Open Access

A subset of patients with MSS/MSI‑low‑colorectal cancer showed increased CD8(+) TILs together with up‑regulated IFN‑γ

  • Authors:
    • Tomohiro Kikuchi
    • Kosaku Mimura
    • Hirokazu Okayama
    • Yuko Nakayama
    • Katsuharu Saito
    • Leo Yamada
    • Eisei Endo
    • Wataru Sakamoto
    • Shotaro Fujita
    • Hisahito Endo
    • Motonobu Saito
    • Tomoyuki Momma
    • Zenichiro Saze
    • Shinji Ohki
    • Koji Kono
  • View Affiliations

  • Published online on: October 2, 2019     https://doi.org/10.3892/ol.2019.10953
  • Pages: 5977-5985
  • Copyright: © Kikuchi et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY_NC 4.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

A small subset of patients with proficient mismatch repair (pMMR)/microsatellite stable (MSS)‑colorectal cancer (CRC) benefit from immunotherapy with anti‑programmed cell death 1 (PD‑1)/programmed death ligand 1 (PD‑L1) blockade. Therefore, the aim of the current study was to evaluate the immune status of patients with pMMR/microsatellite instability‑low (MSI‑L)/MSS‑CRC and deficient MMR (dMMR)/MSI‑high (MSI‑H)‑CRC in order to identify responders to anti‑PD‑1/PD‑L1 inhibitors. The current study used a dataset downloaded from The Cancer Genome Atlas (TCGA) as well as 219 clinical tissue samples to investigate the infiltrating grade of cluster of differentiation (CD) 4 and CD8 tumor infiltrating lymphocytes (TILs), the expression levels of PD‑L1 and PD‑L2, the interferon‑γ (IFN‑γ) and CD8 T effector gene signatures, and the phosphorylated signal transducer and activator of transcription 1 (p‑STAT1) status in patients with pMMR/MSI‑L/MSS‑CRC and dMMR/MSI‑H‑CRC. Analysis of TCGA dataset revealed that the mRNA expression levels of PD‑L1 and PD‑L2, the IFN‑γ gene signature and the CD8 T effector gene signature were significantly upregulated in MSI‑H tumors compared with MSI‑L/MSS tumors. Additionally, a subpopulation of patients with upregulation of the IFN‑γ and CD8 T effector gene signatures was observed in those with MSI‑L/MSS‑CRC. Immunohistochemical staining of the clinical samples revealed a subpopulation of patients with pMMR‑CRC that were positive for PD‑L1 and p‑STAT1, and whom had levels of elevated CD8(+) and CD4(+) TILs infiltration similar to those observed in patients with dMMR‑CRC. The results obtained in the current study suggested that a subpopulation of patients with MSI‑L/MSS‑CRC and pMMR‑CRC with upregulated IFN‑γ and CD8 T effector gene signatures may benefit from immunotherapy with antibodies against PD‑1 and PD‑L1.

Introduction

Colorectal cancer (CRC) was the third most common type of cancer and the fourth leading cause of cancer-associated mortality worldwide in 2012 (1,2). CRC is classified into two categories: i) The microsatellite instability-high (MSI-H) group, which is caused by defects in the DNA mismatch repair (MMR) system and accounts for ~15% of tumors; and ii) the microsatellite stable (MSS) group, which exhibits chromosomal instability and accounts for the remaining 85% of tumors (35). At the protein level, MSI-H is similar to deficient MMR (dMMR) status, while MSI-low (MSI-L)/MSS are similar to proficient MMR (pMMR) status (6,7).

Immunotherapy with anti-programmed cell death 1 (PD-1)/programmed death ligand 1 (PD-L1) monoclonal antibodies (mAbs) is a standard therapeutic strategy in various types of cancer, including lung cancer, gastric cancer and renal cell carcinoma (810). Although the results of initial trials of immune checkpoint inhibitors (ICIs) in CRC were not particularly promising (11,12), dMMR/MSI-H patients with CRC are currently considered to be candidates for anti-PD-1/PD-L1 immunotherapy (13,14). In a phase II clinical trial, treatment with the anti-PD-1 mAb pembrolizumab resulted in a significant clinical response in patients with dMMR-CRC, but not those with pMMR-CRC (13). Similar results were observed in dMMR/MSI-H tumors across 12 different cancer types (15). Furthermore, a recent phase II trial demonstrated the clinical benefit of the PD-1 mAb nivolumab in patients with dMMR/MSI-H-CRC (14). Therefore, immunotherapy with ICIs targeting the PD-1/PD-L1 axis may be a promising treatment option for patients with dMMR/MSI-H solid tumors of varying histological origins.

