Annals of Oncology Advance Access published November 16, 2015 1

Strategies for clinical implementation of TNM-immunoscore in resected non-small cell lung cancer

T. Donnem1,2, T.K. Kilvaer1, S. Andersen1, E. Richardsen3,4, EE. Paulsen1,2, S. M. Hald2, S. Al-Saad3,4, O.T. Brustugun5, A. Helland5,6, M. Lund-Iversen7, S. Solberg8, B.H. Gronberg9,10, S.G.F. Wahl11, L. Helgeland12, O. Fløtten13, M. Pohl14, K. Al-Shibli15, T.M. Sandanger16, F. Pezzella17, LT. Busund3,4,†, R. M. Bremnes1,2,†

Department of Oncology, University Hospital of North Norway, Norway

2

Institute of Clinical Medicine, The Arctic University of Norway, Norway

3

Department of Clinical Pathology, University Hospital of North Norway, Norway

4

Institute of Medical Biology, The Arctic University of Norway, Norway

5

Department of Oncology, Oslo University Hospital, The Norwegian Radium Hospital, Oslo,

Norway 6

Department of Cancer Genetics, Oslo University Hospital, The Norwegian Radium Hospital,

Oslo, Norway 7

Department of Pathology, Oslo University Hospital, The Norwegian Radium Hospital, Oslo,

Norway 8

Department of Cardiothoracic Surgery, Oslo University Hospital, Rikshospitalet, Oslo,

Norway 9

The Cancer Clinic, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway

10

European Palliative Care Research Centre, Department of Cancer Research and Molecular

Medicine, Norwegian University of Science and Technology, Trondheim, Norway 11

Department of Pathology and Medical Genetics, St. Olavs Hospital - Trondheim University

Hospital, Trondheim, Norway © The Author 2015. Published by Oxford University Press on behalf of the European Society for Medical Oncology. All rights reserved. For permissions, please email: [email protected].

Downloaded from http://annonc.oxfordjournals.org/ by guest on November 19, 2015

1

2 12

Department of Pathology, Haukeland University Hospital, Bergen, Norway

13

Department of Thoracic Medicine, Haukeland University Hospital, Bergen, Norway

14

Department of Oncology, Rigshospitalet, Copenhagen, Denmark

15

Department of Pathology, Nordland Hospital, Bodo, Norway

16

Department of Community Medicine, The Artic University of Tromso, Norway

17

Nuffield Department of Clinical Laboratory Sciences, University of Oxford, John Radcliffe

Hospital, Oxford, UK

Oncology, The Arctic University of Norway / University Hospital of North Norway, 9038 Tromso, Norway, Telephone: +47 77645427 / +47 77626000, Fax: +47 77626779, E-mail: [email protected] †These

authors have contributed equally to this paper.

Downloaded from http://annonc.oxfordjournals.org/ by guest on November 19, 2015

Corresponding author: Professor Tom Donnem, Institute of Clinical Medicine / Department of

3

ABSTRACT

Immunoscore is a prognostic tool defined to quantify in situ immune cell infiltrates, and appears highly promising as a supplement to the TNM classification of various tumors. In colorectal cancer, an international task force has initiated prospective multicenter studies aiming to implement TNM-Immunoscore (TNM-I) in a routine clinical setting. In breast cancer, recommendations for the evaluation of tumor infiltrating lymphocytes (TILs) have

potential obstacles related to implementing TNM-I into the clinic. Diverse methods may be needed for different malignancies and even within each cancer entity. Nevertheless, a uniform approach across malignancies would be advantageous. In non-small cell lung cancer (NSCLC), there are several previous reports indicating an apparent prognostic importance of TILs, but studies on TILs in a TNM-I setting are sparse and no general recommendations are made. However, recently published data is promising, evoking a realistic hope of a clinical useful NSCLC TNM-I. This review will focus on the TNM-I potential in NSCLC and propose strategies for clinical implementation of a TNM-I in resected NSCLC.

Key Words: Immunoscore, T-cells, TNM-I, NSCLC, lung cancer

Key message: This Review focuses on the TNM-Immunoscore (TNM-I) potential in nonsmall cell lung cancer (NSCLC) and proposes strategies for clinical implementation of a TNM-I in resected NSCLC.

