• No results found

University of Groningen Circulating tumor cells and the micro-environment in non-small cell lung cancer Tamminga, Menno

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Circulating tumor cells and the micro-environment in non-small cell lung cancer Tamminga, Menno"

Copied!
21
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Circulating tumor cells and the micro-environment in non-small cell lung cancer

Tamminga, Menno

DOI:

10.33612/diss.132713141

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date: 2020

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Tamminga, M. (2020). Circulating tumor cells and the micro-environment in non-small cell lung cancer. University of Groningen. https://doi.org/10.33612/diss.132713141

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

General discussion

(3)

In this thesis, biomarkers in blood (liquid biopsies) and tumor tissue of non-small cell lung cancer (NSCLC) patients were studied in order to better understand underlying processes and predict tumor responses to treatment. In liquid biop-sies different tumor elements are recognized. Circulating tumor cells (CTC), cir-culating tumor DNA (ctDNA) and tumor derived extracellular vesicles (tdEV) are used as prognostic biomarkers. CTC are often identified by the presence of the epithelial cell adhesion molecule (EpCAM). However, in NSCLC, EpCAM-positive (EpCAM+) CTC are rarely detected. To increase CTC detection, methods to detect EpCAM-negative (EpCAM-) CTC have been developed. An example are microsieves which detect CTC by their larger sizes. In chapter 2 we compared EpCAM+ CTC, EpCAM-CTC, ctDNA and tdEV to see which biomarker outperforms the others in predicting prognosis. With the exception of EpCAM- CTC, all biomarkers identified a similar number of at risk patients (tdEV n=26, CTC n=20, ctDNA n=18). Except for EpCAM- CTC, the biomarkers correlated strongly with each other. Their combina-tion therefore resulted in only a slight increase in prognostic value. This is to be expected as all these biomarkers are derived from the tumor and are represent-ative of tumor growth, aggressiveness and size (1). EpCAM- CTC were previously reported to be prognostic by Hofman et al, conflicting with our findings (2). This might be due to our detection method. We detected EpCAM- CTC in the CellSearch waste after processing for EPCAM+ CTC was completed. In this manner EpCAM- CTC might have been lost, which would have influenced our results.

Studies comparing different entities within liquid biopsies are rare. Sundaresan et al compared detection rates of resistant mutations by conventional biopsies, ctDNA and CTC in NSCLC patients (3). Each method individually detected the resistant mutation in similar proportion of the patients (roughly 50%). Howev-er combining all three methods, 73% of patients had resistant mutations. Each method was able to identify mutations in patients who were negative for all other methods, highlighting that the information is complementary. We know that TKI treatment has a large chance of success when the corresponding mutation is detected in conventional tissue biopsies and that mutations detection by liquid biopsies correspond well with outcomes from the primary tumor (3–10). Unfor-tunately no study has investigated whether TKI response in patients with muta-tions detected in CTC or ctDNA are comparable. Compared to CTC, studies using ctDNA are from more recent times, yet ctDNA has already been implemented in several clinical settings (3,8). CTC would need to provide more information than

(4)

today can be gleaned from ctDNA or the presence of a single mutation before clinical adaptation can be considered.

In chapter 3 and 4 we report the predictive value of CTC. Advanced NSCLC pa-tients treated with either chemotherapy (n=54), targeted therapy (n=34) or immu-notherapy (n=104) had significantly worse tumor response rates when CTC were detected in their peripheral blood. Responses were measured according to the response evaluation criteria in solid tumors (RECIST v1.1). Objective response rate at time of treatment evaluation, often after 6 weeks of treatment was the main clinical endpoint. For patients treated with immunotherapy, durable responses (measured as objective response or stable disease longer than 6 months) were also used. This choice was made, as responses to immunotherapy can be long lasting, even when a patient has only stable disease, which would otherwise be denoted as a non-responder.

The presence of CTC was demonstrated at baseline in 32 % of advanced NSCLC patients and was associated with a lower response rate to all therapies (chemo-therapy≈1.5 times; targeted≈3 times lower; immunotherapy≈2 times lower) com-pared with patients who had no CTC detected. A possible explanation for the decreased response rates is the expression of genes related to chemotherapy resistance (11–14). A subset of CTC had characteristics involved with an immune escape mechanism that may be relevant (15,16). Finally, CTC might represent a more aggressive type of tumor and are therefore associated with faster disease progression and lower response rates (17–19).

Sequential CTC measurements during treatment could further improve the pre-dictive value of CTC. In chapter 4 the change of CTC counts corresponded well with tumor response and survival, as reported previously (15,16,20,21). Hiltermann et al showed that the change in CTC counts was the best predictor for survival, outperforming even the conventionally used computed tomography scans (19). When CTC can still be detected after several weeks of treatment with, they in-dicate therapy failure.

The cohorts treated with different therapies were too small to define whether CTC could be used to differentiate which therapy has the highest chance of success. Larger cohorts with more homogenous populations are essential for a definitive

(5)

answer. Additionally, surface markers on CTC, and mutations identified in CTC might further increase the value they can offer to the clinic (16,20). However, single cell sequencing is still new, and a long way off from clinical application. And as NSCLC are highly heterogeneous, predictions based on a single cell are not always reliable (22). To investigate whether CTC can be used as an alternative for conventional biopsies and reflecting the heterogeneity of the whole tumor, larger amounts of CTC are needed. And larger numbers of CTC could increase their prognostic and predictive value.