In general, dMMR/MSI-H tumors in CRC are thought to respond to ICIs due to their highly immunogenic nature (14). For example, dMMR/MSI-H results in the accumulation of several mutations in microsatellites spread along the genome, and produces neo-antigens that produce a highly antigenic microenvironment with a high density of tumor infiltrating lymphocytes (TILs) (3,5,16,17). Moreover, simultaneous expression of multiple immune checkpoint molecules, including PD-1 and PD-L1, has been reported in dMMR/MSI-H-CRC (3,5,16,17). Consequently, in patients with dMMR/MSI-H-CRC, treatment with ICIs targeting the PD-1/PD-L1 axis reactivates anti-tumor specific T cells that subsequently attack tumor cells.

Previous studies revealed that ICIs targeting the PD-1/PD-L1 axis were ineffective at treating MSS/pMMR-CRC (11,12). Clinical studies have established MSI status as a putative response biomarker for PD-1 blockade, with progression free survival rates of up to 78% reported in patients with MSI-H-CRC, compared with 11% of patients with MSS-CRC (13,17). However, other studies have demonstrated that ~10% of patients with MSS/pMMR-CRC exhibit a response to PD-1/PD-L1 inhibitors (13,17). Based on the aforementioned clinical evidence, the aim of the current study was to compare the immune statuses of patients with MSI-L/MSS or pMMR-CRC with MSI-H or dMMR-CRC, in order to identify responders with MSI-L/MSS or pMMR-CRC as distinct biomarker-defined populations. The current study therefore investigated the infiltrating grade of CD4 and CD8 TILs, the expression levels of PD-L1 and PD-L2, the interferon-γ (IFN-γ) and CD8 T effector gene signatures, and the phosphorylated signal transducer and activator of transcription 1 (p-STAT1) status in patients with MSI-L/MSS or pMMR- CRC compared with those with MSI-H or dMMR-CRC.

Materials and methods

The Cancer Genome Atlas (TCGA) dataset analysis

Level 3 Illumina RNA-Seq data (RNA-Seq V2 RSEM normalized) for colon adenocarcinoma (COAD) and rectal adenocarcinoma (READ) in TCGA were downloaded through the cBioPortal (www.cbioportal.org) in July 2018 (18) and consisted of 342 samples. MSI testing data (MSI-H, MSI-L or MSS) for each TCGA tumor were obtained from a previous study by Liu et al (19). The current study analyzed multi-gene expression signatures, including the CD8 T effector gene signature [CD8A molecule (CD8A), CD8B molecule (CD8B), eomesodermin (EOMES), granzyme A (GZMA), granzyme B (GZMB), interferon γ (IFNG) and perforin 1 (PRF1)] and the IFN-γ gene signature [indoleamine 2,3-dioxygenase 1 (IDO1), C-X-C motif chemokine ligand (CXCL) 9 and 10, major histocompatibility complex class II DR α (HLA-DRA), STAT1 and IFNG] (20,21). The IFN-γ gene signature was identified from three clinical studies investigating pembrolizumab, and focused on genes associated with antigen presentation and cytotoxic activity in 220 patients with nine types of cancer (21). The CD8 T effector gene signature was previously established to identify activated T cells such as cytotoxic T lymphocytes (20). The signature scores were calculated by averaging the expression levels of the genes included in each signature (22), excluding HLA-DRA expression values, as these were not available in TCGA RNA-seq data.

Patient samples

Formalin-fixed paraffin-embedded tissue samples from 219 patients with primary CRC, who had undergone surgical resection without preoperative chemotherapy or radiotherapy in Fukushima Medical University Hospital (Fukushima, Japan) between January 2007 and December 2013 were analyzed in the current study. A total of 138 men and 81 women (mean age, 67.8±12.4 years; age range, 27–94 years), were included (Table I). Patients with stage 0 CRC were excluded. Clinical and pathological data were retrospectively obtained from medical records, with the last follow-up in April 2017 (Table I).

Table I.

Clinical features of the patients (n=219).

Table I.

Clinical features of the patients (n=219).

Clinical featuresNumber of patients
Sex
  Male138
  Female81
TNM stagea
  I49
  II72
  III74
  IV24
Degree of differentiation
  Moderate/high differentiation204
  Poor differentiation/undifferentiated15
Lymphatic metastasisa
  N0126
  N1-393
Degree of invasiona
  T125
  T239
  T395
  T460

{ label (or @symbol) needed for fn[@id='tfn1-ol-0-0-10953'] } TNM, tumor-node-metastasis.

a The Japanese Classification of Colon and Rectum Carcinoma was defined according to the Japanese Society of the Colon and Rectum (The 9th Edition).