Downloaded from http://annonc.oxfordjournals.org/ by guest on November 19, 2015

been proposed by an international working group. Regardless of promising results, there are

4

Introduction Lung cancer is the leading cause of cancer-related morbidity and mortality worldwide [1]. In addition, lung cancer affects also younger patients, and is responsible for as many lost lifeyears as colorectal, breast and prostate cancers combined [2]. Lung cancer is divided in smallcell lung cancer (SCLC) and non-small cell lung cancer (NSCLC), the latter representing approximately 85% of all lung cancer patients [3]. About 20-25% of NSCLC are resectable (stage I-IIIA) at the time of diagnosis and potentially curable. There is a considerable

current 7th edition of the lung cancer staging system based on an initiative undertaken by the International Association for the Study of Lung Cancer (IASLC) [4]. In the TNM staging system there is, however, evidently room for improvement as clinical outcome vary significantly among patients with the same pathological stage (pStage). In resected breast and colorectal cancer patients there is a similar picture. The cancers may be potentially curable, but whereas the TNM classification is important in clinical decision making, the outcomes vary significantly within each TNM stage. Common for all these malignancies is the present interest in the immune contexture and the influence of tumor infiltrating immune cells on clinical outcome [5,6]. Different types of infiltrating immune cells have diverse effects on tumor progression and clinical outcome according to context and cancer type [5]. Interestingly, the most consistent positive prognostic impact has been shown for T-cells, especially cytotoxic T-cells, memory T-cells and T helper cells-1 [6]. Two commendable NSCLC-specific reviews on this topic were recently published [7,8]. These papers discuss the prognostic impact of different immune cells and the prognostic and potential predictive impact of immune checkpoint T-cell markers as programmed death -1 (PD1), PD-ligand 1 (PD-L1) and cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4).

Downloaded from http://annonc.oxfordjournals.org/ by guest on November 19, 2015

difference in prognosis between stage I and stage IIIA patients (Figure 1) as shown in the

5

In colorectal cancer, an impressive diagnostic development has been achieved by using assessment of infiltrating immune T cells in cancer tissue to create an “Immunoscore” as an essential important supplement to TNM, designated TNM-Immune (TNM-I) [9-11]. A worldwide task force has ongoing prospective multicenter studies aiming to implement TNMI in clinical decision making [9-11]. A similar international working group has evaluated the role of tumor infiltrating lymphocytes (TILs) in breast cancer and recently published their recommendations [12]. However, as shown in Table 1, there are major differences in the colorectal and the breast cancer approach. The diverse strategies in colorectal cancer and

specific. On the other hand, it may also reflect that there are several ways of evaluating potentially similar mechanisms. Further efforts to standardize and optimize the TNM-I are therefore warranted. In NSCLC, many comprehensive studies have evaluated the prognostic impact of TILs (Table 2). However, to our knowledge only one recently published study has focused on TNM-I in a NSCLC setting where the difference in prognostic impact across each pStage is emphasized [13]. A prospective multicenter NSCLC TNM-I study is planned and central issues with regard to implementing TNM-I into clinical decision making are discussed below.

Immune cell markers of interest Interestingly, the task force in colorectal cancer recommends specific TIL immune markers (CD3 and CD8), while the breast cancer working group endorses hematoxylin & eosin (H&E) staining and morphological evaluation of TILs for their immunoscore [9,11,12]. In colorectal cancer, CD45RO have also shown promising results, but due to background staining and loss of antigenicity in stored sections it was decided to use the two most successful stainings (CD3 and CD8) in the ongoing prospective multicenter colorectal study [11].

Downloaded from http://annonc.oxfordjournals.org/ by guest on November 19, 2015

breast cancer indicate that that the prognostic impact of TILs may be cancer type and context

6

Based on experiences from colorectal cancer and breast cancer, CD3, CD8, CD45RO positive cells as well as TILs evaluated by H&E staining and morphology has been the focus in the search for candidate markers in NSCLC. CD4 has also been included as it has shown prognostic impact in NSCLC in several studies. CD4 T-helper cells differentiate into Th1, Th2, Th17 and regulatory T-cells (Tregs). Several studies have assessed prognostic roles of these T cell markers in lung cancer [6,14], but due to conflicting prognostic impact in both lung cancer and in several other malignancies, these have not been included in this review [6]. Using the following search criteria: i) Stage I-IIIA NSCLC patients, ii) studies published

assessments of TILs, we identified 15 eligible studies (Table 2) [13,15-28]. Five of these included CD3 [15,21,22,26,28], four CD4 [16,22,27,28], 11 CD8 [13,16-18,20-22,24,26-28], one CD45RO [28] and four evaluating TILs morphologically by H&E staining [19,21,23,25]. The most promising candidate marker so far seems to be CD8. The results are quite consistent showing that a high number of infiltrating CD8+ TILs is a positive prognostic factor regardless of tissue compartments evaluated, staining/scoring methods or endpoints used. There is some variation in the statistical strength (from being a strong independent prognostic variable to show significance in only univariate analyses or just indicating a trend), but none of the examined studies did show a negative prognostic impact of CD8+ TILs. A large comprehensive study by Suzuki et al did not show any prognostic trend for CD8, but they included exclusively NSCLC adenocarcinomas stage I [28]. Reassuringly, the two most recent studies which included both test and validation cohorts did show a strong independent positive prognostic impact of CD8+ TILs [13,26]. To our knowledge, merely our recent study have included the results in a TNM-I context. As presented in Figure 1, the stromal CD8+ density adds valuable additional prognostic impact within each pathological stage (pStage) [13].