In chapter 5 we investigated the origin and release of CTC from the primary tumor in the lung and the natural filtering of CTC in the blood circulation. CTC detect-ed in the peripheral veins by CellSearch have consistently been associatdetect-ed with survival and recurrence of disease after surgery for several cancer types, in-cluding lung cancer (2,23–26). Measuring CTC closer to the original tumor, e.g. the draining pulmonary vein, CTC can be detected in high numbers (14,27–34). Crosbie et al suggested that the location of removal might be the microcircula-tion (34). We identified that removal of CTC occurs in a more central locamicrocircula-tion as CTC counts were already strongly decreased in the radial artery (so before the microcirculation). CTC frequencies in the radial artery closely resembled those found in the peripheral veins. As such it appears that the central compartment plays a major role in CTC clearance, and it is unlikely that the microcirculation is involved therein. Possibly the blood environment combined with shearing forces of the circulation (and heart) destroy the majority of newly released CTC. We also observed in chapter 5 that during surgery most of the epithelial like cells in the pulmonary vein were non-malign. The majority of circulating epithelial cells showed no structural genetic changes (copy number alterations of the number of chromosomes). Morphologically there were also differences between CTC ‘nor-mally’ detected in NSCLC patients and the cells identified in the pulmonary vein. CTC identified in the pulmonary vein often were larger, with stronger expression of cytokeratin. Apparently most of these non-malign epithelial cells are eliminated in the central compartment because the counts in the radial artery were already strongly diminished and morphologically resembled ‘normal’ CTC more strongly. The CellSearch is a very sensitive system which can identify a few epithelial cells in a background of millions of normal blood cells. For peripheral blood it is a well

(6)

validated system. The CTC detected in the peripheral blood of patients are an important predictor of survival and many studies have reported that these cell (when not detected during surgery) harbor mutations (35–41). However, our find-ings would explain that even in healthy patients CTC can occasionally be found (42). The presence of non-malign cells in the pulmonary vein also explains why previous studies found that CTC measured in the pulmonary vein were not supe-rior in predicting recurrence of disease after surgery compared to those isolated from a peripheral vein (29,32,34).

Our findings indicate that care has to be taken when using the CellSearch beyond the limits of its validation for which the FDA clearance was given. The CellSearch system is a very sensitive and reliable method to identify CTC in the peripheral blood when the patient is not undergoing surgery, but can falsely identify cells as CTC in the pulmonary circulation. This might also explained why CTC identi-fied in the peripheral vein have a stronger association with survival than those detected in the pulmonary vein (2,26).

Unfortunately CTC detection in the peripheral blood is too often negative in NSCLC patients. Even in patients with CTC, the number of CTC in 10mL of blood is very low (1,2). To increase the number of detectable CTC, the screened volume of blood should be increased. As shown in chapter 6, diagnostic leukapheresis (DLA) can be used to process the total body blood volume in 1-3 hours. DLA separates the blood components from one another, allowing for the sequestering of CTC (to-gether with the mononuclear cell fraction). Stoecklein et al and Fischer et al were the first to implement this method to increase CTC in metastatic breast cancer (43,44). Subsequently these findings have been confirmed in different cohorts of breast and prostate cancer patients (35,45,46). Our study is the first performed in a NSCLC population. CTC counts in DLA products corrected for lymphocytes approached counts detected in the peripheral blood. In our study DLA-CTC were stronger associated with survival than peripheral vein blood-CTC. A change in their counts after treatment correlated well with tumor response to therapy. As such, DLA can be used to increase CTC detection, and improves their predictive and prognostic value.

One of the current limitations is that only small volumes of DLA product can be screened with CellSearch. The CellSearch method can at most process 2×108

(7)

kocytes. As the DLA products contain around 10 times as many leukocytes as the peripheral blood, CellSearch can only process a few milliliters. With a leukocyte depletion step, e.g. the Rosette-SEP, a larger volume of DLA product could be processed. Unfortunately, many CTC were lost with this procedure, and prog-nostic and predictive values decreased. Therefore, this procedure was not de-veloped further.

Systems using physical characteristics of CTC to distinguish them from other blood cells, specifically their size and rigidity, might be less restricted in the volume of DLA product that can be processed. Two such systems, the Vycap mi-crosieves and the Isolation of Epithelial Tumor cells (ISET) were tested in chapter 7 and 8, respectively. Vycap microsieves are capable of filtering the DLA product and could identify CTC, but were prone to clogging. Especially the 5μm pores mi-crosieves could not process even a limited volume of DLA product (1.1-2.3 mL). Longer sample fixation times were a likely contributor. It seems that Vycap mi-crosieves are only capable of processing the DLA product efficiently within 24 hours after fixation. This requires laboratory procedures near the clinical setting, which is not always possible. This loss of CTC observed with the Vycap micro-sieves using DLA was also reported from patients with breast cancer (45). In chapter 8 we established a protocol for the ISET method to effectively filter DLA products. Up to 10 mL of DLA product could be processed after fixation. CTC were identified in the majority of patients with immunological staining and in higher counts per mL DLA compared to detection with CellSearch. Encourag-ingly, EpCAM+ CTC were identified in similar proportions and concentrations for both ISET and Cellsearch. This implies two important findings: The CellSearch is highly efficient in identifying EpCAM+ CTC even in low volumes. And no relevant loss of EpCAM+ CTC by the ISET seemed to occur when processing DLA product of NSCLC patients (47).