Immunohistochemistry (IHC)

Tissue blocks were cut into 4-µm-thick sections and were subsequently deparaffinized by three washes of 3 min with xylene at room temperature and rehydrated by decreasing gradient of ethanol at room temperature. Sections were incubated with 0.3% hydrogen peroxide in methanol for 30 min at room temperature to block endogenous peroxidase activity. For CD4, CD8 and forkhead box P3 (FOXP3) staining, antigen retrieval was performed by autoclaving the sections for 5 min at 105°C in target retrieval solution (pH 9.0; Dako; Agilent Technologies, Inc.). For PD-L1 staining, antigen retrieval was performed by autoclaving the sections for 10 min at 120°C in target retrieval solution (pH 9.0). For p-STAT1 staining, antigen retrieval was performed by autoclaving the sections for 5 min at 105°C in citrate buffer solution (pH 6.0). Tissue sections were subsequently incubated with the following primary antibodies directed against CD4 (clone 4B12; cat. no. M7310; 1:100; Dako; Agilent Technologies, Inc.), CD8 (clone C8/144B; cat. no. M7103; 1:100; Dako; Agilent Technologies, Inc.), FOXP3 (clone 236A/E7; cat. no. ab20034; 1:200; Abcam), PD-L1 (clone E1L3N; cat. no. 13684S; 1:400; Cell Signaling Technology, Inc.) and p-STAT1 (clone D3B7; cat. no. 8826S; 1:800; Cell Signaling Technology, Inc.) at 4°C overnight. Following the primary antibody incubation, the sections were incubated with horseradish peroxidase (HRP)-conjugated anti-rabbit or anti-mouse secondary antibodies (Envision + System-HRP; cat. no. K4003 or K4001; ready to use; Dako; Agilent Technologies, Inc.) for 30 min at room temperature. The sections were then stained with diaminobenzidine (Dojindo Molecular Technologies, Inc,) at room temperature for 10 min and subsequently counterstained with Mayer's hematoxylin solution (cat. no. 131-09665; Wako Pure Chemical Industries, Ltd.) at room temperature for 1 min. Sections in which the primary antibodies were replaced with PBS were used as negative controls. Esophagus and stomach cancer tissue sections served as positive controls (23).

Assessment of IHC staining

IHC analysis was performed by four independent observers (TK, KS, LY and EE) who were blinded to the clinical data. The TILs at the invasive front region of the tumor were reviewed in four independent areas using light microscope at a magnification of ×400. PD-L1 expression was evaluated by assessing membranous staining without cytoplasmic staining and tumor specimens were considered to be PD-L1-positive when >1% of the tumor cells exhibited membranous staining of any intensity (24). p-STAT1 staining was evaluated by assessing nuclear staining of the tumor cells. Positive staining for p-STAT-1 was defined by the presence of positive nuclear staining in tumor cells. CD4, CD8 and FOXP3 expression was evaluated by counting the number of stained lymphocytes from four independent areas.

Determination of MMR status

IHC for MMR proteins, including mutL homolog 1, mutS homolog 2 and 6, and PMS1 homolog 2 mismatch repair system component, was performed as previously described (25). Loss of a MMR protein was defined as the absence of nuclear staining of tumor cells in the presence of positive nuclear staining of normal colon mucosa cells or stromal cells. Tumors demonstrating the loss of at least one MMR protein were designated as dMMR, and tumors with intact MMR protein expression as pMMR.

Statistical analysis

An unpaired Student's t-test was used to compare two groups and one-way analysis of variance followed by a Tukey's post hoc test was used to compare multiple groups. The data are presented as the means ± standard deviation. The correlation of IFN-γ or CD8 T effector gene signatures with the expression of PD-L1 and PD-L2, STAT1 and Janus-activated kinase (JAK) 1 or 2, and the correlation of the IFN-γ gene signature with the CD8 T effector gene signature were assessed using scatter diagrams and Pearson's correlation test. Associations between p-STAT1 and PD-L1 expression were assessed using the χ2 test. All statistical analyses were conducted using GraphPad Prism software (version 7.0; GraphPad Software, Inc.). P-values were two-sided and P<0.05 was considered to indicate a statistically significant difference.