Downloaded from http://annonc.oxfordjournals.org/ by guest on November 19, 2015

between 2000- and March 2015, iii) more than 200 included patients and iv) in situ

7

Another promising marker is CD3. As a pan T-cell marker it is not surprising that a high CD3+ TILs density would indicate a positive prognostic impact in the same way as evaluating TILs by H&E staining and morphology. CD4 T-helper cells show basically the same results as CD3 and CD8, but may be considered unspecific as it stains both regulatory (Tregs) and tumor suppressing T cells. Nevertheless, as a prognostic factor it is promising. To our knowledge CD45RO (memory T-cells) is only evaluated in one large NSCLC study by Suzuki et al.[28]. As this study included stage I adenocarcinomas patients only, and it showed highly significant impact in colorectal cancer, CD45RO deserves to be further elucidated in

TIL location and compartments within the tumor The presence of TILs related to compartments within the tumor is frequently a matter of debate and the terminology used is often ambiguous. Essentially, compartments mentioned are either intra-epithelial (often named “tumor” or “cancer nests”) or stromal (called “intratumoral stroma” or “tumor-related stroma”). These compartments may be located in the center or/and at the invasive margin of the tumor. In addition, lymphocytes may be located in adjacent tertiary lymphoid structures [6,16]. In colorectal cancer, the TNM-I score differentiates between central tumor and invasive margin, but the selected areas for TIL assessment (both in the central tumor and in the invasive margin) includes lymphocytes from both intra-epithelial and stromal compartments [9-11]. The breast cancer recommendations endorse solely the stromal compartment (i.e. area occupied by mononuclear inflammatory cells over the total intratumoral stroma) within the borders of the invasive margin [12]. In NSCLC prognostic studies (Table 2), the compartment of interest has often been described as intra-epithelial, stromal or a combination of these two. In some cases the intra-

Downloaded from http://annonc.oxfordjournals.org/ by guest on November 19, 2015

NSCLC [29,30].

8

epithelial and the stromal compartment have been evaluated separately. However, whether the calculated lymphocytes are located in the invasive margin or in the central parts of tumor is generally not described. In our study [13], we considered the same approach as Galon et al. as both intra-epithelial and stromal CD8+ expression turned out as positive prognostic factors in our test cohort, but the stromal expression alone showed a better prognostic impact than the combination of these two closely related compartments. Planning a prospective study, we are considering using the total expression for the simplicity and potentially easier use of computer based analysis. Basically, we have to establish an approach fitting the NSCLC biology as well

work by Galon et al. To our knowledge, only our recently published study has addressed the issue of central tumor versus invasive margin in NSCLC TNM-I [13]. In one of our cohorts the localization of each tissue core was clearly noted and, interestingly, the prognostic impact of stromal CD8+ TILs appeared to be strongest when located in the invasive margin. When it comes to evaluation of different compartments (e.g. intraepithelial versus stromal) Schalper et al. has taken this issue a step further by using multiplexed quantitative fluorescence (QIF) [26]. QIF score of each fluorescence channel in the tumor and/or stromal compartment was calculated by dividing the target TIL marker pixel intensity by the area of desired compartment defined by the cytokeratin positivity (i.e. intra-epithelial compartment), absence of cytokeratin positivity (i.e. stromal compartment) or DAPI positive cells (e.g. including both intraepithelial and stromal compartments). They advocate for this method and state that differentiating between intra-epithelial and stromal staining is pivotal in optimizing the assessment of the prognostic impact of CD8+ TILs in NSCLC. This is in contrast to the colorectal TNM-I approach where scoring was a mix between these two compartments. Based on our findings, we believe that both approaches may have strengths and weaknesses. Our data indicate a stronger impact based on assessments in the stromal compartment for CD8+

Downloaded from http://annonc.oxfordjournals.org/ by guest on November 19, 2015

as possible. In this process some solutions may differ from the impressive TNM-I pioneer

9

density, supporting Schalper et al, but localization (invasive margin) is definitively also an issue in NSCLC TNM-I. Combining these strategies may be a sound solution, but further research is warranted to clarify these important questions.