Furthermore with an adapted live cell protocol up to 20 ml, DLA product could be processed with an ISET filter. The number of leukocytes within this volume of DLA product was around 10-15 times the number of leukocytes that can be han-dled by CellSearch. All 8 processed samples had EpCAM+ cells. Six patients had 30 or more CTC and with three over 100 CTC, resembling the numbers as extrap-olated by Stoecklein et al (44,48). This means that using DLA, in 75% of patients

(8)

we identified sufficient CTC for genomic and functional testing, even though we only processed a part (15-25%) of the total DLA product.

Due to the costs of the DLA procedure, CTC enumeration and analysis, at this point of time implementation may be clinically viable in two situations: in patients undergoing curative surgery to identify patients likely at risk for recurrence; in patients in whom original (recurrent) biopsies failed to obtained enough material for a diagnosis.

Between 30 and 40 percent of patients undergoing surgery have recurrence of disease (49). Identification of patients at risk for recurrence is still difficult. CTC measured in the peripheral blood have been linked to recurrence and could there-fore be used to stratify patients in high and low risk groups with therapeutic consequences (2,23–26).

DLA to obtain sufficient CTC could play a role in both these cases by increasing predictive and prognostic values. However, this would also increase costs. Eco-nomic health care cost calculation are lacking today to evaluate the benefits and cost effectiveness.

DLA could prove useful in the patients whom have not enough tumor cells ex-tracted by conventional biopsies. And up to 25% of NSCLC cannot be biopsied, or provide enough material for evaluation and diagnoses (50). Especially fine needle aspirations often do not obtain enough tumor cells for analysis. It is also less inva-sive than a mediastinoscopy. Finally, CTC measurements might be useful for pa-tients receiving hematopoietic stem cell transplants or cell products. In papa-tients with breast cancer who received bone marrow transplants, only the presence of CTC in the apheresis product corresponded to recurrence of disease (51). With the increased popularity of cellular therapies, and the ease to simultaneously collect the CTC fraction, screening for CTC in the blood products might be new approach to further improve methods to prevent relapses.

Next to the circulating blood derived biomarkers we investigated the role of biomarkers in the tumor microenvironment of NSCLC. In chapter 9 and 10 we used publicly available data to explore the immune infiltrate and the expression of immune genes and associations with survival. The immune system and the

(9)

immune infiltrate in the tumor are of great importance for the response to im-munotherapy (52–55). While data of patients treated with imim-munotherapy are at this moment available in insufficient quantities in the public domain, data about treatment naïve patients are available and can provide insight in immune com-positions that are associated with survival. Gene expression differences of gene groups involved in antigen presentation, co-stimulation, inhibition and cytokine production between NSCLC samples and normal tissue can provide clues about the mechanisms employed by cancer to escape the immune system. In chapter 9, we have focused on the differences in DNA methylation and RNA expression between the two major subtypes of NSCLC and whether these differences were involved in an effect on immune function. As shown before, the genome in NSCLC was hypomethylated when compared to normal tissue (56,57). Hypomethylation of cancer-related genes is a major factor in driving the tumor cells to proliferate. Adenocarcinoma showed a higher methylation than squamous cell lung carcino-ma. These histological subtypes could be differentiated by methylation and by RNA expression. Expression differences in five genes (KRT5, DSC3, DSG3, TP63,

CALML3) combined could correctly classify 95% of patients in adenocarcinoma

or squamous cell carcinoma. Subtype differentiating genes were mostly involved in extracellular matrix and cell structure which are already clinically used by pa-thologists to classify cancer subtypes, highlighting the robustness of the study. None of the genes or methylation probes that were involved in histological sub-types were involved in immune functions. When comparing the RNA expression of tumor samples with matched non-cancerous tissue, we observed that NSCLC samples showed a large expression variation but a clearly decreased expression of antigen presenting and antigen processing genes. No changes were observed in co-stimulatory, co-inhibitory, and cytokine production gene groups. In NSCLC tumor cells the central immune defect is located in the antigen presenting gene group.

In chapter 10 the immune infiltrate composition in mostly early stage (pretreat-ment) NSCLC patients was assessed by means of CIBERSORT, which has a high concordance with immune cells measured by FACS and histology examination (58). The immune microenvironment was mostly composed of macrophages, plasma cells, CD8 T-cells, resting CD4 T-cells and memory B-cells. The differ-ences between the two major histological subtypes of NSCLC, adenocarcinoma and squamous cell carcinoma were minimal. However, cell fractions that show

(10)

significant differences between subtypes also showed differences in their asso-ciations with survival. For instance, the neutrophil fraction was strongly associ-ated with shorter survival for adenocarcinoma patients, but had no association with survival in squamous cell carcinoma. The fractions of memory B-cells and naïve CD4 T-cells were associated with better survival in adenocarcinoma, but worse survival in squamous cell carcinoma. These differences in survival were reflected in corresponding differences in the immune composition. Therefore, the pooling of NSCLC without stratification for histological subtype might con-found associations with survival and should be avoided.