Results

A subset of patients with MSI-L/MSS-CRC exhibit IFN-γ and CD8 T effector gene signature upregulation

TCGA COADREAD dataset consisted of 342 samples, including 51 MSI-H (14.9%) and 291 MSI-L/MSS (85.1%) tumor samples. The mRNA expression levels of PD-L1 and PD-L2 were significantly upregulated in MSI-H tumors compared with MSI-L/MSS tumors (Fig. 1A). The IFN-γ gene signature consisting of IDO1, CXCL10, CXCL9, STAT1 and IFNG, and the CD8 T effector gene signature consisting of CD8A, CD8B, EOMES, GZMA, GZMB, IFNG and PRF1 were subsequently assessed. The two gene signatures were more prevalent in MSI-H tumors compared with MSI-L/MSS tumors (Fig. 1A). There was a significant variation in the IFN-γ gene signature in the MSS/MSI-L group. A subpopulation with a high IFN-γ gene signature in the MSS/MSI-L group, with expression levels of the IFN-γ gene signature similar to those in the MSI-H group was identified (Fig. 1B). Similarly, there was a subset of patients with a high CD8 T effector gene signature in the MSS/MSI-L group, with expression levels similar to those in the MSI-H group (Fig. 1B). The results obtained in the current study suggest that there is a subpopulation of patients with upregulated CD8 T effector and IFN-γ gene signatures in MSI-L/MSS-CRC.

Correlations of PD-L1 and PD-L2 with the IFN-γ and CD8 T effector gene signatures

There were significant positive correlations between the IFN-γ gene signature and PD-L1, between the IFN-γ gene signature and PD-L2, between the CD8 T effector gene signature and PD-L1, between the CD8 T effector gene signature and PD-L2, and finally between the CD8 T effector and IFN-γ gene signatures in all 342 CRC cases (Fig. 2A). The current study demonstrated that a subset of patients with CD8 T effector and IFN-γ gene signature upregulation in the MSI-L/MSS-CRC group exhibited upregulation of immune checkpoint molecules, including PD-L1 and PD-L2.

In the present study, there were significant positive correlations between the IFN-γ or CD8 T effector gene signatures and STAT1 or JAK1/2 (Fig. 2B). Furthermore, STAT1 and JAK2 mRNA expression levels were significantly upregulated in MSI-H tumors compared with MSI-L/MSS tumors (Fig. 2C). These findings suggested that the activation of IFN-γ signaling is associated with IFN-γ production and CD8 T cell infiltration within the tumor microenvironment, and that IFN-γ signaling is significantly activated in MSI-H tumors compared with MSI-L/MSS tumors. Since the STAT1 mRNA expression level was correlated to the highest degree with the IFN-γ and CD8 T effector gene signatures (Fig. 2B), and the binding of IFN-γ to its cognate receptor leads to the phosphorylation-dependent activation of STAT1 (26), the p-STAT1 expression level in clinical samples was analyzed to investigate the effect of IFN-γ in the tumor microenvironment.

A subset of patients in the pMMR-CRC group exhibit increased CD8 and CD4 infiltration

In the current study, a total of 219 patients received colorectal surgery (Table I). IHC was used to classify patients based on MMR protein expression, and a total of 18 (8.2%) and 201 (91.8%) patients were identified to have dMMR- and pMMR-CRC, respectively. Representative IHC staining for MMR proteins are presented in Fig. 3A. Compared with TCGA COADREAD database, the prevalence of dMMR-CRC in the current study was low (14.9 vs. 8.2%). The immune status, including PD-L1 and p-STAT1 in the tumors, and CD4(+), CD8(+) and FOXP3(+) TILs in the tumor microenvironments, was evaluated by IHC as presented in Fig. 3B. There were significant associations between PD-L1 expression and the grade of CD4(+) or CD8(+) TIL infiltration; however, there was no significant association between PD-L1 expression and FOXP3(+) TIL infiltration (Fig. 4A-C). Furthermore, there was a tendency towards a positive association between PD-L1 and p-STAT1 expression levels (Fig. 4D). The immune parameters of patients with dMMR- and pMMR-CRC were compared, a subpopulation of PD-L1(+) and p-STAT1(+) patients with pMMR-CRC (n=26) showed increasing grades of infiltrating CD4(+) or CD8(+) TILs compared with pMMR/others including pMMR-CRC without a subpopulation of PD-L1(+) and p-STAT1(+) patients, which were similar to the levels seen in patients with dMMR-CRC (Fig. 4E and F). Although the number of FOXP3(+) TILs was increased in patients with pMMR-CRC with PD-L1(+) and p-STAT1(+) compared with patients with dMMR-CRC, there were no significant difference between pMMR-CRC/others and dMMR- or pMMR-CRC with PD-L1(+) and p-STAT1(+) regarding FOXP3 TIL infiltration (Fig. 4G). The aforementioned observations in the clinical cohort suggested that there was a subset of patients with pMMR-CRC with elevated CD8(+) and CD4(+) TIL infiltration.