Tissue microarrays versus whole slides in the assessment of TNM-I Closely related to the compartment/localization issue is the debate whether tissue microarrays (TMAs) are adequately representative or inferior to whole slides when it comes to reflecting the infiltration of lymphocytes in the tumor. The colorectal world task force argues that for

well as the invasive part. However, they are evaluating a total of only four selected areas within the whole slide, not unlike a TMA approach. In the breast cancer recommendations they also initially preferred whole slides morphological assessment (H&E staining), but concluded that TMA may be a good option for future studies, particularly for the rapid evaluation of large cohorts using immunohistochemistry based assessments of TILs subsets. More research on this issue is needed in NSCLC. Several studies have shown that TMA results largely correlate with whole slide findings [31]. The numbers of cores, their size, and the localization they are taken from are of pivotal importance. It is, however, worth mentioning that in these correlation studies, the gold standard is usually whole slides. As a tumor is three- and not two-dimensional, one may ask how representative is a slide for the whole tumor. This is a highly relevant question also in a TNM-I setting and needs further attention. Another relevant matter is the thickness of the tissue sections. In cases where the score is based on a ratio between an actual marker and the number of other nucleated cells, this is not a problem. However, when the score is based on the number of marked cell per area this

Downloaded from http://annonc.oxfordjournals.org/ by guest on November 19, 2015

routine practice purposes whole slides is favorable as it includes areas from both the center as

10

may be challenging. Most slides are 4-µm or 5-µm thick, but even this minor difference will in average lead to a result discrepancy of about 20 percent.

Challenges related to scoring – pathologist or computer? In colorectal cancer, Galon et al. advocate the use of digital scoring as computer-assisted image analyses provides important advantages [9,11]. Due to the fact that validation studies have demonstrated high concordance between digital and manual scoring the authors state that digital scoring is pivotal to facilitate routine pathology and to speed up the process of

approaches, while promising, have not been published in large series with consistent methodology and, hence, is an important area for further research. Digital imaging may further improve the reproducibility in the assessment of percentages of immune positive area compared to visual semiquantitative estimation. This is also the key message in a published study evaluating TILs in colorectal cancer, focusing on this issue [32]. However, in the same paper the authors also state that the semiquantitative scoring showed an excellent reproducibility (T-cell markers CD3 and FoxP3 were evaluated) and conclude that computer-assisted image analysis provides a valuable alternative to semiquantitative assessments in immunohistochemically stained sections. It is important to note that manual exact count is the gold standard that both the semiquantitative and the computer-based analysis are compared against. As mentioned, digital scoring analysis is less time-consuming when finally established. This is the case when the area to be evaluated is well defined and the computer counts the number of stained cells. In NSCLC, stromal and intra-epithelial compartments are very closely related. Consequently, evaluation of only one of these compartments is regarded challenging. Hence, in a digital setting a pathologist will manually have predefined the correct

Downloaded from http://annonc.oxfordjournals.org/ by guest on November 19, 2015

quantification. In contrast, the breast cancer recommendations conclude that digital scoring

11

compartments. Thus in a prospective study one may as Galon et al. consider not to evaluate the TIL markers in the intra-epitelial and stromal compartment separately. Several wellperformed NSCLC studies have already used a computer-based scoring approach when evaluating TIL specific markers [17,18,20,24,26], and the previously mentioned comprehensive study by Schalper et al. addressed the issue of different compartments (intraepithelial versus stromal). Their method seems promising and further testing with a digital approach in prospective studies is needed before concluding on this matter. A major advantage with digital analyses (as for the more time-consuming exact count)

multivariate prediction models without any cut-offs. Continuous data are also much more flexible in an explorative study where cut-off is to be decided. Median, percentiles and “best p-value” are frequently used cut-off strategies, all with potential strengths and weaknesses. In any case, cut-offs must be predefined and validated in several cohorts prior to entering a prospective multicenter study and, finally, potentially be used in NSCLC TNM-I clinical decision making.

Future directions There are challenges related to translating promising prognostic immunoscore results into a TNM-I clinical setting. For the time being the world task force in colorectal cancer seems to be closest in reaching such a goal. The breast cancer working group recently has published some general recommendations. They acknowledge the immense complexity of lymphocyte assessment and suggest that closer molecular characterization of the TIL infiltrates may add both sensitivity and specificity to the predictive value of morphologically recognized TILs [12]. In NSCLC, the interest for TILs as a prognostic tool is increasing, but a shift from

Downloaded from http://annonc.oxfordjournals.org/ by guest on November 19, 2015

compared to a semi quantitative score is that a continuous data variable can be used directly in