We also investigated the cell composition in current smokers and (former) non-smokers. The immune composition of (former)smokers encompassed higher fractions of regulatory T-cells, follicular helper T-cells, neutrophils and M2 mac-rophages. All these fractions were associated with a shorter survival. Around 80% of lung cancer cases can be attributed to smoking. It is believed that smoking induces inflammation and may influence the functionality of the immune system, as smokers suffer more often from (more serious) infections (59–61). Yet smok-ing is associated with an increased chance to respond to immunotherapy. The exact cause is unknown, but it could be because of increased tumor mutational burden associated with high levels of neo-antigens, or because PD-L1 therapy is mostly effective in reactivating exhausted effector cells which might be more present in smokers (61–64).

Future prospects

In up to 25% of NSCLC no biopsies could be obtained, or the biopsies do not harbor enough material for evaluation and diagnoses (50). In a time where per-sonalized treatment is becoming the norm, this lack of information can hamper treatment-decision-making. CTC could be a useful alternative to tumor biopsies. Their numbers provide additional information regarding the prognosis but also on the chance to respond to treatment. Studies in NSCLC comparing CTC to other liquid biopsies are still lacking, but CTC seem to be identified in similar proportion of patients and have similar prognostic value as ctDNA in our study (chapter 2). DLA increases the number of patients with sufficient CTC detected for diagno-sis, and increases their predictive and prognostic value. DLA based CTC analysis

(11)

can be considered if no tissue is available with routine diagnostic approaches. However it is unsuitable for sequential monitoring of CTC counts in the whole NSCLC population. Costs of DLA need to be decreased, or other methods to screen larger volumes of blood for CTC are needed. The current technology has not been developed to the state that it will be implemented in routine practice. Finally, currently we only enumerate CTC counts. We have shown that they can be used for single cell whole genome sequencing to detect copy number varia-tions and structural DNA variavaria-tions such as loss of chromosomal arms. Others have shown that specific mutations can be identified (65–67). However, for clin-ical use it is important to evaluate the full spectrum of targetable mutations. Currently sequencing analyses on a single cell level is being developed at a rapid pace, and even RNA sequencing is becoming available, opening up more options in the future (68). These single cell analyses are not yet used clinically. However, when CTC can be isolated from larger volumes of DLA, or purified to a greater extent, sequencing can be performed on the DNA from pooled CTC cell suspen-sions. Already the ISET is capable of isolating a large number of CTC, but it is a labor intensive method.

Surface markers can also increase the prognostic and predictive values of CTC as shown before (20,69). The microwell system of Vyap (which was not investi-gated in this thesis) is capable of isolating single cells when only a small number of cells need to be processed (70–72). The isolated single CTC can then be used for genomic analyses, and even functional tests and protein production (70–72). Hopefully, CTC enumerations will be adapted to include genomic and functional analyses to provide all this information.

In tissue biopsies, neutrophils, memory B-cells, CD4 T-cells and regulatory T-cells should be assessed for their different functions depending on the histological subtypes and smoking behavior. Already, different immune scores based on cell counts on the peripheral blood have been developed but with limited efficacy (73). If these immune scores could be improved, they would provide another useful marker for treatment efficacy.

All in all, in the blood compartment CTC from NSCLC patients are a very prom-ising biomarker with multiple applications. We have studied different methods to increase the number of CTC. However, currently we are still unable to utilize

(12)

them to their full potential in NSCLC. Technological advances have to be made first. Until then, CTC can be used for the patients in whom conventional biopsies did not provide the required genomic information.

In the tumor itself the primary defect is located in the decreased expression of antigen presenting and antigen processing genes involved in resistance. In its microenvironment different immune related cells are associated with survival. The predictive values of immune scores for the response to therapy will be fur-ther developed.

(13)

References

1. Heitzer E, Haque IS, Roberts CES, Speicher MR. Current and future perspectives of liquid biopsies in genomics-driven oncology. Nat Rev Genet. 2019;20(2):71–88. 2. Hofman V, Ilie MI, Long E, Selva E, Bonnetaud C, Molina T, et al. Detection of

cir-culating tumor cells as a prognostic factor in patients undergoing radical surgery for non-small-cell lung carcinoma: comparison of the efficacy of the CellSearch AssayTM and the isolation by size of epithelial tumor cell method. Int J Cancer.

2010;129(7):1651–60.

3. Sundaresan TK, Sequist L V., Heymach J V., Riely GJ, Janne PA, Koch WH, et al. Detection of T790M, the acquired resistance EGFR mutation, by tumor biopsy versus noninvasive blood-based analyses. Clin Cancer Res. 2015;1–9.

4. Mok T, Wu YL, Lee JS, Yu CJ, Sriuranpong V, Sandoval-Tan J, et al. Detection and dynamic changes of EGFR mutations from circulating tumor DNA as a predictor of survival outcomes in NSCLC Patients treated with first-line intercalated erlotinib and chemotherapy. Clin Cancer Res. 2015;21(14):3196–203.

