+ All Categories
Home > Documents > Intratumoral CD3 and CD8 T-cell Densities Associated with ... · Research Article Intratumoral CD3...

Intratumoral CD3 and CD8 T-cell Densities Associated with ... · Research Article Intratumoral CD3...

Date post: 05-Jun-2020
Category:
Upload: others
View: 9 times
Download: 0 times
Share this document with a friend
13
Research Article Intratumoral CD3 and CD8 T-cell Densities Associated with Relapse-Free Survival in HCC Andrew Gabrielson 1 , Yunan Wu 2 , Hongkun Wang 1 , Jiji Jiang 1 , Bhaskar Kallakury 3 , Zoran Gatalica 4 , Sandeep Reddy 4 , David Kleiner 5 , Thomas Fishbein 6 , Lynt Johnson 7 , Eddie Island 6 , Rohit Satoskar 6 , Filip Banovac 8 , Reena Jha 8 , Jaydeep Kachhela 1 , Perry Feng 1 , Tiger Zhang 1 , Anteneh Tesfaye 1 , Petra Prins 1 , Christopher Loffredo 1 , John Marshall 1 , Louis Weiner 1 , Michael Atkins 1 , and Aiwu Ruth He 1 Abstract Immune cells that inltrate a tumor may be a prognostic factor for patients who have had surgically resected hepatocel- lular carcinoma (HCC). The density of intratumoral total (CD3 þ ) and cytotoxic (CD8 þ ) T lymphocytes was measured in the tumor interior and in the invasive margin of 65 stage I to IV HCC tissue specimens from a single cohort. Immune cell density in the interior and margin was converted to a binary score (0, low; 1, high), which was correlated with tumor recurrence and relapse-free survival (RFS). In addition, the expression of programmed death 1 (PD-1) and programmed death ligand 1 (PD-L1) was correlated with the density of CD3 þ and CD8 þ cells and clinical outcome. High densities of both CD3 þ and CD8 þ T cells in both the interior and margin, along with corresponding Immunoscores, were signicantly associ- ated with a low rate of recurrence (P ¼ 0.007) and a prolonged RFS (P ¼ 0.002). In multivariate logistic regression models adjusted for vascular invasion and cellular differentiation, both CD3 þ and CD8 þ cell densities predicted recurrence, with odds ratios of 5.8 [95% condence interval (CI), 1.621.8] for CD3 þ and 3.9 (95% CI, 1.114.1) for CD8 þ . Positive PD-L1 staining was correlated with high CD3 and CD8 density (P ¼ 0.024 and 0.005, respectively) and predicted a lower rate of recurrence (P ¼ 0.034), as well as prolonged RFS (P ¼ 0.029). Immuno- score and PD-L1 expression, therefore, are useful prognostic markers in patients with HCC who have undergone primary tumor resection. Cancer Immunol Res; 4(5); 112. Ó2016 AACR. Introduction Surgical resection is frequently used as a curative treatment for patients with hepatocellular carcinoma (HCC) who have well- compensated liver function and no signicant portal hyperten- sion. However, the rate of HCC recurrence within 5 years of resection has been reported as ranging from 34% to 100% in heterogeneous patient populations (1). Conventional methods of assessing clinical and pathologic risk in patients with HCC typ- ically involve excision of the primary tumor, followed by histo- pathologic analysis of tissue samples collected during the resec- tion procedure. The extent of tumor burden and aggressive phenotype of cancer cells have been integrated in the American Joint Committee on Cancer/Union of International Cancer Con- troltumornodemetastasis (AJCC/UICC-TNM) tumor staging classication method, which categorizes the extent of the tumor burden (T), the presence of tumor-derived cells in proximal lymph nodes (N), and the degree of metastasis (M; refs. 24). This classication has been adopted to predict relapse-free sur- vival (RFS), disease-specic survival (DSS), and overall survival (OS) rates in patients after curative resection (5). Recently, addi- tional tumor cell characteristics such as abnormal nuclear mor- phology, altered expression of oncogenes (P53, c-myc, b-catenin, hMSH2), and increased intratumoral vascular density have been suggested as predictive of HCC recurrence, RFS, and OS (6). However, few such relationships have been independently validated. Although the AJCC/UICC-TNM system provides information regarding patient prognosis after curative resection of HCC, large variations in RFS and OS exist among patients with the same stage (pTNM) of disease (7). Some patients with advanced-stage cancer (III/IV) can remain free of cancer progression for years, whereas patients with early-stage cancer (I/II) can experience aggressive tumor recurrence, progression, metastases, and death following apparent complete surgical resection. The AJCC/UICC-TNM sys- tem only describes tumor burden on a macro scale; it does not provide any detailed information on features of HCC biology nor the microenvironment in which HCC proliferates (8). 1 Georgetown Lombardi Comprehensive Cancer Center, Division of Hematology and Oncology, Washington, District of Columbia. 2 First Hospital of Hunan University of Chinese Medicine, Changsha City, Hunan Province, People's Republic of China. 3 Department of Pathol- ogy, Georgetown University Hospital,Washington, District of Colum- bia. 4 Caris Life Science, Irving, Texas. 5 Center for Cancer Research, National Cancer Institute, Bethesda, Maryland. 6 Medstar Transplant Institute, Georgetown University Hospital, Washington, District of Columbia. 7 Department of Surgery, Georgetown University Hospital, Washington, District of Columbia. 8 Department of Radiology, George- town University Hospital, Washington, District of Columbia. Note: Supplementary data for this article are available at Cancer Immunology Research Online (http://cancerimmunolres.aacrjournals.org/). A. Gabrielson and Y. Wu contributed equally to this article. Corresponding Author: Aiwu Ruth He, Lombardi Comprehensive Cancer Center, Department of Oncology and MedStar Georgetown University Hospital Division of Hematology/Oncology, 3800 Reservoir Road, NW, Washington, DC 20007. Phone: 202-444-8642; Fax: 2024-444-9429; E-mail: [email protected] doi: 10.1158/2326-6066.CIR-15-0110 Ó2016 American Association for Cancer Research. Cancer Immunology Research www.aacrjournals.org OF1 on June 11, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from Published OnlineFirst March 11, 2016; DOI: 10.1158/2326-6066.CIR-15-0110
Transcript
Page 1: Intratumoral CD3 and CD8 T-cell Densities Associated with ... · Research Article Intratumoral CD3 and CD8 T-cell Densities Associated with Relapse-Free Survival in HCC Andrew Gabrielson1,Yunan

Research Article

Intratumoral CD3 and CD8 T-cell DensitiesAssociated with Relapse-Free Survival in HCCAndrew Gabrielson1, Yunan Wu2, Hongkun Wang1, Jiji Jiang1, Bhaskar Kallakury3,Zoran Gatalica4, Sandeep Reddy4, David Kleiner5, Thomas Fishbein6, Lynt Johnson7,Eddie Island6, Rohit Satoskar6, Filip Banovac8, Reena Jha8, Jaydeep Kachhela1,Perry Feng1, Tiger Zhang1, Anteneh Tesfaye1, Petra Prins1, Christopher Loffredo1,John Marshall1, Louis Weiner1, Michael Atkins1, and Aiwu Ruth He1

Abstract

Immune cells that infiltrate a tumor may be a prognosticfactor for patients who have had surgically resected hepatocel-lular carcinoma (HCC). The density of intratumoral total(CD3þ) and cytotoxic (CD8þ) T lymphocytes was measuredin the tumor interior and in the invasive margin of 65 stage I toIV HCC tissue specimens from a single cohort. Immune celldensity in the interior and margin was converted to a binaryscore (0, low; 1, high), which was correlated with tumorrecurrence and relapse-free survival (RFS). In addition, theexpression of programmed death 1 (PD-1) and programmeddeath ligand 1 (PD-L1) was correlated with the density of CD3þ

and CD8þ cells and clinical outcome. High densities of bothCD3þ and CD8þ T cells in both the interior and margin, along

with corresponding Immunoscores, were significantly associ-ated with a low rate of recurrence (P ¼ 0.007) and a prolongedRFS (P ¼ 0.002). In multivariate logistic regression modelsadjusted for vascular invasion and cellular differentiation, bothCD3þ and CD8þ cell densities predicted recurrence, with oddsratios of 5.8 [95% confidence interval (CI), 1.6–21.8] for CD3þ

and 3.9 (95% CI, 1.1–14.1) for CD8þ. Positive PD-L1 stainingwas correlated with high CD3 and CD8 density (P ¼ 0.024 and0.005, respectively) and predicted a lower rate of recurrence(P ¼ 0.034), as well as prolonged RFS (P ¼ 0.029). Immuno-score and PD-L1 expression, therefore, are useful prognosticmarkers in patients with HCC who have undergone primarytumor resection. Cancer Immunol Res; 4(5); 1–12. �2016 AACR.