Discussion

There are several potential biomarkers to predict response to ICIs, including tumor mutation burden (TMB), MSI status, PD-L1 expression, CD8 infiltration and the IFN-γ gene signature (2733). Several clinical studies have established MSI status as a putative response biomarker for PD-1 blockade, and immunotherapy with ICIs against the PD-1/PD-L1 axis is clinically approved for dMMR/MSI-H solid tumors of varying histological origin (13,14,17). dMMR/MSI-H tumors are thought to respond to ICIs for a number of reasons. dMMR/MSI-H tumors have an antigenic microenvironment with a high density of TILs and express multiple immune checkpoint molecules, including PD-1 and PD-L1, due to mutation-induced neo-antigens (3,5,16,17). The present study used clinical samples and TCGA database to reveal that dMMR/MSI-H tumors have immunogenic tumor microenvironments and exhibit upregulated PD-L1 and PD-L2 expression levels, and an increase in p-STAT1, and increased infiltration of CD8 T cells compared with pMMR/MSS tumors, and that these factors are closely associated with IFN-γ production within the tumor microenvironment. Since the binding of IFN-γ to its cognate receptor leads to the phosphorylation-dependent activation of JAK1/2 and STAT1, and that p-STAT1 forms subsequently homodimers that translocate to the nucleus and activate the transcription of IFN-γ-stimulated genes (26), p-STAT1 expression was investigated in the present study. These observations are in line with previous studies investigating gastric cancer, which reported that the upregulation of PD-L1 and PD-L2 is significantly associated with IFN-γ production and CD8 T cell infiltration within the tumor microenvironment (34,35).

However, previous studies revealed that in pMMR/MSS-CRC, a small subset of patients may still benefit from immunotherapy with anti-PD-1/PD-L1 antibodies (1315). Therefore, the identification of distinct biomarker-defined populations with pMMR/MSS-CRC may improve patient outcome. The present study revealed a subset of patients with MSI-L/MSS-CRC with upregulated CD8 T effector and IFN-γ gene signatures, as seen in MSI-H-CRC. Previous studies reported that the number of CD8+ TILs was increased in MSI-CRC compared with MSS-CRC (3638). Also, De Smedt et al (36) used IHC to demonstrate that there were no significant differences in the number of CD4(+) TILs in MSI- or MSS-CRC; however, Boissière-Michot et al (37) reported that the density of CD4(+) TILs was increased in MSI-CRC compared with MSS-CRC. Pagès et al (38) revealed that 21% of patients with MSS tumors had high immunoscore densities of CD3+ and CD8+ TILs, similar to MSI tumors. The present study identified a subset of patients with pMMR-CRC with increased CD8(+) and CD4(+) TIL infiltration, as seen in dMMR-CRC. The aforementioned patients may benefit from immunotherapy with anti-PD-1/PD-L1 mAbs.

It is well known that regulatory T (Treg) cells express FOXP3 and suppress antitumor immune responses (3943). In patients with CRC, the number of Treg cells (which suppress the activity of tumor antigen-specific T cells) is increased in the tumor microenvironment (4446). Although several studies have evaluated the association between Treg cells and MSI status, the results were controversial (37,3941,47). Differences in these studies may be attributed to the use of different methodologies, such as PCR and IHC, the types of lesions analyzed, including invasive front and surface of tumor, and the phenotypic diversity of Treg cells (48). Further investigation is required to clarify the association between Treg cells and MSI or MMR status in CRC.

A recent study demonstrated that in a large cohort of patients with CRC, those with high TMB accounted for 3% of MSS-CRC cases (49), indicating that a subset of patients with MSS-CRC with high TMB may benefit from treatment with ICIs. As the TMB status was not evaluated in the present study, it is currently unclear whether the subpopulation of patients with MSI-L/MSS-CRC with upregulated CD8 T effector and IFN-γ gene signatures corresponds with high mutation burden groups. Furthermore, conclusions about the responsiveness to ICI therapies cannot be drawn in the current study. Further investigations to clarify the association between TMB and immune profiling in MSI-L/MSS-CRC, as well as the clinical response in the identified subpopulation, are required.