5. Mok TS, Wu Y, Thongprasert S, Yang C, Saijo N, Sunpaweravong P, et al. Gefitin-ib or Carboplatin–Paclitaxel in Pulmonary Adenocarcinoma. N Engl J Med. 2009;361(10):947–57.

6. Mok TS, Wu Y-L, Ahn M-J, Garassino MC, Kim HR, Ramalingam SS, et al. Osimerti-nib or Platinum–Pemetrexed in EGFR T790M–Positive Lung Cancer. N Engl J Med. 2017;376(7):629–40.

7. Oxnard GR, Thress KS, Alden RS, Lawrance R, Paweletz CP, Cantarini M, et al. As-sociation between plasma genotyping and outcomes of treatment with osimertinib (AZD9291) in advanced non-small-cell lung cancer. J Clin Oncol. 2016;34(28):3375–82. 8. Mostert B, Jiang Y, Sieuwerts AM, Wang H, Bolt-De Vries J, Biermann K, et al. KRAS

and BRAF mutation status in circulating colorectal tumor cells and their correlation with primary and metastatic tumor tissue. Int J Cancer. 2013;133(1):130–41. 9. Punnoose E a., Atwal S, Liu W, Raja R, Fine BM, Hughes BGM, et al. Evaluation of

circulating tumor cells and circulating tumor DNA in non-small cell lung cancer: Association with clinical endpoints in a phase II clinical trial of pertuzumab and erlotinib. Clin Cancer Res. 2012;18(8):2391–401.

10. Ilie M, Long E, Butori C, Hofman V, Coelle C, Mauro V, et al. ALK-gene rearrangement: A comparative analysis on circulating tumour cells and tumour tissue from patients with lung adenocarcinoma. Ann Oncol. 2012;23(11):2907–13.

(14)

11. Papadaki MA, Stoupis G, Theodoropoulos PA, Mavroudis D, Georgoulias V, Agelaki S. Circulating tumor cells with stemness and epithelial-to-mesenchymal transition features are chemoresistant and predictive of poor outcome in metastatic breast cancer. Mol Cancer Ther. 2018;molcanther.0584.2018.

12. Chebouti I, Kasimir-Bauer S, Buderath P, Wimberger P, Hauch S, Kimmig R, et al. EMT-like circulating tumor cells in ovarian cancer patients are enriched by plati-num-based chemotherapy. Oncotarget. 2017;8(30):48820–31.

13. Hyun K-A, Goo K-B, Han H, Sohn J, Choi W, Kim S-I, et al. Epithelial-to-mesenchymal transition leads to loss of EpCAM and different physical properties in circulating tumor cells from metastatic breast cancer. Oncotarget. 2016;7(17):24677–87. 14. Murlidhar V, Reddy RM, Fouladdel S, Zhao L, Ishikawa MK, Grabauskiene S, et al.

Poor Prognosis Indicated by Venous Circulating Tumor Cell Clusters in Early-Stage Lung Cancers. Cancer Res. 2017;77(18):5194–207.

15. Raimondi C, Carpino G, Nicolazzo C, Gradilone A, Gianni W, Gelibter A, et al. PD-L1 and epithelial-mesenchymal transition in circulating tumor cells from non-small cell lung cancer patients: A molecular shield to evade immune system? Oncoim-munology. 2017;6(12):1–5.

16. Nicolazzo C, Raimondi C, Mancini M, Caponnetto S, Gradilone A, Gandini O, et al. Monitoring PD-L1 positive circulating tumor cells in non-small cell lung cancer pa-tients treated with the PD-1 inhibitor Nivolumab. Sci Rep. 2016;6(July):1–8.

17. Budd GT, Cristofanilli M, Ellis MJ, Stopeck A, Borden E, Miller MC, et al. Circulating tumor cells versus imaging - Predicting overall survival in metastatic breast cancer. Clin Cancer Res. 2006;12(21):6403–9.

18. De Giorgi U, Valero V, Rohren E, Mego M, Doyle G V., Miller MC, et al. Circulating tumor cells and bone metastases as detected by FDG-PET/CT in patients with metastatic breast cancer. Ann Oncol. 2009;21(1):33–9.

19. Hiltermann TJN, Pore MM, van den Berg A, Timens W, Boezen HM, Liesker JJW, et al. Circulating tumor cells in small-cell lung cancer: A predictive and prognostic factor. Ann Oncol. 2012;23(11):2937–42.

20. Ilié M, Szafer-Glusman E, Hofman V, Chamorey E, Lalvée S, Selva E, et al. Detection of PD-L1 in circulating tumor cells and white blood cells from patients with advanced non-small-cell lung cancer. Ann Oncol. 2018;29(1):193-9.

21. Breitenbuecher F, Hoffarth S, Worm K, Cortes-Incio D, Gauler TC, Köhler J, et al. Development of a highly sensitive and specific method for detection of circulating tumor cells harboring somatic mutations in non-small-cell lung cancer patients. PLoS One. 2014;9(1):e85350.