IntroductionSurgical resection is frequently used as a curative treatment for

patients with hepatocellular carcinoma (HCC) who have well-compensated liver function and no significant portal hyperten-sion. However, the rate of HCC recurrence within 5 years ofresection has been reported as ranging from 34% to 100% inheterogeneous patient populations (1).Conventionalmethodsofassessing clinical and pathologic risk in patients with HCC typ-

ically involve excision of the primary tumor, followed by histo-pathologic analysis of tissue samples collected during the resec-tion procedure. The extent of tumor burden and aggressivephenotype of cancer cells have been integrated in the AmericanJoint Committee on Cancer/Union of International Cancer Con-trol–tumor–node–metastasis (AJCC/UICC-TNM) tumor stagingclassification method, which categorizes the extent of the tumorburden (T), the presence of tumor-derived cells in proximallymph nodes (N), and the degree of metastasis (M; refs. 2–4).This classification has been adopted to predict relapse-free sur-vival (RFS), disease-specific survival (DSS), and overall survival(OS) rates in patients after curative resection (5). Recently, addi-tional tumor cell characteristics such as abnormal nuclear mor-phology, altered expression of oncogenes (P53, c-myc, b-catenin,hMSH2), and increased intratumoral vascular density have beensuggested as predictive of HCC recurrence, RFS, and OS (6).However, few such relationships have been independentlyvalidated.

Although the AJCC/UICC-TNM system provides informationregarding patient prognosis after curative resection of HCC, largevariations in RFS andOS exist among patients with the same stage(pTNM) of disease (7). Some patients with advanced-stage cancer(III/IV) can remain free of cancer progression for years, whereaspatients with early-stage cancer (I/II) can experience aggressivetumor recurrence, progression, metastases, and death followingapparent complete surgical resection. The AJCC/UICC-TNM sys-tem only describes tumor burden on a macro scale; it does notprovide any detailed information on features of HCC biology northe microenvironment in which HCC proliferates (8).

1Georgetown Lombardi Comprehensive Cancer Center, Division ofHematology and Oncology, Washington, District of Columbia. 2FirstHospital of Hunan University of Chinese Medicine, Changsha City,Hunan Province, People's Republic of China. 3Department of Pathol-ogy, Georgetown University Hospital,Washington, District of Colum-bia. 4Caris Life Science, Irving, Texas. 5Center for Cancer Research,National Cancer Institute, Bethesda, Maryland. 6Medstar TransplantInstitute, Georgetown University Hospital, Washington, District ofColumbia. 7Department of Surgery, Georgetown University Hospital,Washington,District of Columbia. 8DepartmentofRadiology,George-town University Hospital,Washington, District of Columbia.

Note: Supplementary data for this article are available at Cancer ImmunologyResearch Online (http://cancerimmunolres.aacrjournals.org/).

A. Gabrielson and Y. Wu contributed equally to this article.

Corresponding Author: Aiwu Ruth He, Lombardi Comprehensive CancerCenter, Department of Oncology and MedStar Georgetown University HospitalDivision of Hematology/Oncology, 3800 Reservoir Road, NW, Washington,DC 20007. Phone: 202-444-8642; Fax: 2024-444-9429; E-mail:[email protected]

doi: 10.1158/2326-6066.CIR-15-0110

�2016 American Association for Cancer Research.

CancerImmunologyResearch

www.aacrjournals.org OF1

on June 11, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst March 11, 2016; DOI: 10.1158/2326-6066.CIR-15-0110

Page 2: Intratumoral CD3 and CD8 T-cell Densities Associated with ... · Research Article Intratumoral CD3 and CD8 T-cell Densities Associated with Relapse-Free Survival in HCC Andrew Gabrielson1,Yunan

Experimental data have already explored the concept of immu-nosurveillance in mouse cancer models (9, 10), but this has onlyrecently been applied to human cancers as a prognostic tool. Thepresence of immune cells flanking tumor tissue reflects a distinctunderlying biology of the tumor and may have a great impact oncancer cells in their response to secreted growth factors, inflam-matory mediators, and immune response elements (11). Inhumans, the presence of tumor-infiltrating lymphocytes (TIL)has been associated with a favorable prognosis (12–19).

Programmed death ligand 1 (PD-L1) is a 40-kDa type 1transmembrane protein that has been speculated to play a majorrole in suppressing the immune system during particular eventssuch as pregnancy, tissue allografts, autoimmune disease, andother disease states, such as hepatitis and cancer. Normally, theimmune system reacts to foreign antigens where there is someaccumulation in the lymph nodes or spleen, which triggers aproliferation of antigen-specific CD8þ T cells. The formation ofPD-1 receptor/PD-L1 or B7.1 receptor/PD-L1 ligand complexestransmits an inhibitory signal that reduces the proliferation ofthese CD8þ T cells in the lymph nodes (2, 20). Interactionbetween surface molecules and different immune cells regulatesthe antitumor immune response (21).

The tumor immune microenvironment has been defined byAngell andGalon (22) as the type, functional orientation, density,and location of adaptive immune cells within distinct tumorregions [tumor interior (TI) and the invasive margin (IM)]. Inorder to quantify the immune microenvironment and apply itto a heterogeneous group of patients, the authors proposed the"Immunoscore." Evaluation of the Immunoscore in patients withcolorectal cancer has demonstrated that it is a powerful prognostictool and may supplement the already well-established AJCC/UICC-TNM staging system (23). T-cell subpopulations (CD3þ

T lymphocytes, CD8þ cytotoxic T lymphocytes, and CD45ROþ

memory T cells) were studied and correlated with cancer recur-rence and OS in patients with colorectal cancer (23). Because ofbackground staining and loss of antigenicity in stored sectionsstained for CD45RO, the two easiest membrane stains, CD3 andCD8, were used by theworldwide Immunoscore consortiumwiththe support of the Society for Immunotherapy of Cancer (SITC)for validation in clinical practice. The Immunoscore can beobtained by performing histopathologic analysis of tumor tissuesand by applying a rubric of high/low density values to the tumor-infiltrating immune cells, with total (CD3þ) and cytotoxic(CD8þ) T lymphocytes being measured in the aforementionedstudy (23).

The prognostic value of the Immunoscore was demonstrated inpatients (n ¼ 602) with early-stage (I/II) colorectal cancer topredict OS and risk of recurrence. Five years after diagnosis, only4.8% of patients with high Immunoscores of 3 or 4 had tumorrecurrence with an OS rate of 86.2%. However, patients withImmunoscores of 0 or 1 had a tumor recurrence rate of 75% andfaced an OS rate of 27.5% (24). Spranger and colleagues reportedthat T-cell infiltration is completely excluded from tumors withhigh levels of b-catenin activity in animal models, possibly due tolack of chemokine secretion and recruitment of T cells into thetumor (25).

In the current study, the clinical utility of the Immunoscoremethodology was extended to assess the risk of recurrence andRFS in patients diagnosedwith early- and late-stageHCCwhohadreceived curative resection. A secondary aim of this study wasto identify clinicopathologic factors that were associated with

immune infiltration and the corresponding Immunoscore. Byidentifying factors that influence tumor immune infiltration, wehope to better define risk of HCC recurrence after curative resec-tion and, ultimately, improve the clinical management of high-risk patients.