12

promising retrospective studies to larger prospective multicenter studies with focus on clinical use is warranted. So far stromal CD8 seems to be the most promising candidate in NSCLC as several studies have shown its positive prognostic impact and the fact that additional impact can be clearly demonstrated across each pStage (Figure 1). In addition CD3, CD4 and CD45RO are potentially candidate markers to be included in a prospective study, but validated results on the prognostic impact of these markers according to pStage are so far sparse. As there is a close connection between these markers and mechanisms related to new

checkpoint inhibitors targeting CTLA-4/PD1/PD-L1 are highly in need of valid predictive markers. PD-L1 expression by immunohistochemistry (IHC) has been found to predict response to PD1 and PD-L1 inhibitors, but patient with a low PD-L1 score may still respond to these antibodies [7]. As the TNM-I is a pathological TNM-I (pTNM-I), it may only be used as a predictor of treatment response in a postoperative setting. Yet, evaluating TNM-I as a predictive tool in an adjuvant immunotherapy setting is highly interesting. Nevertheless, both as a prognostic and potentially as a predictive tool, the IHC procedure and the scoring systems need to be standardized. The greatest challenge may be the heterogeneity of TILs in the tumor. Besides, the reproducibility and reliability have to be significantly improved as we move from predicting survival in large cohorts to testing sensitivity and specificity at a patient level. However, as the prognostic impact of TILs in NSCLC is promising the aim should be to develop a clinically useful NSCLC TNM-I by refining the current knowledge combined with prospective multicenter studies.

Downloaded from http://annonc.oxfordjournals.org/ by guest on November 19, 2015

targeted immunotherapy, there is a potential predictive value of TNM-I as well. Immune

13

Acknowledgements We want to thank the Norwegian Cancer Society and Northern Norway Health Region Authority for financial support. Conflict of interest No conflict of interest.

Figure 1. From TNM to TNM-Immunoscore (TNM-I) in Non-Small Cell Cancer. The American Joint Committee of Cancer (AJCC) and the Union Internationale Contre le Cancer (UICC) define and periodically update the TNM classification system. In lung cancer, the current 7th edition of the lung cancer staging system is based on an initiative undertaken by the International Association for the Study of Lung Cancer (IASLC). (A) Five-year overall survival (OS) rates in 797 resected, stage I-IIIA, NSCLC patients from four different Scandinavian cohorts is comparable to IASLC data [4,13]. The combination of stromal CD8 expression (B) and pathological stage (C) survival data [13] results in a TNM-Immunoscore (D) adding significant prognostic impact across each pathological stage.

Downloaded from http://annonc.oxfordjournals.org/ by guest on November 19, 2015

Legend of figure

14

References 1. Siegel R, Naishadham D, Jemal A. Cancer statistics, 2012. CA Cancer J Clin 2012; 62: 10-29. 2. Brustugun OT, Moller B, Helland A. Years of life lost as a measure of cancer burden on a national level. Br J Cancer 2014; 111: 1014-20. 3. Goldstraw P, Ball D, Jett JR et al. Non-small-cell lung cancer. Lancet 2011; 378: 1727-40. 4. Goldstraw P, Crowley J, Chansky K et al. The IASLC Lung Cancer Staging Project: proposals for the revision of the TNM stage groupings in the forthcoming (seventh) edition of the TNM Classification of malignant tumours. J Thorac Oncol 2007; 2: 706-14.

chronic inflammation at the tumor site on cancer development, progression, and prognosis: emphasis on non-small cell lung cancer. J Thorac Oncol 2011; 6: 824-33. 6. Fridman WH, Pages F, Sautes-Fridman C et al. The immune contexture in human tumours: impact on clinical outcome. Nat Rev Cancer 2012; 12: 298-306. 7. Anagnostou VK, Brahmer JR. Cancer immunotherapy: a future paradigm shift in the treatment of non-small cell lung cancer. Clin Cancer Res 2015; 21: 976-84. 8. Remark R, Becker C, Gomez JE et al. The non-small cell lung cancer immune contexture. A major determinant of tumor characteristics and patient outcome. Am J Respir Crit Care Med 2015; 191: 377-90. 9. Galon J, Pages F, Marincola FM et al. Cancer classification using the Immunoscore: a worldwide task force. J Transl Med 2012; 10: 205. 10. Galon J, Angell HK, Bedognetti D et al. The continuum of cancer immunosurveillance: prognostic, predictive, and mechanistic signatures. Immunity 2013; 39: 11-26. 11. Galon J, Mlecnik B, Bindea G et al. Towards the introduction of the 'Immunoscore' in the classification of malignant tumours. J Pathol 2014; 232: 199-209.