(15)

22. Hensley CT, Faubert B, Yuan Q, Lev-Cohain N, Jin E, Kim J, et al. Metabolic Heter-ogeneity in Human Lung Tumors. Cell. 2016;164(4):681–94.

23. Ou H, Huang Y, Xiang L, Chen Z, Fang Y, Lin Y, et al. Circulating Tumor Cell Phenotype Indicates Poor Survival and Recurrence After Surgery for Hepatocellular Carcinoma. Dig Dis Sci. 2018;63(9):2373–80.

24. Van Dalum G, Van Der Stam GJ, Tibbe AGJ, Franken B, Mastboom WJB, Vermes I, et al. Circulating tumor cells before and during follow-up after breast cancer surgery. Int J Oncol. 2015;46(1):407–13.

25. Cueto Ladrón de Guevara A, Serrano MJ, Sánchez-Palencia Ramos A, Delga-do-Rodríguez M, Expósito Ruiz M, Ruiz Zafra J, et al. Association of circulating tumour cells with early relapse and 18F-fluorodeoxyglucose positron emission to-mography uptake in resected non-small-cell lung cancers†. Eur J Cardio-Thoracic Surg. 2017;52(1):55–62.

26. Bayarri-Lara C, Ortega FG, Guevara ACL de, Puche JL, Zafra JR, Miguel-Pérez D de, et al. Circulating tumor cells identify early recurrence in patients with non-small cell lung cancer undergoing radical resection. PLoS One. 2016;11(2):1–14.

27. LV C, ZHAO B, WANG L, ZHANG P, MA Y, WANG Y, et al. Detection of circulating tumor cells in pulmonary venous blood for resectable non-small cell lung cancer. Oncol Lett. 2018;15(1):1103–12.

28. Reddy RM, Murlidhar V, Zhao L, Grabauskiene S, Zhang Z, Ramnath N, et al. Pulmo-nary venous blood sampling significantly increases the yield of circulating tumor cells in early-stage lung cancer. J Thorac Cardiovasc Surg. 2016;151(3):852–8. 29. Okumura Y, Tanaka F, Yoneda K, Hashimoto M, Takuwa T, Kondo N, et al. Circulating

Tumor Cells in Pulmonary Venous Blood of Primary Lung Cancer Patients. Ann Thorac Surg 2009;87(6):1669–75.

30. Chudasama D, Burnside N, Beeson J, Karteris E, Rice A, Anikin V. Perioperative detection of circulating tumour cells in patients with lung cancer. Oncol Lett. 2016;14(2):1281–6.

31. Sawabata N, Funaki S, Hyakutake T, Shintani Y, Fujiwara A, Okumura M. Perioper-ative circulating tumor cells in surgical patients with non-small cell lung cancer: does surgical manipulation dislodge cancer cells thus allowing them to pass into the peripheral blood? Surg Today. 2016;46(12):1402–9.

32. Li Y, Cheng X, Chen Z, Liu Y, Liu Z, Xu S. Circulating tumor cells in peripheral and pulmonary venous blood predict poor long-term survival in resected non-small cell lung cancer patients. Sci Rep. 2017;7(1):1–8.

(16)

33. Hashimoto M, Tanaka F, Yoneda K, Takuwa T, Matsumoto S, Okumura Y, et al. Signifi-cant increase in circulating tumour cells in pulmonary venous blood during surgical manipulation in patients with primary lung cancer. Interact Cardiovasc Thorac Surg. 2014;18(6):775–83.

34. Crosbie PAJ, R. Shah PK, Zhou C, Morris K, Tugwood J, Booton R, et al. Circulat-ing Tumor Cells Detected in the Tumor-DrainCirculat-ing Pulmonary Vein Are Associated with Disease Recurrence after Surgical Resection of NSCLC. J Thorac Oncol. 2016;11(10):1793–7.

35. Lambros MB, Seed G, Sumanasuriya S, Gil V, Crespo M, Fontes M, et al. Single-Cell Analyses of Prostate Cancer Liquid Biopsies Acquired by Apheresis. Clin Cancer Res. 2018 Nov 15 [cited 2019 Mar 6];24(22):5635–44.

36. Gasch C, Bauernhofer T, Pichler M, Langer-Freitag S, Reeh M, Seifert AM, et al. Heterogeneity of epidermal growth factor receptor status and mutations of KRAS/ PIK3CA in circulating tumor cells of patients with colorectal cancer. Clin Chem. 2013;59(1):252–60.

37. Swennenhuis JF, Tibbe AGJ, Levink R, Sipkema RCJ, Terstappen LWMM. Charac-terization of circulating tumor cells by fluorescence in situ hybridization. Cytom Part A. 2009;75(6):520–7.

38. Pailler E, Oulhen M, Borget I, Remon J, Ross K, Auger N, et al. Circulating tumor cells with aberrant ALK copy number predict progression-free survival during crizotin-ib treatment in ALK-rearranged non-small cell lung cancer patients. Cancer Res. 2017;77(9):2222–30.

39. Ni X, Zhuo M, Su Z, Duan J, Gao Y, Wang Z, et al. Reproducible copy number varia-tion patterns among single circulating tumor cells of lung cancer patients. Pnas. 2013;110(52):21083–8.