Materials and MethodsPatients and database

The records of 65 consecutive patients with stage I to IV HCCwho underwent primary tumor resection at the Medstar/George-townUniversity Hospital/Lombardi Comprehensive Cancer Cen-ter (Washington, DC) between 2006 and 2015 were reviewed.Postoperative treatment was in accord with generally acceptedguidelines, and themean duration of follow-up was 39.7months(range, 9–84months). RFS was defined as the length of time fromresection of the primary tumor to documented disease progres-sion. Patient tumor samples were obtained from the RueschCenter for the Cure for Gastrointestinal Cancers' Tissue Bank atLombardi. Patients provided consent for tissue banking prior tosurgical resection of their HCC. A secure database with propersafeguards was constructed for the management of patient data.An Institutional Review Board approved the ethical, legal, andsocial implications of the project.

IHC and semiquantitative analysisPathologic slides from paraffin blocks prepared with surgical

tissue specimenwere stainedwithmonoclonal antibodies to CD3(polyclonal rabbit anti-human; Dako North America Inc.; catalogno. A0452), CD8 (monoclonal mouse anti-human; Dako NorthAmerica Inc.; catalog no. M7103), PD-1 (EH12.1 from BD Bios-cences/Pharmingen; catalog no. 561273), PD-L1 (SP142, SpringBiosciences; catalog no. M4420; SP263, Roche/Ventana MedicalSystems; catalog no. 790-4905), PD-L1_IHC_22C3 (companiondiagnostics kit antibody from Dako North America, Inc.), andb-catenin (Millipore; catalog no. 05-665). All slides were thenstained with hematoxylin and eosin (H&E). Slides stained forCD3þ, CD8þ, PD-1þ, and PD-L1þ cells were first evaluated by aclinical pathologist. The TI, IM, and noncancerous liver paren-chyma were marked for biomarker imaging. Expression of b-cate-nin expression was determined by intensity (0, þ1, þ2, or þ3)and distribution (þ1 as less than 10%of cells,þ2 as 10%–50%ofcells, and þ3 as >50% of cells).

Image capture and quantificationAfter the TI and IM were reviewed, images were captured in

areas where TIL density was focally high, as well as in regionswhere the TIL density was representative of the TI and IM as awhole; this was done under high-power magnification (�20).Images were captured as a spectral cube with a Nikon E600Epifluorescence microscope and Nuance FX Multiplex BiomarkerImaging. For both CD3þ- and CD8þ-stained slides, two images ofhigh cell density each were taken of the TI and IM. Computer-assisted image analysis was conducted using ImageJ v1.48, apublic domain, Java-based, image processing program developedat theNIH. Five built-in functions of ImageJ were used tomeasurethe number of positively stained cells per tissue surface unit insquare millimeters (see Supplementary Fig. S1 for a flowchart ofthe image analysis process, Supplementary Fig. S2 for an exampleof images obtained using computer-assisted evaluation of infil-trate densities, and Supplementary Fig. S3 for ImageJ instructionsand plug-in code).

Gabrielson et al.

Cancer Immunol Res; 4(5) May 2016 Cancer Immunology ResearchOF2

on June 11, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst March 11, 2016; DOI: 10.1158/2326-6066.CIR-15-0110

Page 3: Intratumoral CD3 and CD8 T-cell Densities Associated with ... · Research Article Intratumoral CD3 and CD8 T-cell Densities Associated with Relapse-Free Survival in HCC Andrew Gabrielson1,Yunan

The median immune cell density was used to stratify patientsinto groups based on the degree of tumor infiltration. Thismethod was independently validated by Angell and Galon intheir evaluation of the Immunoscore in colorectal cancer and wasthus adopted for the current study (22). The cutoff threshold forCD3þ andCD8þ cell density was determined to be 273 cells/mm2

and 217.5 cells/mm2, respectively (Supplementary Fig. S4). Basedon the established threshold, each patientwas given a binary score(0 as low, 1 as high) for each immune cell type (CD3þ andCD8þ)in each tumor region (TI and IM). An Immunoscore for eachpatient was obtained by summation of the four binary scorevalues, the scale being from 0 to 4. Five patient groups weredefined: patients with low densities of CD3þ and CD8þ T cells inboth tumor regions (All-Low)were classified as Im0; patientswithone high (1-High) density for one marker were classified as Im1;and patients with two (2-High), three (3-High), and four (All-High) among these two markers were classified as Im2, Im3, andIm4, respectively. Patients with a high degree of intratumoralimmune cell infiltration were those with an Immunoscore of 3þ,and patients with a low degree of immune cell infiltration werethose with an Immunoscore of �2. The corresponding Immuno-score values were correlated with tumor recurrence and RFS, aswell as being compared with the pathologic factors currentlyindicative of tumor invasiveness, such as vascular invasion, ele-vated a-fetoprotein (AFP), and advanced pTNM stage.

Statistical analysisDescriptive statistics were used to summarize patients' demo-

graphic information. The c2 test or Fisher exact test was used toexamine the association between categorical variables. For anal-ysis, patients were grouped by CD3þ density, CD8þ density, andImmunoscore. The nonparametric Kaplan–Meier methodologywas used to analyze the RFS time, and the log-rank test was used tocompare survival curves. Multivariate logistic regression modelswere fit to look at the effect of CD3þ and CD8þ density on theodds of tumor recurrence, when adjusting for other significantcovariates.

ResultsDemographics of HCC patients treated with primary surgery

The demographic and clinical characteristics of the registeredcohort treated at MedStar Georgetown University Hospital areshown in Table 1. Themean age was 61 years (range, 30–86), andthe patient group was predominantly male (77%). Regarding theetiology of HCC, 16.9% of patients had viral hepatitis B (HBV),32.3%patients had viral hepatitis C (HCV), 12.3%of patients hadcoinfection of HBV and HCV, and 38.4% of the patients hadneither virus. Using the Kaplan–Meier methodology, the estimat-ed probability of RFS following HCC was found to be 63% at 1year and 59% at 3 years, and the estimated probability of OS was82% at 1 year and 57% at 3 years.

Pathology review and IHC characteristicsSlides stained with CD3 and CD8 antibodies revealed a wide

spectrum of immune infiltrate densities in the TI and IM tumorregions (Fig. 1 and Fig. 2A, B, E, and G). The H&E stain of thetumor is shown in Fig. 2D, and the negative control for CD3and CD8 staining are shown in Fig. 2F and H, respectively. Thepattern of TILs in each region fell into 3 categories: (i) densenodular clusters of lymphocytes, (ii) diffuse, evenly distributed

lymphocytes, and (iii) rare or scattered lymphocytes (seeFig. 1A, B, and E, respectively). Clusters of lymphocytes arefound from the IM to the TI in the first two patterns of TILs(Fig. 1C and D, migration marked by red arrows), whereas thereare no obvious TILs between the IM and TI in the third patternof TIL (Fig. 1F). Bivariate analysis showed no statisticallysignificant association between the pattern of TIL and TNMstaging (P ¼ 0.219 for CD3, 0.377 for CD8), level of differen-tiation (P ¼ 0.054 for CD3, 0.283 for CD8), or vascularinvasion (P ¼ 0.096 for CD3, 0.104 for CD8; SupplementaryFig. S5). Similarly, the pattern of TILs showed no associationwith RFS or OS in either immune cell type (Supplementary

Table 1. Demographics of the HCC patient cohort

Characteristics

Age, years (mean, min–max) 61 (30–86)Sex, n (%)Female 15 (23.08)Male 50 (76.92)

Ethnicity, n (%)Missing 2 (3.08)African American 23 (35.38)Asian 19 (29.23)Caucasian 21 (32.31)

Tumor size, n (%)Small (size � 3 cm) 21 (32.31)Medium (3 < size � 5 cm) 9 (13.85)Large (size > 5 cm) 35 (53.85)

Tumor stage, n (%)Stage I 37 (56.92)Stage II 13 (20.00)Stage III 14 (21.54)Stage IV 1 (1.54)

Grade, n (%)1 6 (9.23)2 24 (36.92)3 29 (44.62)4 6 (9.23)

Serum AFP, n (%)Missing 36 (55.38)Low (serum AFP � 500) 23 (35.38)High (serum AFP > 500) 6 (9.23)

Differentiation, n (%)Poor 23 (35.38)Moderate 21 (32.31)Well 19 (29.23)Undifferentiated 2 (3.08)

Etiology, n (%)Hep B 11 (16.92)Hep C 21 (32.31)Hep B and Hep C 8 (12.31)Other 25 (38.46)

Vascular invasion, n (%)Yes 21 (32.31)No 44 (67.69)

Tumor thrombosis, n (%)Yes 15 (23.08)No 50 (76.92)

Cirrhosis, n (%)Yes 23 (35.38)No 42 (64.62)

Recurrence, n (%)Yes 19 (29.23)No 46 (70.77)

Alive, n (%)Yes 46 (70.77)No 19 (29.23)

Intratumoral CD3/CD8 Density Predicts Relapse in HCC

www.aacrjournals.org Cancer Immunol Res; 4(5) May 2016 OF3

on June 11, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst March 11, 2016; DOI: 10.1158/2326-6066.CIR-15-0110

Page 4: Intratumoral CD3 and CD8 T-cell Densities Associated with ... · Research Article Intratumoral CD3 and CD8 T-cell Densities Associated with Relapse-Free Survival in HCC Andrew Gabrielson1,Yunan

Fig. S6). Tumors with different staging, differentiation, andvascular invasion were found to have either pattern of TILs.