While the MSS/TMB-high group may benefit from treatment with ICIs, there are several unresolved issues to accurately identify patients with MSS/TMB-high status. TMB analysis from whole exome sequencing is not widely available, as it is a time and cost intensive method (50,51). More importantly, it has recently been shown that patients with metastatic MSI-H/dMMR-CCR who exhibit primary resistance to ICIs may have been misdiagnosed (52). In other words, patients who had been initially diagnosed as MSI-H were actually MSS, indicating that MSI-testing is currently not accurate enough and not fully established. Furthermore, Wang et al (53) reported that patients with a polymerase ε-mutation present a hyper-mutated tumor phenotype caused by high frequent base substitution mutations, without necessarily giving rise to the short tandem repeat signature identified through MSI-H testing. In light of the aforementioned complexities, multiple predictive biomarkers are required to accurately select ICI responders with MSS/pMMR-CRC. The evaluation of CD8 T effector and IFN-γ gene signatures described in the current study may be used as part of a combination testing strategy for ICI responder identification.

The present study demonstrated that a subset of patients with upregulated CD8 T effector and IFN-γ gene signatures MSI-L/MSS- or pMMR-CRC may benefit from immunotherapy with anti-PD-1/PD-L1 mAbs.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets generated and/or analyzed during the current study are available in The Cancer Genome Atlas and were downloaded via cBioPortal (www.cbioportal.org).

Authors' contributions

KM and KK conceived and designed the study. HO and YN analyzed TCGA dataset. KS, LY, EE, WS, SF, HE, MS, TM, ZS and SO contributed to the acquisition of the patient samples. TK, KS, LY and EE performed and evaluated the IHC staining. TK, KM, HO, YN, WS, SF, HE, MS, TM, ZS and SO analyzed the patient data. TK, HO, KM and KK drafted the manuscript. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

All procedures were conducted in accordance with the Helsinki Declaration and were approved by the responsible committee on human experimentation at Fukushima Medical University (Reference 2847). Written informed consent was obtained from all patients.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

CRC

colorectal cancer

dMMR

deficient mismatch repair

ICIs

immune checkpoint inhibitors

IHC

Immunohistochemistry

IFN-γ

interferon-γ

JAK

Janus-activated kinase

MSI-H

microsatellite instability-high

MSI-L

MSI-low

MSS

microsatellite stable

mAbs

monoclonal antibodies

p-STAT1

phosphorylated signal transducer and activator of transcription 1

pMMR

proficient MMR

PD-1

programmed cell death 1

PD-L1

programmed death ligand 1

Treg

regulatory T

TCGA

The Cancer Genome Atlas

TILs

tumor infiltrating lymphocytes

TMB

tumor mutation burden

References

1 

Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D and Bray F: Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 136:E359–E386. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2016. CA Cancer J Clin. 66:7–30. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Brenner H, Kloor M and Pox CP: Colorectal cancer. Lancet. 383:1490–1502. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Cancer Genome Atlas Network, . Comprehensive molecular characterization of human colon and rectal cancer. Nature. 487:330–337. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Kloor M and von Knebel Doeberitz M: The immune biology of microsatellite-unstable cancer. Trends Cancer. 2:121–133. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Koopman M, Kortman GA, Mekenkamp L, Ligtenberg MJ, Hoogerbrugge N, Antonini NF, Punt CJ and van Krieken JH: Deficient mismatch repair system in patients with sporadic advanced colorectal cancer. Br J Cancer. 100:266–273. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Sargent DJ, Marsoni S, Monges G, Thibodeau SN, Labianca R, Hamilton SR, French AJ, Kabat B, Foster NR, Torri V, et al: Defective mismatch repair as a predictive marker for lack of efficacy of fluorouracil-based adjuvant therapy in colon cancer. J Clin Oncol. 28:3219–3226. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, Chow LQ, Vokes EE, Felip E, Holgado E, et al: Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med. 373:1627–1639. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Kang YK, Boku N, Satoh T, Ryu MH, Chao Y, Kato K, Chung HC, Chen JS, Muro K, Kang WK, et al: Nivolumab in patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens (ONO-4538-12, ATTRACTION-2): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 390:2461–2471. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Escudier B, Motzer RJ, Sharma P, Wagstaff J, Plimack ER, Hammers HJ, Donskov F, Gurney H, Sosman JA, Zalewski PG, et al: Treatment beyond progression in patients with advanced renal cell carcinoma treated with nivolumab in CheckMate 025. Eur Urol. 72:368–376. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K, et al: Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 366:2455–2465. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB, et al: Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 366:2443–2454. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, Skora AD, Luber BS, Azad NS, Laheru D, et al: PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 372:2509–2520. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Overman MJ, McDermott R, Leach JL, Lonardi S, Lenz HJ, Morse MA, Desai J, Hill A, Axelson M, Moss RA, et al: Nivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer (CheckMate 142): An open-label, multicentre, phase 2 study. Lancet Oncol. 18:1182–1191. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, Lu S, Kemberling H, Wilt C, Luber BS, et al: Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 357:409–413. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Bauer K, Nelius N, Reuschenbach M, Koch M, Weitz J, Steinert G, Kopitz J, Beckhove P, Tariverdian M, von Knebel Doeberitz M and Kloor M: T cell responses against microsatellite instability-induced frameshift peptides and influence of regulatory T cells in colorectal cancer. Cancer Immunol Immunother. 62:27–37. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Dudley JC, Lin MT, Le DT and Eshleman JR: Microsatellite instability as a biomarker for PD-1 blockade. Clin Cancer Res. 22:813–820. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun Y, Jacobsen A, Sinha R, Larsson E, et al: Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 6:pl12013. View Article : Google Scholar : PubMed/NCBI