40. Su Z, Wang Z, Ni X, Duan J, Gao Y, Zhuo M, et al. Inferring the Evolution and Pro-gression of Small-Cell Lung Cancer by Single-Cell Sequencing of Circulating Tumor Cells. Clin Cancer Res. 2019;clincanres.3571.2018.

41. Carter L, Rothwell DG, Mesquita B, Smowton C, Leong HS, Fernandez-Gutierrez F, et al. Molecular analysis of circulating tumor cells identifies distinct copy-number profiles in patients with chemosensitive and chemorefractory small-cell lung cancer. Nat Med 2016;(November). A

42. Allard WJ, Matera J, Miller MC, Repollet M, Connelly MC, Rao C, et al. Tumor Cells Circulate in the Peripheral Blood of All Major Carcinomas but not in Healthy Subjects or Patients With Nonmalignant Diseases Tumor Cells Circulate in the Peripheral Blood of All Major Carcinomas but not in Healthy Subjects or Patients With Non. Clin Cancer Res. 2005;10(October):6897–904.

(17)

43. Fischer JC, Niederacher D, Topp SA, Honisch E, Schumacher S, Schmitz N, et al. Diagnostic leukapheresis enables reliable detection of circulating tumor cells of nonmetastatic cancer patients. PNAS. 2013;110(41):16580–5.

44. Stoecklein NH, Fischer JC, Niederacher D, Terstappen LWMM. Challenges for CTC-based liquid biopsies: low CTC frequency and diagnostic leukapheresis as a potential solution. Expert Rev Mol Diagn. 2015;7159(December 2015):14737159.2016.1123095. 45. Andree KC, Mentink A, Zeune LL, Terstappen LWMM, Stoecklein NH, Neves RP, et

al. Toward a real liquid biopsy in metastatic breast and prostate cancer: Diagnostic LeukApheresis increases CTC yields in a European prospective multicenter study (CTCTrap). Int J Cancer . 2018 Nov 15;143(10):2584–91.

46. Fehm TN, Meier‐Stiegen F, Driemel C, Jäger B, Reinhardt F, Naskou J, et al. Diag-nostic leukapheresis for CTC analysis in breast cancer patients: CTC frequency, clinical experiences and recommendations for standardized reporting. Cytom Part A. 2018 Dec 14;93(12):1213–9.

47. Massard C, Oulhen M, Le Moulec S, Auger N, Foulon S, Abou-Lovergne A, et al. Phe-notypic and genetic heterogeneity of tumor tissue and circulating tumor cells in patients with metastatic castration-resistant prostate cancer: a report from the PETRUS prospective study. Oncotarget. 2016;7(34).

48. Coumans FAW, Ligthart ST, Uhr JW, Terstappen LWMM. Challenges in the enumer-ation and phenotyping of CTC. Clin Cancer Res. 2012;18(20):5711–8.

49. Cruz C, Afonso M, Oliveiros B, Pêgo A. Recurrence and Risk Factors for Relapse in Patients with Non-Small Cell Lung Cancer Treated by Surgery with Curative Intent. Oncol. 2017;92(6):347–52.

50. Kerner GSM a, Schuuring E, Sietsma J, Hiltermann TJN, Pieterman RM, de Leede GPJ, et al. Common and rare EGFR and KRAS mutations in a Dutch non-small-cell lung cancer population and their clinical outcome. PLoS One. 2013;8(7):e70346. 51. Solano C, Badia B, Lluch A, Marugan I, Benet I, Arbona C, et al. Prognostic

signifi-cance of the immunocytochemical detection of contaminating tumor cells (CTC) in apheresis products of patients with high-risk breast cancer treated with high-dose chemotherapy and stem cell transplantation. Bone Marrow Transplant. 2001 Feb 17;27(3):287–93.

52. Noguchi T, Ward JP, Gubin MM, Arthur CD, Lee SH, Hundal J, et al. Temporally Distinct PD-L1 Expression by Tumor and Host Cells Contributes to Immune Escape. Cancer Immunol Res. 2017;5(2):106–17.

53. Juneja VR, McGuire KA, Manguso RT, LaFleur MW, Collins N, Haining WN, et al. PD-L1 on tumor cells is sufficient for immune evasion in immunogenic tumors and inhibits CD8 T cell cytotoxicity. J Exp Med. 2017;214(4):895–904.

(18)

54. Wu SP, Liao RQ, Tu HY, Wang WJ, Dong ZY, Huang SM, et al. Stromal PD-L1–Positive Regulatory T cells and PD-1–Positive CD8-Positive T cells Define the Response of Different Subsets of Non–Small Cell Lung Cancer to PD-1/PD-L1 Blockade Immu-notherapy. J Thorac Oncol. 2018;13(4):521–32.

55. Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, et al. Mutational landscape determines sensitivity to PD-1 blockade in non–small cell lung cancer. Science (80- ). 2016;348(6230):124–8.

56. Kettunen E, Hernandez-Vargas H, Cros MP, Durand G, Le Calvez-Kelm F, Stuopelyte K, et al. Asbestos-associated genome-wide DNA methylation changes in lung cancer. Int J Cancer. 2017;141(10):2014–29.