The number of PD-1–positive TILs varied from case to case,ranging from 0 to >1,000 cells per �10 magnification field. Amoderate number of PD-1–positive TILs (about 160 cells per�20 magnification field) were both within the TI and at the IM(Fig. 2K). In the 60 HCC samples evaluated, only two samplesshowed positive PD-L1 stain in cancer cells; 19 samples showedPD-L1 expression within or around the tumor, but in nonma-lignant (noncancer) cells (Fig. 2C, I, and J), presumably mono-nuclear cells such as macrophages, dendritic cells, and lympho-cytes; and 39 samples showed no PD-L1 stain. The IHC analysisof PD-L1 expression using three antibodies (SP142, SP263, andPD-L1_IHC_22C3) gave identical results. PD-L1 expressionshowed a positive correlation with CD3þ and CD8þ cell den-sities. Fourteen of 31 HCC samples with high CD3þ densityshowed PD-L1 expression, whereas only five of 29 HCC sampleswith low CD3þ density showed PD-L1 expression (Fisher exacttest P ¼ 0.011). Fifteen of 31 HCC samples with high CD8þ

density showed PD-L1 expression, whereas only four of 28 HCCsamples with low CD8þ density showed PD-L1 expression(Fisher exact test P ¼ 0.003). Similarly, CD3þ cell densityshowed a positive correlation with CD8þ cell densities. Twen-ty-two of 32 HCC samples with high CD3þ density showed highCD8þ cell density, whereas only 10 of 31 HCC samples with low

CD3þ density showed high CD8þ cell density (Fisher exact testP ¼ 0.0055). IHC analysis of b-catenin expression showed avariable level of cytoplasmic, membranous, and nuclear stain-ing (Fig. 2L). Expression of b-catenin and CD3þ or CD8þ celldensity was not correlated (P ¼ 0.432, P ¼ 0.227, respectively,as shown in Supplementary Fig. S7).

Association between clinical risk factors and recurrenceEstablished risk factors for HCC invasiveness include tumor

size, tumor stage, tumor grade, serum AFP level, cellular differ-entiation, HCV or HBV infection, vascular invasion, and thepresence of liver cirrhosis (26–29). These elementswere evaluatedin our HCC patient cohort. However, bivariate analysis revealedthat only tumor cell differentiation, grade, and the presence ofvascular invasion were significantly correlated with HCC recur-rence (Table 2). Patients with elevated serum AFP and advancedtumor stage showed an increased rate of recurrence; however, theassociation was not statistically significant. Tumor stage did notcorrelate with recurrence, but correlated with vascular invasionand cellular differentiation (as shown in Supplementary Fig. S8).Patientswithwell-differentiated tumors experienced a significant-ly reduced incidence ofHCC recurrence: 5%ofwell-differentiatedtumors recurred after primary resection compared with 33% and48% for moderate and poorly differentiated tumors, respectively(P ¼ 0.011). Furthermore, patients with HCC whose tumors did

HCC TI (×20)A B HCC TI (×20)

HCC IM (×20) HCC IM (×20)

HCC TI (×20) HCC IM (×20)

DC

E F

Figure 1.Representative images ofCD3þTILs inHCC fromdifferent patient samples. A,B, and E are focused on the interior ofthe tumors, whereas C, D, and F arefocused on the invasive margin. Thepattern of TILs in each region fell intothree categories: dense nodularclusters of lymphocytes as in A;diffuse, evenly distributedlymphocytes as in B; or rare, orscattered, lymphocytes as in E. Sometumors had clusters of TILs from the IMto the TI, as in C and D, marked by redarrows, whereas others had noobvious TILs, as in F.

Gabrielson et al.

Cancer Immunol Res; 4(5) May 2016 Cancer Immunology ResearchOF4

on June 11, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst March 11, 2016; DOI: 10.1158/2326-6066.CIR-15-0110

Page 5: Intratumoral CD3 and CD8 T-cell Densities Associated with ... · Research Article Intratumoral CD3 and CD8 T-cell Densities Associated with Relapse-Free Survival in HCC Andrew Gabrielson1,Yunan

not reveal vascular invasion also faced a reduction in the rate ofrecurrence: those exhibiting tumor vascular invasion recurred in48% of cases compared with only 22% in those without vascularinvasion (P ¼ 0.040).

Impact of the immune response on recurrence and RFSThe densities of total (CD3þ) and cytotoxic T (CD8þ) TILs were

examined in the TI and IM regions. Statistically significant asso-ciations were observed between the individual densities of CD3þ

and CD8þ cells in the distinct tumor regions and frequency ofHCC recurrence (Table 2). Patients with a high density of CD3þ

immune infiltrates in the TI and IMexperienced recurrence of theirHCC in only 15%of cases comparedwith 44% in thosewith a lowCD3þ cell density (P ¼ 0.027). Similarly, 15% of patients with ahigh density of CD8þ immune infiltrates in the TI and IMexperienced recurrence of their HCC compared with 45% in thosewith a low CD8þ cell density (P ¼ 0.014). According to theImmunoscore grouping method outlined above, 27%, 14%,16%, 16%, and 27% of the patients were classified as Im0, Im1,Im2, Im3, and Im4, respectively. The frequency ofHCC recurrencein each Immunoscore subgroupwas as follows: 65% for Im0, 22%

for Im1, 10% for Im2, 10% for Im3, and 11% for Im4. Thiscombined analysis revealed that patients with a high density ofCD3þ and CD8þ cells in one or both tumor regions (TI or IM)experienced a significant reduction in the rate of HCC recurrence(P¼ 0.007). In keeping with the positive correlation between PD-L1 expression and CD3þ and CD8þ TIL density, the lack of PD-L1expression also predicts higher risk of HCC recurrence. Fifteen of39 HCC samples lacking PD-L1 expression recurred, while only 2of 19 HCC samples with PD-L1 expression recurred (P ¼ 0.034).

The densities of CD3þ and CD8þ immune cell populationswere also examined in relation to RFS times, our primary end-point. Different permutations of CD3þ and CD8þ cell popula-tions were used in the analysis: CD3þ density in each tumorregion alone, CD8þ in each tumor region alone, total CD3þ

density in the TI þ IM, total CD8þ density in the TI þ IM, andthe combined Immunoscore measuring both cell types in bothtumor regions (Fig. 3). Kaplan–Meier survival curves for averageCD3þ (Fig. 3A and B) and CD8þ (Fig. 3C and D) cell density ineach tumor region alone were first assessed. When each cellpopulation (CD3TI, CD3IM, CD8TI, CD8IM) was analyzed inde-pendently of one another, RFS time between patients with high

H&E x2

PD-L1 x2

CD3 x4TI IM

CD8 x4

IMTI

CD3 x20

CD8 x20

CD3-nega�ve control x20

β-catenin x20

PD-L1 x20

PD-1 x20

CD8-nega�ve control x20

PD-L1 x20

A E

D

C

B

F

HG

LK

JI

Figure 2.IHC staining of CD3 (A and E), CD8 (Band G), PD-L1 (C and I), negativecontrols (E and H), PD-1 (K), orb-catenin (L) in a lesion from an HCCsurgical sample (A–D) or in an HCCnodule, shown at �20 magnification(E–L).