19 

Liu Y, Sethi NS, Hinoue T, Schneider BG, Cherniack AD, Sanchez-Vega F, Seoane JA, Farshidfar F, Bowlby R, Islam M, et al: Comparative molecular analysis of gastrointestinal adenocarcinomas. Cancer Cell. 33:721–735.e8. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Wallin JJ, Bendell JC, Funke R, Sznol M, Korski K, Jones S, Hernandez G, Mier J, He X, Hodi FS, et al: Atezolizumab in combination with bevacizumab enhances antigen-specific T-cell migration in metastatic renal cell carcinoma. Nat Commun. 7:126242016. View Article : Google Scholar : PubMed/NCBI

21 

Ayers M, Lunceford J, Nebozhyn M, Murphy E, Loboda A, Kaufman DR, Albright A, Cheng JD, Kang SP, Shankaran V, et al: IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest. 127:2930–2940. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Nakayama Y, Mimura K, Tamaki T, Shiraishi K, Kua LF, Koh V, Ohmori M, Kimura A, Inoue S, Okayama H, et al: Phospho-STAT1 expression as a potential biomarker for anti-PD-1/anti-PD-L1 immunotherapy for breast cancer. Int J Oncol. 54:2030–2038. 2019.PubMed/NCBI

23 

Ashizawa M, Okayama H, Ishigame T, Thar Min AK, Saito K, Ujiie D, Murakami Y, Kikuchi T, Nakayama Y, Noda M, et al: miRNA-148a-3p regulates immunosuppression in DNA mismatch repair-deficient colorectal cancer by targeting PD-L1. Mol Cancer Res. 17:1403–1413. 2019.PubMed/NCBI

24 

Herbst RS, Baas P, Kim DW, Felip E, Pérez-Gracia JL, Han JY, Molina J, Kim JH, Arvis CD, Ahn MJ, et al: Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): A randomised controlled trial. Lancet. 387:1540–1550. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Noda M, Okayama H, Tachibana K, Sakamoto W, Saito K, Thar Min AK, Ashizawa M, Nakajima T, Aoto K, Momma T, et al: Glycosyltransferase gene expression identifies a poor prognostic colorectal cancer subtype associated with mismatch repair deficiency and incomplete glycan synthesis. Clin Cancer Res. 24:4468–4481. 2018. View Article : Google Scholar : PubMed/NCBI

26 

Khodarev NN, Roizman B and Weichselbaum RR: Molecular pathways: Interferon/stat1 pathway: Role in the tumor resistance to genotoxic stress and aggressive growth. Clin Cancer Res. 18:3015–3021. 2012. View Article : Google Scholar : PubMed/NCBI

27 

McGranahan N, Furness AJ, Rosenthal R, Ramskov S, Lyngaa R, Saini SK, Jamal-Hanjani M, Wilson GA, Birkbak NJ, Hiley CT, et al: Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science. 351:1463–1469. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Postow MA, Callahan MK and Wolchok JD: Immune checkpoint blockade in cancer therapy. J Clin Oncol. 33:1974–1982. 2015. View Article : Google Scholar : PubMed/NCBI