57. Van Tongelen A, Loriot A, De Smet C. Oncogenic roles of DNA hypomethylation through the activation of cancer-germline genes. Cancer Lett. 2017;396:130–7. 58. Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, et al. Robust enumeration

of cell subsets from tissue expression profiles. Nat Methods. 2015;12(MAY 2014):1–10. 59. Mehta H, Nazzal K, Sadikot RT. Cigarette smoking and innate immunity. Inflammation

Research. 2008.

60. Desrichard A, Kuo F, Chowell D, Lee KW, Riaz N, Wong RJ, et al. Tobacco smoking-as-sociated alterations in the immune microenvironment of squamous cell carcinomas. J Natl Cancer Inst. 2018;

61. Khuder SA. Effect of cigarette smoking on major histological types of lung cancer in men. Lung Cancer. 1998;22(1):15–21.

62. Wei SC, Levine JH, Cogdill AP, Zhao Y, Anang NAAS, Andrews MC, et al. Distinct Cellular Mechanisms Underlie Anti-CTLA-4 and Anti-PD-1 Checkpoint Blockade. Cell. 2017;170(6):1120-1133.e17.

63. Collisson EA, Campbell JD, Brooks AN, Berger AH, Lee W, Chmielecki J, et al. Compre-hensive molecular profiling of lung adenocarcinoma. Nature. 2014;511(7511):543–50. 64. Hammerman PS, Lawrence MS, Voet D, Jing R, Cibulskis K, Sivachenko A, et al.

Comprehensive genomic characterization of squamous cell lung cancers. Nature. 2012;489(7417):519–25.

65. Marchetti A, Del Grammastro M, Felicioni L, Malatesta S, Filice G, Centi I, et al. As-sessment of EGFR Mutations in Circulating Tumor Cell Preparations from NSCLC Patients by Next Generation Sequencing: Toward a Real-Time Liquid Biopsy for Treatment. PLoS One. 2014;9(8):e103883.

66. Guibert N, Pradines A, Casanova A, Farella M, Keller L, Soria JC, et al. Detection and monitoring of the BRAF mutation in circulating tumor cells and circulating tumor DNA in BRAF-mutated lung adenocarcinoma. J Thorac Oncol. 2016;11(9):e109–12.

(19)

67. Guibert N, Pradines A, Farella M, Casanova A, Gouin S, Keller L, et al. Monitoring KRAS mutations in circulating DNA and tumor cells using digital droplet PCR during treatment of KRAS-mutated lung adenocarcinoma. Lung Cancer. 2016;100:1–4. 68. Tang F, Barbacioru C, Wang Y, Nordman E, C L, Xu N, et al. mRNA-Seq

whole-tran-scriptome analysis of a single cell. Nat met. 2009;6(5):377–82.

69. Wang X, Sun Q, Liu Q, Wang C, Yao R, Wang Y. CTC immune escape mediated by PD-L1. Med Hypotheses. 2016;93:138–9.

70. Swennenhuis JF, Tibbe AGJ, Stevens M, Katika MR, van Dalum J, Duy Tong H, et al. Self-seeding microwell chip for the isolation and characterization of single cells. Lab Chip. 2015;15(14):3039–46.

71. Oomens L, Passanha F, Kraan J, Andree K, Abali F, Terstappen L, et al. Self-Seeding Microwells to Isolate and Assess the Viability of Single Circulating Tumor Cells. Int J Mol Sci. 2019;20(3):477.

72. Abali F, Stevens M, Tibbe AGJ, Terstappen LWMM, van der Velde PN, Schasfoort RBM. Isolation of single cells for protein therapeutics using microwell selection and Surface Plasmon Resonance imaging. Anal Biochem. 2017 Aug 15;531:45–7. 73. Minami S, Ihara S, Komuta K. Gustave Roussy Immune Score Is a Prognostic Factor

for Chemotherapy-Naive Pulmonary Adenocarcinoma With Wild-Type Epidermal Growth Factor Receptor. World J Oncol. 2019;10(1):55–61.

(20)
(21)

Referenties

GERELATEERDE DOCUMENTEN

Therefore, the aim of this multicenter snapshot study was to evaluate the impact of OP on per- ineal wound healing, presacral abscess formation, prevention of small bowel

The question remains whether alterations in HPA axis are the result of abnormal pain perception or that CP can be seen as a consequence of HPA axis dysfunction (Adler &

Chapter 4 Circulating tumor cells in advanced non-small cell lung cancer patients are associated with worse tumor response to checkpoint inhibitors. Journal for immunotherapy

PD-L1 and epithelial-mesenchymal transition in circulating tumor cells from non-small cell lung cancer patients: A molecular shield to evade immune system.

We studied the relation between overall survival (OS) and the presence of four cancer biomarkers from a single blood draw in advanced NSCLC patients: EpCAM high circulating

In this study we showed that the presence of CTC before therapy is a risk factor for worse tumor response rates and survival in advanced non-small cell lung cancer, irrespective

Percentage of advanced non-small cell lung cancer (NSCLC) patients with an early response (partial and complete response according to the revised response evaluation criteria in

For patients undergoing an open thoracotomy, 7.5 mL of blood was drawn from the radial artery at the start of surgery (baseline, T0), followed by blood draws from both the