Intratumoral CD3/CD8 Density Predicts Relapse in HCC

www.aacrjournals.org Cancer Immunol Res; 4(5) May 2016 OF5

on June 11, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst March 11, 2016; DOI: 10.1158/2326-6066.CIR-15-0110

Page 6: Intratumoral CD3 and CD8 T-cell Densities Associated with ... · Research Article Intratumoral CD3 and CD8 T-cell Densities Associated with Relapse-Free Survival in HCC Andrew Gabrielson1,Yunan

Figure 3.Kaplan–Meier curves comparing RFS in patientswith high and low intratumoral CD3 cell density in the TI (A) and IM (B), as well as high and low intratumoral CD8 celldensity in the TI (C) and IM (D). Patients were stratified in high and low groups using median immune cell density as the threshold. Kaplan–Meier curvescomparing RFS in patientswith high and low total intratumoral CD3 density (CD3TIþ CD3IM) and total CD8 density (CD8TIþCD8IM), usingmedian cutoff points asthe threshold (E and F, respectively). Kaplan–Meier curves comparing RFS in patients with different Immunoscores (G). Kaplan–Meier curves comparingRFS in patients with and without PD-L1 expression (H).

Gabrielson et al.

Cancer Immunol Res; 4(5) May 2016 Cancer Immunology ResearchOF6

on June 11, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst March 11, 2016; DOI: 10.1158/2326-6066.CIR-15-0110

Page 7: Intratumoral CD3 and CD8 T-cell Densities Associated with ... · Research Article Intratumoral CD3 and CD8 T-cell Densities Associated with Relapse-Free Survival in HCC Andrew Gabrielson1,Yunan

Figure 4.Kaplan–Meier curves comparing OS in patients with high and low intratumoral CD3 cell density in the TI (A) and IM (B), as well as high and low intratumoral CD8 celldensity in the TI (C) and IM (D). Patients were stratified in high and low groups using median immune cell density as the threshold. Kaplan–Meier curvescomparing OS in patients with high and low total intratumoral CD3 density (CD3TIþ CD3IM) and total CD8 density (CD8TIþ CD8IM), using median cutoff points asthe threshold (E and F, respectively). Kaplan–Meier curves comparing OS in patients with different Immunoscores (G). Kaplan–Meier curves comparingOS in patients with and without PD-L1 expression (H).

Intratumoral CD3/CD8 Density Predicts Relapse in HCC

www.aacrjournals.org Cancer Immunol Res; 4(5) May 2016 OF7

on June 11, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst March 11, 2016; DOI: 10.1158/2326-6066.CIR-15-0110

Page 8: Intratumoral CD3 and CD8 T-cell Densities Associated with ... · Research Article Intratumoral CD3 and CD8 T-cell Densities Associated with Relapse-Free Survival in HCC Andrew Gabrielson1,Yunan

and low immune cell density in either region were statisticallysignificantly different (CD3TIP¼0.044,CD3IMP<0.001,CD8TIP ¼ 0.002, CD8IM P ¼ 0.01). RFS was then correlated with thetotal CD3þ cell density, total CD8þ cell density and the corre-sponding Immunoscore value. The Kaplan–Meier curves (Fig. 3E)revealed a strong positive association between high total CD3þ

cell density TIþ IM and prolonged RFS (P¼ 0.006). The Kaplan–Meier curves (Fig. 3F) for total CD8þ cell density TIþ IM yielded asimilar but less significant association with RFS (P¼ 0.01). Lastly,Kaplan–Meier curves for the Immunoscore were constructed toprovide a more comprehensive picture of the two immune cellpopulations as they relate to survival. The curves (Fig. 3G) showthree distinct patient groups with statistically significant differ-ences in RFS times (P¼ 0.002). Patients with an Immunoscore of0 experienced the worst postoperative outcome, with all patientshaving disease recurrence within 2 years of follow-up. The Immu-noscore remained significantly associated with recurrence andRFS, whereas several clinicopathologic factors, including TNMstaging, did not reach significance. PD-L1 expression was also

examined in relation to RFS times. The RFS time between patientswith and without PD-L1 expression was significantly different(Fig. 3H; P ¼ 0.029).

Immunoscore predicts risk of recurrenceAnalysis of possible links between immune infiltration and

clinical risk factors is shown in Table 3. Viral hepatitis B and Cwere the predominant etiologies for HCC in our study popu-lation; however, the densities of CD3þ and CD8þ TILs amongpatients with or without HBV and HCV infection were notsignificantly different. Similarly, vascular invasion, cellulardifferentiation, and cancer stage showed no significant associ-ation with CD3þ and CD8þ cell densities in the TI and IM(Table 3). The expression of PD-L1 showed no significantassociation with vascular invasion, cellular differentiation, ortumor stage (Table 3). Because vascular invasion and differen-tiation were the only clinical risk factors that strongly correlatedwith HCC recurrence in our bivariate analysis, we focused onthese factors in the multivariate analysis.

Table 2. Association between HCC recurrence and patient demographics and clinicopathologic factors

RecurrencePredictor Variables Yes, n No, n P

Sex Female 3 12 0.522Male 16 34

Ethnicity African American 5 18Asian 7 12 0.58Caucasian 7 14

Etiology HBV and HCV 2 6HBV only 2 9HCV only 9 12 0.447Other 6 19

Tumor grade 1 1 52 2 223 15 14 0.0034 1 5

Serum AFP High (AFP > 500 ng/mL) 7 16 0.164Low (AFP � 500 ng/mL) 4 2

Tumor size Small (Size � 3 cm) 4 18Medium (3 cm < Size � 5 cm) 3 14 0.485Large (Size > 5 cm) 12 12

Tumor stage I 8 29II 6 7III 4 10 0.143IV 1 0

Cirrhosis Yes 6 11 1.00No 9 18

Vascular invasion Yes 10 11 0.040No 9 35

Differentiation Well 1 18Moderate 7 14Poor 11 12 0.011Undifferentiated 0 2

PD-1 High 7 22 0.573Low 10 21

PD-L1 Negative 15 24 0.034Positive 2 17

CD3 density in TI and IM High 5 27 0.027Low 14 18

CD8 density in TI and IM High 5 27 0.014Low 14 17

Immunoscore: CD3 þ CD8 density in TI and IM 0 (All-Low) 11 61 (1-High) 2 72 (2-High) 1 9 0.0073 (3-High) 1 94 (All-High) 2 15

Gabrielson et al.

Cancer Immunol Res; 4(5) May 2016 Cancer Immunology ResearchOF8

on June 11, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst March 11, 2016; DOI: 10.1158/2326-6066.CIR-15-0110

Page 9: Intratumoral CD3 and CD8 T-cell Densities Associated with ... · Research Article Intratumoral CD3 and CD8 T-cell Densities Associated with Relapse-Free Survival in HCC Andrew Gabrielson1,Yunan

In order to assess the impact of CD3þ and CD8þ cell densitieson the tumor recurrence, when adjusting for the significantcovariates in the bivariate analysis (specifically, vascular invasionand differentiation), multivariate logistic regression models werefit. The adjusted odds ratio (OR) of HCC recurrence was 5.8 [95%confidence interval (CI), 1.6–21.8] for CD3þ cell density. Simi-larly significant results were obtained for CD8þ density (theadjusted OR was 3.9; 95% CI, 1.1–14.1). The results showed thatafter adjusting for other covariates, CD3þ and CD8þ cell densitiesremained as significant risk factors for predicting tumor recur-rence. Thus, the Immunoscore could predict risk of recurrenceindependently of the only risk factors that demonstrated predic-tive value (vascular invasion and cellular differentiation) in ourdata set. As a result, the Immunoscore seems to be a highlysignificant prognostic factor in the cohort of patients with HCCtreated with primary surgery.

Given the great impact of CD3þ and CD8þ cell densities, andPD-L1 expression on tumor recurrence and RFS, we examined therelation between CD3þ and CD8þ cell density and OS. We foundthat TIL or PD-L1 expression density and OS in patients aftercurative resection were not significantly correlated (Fig. 4).

