ. Absence of chromosome 17 polysomy inbreast cancer: analysis by CEP17 chromogenic in situ hybridization and multiplex ligation-dependent probe amplification. Breast Cancer Res Treat. 2010; 120: 1–7. pmid:19760503
  30. 30. Gunn S, Yeh IT, Lytvak I, Tirtorahardjo B, Dzidic N, Zadeh S, et al. Clinical array-based karyotyping of breast cancer with equivocal HER2 status resolves gene copy number and reveals chromosome 17 complexity. BMCCancer. 2010; 10: 396.
  31. 31. Murthy SS, Sandhya DG, Ahmed F, Goud KI, Dayal M, Suseela K, et al. Assessment of HER2/Neu status by fluorescence in situ hybridization inimmunohistochemistry-equivocal cases of invasive ductal carcinoma and aberrant signal patterns: a study at a tertiary cancer center. Indian J Pathol Microbiol. 2011; 54:532–538. pmid:21934215
  32. 32. Sneige N, Liu B, Yin G, Gong Y, Arun BK. Correlation of cytologic findings and chromosomal instability detected by fluorescence in situ hybridization in breast fine-needle aspiration specimens from women at high risk for breast cancer. Mod Pathol. 2006; 19: 622–629. pmid:16528376
  33. 33. Vanden Bempt I, Van Loo P, Drijkoningen M, Neven P, Smeets A, Christiaens MR, et al. Polysomy 17 in breast cancer: clinicopathologic significance and impact on HER-2 testing. J Clin Oncol. 2008; 26: 4869–4874. pmid:18794552
  34. 34. Kim A, Shin HC, Bae YK, Kim MK, Kang SH, Lee SJ, et al. Multiplication of Chromosome 17 Centromere Is Associated with Prognosis in Patients with Invasive Breast Cancers Exhibiting Normal HER2 and TOP2A Status. J Breast Cancer. 2012; 15:24–33. pmid:22493625
  35. 35. Krishnamurti U, Hammers JL, Atem FD, Storto PD, Silverman JF. Poor prognostic significance of unamplified chromosome 17 polysomy in invasive breast carcinoma. Mod Pathol. 2009; 22: 1044–1048. pmid:19396150
  36. 36. Bartlett AI, Starcyznski J, Robson T, Maclellan A, Campbell FM, van de Velde CJ, et al. Heterogeneous HER2 gene amplification: impact on patient outcome and a clinically relevant definition. Am J Clin Pathol. 2011; 13: 266–274.
  37. 37. Bartlett JM, Campbell FM, Mallon EA. Determination of HER2 amplification by in situ hybridization: when should chromosome 17 also be determined? Am J Clin Pathol. 2008; 130: 920–926. pmid:19019769
  38. 38. Torrisi R, Rotmensz N, Bagnardi V, Viale G, Curto BD, Dell'orto P, et al. HER2 status inearly breast cancer: relevance of cell staining patterns, gene amplification and polysomy 17. Eur J Cancer. 2007; 43:2339–2344. pmid:17855075
  39. 39. Sapino A, Maletta F, Verdun di Cantogno L, Macrì L, Botta C, Gugliotta P, et al. Gene status in HER2 equivocal breast carcinomas: impact of distinct recommendations and contribution of a polymerase chain reaction-based method. Oncologist. 2014; 19:1118–1126. pmid:25323485