Downloaded from http://annonc.oxfordjournals.org/ by guest on November 19, 2015

5. Bremnes RM, Al-Shibli K, Donnem T et al. The role of tumor-infiltrating immune cells and

15 12. Salgado R, Denkert C, Demaria S et al. The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an International TILs Working Group 2014. Ann Oncol 2015; 26: 259-71. 13. Donnem TP, Hald SM, Paulsen EE et al. Stromal CD8+ T Cell Density - A Promising Supplement to TNM staging in Non-Small Cell Lung Cancer. Clin Cancer Res 2015; 21: 2635-43. 14. Duan MC, Zhong XN, Liu GN et al. The Treg/Th17 paradigm in lung cancer. J Immunol Res 2014; 2014: 730380. 15. Al-Shibli K, Al-Saad S, Andersen S et al. The prognostic value of intraepithelial and stromal

16. Al-Shibli KI, Donnem T, Al-Saad S et al. Prognostic effect of epithelial and stromal lymphocyte infiltration in non-small cell lung cancer. Clin Cancer Res 2008; 14: 5220-7. 17. Alifano M, Mansuet-Lupo A, Lococo F et al. Systemic inflammation, nutritional status and tumor immune microenvironment determine outcome of resected non-small cell lung cancer. PLoS One 2014; 9: e106914. 18. Goc J, Germain C, Vo-Bourgais TK et al. Dendritic cells in tumor-associated tertiary lymphoid structures signal a Th1 cytotoxic immune contexture and license the positive prognostic value of infiltrating CD8+ T cells. Cancer Res 2014; 74: 705-15. 19. Horne ZD, Jack R, Gray ZT et al. Increased levels of tumor-infiltrating lymphocytes are associated with improved recurrence-free survival in stage 1A non-small-cell lung cancer. J Surg Res 2011; 171: 1-5. 20. Ilie M, Hofman V, Ortholan C et al. Predictive clinical outcome of the intratumoral CD66bpositive neutrophil-to-CD8-positive T-cell ratio in patients with resectable nonsmall cell lung cancer. Cancer 2012; 118: 1726-37. 21. Johnson SK, Kerr KM, Chapman AD et al. Immune cell infiltrates and prognosis in primary carcinoma of the lung. Lung Cancer 2000; 27: 27-35.

Downloaded from http://annonc.oxfordjournals.org/ by guest on November 19, 2015

CD3-, C. APMIS 2010; 118: 371-82.

16 22. Kayser G, Schulte-Uentrop L, Sienel W et al. Stromal CD4/CD25 positive T-cells are a strong and independent prognostic factor in non-small cell lung cancer patients, especially with adenocarcinomas. Lung Cancer 2012; 76: 445-51. 23. Kilic A, Landreneau RJ, Luketich JD et al. Density of tumor-infiltrating lymphocytes correlates with disease recurrence and survival in patients with large non-small-cell lung cancer tumors. J Surg Res 2011; 167: 207-10. 24. Kim MY, Koh J, Kim S et al. Clinicopathological analysis of PD-L1 and PD-L2 expression in pulmonary squamous cell carcinoma: Comparison with tumor-infiltrating T cells and the

25. Ruffini E, Asioli S, Filosso PL et al. Clinical significance of tumor-infiltrating lymphocytes in lung neoplasms. Ann Thorac Surg 2009; 87: 365-71. 26. Schalper KA, Brown J, Carvajal-Hausdorf D et al. Objective measurement and clinical significance of TILs in non-small cell lung cancer. J Natl Cancer Inst 2015; 107. 27. Sterlacci W, Wolf D, Savic S et al. High transforming growth factor beta expression represents an important prognostic parameter for surgically resected non-small cell lung cancer. Hum Pathol 2012; 43: 339-49. 28. Suzuki K, Kadota K, Sima CS et al. Clinical impact of immune microenvironment in stage I lung adenocarcinoma: tumor interleukin-12 receptor beta2 (IL-12Rbeta2), IL-7R, and stromal FoxP3/CD3 ratio are independent predictors of recurrence. J Clin Oncol 2013; 31: 490-8. 29. Pages F, Kirilovsky A, Mlecnik B et al. In situ cytotoxic and memory T cells predict outcome in patients with early-stage colorectal cancer. J Clin Oncol 2009; 27: 5944-51. 30. Pages F, Berger A, Camus M et al. Effector memory T cells, early metastasis, and survival in colorectal cancer. N Engl J Med 2005; 353: 2654-66. 31. Kallioniemi OP, Wagner U, Kononen J et al. Tissue microarray technology for high-throughput molecular profiling of cancer. Hum Mol Genet 2001; 10: 657-62.

Downloaded from http://annonc.oxfordjournals.org/ by guest on November 19, 2015

status of oncogenic drivers. Lung Cancer 2015; 88: 24-33.

17 32. Vayrynen JP, Vornanen JO, Sajanti S et al. An improved image analysis method for cell counting lends credibility to the prognostic significance of T cells in colorectal cancer. Virchows Arch 2012; 460: 455-65.