DiscussionHCC is one of the most common forms of cancer and remains

the second leading cause of cancer-related death worldwide (26).In areas with a high prevalence of HCC and a limited organsupply, surgical resection of HCC in patients with well-compen-sated liver function remains the primary curative treatmentoption. Unfortunately, the rate of HCC recurrence after surgicalintervention is high, and no effective treatment is available to

lower the risk of recurrence. Identification of biomarkers thatpredict recurrence is urgently needed. With a better understan-ding of the mechanism by which cancer recurs, new therapeutictargets that prevent recurrence may be revealed. As previouslyreported, the clinicopathologic factors, namely, serum AFP (27),vascular invasion (28, 29), and tumor stage (30), can predictrecurrence and poor outcome. However, these factors focus spe-cifically on the differences among HCC tumors and do notaccount for the interaction between the tumor and the hostimmune response. Improved prognostic and predictive markersfor HCC recurrence after primary resection that incorporate thisinteraction are needed.

Our study addresses this need. We showed that the intratu-moral immune cell densities of CD3þ and CD8þ cells with theirassociated Immunoscore were significantly associated with recur-rence and RFS independently of other predictive clinicopatho-logic factors, such as vascular invasion and cellular differentiation.The effect of CD3þ and CD8þ density in predicting recurrencewas statistically significant when adjusting for other covariatesin the multivariate logistic model, even with the small samplesize of the study.

Combined analysis revealed that patients with a high density ofCD3þ and CD8þ cells in one or both tumor regions (TI or IM)experienced a significant reduction in the rate of HCC recurrence(P ¼ 0.002). The Kaplan–Meier curves for CD3þ and CD8þ cellpopulations combined as an Immunoscore define three groupswith distinct RFS times (based on Immunoscores 0, 1, and 2, and3 and 4). Whereas 65% of patients with an Immunoscore of0 experienced recurrence, 22% of patients with an Immunscoreof 1 or 2, and only 10% of patients with high Immunoscores of3, 4, or 5 experienced recurrences. This suggests that both CD3þ

Table 3. Bivariate analysis demonstrated no association betweenCD3þ andCD8þ cell density and vascular invasion (A), cellular differentiation (B), etiology (C), andtumor stage (D). However, an association between CD3þ and CD8þ cell density and PD-L1 þ cells was apparent

A. Vascular invasionYes No P

CD3 density in TI and IM High 5 (24%) 16 (76%) 0.74Low 7 (30%) 16 (70%)

CD8 density in TI and IM High 4 (19%) 17 (81%) 0.31Low 8 (36%) 14 (64%)

PD-L1 expression Positive 6 (32%) 13 (68%) 0.18Negative 12 (31%) 27 (69%)

B. DifferentiationWell Moderate Poor P

CD3 density in TI and IM High 9 (43%) 8 (38%) 4 (19%) 0.48Low 7 (30%) 13 (57%) 3 (13%)

CD8 density in TI and IM High 10 (48%) 6 (28%) 5 (24%) 0.04Low 5 (23%) 15 (68%) 2 (9%)

PD-L1 expression Positive 5 (29%) 4 (24%) 8 (47%) 0.15Negative 12 (31%) 15 (38%) 12 (31%)

C. EtiologyHBV HCV HBV and HCV Other P

CD3 density in TI and IM High 7 (70%) 7 (50%) 2 (50%) 5 (32%) 0.29Low 3 (30%) 7 (50%) 2 (50%) 11 (68%)

CD8 density in TI and IM High 6 (60%) 7 (50%) 3 (75%) 5 (33%) 0.39Low 4 (40%) 7 (50%) 1 (25%) 10 (67%)

D. StageI II III IV P

CD3 density in TI and IM High 13 (50%) 4 (50%) 4 (44%) 0 (0%) 1.00Low 13 (50%) 4 (50%) 5 (56%) 1 (100%)

CD8 density in TI and IM High 16 (62%) 2 (25%) 3 (37%) 0 (0%) 0.17Low 10 (38%) 6 (75%) 5 (63%) 1 (100%)

PD-L1 expression Positive 11 (58%) 6 (32%) 2 (10%) 0 (0%) 0.12Negative 23 (59%) 6 (15%) 9 (23%) 1 (3%)

www.aacrjournals.org Cancer Immunol Res; 4(5) May 2016 OF9

Intratumoral CD3/CD8 Density Predicts Relapse in HCC

on June 11, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst March 11, 2016; DOI: 10.1158/2326-6066.CIR-15-0110

Page 10: Intratumoral CD3 and CD8 T-cell Densities Associated with ... · Research Article Intratumoral CD3 and CD8 T-cell Densities Associated with Relapse-Free Survival in HCC Andrew Gabrielson1,Yunan

and CD8þ cell populations contribute to the antitumor immuneresponse, and the high density of both CD3þ and CD8þ T cellsinfiltrating the tumor generates a better antitumor response inthe prevention of cancer recurrence compared with high den-sities of CD3þ or CD8þ T cells alone. Future studies with a largersample size and an independent cohort of patients are warrantedto confirm these findings.

Previous studies have demonstrated an association betweenthe presence of ectopic lymph node–like structures residingwithin the parenchyma of solid tumors and improved survivaltimes in patients with colorectal cancer (31, 32). In our currentstudy focusing on HCC, we observed high densities of TILs indense nodular clusters as well as diffuse, evenly distributedpopulations. Both arrangements of TILs seem to be protectivein HCC. In the small dataset, the strongest predictor of nocancer recurrence was a high density of CD3þ and/or CD8þ

cells within the parenchymal and invasive margin of the tumor.A possible difference in the amount of protection may existbetween nodular clusters and the diffuse type of TILs, but thiswas not evident in our dataset. It is also possible that the mixedpopulation of immune cells flanking the tumor was sufficientto generate an immune response against cancer, and that thepresence of ectopic lymph node–like structures in the form ofdense nodular clusters was not required for an antitumorimmune response in HCC.

It was surprising that only 2 of 60 HCC cases showed positivePD-L1 staining in cancer cells from biopsy samples. Nineteensamples showed PD-L1 expression in or around the tumor butonly in nonmalignant (noncancer) cells, presumably mononu-clear cells such as macrophages, dendritic cells, and lymphocytes.PD-L1 expression was positively correlated with CD3þ and CD8þ

TIL density. Tumor escape from immune-mediated destructionhas been associated with immunosuppressive mechanisms thatinhibit T-cell activation. It has been reported that a subset of Tcell–inflamed tumors showed high expression of three definedimmunosuppressive mechanisms: indoleamine-2,3-dioxygenase(IDO), PD-L1/B7-H1, and FoxP3þ regulatory T cells (Treg). Thissuggests that these inhibitory pathways might serve as negativefeedback mechanisms that follow, rather than precede, CD8þ T-cell infiltration (33). Mechanistic studies in mice revealed thatupregulated expression of IDO and PD-L1, as well as recruitmentof Tregs, in the tumor microenvironment depended on thepresence of CD8þ T cells. The former was driven by IFNg , andthe latter by a production of CCR4-binding chemokines alongwith a component of induced proliferation. Cancer immunother-apy approaches that target negative regulatory immune check-points, such as PD-L1, have been shown to preferentially benefitpatients with a preexisting T cell–inflamed tumor microenviron-ment (21). Furthermore, high PD-L1 expression has been corre-lated with improved response to immune checkpoint inhibitors,compared with low PD-L1 expression (34). High PD-L1 expres-sion in nonmalignant cells in or around the tumor, with noexpression in malignant HCC cells, may be correlated withimproved response to immune checkpoint inhibitors. Nivolu-mab, a fully human IgG4monoclonal antibody PD-1 inhibitor, iscurrently being evaluated in patients with advanced HCC(NCT01658878), and was reported to have promising clinicalactivity (35). Biomarker analysis for the HCC samples in thisstudy may be available in a few years.

High densities of immune cell infiltrates appeared to decreasethe risk of cancer recurrence, yet the reason why some patients

have high densities of immune cell infiltrates and others do not isnot well understood. It has been speculated that both host andtumor contribute to the infiltration of immune cells. Accordingly,functional polymorphisms in Fas and Fas ligand (FasL) of thehost T lymphocytes were reported to contribute to increasedapoptosis of TILs and enhanced risk of breast cancer progression(36). Polymorphisms that lead to constitutive expression of deathreceptor ligands, such as FasL, may cause tumor cells to adopt anactivation-induced cell deathmechanism (induction of apoptosisof previously activated T cells on subsequent encounters with anantigen) to delete the attacking antitumor T cells through theinduction of apoptosis via death receptor and death receptorligand interactions (37).