Downloaded from http://annonc.oxfordjournals.org/ by guest on November 19, 2015

18

Downloaded from http://annonc.oxfordjournals.org/ by guest on November 19, 2015

Table 1. At a glance - recommondations TNM-Imunnoscore (TNM-I) in colorectal cancer and breast cancer and status in non-small cell lung cancer (NSCLC) TNM-I. Breast cancer [12]

NSCLC

Lymphocyte markers

CD3 and CD8

Tumor infiltrating lymphocytes, TILs by H&E staining. All mononuclear cells should be stained

Stromal CD8 most conclusive candidate so far. But further studies needed on promising T-cell markers as CD3, CD4, CD45RO

Compartment

Both stromal and intraepithelial CD8 and CD3 positive cells included

Stromal compartment

Both stromal and intraepithelial compartment is promising. Potentially a combination of both

Invasive margin versus tumor center

Both invasive margin and central areas included

Borders of the invasive margin should be evaluated

Only one study has addressed this issue. For stromal CD8, invasive margin looks most promising

Score

Score based on a combination of CD3 and CD8 score in both central tumor and invasive margin

TILs should be assessed as a continuous parameter. No formally recommendation for a clinically relevant threshold(s) can be given at this stage

Too premature to conclude. Stromal CD8 looks promising – see Figure 1

TMA versus whole slides

Whole slides recommended, but selected areas from invasive margin and central tumor are scored

Full sections are preferred over biopsies whenever possible, but cores may be used in pre-therapeutic neoadjuvant setting

Promising results based on TMA, but whole slides may be preferred to better address localization - central tumor versus invasive margin

Manual versus digital scoring

Digital (computer based) scoring recommended

Manual scoring ok, digital scoring important in further research

Manual scoring shows clearly significant results across each pStage, but digital scoring may be more precise and less timeconsuming when established

Continuous versus semiquantitative variable

Scored as a continuous variable, but redefined as a semiquantitative variable

Ideally should be scored as a continuous variable (% stromal TILs). However, pathologists should report their scores in as much detail as the pathologist feel comfortable with

Semiquantitative scoring seems promising, but continuous variable may be preferred in the scoring to best decide cut-off

Formally recommended threshold

Immunscore established – score 1 to 4 based on the above mentioned combination

No

No

Downloaded from http://annonc.oxfordjournals.org/ by guest on November 19, 2015

Colorectal cancer [9, 11]

Downloaded from http://annonc.oxfordjournals.org/ by guest on November 19, 2015

Table 2. Characteristics of non-small cell cancer studies between 2000 and March 2015 with focus on in situ assessment of tumor infiltrating lymphocytes. Study

Year

N

Histology

pStage

Marker

Compartment

Invasive margin vs. tumor center addressed Yes

TMA vs. WS

- Scoring approach - Digital versus manual scoring - Cut-off

Donnem T et al [13]

2015

797

ADC, SCC, LCC

I-IIIA

CD8

Stromal

TMA

“Representative intratumoral areas” (mix intra-epithelial and stromal). Intra-epithelial, stromal and total.

No

TMA

- Percentages CD8+ / total amount of nucleated cells - Manual - Predefined three-tiered cut-off - CD8+ TILs per unit area (mm2) - Digital - Median cut-off

Kim MY et al [24]

2015

331

SCC

I-III

CD8, PD1, PDL1

Schalper KA et al [26]

2015

552

ADC, SCC, Other

I-IV

CD3, CD8, CD20

No

TMA

Alifano M et al [17]

2014

303

ADC, SCC, LCC, Others

I-IV

CD8, CD1A

Tumor nests (intra-epithelial) and stromal

No

WS

Goc J et al [18]

2014

458

ADC, SCC, Others

I-IV

DC, CD8

Tumor nests (intra-epithelial) and stromal

No

WS

Suzuki K et al [28]

2013

956

ADC

pStage I only

Tumor nests (intra-epithelial) and stromal

No

TMA

ADC, SCC, LCC

I-IV

FoxP3/C D3 ratio, CD4,CD8, CD45RO CD3, CD3/CD8 CD4/ CD25

Kayser G et al [22]

2012

232

Intra-epithelial and stromal

No

TMA

- Quantitative measurement of fluorescent signal (QIF) calculated by dividing the target TIL marker pixel intensity by the area of desired compartment. - Digital and manual - Cut-off: tertiles and categorized - Intra-tumoral density of CD8+ T-cells (Both stromal area and tumor nests gave significant associations, but they decided to report findings only from tumor cells nests) - Digital - “Minimum p-value approach”. Cut-off: 96 cells/mm2 - Absolute number positive cells/mm2 - Digital and manual -“Minimum p-value approach” Cut-off: Tumor nests: 114 cells/mm2 Stroma: 383 cells/ mm2 - Marked cells/field - Manual - Three-tiered score categorized in high and low - The absolute number of each IHC detectable subgroup/core - Manual - Median cut-off

Validation cohort

Prognostic impact (PI)

PI across each stage/TNM addressed Yes Significant within each pStage No

Yes

Independent positive PI

No

Significant positive PI in univariate with DFS as endpoint

Yes

CD8 independent positive PI in both cohorts. CD3 independent positive in one cohort

No

No

CD8 independent positive PI in total material and combined stage I-II

Partly

No

Both intra-epithelial and stromal CD8 significant positive PI in univariate

No

Yes

No marker significant PI in univariate

No

Stromal CD3 positive PI in univariate. Stromal CD4/CD25 independent PI

Included pStage I patients only No

Year

N

Histology

pStage

Marker

Compartment

Invasive margin vs. tumor center addressed

TMA vs. WS

- Scoring - Digital vs. manual - Cut-off

Ilie M et al [20]

2012

632

ADC, SCC, LCC, Others

I-III

CD8, CD8/ CD66b ratio

Intratumoral (mix Intraepithelial and stromal)

Central areas of the tumor

TMA

Sterlacci et al [27]

2012

383

I-IV

CD4 CD8

Intra-epithelial

No

TMA

Horne ZD et al [19]

2011

273

ADC, SCC, LCC ADC, SCC, LCC

1A

TILs

Combination tumor nests (intra-epithelial) and stromal

No

WS

Kilic A et al [23]

2011

219

ADC, SCC, Other

1A-1B

TILs

No

WS

Al-Shibli et al [15]

2010

335

ADC, SCC, LCC

I-IIIA

CD3, CD117, CD 138

Combination tumor nests (intra-epithelial) and stromal Intra-epithelial and stromal

No

TMA

Ruffini et al [25]

2009

1290

ADC, SCC, LCC, Others

I-IIIA

TILs

Intra-epithelial

No

WS

Al-Shibli et al [16]

2008

335

ADC, SCC, LCC

I-IIIA

CD8, CD4

Intra-epithelial and stromal

No

TMA

Johnson SK et al [21]

2000

710 (95)

I-III

TILs CD3 CD8

Intra-epithelial and stromal

No

WS

- Density as the number of positive cells per unit of tissue surface area - Digital - Median cut-off. The median ± SD was 110 ± 142 cells - Number of positive cells/area - Manual - Median cut-off - Subjective low (scattered), moderate and high (intense) TIL infiltration - Manual - Cut-off: TIL - : low infiltration, TIL+: modest and high infiltration - “Routine protocol by an expert academic pathologist; group 1, none to mild, or group 2, moderate to serve infiltrate” - Manual - Stroma, high if they represented > 50% of nucleated cells in stroma. Intraepithelial, high if they represented more than 1% of nucleated cells -Manual - TILs were evaluated morphologically, only in a subset of 21 patients IHC - Manual - Cut-off: Low: < 20% of cells, high: ≥ 20% of cells - Percentages marker / total amount of nucleated cells - Manual - Cut-off: CD4, intraepithelial ≥ 5% , CD4, stromal ≥ 25%, CD8, intraepithelial > 5%, CD8, stromal > 50% - In 710 patients: Morphologically TIL by H&E staining. Semiquantitative score. In subset of 95 patients: CD3 and CD8, intratumoral, low if < 10%, high if ≥ 10% cells per high power field - Manual and digital

Downloaded from http://annonc.oxfordjournals.org/ by guest on November 19, 2015

Study

Validation cohort

Prognostic impact (PI)

PI across each stage/TNM addressed

No

CD8 not significant PI in univariate. CD66b/CD8 ratio independent positive PI CD4/CD8 ratio positive PI for ADC, in stage IA only Significant positive PI with recurrence free survival as endpoint

No

No

Positive PI in univariate

Stage IA and IB only

No

Stromal CD3 independent positive PI. Stromal CD3 PI in pStage IIIA, trend in I and II Independent positive PI in the total cohort and in SCC. Univariate: pStage I, SCC Stromal CD4 and CD8 independent positive prognostic factors. Intraepithelial PI in univariate. TIL total cohort not significant. Subset 95 patients, intraepithelial CD3 positive PI in univariate

Partly

No

No

No

No

No

Partly

Stage IA only

Partly

No

No

Abbreviations: SCC, squamous cell carcinoma; LCC, large-cell carcinoma; ADC, adenocarcinoma; TILs, tumor infiltrating lymphocytes; PD1, programmed death -1; PDL1, PD-ligand 1; TMA, tissue microarrays; WS, whole slides; N, number of patients; PI, prognostic impact; DFS, disease-free survival.

Strategies for clinical implementation of TNM-Immunoscore in resected nonsmall-cell lung cancer.

Immunoscore is a prognostic tool defined to quantify in situ immune cell infiltrates and appears highly promising as a supplement to the tumor-node-me...
565B Sizes 0 Downloads 10 Views