Analysis of humanmelanoma cell lines has revealed that manytumors without immune infiltration show alterations in theWnt/b-catenin signaling pathway (25). Tumor cells with active b-cate-nin showed reduced expression of the chemokines CCL4 andCXCL1 and were thus associated with a diminished antitumor T-cell response (25). The effect of tumor b-catenin on the antitumorT-cell response may be tumor type–dependent because the HCCdata in this study demonstrate no association between b-cateninexpression and TIL density. However, we did not measure b-cate-nin activity, which might differ from its expression. TILs inglioblastoma were also associated with specific genomic altera-tions, and TILs were enriched in glioblastomas of the mesenchy-mal class, strongly associated with mutations in NF1 and RB1.Conversely, TILs were depleted in epidermal growth factor recep-tor–amplified and homozygous PTEN-deleted tumors (38). Theheterogeneity of immune cell infiltration among patients may bea consequence of many such genomic alterations and may differdepending on the tumor type.

In addition, the high mutational frequency found withintumors raises the possibility that T cells may preferentially invadetumors in patients whose T cells recognize mutated epitopesfound within the tumor tissue. The rate of somatic mutationsseems to correlate with tumor response to immune checkpointblockers (39). Characterizing commonly mutated epitopes mayprovide targets for novel antitumor vaccines (40).

Identification of the defects that lead to low intratumoralimmune cell density may also provide novel therapeutic targetsfor anticancer treatment. Type I IFNs are implicated in theinnate immune sensing of immunogenic tumors, leading toadaptive T-cell responses (41). A recent breakthrough in cancerimmunology suggests that the stimulator of interferon genes(STING) complex is required for sensing of cancer cell–derivedDNA and activation of dendritic cells, leading to CD8þ T-cellpriming against tumors (42). The defective STING signalingpathway may result in low intratumoral immune cell densityand a poor antitumor immune response. Multiple STINGactivators are being developed in promoting innate immunesensing and adaptive T-cell response against cancer (43). Thecombination of STING activators with inhibitors to immunecheckpoints and/or antitumor vaccines may provide promisingstrategies for cancer treatment. The efficacy of immune check-point inhibition against HCC has been suggested in a phase Iclinical trial that evaluated anti–PD-1 therapy (nivolumab),and a phase II clinical trial that evaluated an anti–CTLA-4therapy (tremelimumab) in patients with HCC (35, 44). It ispossible that immune infiltrates are not only prognostic, butpredictive as well; identifying patients whose tumors might besensitive to immunotherapy. Immune checkpoint inhibitors

Cancer Immunol Res; 4(5) May 2016 Cancer Immunology ResearchOF10

Gabrielson et al.

on June 11, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst March 11, 2016; DOI: 10.1158/2326-6066.CIR-15-0110

Page 11: Intratumoral CD3 and CD8 T-cell Densities Associated with ... · Research Article Intratumoral CD3 and CD8 T-cell Densities Associated with Relapse-Free Survival in HCC Andrew Gabrielson1,Yunan

would be of particular interest in these patients and could beevaluated in prospective clinical trials.

Disclosure of Potential Conflicts of InterestF. Banovac reports receiving speakers bureau honoraria from Onyx Inc. No

potential conflicts of interest were disclosed by the other authors.

Authors' ContributionsConception and design: A. Gabrielson, R. Jha, P. Feng, J. Marshall, L. Weiner,M. Atkins, A.R. HeDevelopment of methodology: A. Gabrielson, Z. Gatalica, R. Jha, J. Kachhela,P. Feng, T. Zhang, A.R. HeAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): A. Gabrielson, Y. Wu, B. Kallakury, Z. Gatalica,D. Kleiner, E. Island, F. Banovac, P. Feng, T. Zhang, A. Tesfaye, P. Prins,J. Marshall, A.R. HeAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): A. Gabrielson, Y. Wu, H. Wang, J. Jiang, B. Kallakury,D. Kleiner, T. Fishbein, J. Kachhela, P. Feng, C. Loffredo, J. Marshall, L. Weiner,A.R. HeWriting, review, and/or revision of the manuscript: A. Gabrielson, H. Wang,B. Kallakury, S. Reddy, T. Fishbein, L. Johnson, E. Island, R. Satoskar, R. Jha,A. Tesfaye, C. Loffredo, J. Marshall, L. Weiner, M. Atkins, A.R. He

Administrative, technical, or material support (i.e., reporting or organizingdata, constructing databases):A.Gabrielson,D. Kleiner, J. Kachhela, A. Tesfaye,P. Prins, A.R. HeStudy supervision: A.R. He

AcknowledgmentsThe authors thank Marion Hartley, PhD, for editing assistance.

Grant SupportThis work was supported by American Cancer Society grant 118525-MRSG-

10-068-01-TBE (A.R. He). The experiments were carried out with the help ofGeorgetown University Medical Center's (GUMC) Shared Resources (includinghistopathology,microscopy and imaging, genomics and epigenomics, and flowcytometry), which was supported by NIH-P30 CA51008 and NCATS 8 UL1TR000101.

The costs of publication of this article were defrayed in part by thepayment of page charges. This article must therefore be hereby markedadvertisement in accordance with 18 U.S.C. Section 1734 solely to indicatethis fact.

Received April 20, 2015; revised December 21, 2015; accepted January 6,2016; published OnlineFirst March 11, 2016.

References1. Said A, Wells J. Management of heptocellular carcinoma. Minerva Med

2009;100:51–68.2. Locker GY, Hamilton S, Harris J, Jessup JM, Hayes D, Bast RCJr, et al. ASCO

2006 update of recommendations for the use of tumor markers in gastro-intestinal cancer. J Clin Oncol 2006;24:5313–27.

3. Sobin L, Wittekind C. TNM classification of malignant tumors . New York:Wiley-Liss; 2002.

4. Weitz J, Koch M, Debus J, Hohler T, Galle PR, Buchler MW. Colorectalcancer. Lancet 2005;365:153–65.

5. ChanAC, Fan ST,Cheung TT,ChokKS,ChanSC, LoCM, et al. Evaluationofthe seventh edition of the American Joint Committee on Cancer tumour-node-metastasis (TNM) staging system for patients undergoing curativeresection of hepatocellular carcinoma: implications for the development ofa refined staging system. HPB 2013;15:439–48.

6. Lee SC. Prognostic biomarkers for prediction of recurrence of hepatocel-lular carcinoma: current status and future prospects. World J Gastroenterol2014;20:3112–24.

7. Nagtegaal ID, Quirke P, Schmoll HJ. Has the new TNM classification forcolorectal cancer improved care?Nat Rev Clin Oncol 2011;9:119–23.

8. Mlecnik B, Bindea G, Pages F, Galon J. Tumor immunosurveillance inhuman cancers. Cancer Metastasis Rev 2011;30:5–12.

9. Dunn GP, Old LJ, Schreiber RD. The three E's of cancer immunoediting.Annu Rev Immunol 2004;22:329–60.

10. Shankaran V, Ikeda H, White JM, Swanson PE, Old LJ, Bruce AT, et al. IFNgamma and lymphocytes prevent primary tumour development and shapetumour immunogenicity. Nature 2001;410:1107–11.

11. Ascierto ML, De Giorgi V, Liu Q, Bedognetti D, Spivey TL, Chouchane L,et al. An immunologic portrait of cancer. J Transl Med 2011;9:146.

12. Atreya I, Neurath MF. Immune cells in colorectal cancer: Prognosticrelevance and therapeutic strategies. Expert Rev Anticancer Ther 2008;8:561–72.

13. Baier PK, Wimmenauer S, Hirsch T, von Kleist S, Keller H, Farthmann EH,et al. Analysis of the T cell receptor variability of tumor infiltratinglymphocytes in colorectal carcinomas. Tumour Biol 1998;19:205–12.

14. Diederichsen AC, Hjelmborg JB, Christensen PB, Zeuthen J, Fenger C.Prognostic value of the CD4þ/CD8þ ratio of tumour infiltrating lympho-cytes in colorectal cancer and HLA-DR expression on tumor cells. CancerImmunol Immunother 2003;52:423–8.

15. Finn OJ. Cancer immunology. N Engl J Med 2008;358:2704–15.16. Galon J, Costes A, Sanchez-Cabo F, Tranjanoski Z, Fridman WH, Pages F,

et al. Type, density, and location of immune cells within human colorectaltumors predict clinical outcome. Science 2006;313:1960–4.

17. Galon J, Fridman WH, Pages F. The adaptive immunologic microenviron-ment in colorectal cancer: a novel perspective. Cancer Res 2007;67:1883–6.

18. Naito Y, Saito K, Shiiba K, Saigenji K, Nagura H, Ohtani H, et al. CD8þ Tcells infiltrated within cancer cell nests as a prognostic factor in humancolorectal cancer. Cancer Res 1998;58:3491–4.

19. Pag�es F, Berger A, Camus M, Trajanoski Z, Fridman WH, Galon J, et al.Effector memory T cells, early metastasis, and survival in colorectal cancer.N Engl J Med 2005;353:2654–66.

20. Chemnitz JM, Parry RV, Nichols KE, June CH, Riley JL. SHP-1 and SHP-2associate with immunoreceptor tyrosine-based switch motif of pro-grammed death 1 upon primary human T cell stimulation, but onlyreceptor ligation prevents T cell activation. J Immunol 2004;173:945–54.

21. KimYJ. Subverting the adaptive immune resistancemechanism to improveclinical responses to immune checkpoint blockade therapy. OncoImmu-nology 2015;3:e954868.

22. Angell H, Galon J. From the immune contexture to the immunoscore: therole of prognostic and predictive immune markers in cancer. Curr OpinImmunol 2013;25:261–7.

23. Anitei M, Zeitoun G, Mlecnik B, Berger A, Galon J, Pages F. Prognostic andpredictive values of the immunoscore in patients with rectal cancer. ClinCancer Res 2014;20.7:1891–9.

24. Pages F, Kirilovsky A, Mlecnik B, Asslaber M, Bruneval P, Galon J, et al. Insitu cytotoxic and memory T cells predict outcome in patients with earlystage colorectal cancer. J Clin Oncol 2009;27:5944–51.

25. Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic b-catenin signal-ing prevents T cell infiltration and anti-tumor immunity. Nature 2015;231–35.

26. Jemal A, Freddie B, Melissa M, Jacques F, Elizabeth W, David F. Globalcancer statistics. CA Cancer J Clin 2011;61:69–90.

27. Parpart S, Stephanie R, Fei D, Zhao-You T, Wang X. Modulation of MiR-29expression by alpha-fetoprotein is linked to the hepatocellular carcinomaepigenome. Hepatology 2014;60:872–83.

28. Marshall A, Alexander G. Vascular invasion leaves its mark in hepatocel-lular carcinoma. J Hepatol 2011;55:1174–5.

29. Thuluvath P. Vascular invasion is the most important predictor of survivalin hcc, but how do we find it?J Clin Gastroenterol 2009;43:101–2.

30. Shah S, Cleary S, Greig P, Gallinger S. Recurrence after liver resection forhepatocellular carcinoma: risk factors, treatment, and outcomes. Surgery2007;141:330–9.

31. Coppola D, Mul�e J. Ectopic lymph nodes within human solid tumors.J Clin Oncol 2008;26:4369–70.

www.aacrjournals.org Cancer Immunol Res; 4(5) May 2016 OF11

Intratumoral CD3/CD8 Density Predicts Relapse in HCC

on June 11, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst March 11, 2016; DOI: 10.1158/2326-6066.CIR-15-0110

Page 12: Intratumoral CD3 and CD8 T-cell Densities Associated with ... · Research Article Intratumoral CD3 and CD8 T-cell Densities Associated with Relapse-Free Survival in HCC Andrew Gabrielson1,Yunan

32. Dubinett S, Lee J, Sharma S, Mul�e J. Chemokines: can effector cells be re-directed to the site of tumor?Cancer J 2010;167:325–35.

33. Spranger S, Spaapen RM,Meng Y, Ha T, Gajewski TF. Up-regulation of PD-L1, IDO, and T(regs) in the melanoma tumor microenvironment is drivenby CD8(þ) T cells. Sci Transl Med 2013;8:200ra116.

34. Taube JM. Unleashing the immune system: PD-1 and PD-Ls in the pre-treatment tumor microenvironment and correlation with response to PD-1/PD-L1 blockade. Oncoimmunology 2014;11:e963413. eCollection2014.

35. El-Khoueiry A, Ignacio M, Crocenzi T, Sangro B. Phase I/II safety andantitumor activity of nivolumab in patients with advanced hepatocellularcarcinoma (HCC): CA209-040. J Clin Oncol 2015;33suppl; abstr LBA101.

36. Zhang B, Sun T, Xue L, Zhang X, Guo Y, Lin D. Functional polymorphismsin FAS and FASL contribute to increased apoptosis of tumor infiltrationlymphocytes and risk of breast cancer. Carcinogenesis 2007;28:1067–73.

37. Igney FH, Krammer PH. Immune escape of tumors: apoptosis resistanceand tumor counterattack. J Leukoc Biol 2002;71:907–20.

38. Hanane L, Natarajan A, Sieghart W, Sibilia M. EGFR has a tumour-promoting role in liver macrophages during hepatocellular carcinomaformation. Nat Cell Biol 2014;16.972.

39. Rutledge WC, Saltz JH, Moreno CS, Brat DJ. Tumor-infiltrating lympho-cytes in glioblastoma are associated with specific genomic alterations andrelated to transcriptional class. Clin Cancer Res 2013;19:4951–60.

40. Champiat S, Ferte C, Lebel-Binay S, Eggermont A, Soria JC. Exomics andimmunogenics: bridging mutational load and immune checkpoints effi-cacy. Oncoimmunology 2014;3:e27817.

41. Fuertes MB, Kacha AK, Kline J, Woo SR, Murphy KM, Gajewski TF, et al.Host type I IFN signals are required for antitumor CD8þ T cell responsesthrough CD8aþ dendritic cells. J Exp Med 2011;208:2005–16.

42. Woo SR, Fuertes MB, Alegre ML, Gajewski TF. STING-dependent cytosolicDNA sensing mediates innate immune recognition of immunogenictumors. Immunity 2014;41:830–42.

43. Leong EJ, Pardoll D, Dubensky T. Modified STING-activating cyclicdinucleotide derivatives significantly enhance the anti-tumor activityof therapeutic vaccines. J ImmunoTherapy Cancer 2013;1.Suppl 1:O20.

44. Sangro B, Carlos GM, De La Mata M, Inarrairaegui M, Perez-Gracia JL,Melero I, et al. A clinical trial of CTLA-4 blockade with tremelimumab inpatients with hepatocellular carcinoma and chronic hepatitis C. J Hepatol2013;59:81–8.

Cancer Immunol Res; 4(5) May 2016 Cancer Immunology ResearchOF12

Gabrielson et al.

on June 11, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst March 11, 2016; DOI: 10.1158/2326-6066.CIR-15-0110

Page 13: Intratumoral CD3 and CD8 T-cell Densities Associated with ... · Research Article Intratumoral CD3 and CD8 T-cell Densities Associated with Relapse-Free Survival in HCC Andrew Gabrielson1,Yunan

Published OnlineFirst March 11, 2016.Cancer Immunol Res   Andrew Gabrielson, Yunan Wu, Hongkun Wang, et al.   Relapse-Free Survival in HCCIntratumoral CD3 and CD8 T-cell Densities Associated with

  Updated version

  10.1158/2326-6066.CIR-15-0110doi:

Access the most recent version of this article at:

  Material

Supplementary

  .DC1

http://cancerimmunolres.aacrjournals.org/content/suppl/2016/03/11/2326-6066.CIR-15-0110Access the most recent supplemental material at:

   

   

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  Rightslink site. (CCC)Click on "Request Permissions" which will take you to the Copyright Clearance Center's

.http://cancerimmunolres.aacrjournals.org/content/early/2016/04/09/2326-6066.CIR-15-0110To request permission to re-use all or part of this article, use this link

on June 11, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst March 11, 2016; DOI: 10.1158/2326-6066.CIR-15-0110


Recommended