29 

McLaughlin J, Han G, Schalper KA, Carvajal-Hausdorf D, Pelekanou V, Rehman J, Velcheti V, Herbst R, LoRusso P and Rimm DL: Quantitative assessment of the heterogeneity of PD-L1 expression in non-small-cell lung cancer. JAMA Oncol. 2:46–54. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Bhaijee F and Anders RA: PD-L1 expression as a predictive biomarker: Is absence of proof the same as proof of absence? JAMA Oncol. 2:54–55. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Schalper KA, Kaftan E and Herbst RS: Predictive biomarkers for PD-1 axis therapies: The hidden treasure or a call for research. Clin Cancer Res. 22:2102–2104. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Nishino M, Ramaiya NH, Hatabu H and Hodi FS: Monitoring immune-checkpoint blockade: Response evaluation and biomarker development. Nat Rev Clin Oncol. 14:655–668. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Gibney GT, Weiner LM and Atkins MB: Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol. 17:e542–e551. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Mimura K, Kua LF, Shiraishi K, Kee Siang L, Shabbir A, Komachi M, Suzuki Y, Nakano T, Yong WP, So J and Kono K: Inhibition of mitogen-activated protein kinase pathway can induce upregulation of human leukocyte antigen class I without PD-L1-upregulation in contrast to interferon-gamma treatment. Cancer Sci. 105:1236–1244. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Mimura K, Teh JL, Okayama H, Shiraishi K, Kua LF, Koh V, Smoot DT, Ashktorab H, Oike T, Suzuki Y, et al: PD-L1 expression is mainly regulated by interferon gamma associated with JAK-STAT pathway in gastric cancer. Cancer Sci. 109:43–53. 2018. View Article : Google Scholar : PubMed/NCBI

36 

De Smedt L, Lemahieu J, Palmans S, Govaere O, Tousseyn T, Van Cutsem E, Prenen H, Tejpar S, Spaepen M, Matthijs G, et al: Microsatellite instable vs. stable colon carcinomas: Analysis of tumour heterogeneity, inflammation and angiogenesis. Br J Cancer. 113:500–509. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Boissière-Michot F, Lazennec G, Frugier H, Jarlier M, Roca L, Duffour J, Du Paty E, Laune D, Blanchard F, Le Pessot F, et al: Characterization of an adaptive immune response in microsatellite-instable colorectal cancer. Oncoimmunology. 3:e292562014. View Article : Google Scholar : PubMed/NCBI

38 

Pagès F, Mlecnik B, Marliot F, Bindea G, Ou FS, Bifulco C, Lugli A, Zlobec I, Rau TT, Berger MD, et al: International validation of the consensus Immunoscore for the classification of colon cancer: A prognostic and accuracy study. Lancet. 391:2128–2139. 2018. View Article : Google Scholar : PubMed/NCBI

39 

Le Gouvello S, Bastuji-Garin S, Aloulou N, Mansour H, Chaumette MT, Berrehar F, Seikour A, Charachon A, Karoui M, Leroy K, et al: High prevalence of Foxp3 and IL17 in MMR-proficient colorectal carcinomas. Gut. 57:772–779. 2008. View Article : Google Scholar : PubMed/NCBI

40 

Michel S, Benner A, Tariverdian M, Wentzensen N, Hoefler P, Pommerencke T, Grabe N, von Knebel Doeberitz M and Kloor M: High density of FOXP3-positive T cells infiltrating colorectal cancers with microsatellite instability. Br J Cancer. 99:1867–1873. 2008. View Article : Google Scholar : PubMed/NCBI

41 

Salama P, Phillips M, Grieu F, Morris M, Zeps N, Joseph D, Platell C and Iacopetta B: Tumor-infiltrating FOXP3+ T regulatory cells show strong prognostic significance in colorectal cancer. J Clin Oncol. 27:186–192. 2009. View Article : Google Scholar : PubMed/NCBI

42 

Blatner NR, Bonertz A, Beckhove P, Cheon EC, Krantz SB, Strouch M, Weitz J, Koch M, Halverson AL, Bentrem DJ and Khazaie K: In colorectal cancer mast cells contribute to systemic regulatory T-cell dysfunction. Proc Natl Acad Sci USA. 107:6430–6435. 2010. View Article : Google Scholar : PubMed/NCBI

43 

Frey DM, Droeser RA, Viehl CT, Zlobec I, Lugli A, Zingg U, Oertli D, Kettelhack C, Terracciano L and Tornillo L: High frequency of tumor-infiltrating FOXP3(+) regulatory T cells predicts improved survival in mismatch repair-proficient colorectal cancer patients. Int J Cancer. 126:2635–2643. 2010.PubMed/NCBI

44 

Clarke SL, Betts GJ, Plant A, Wright KL, El-Shanawany TM, Harrop R, Torkington J, Rees BI, Williams GT, Gallimore AM and Godkin AJ: CD4+CD25+FOXP3+ regulatory T cells suppress anti-tumor immune responses in patients with colorectal cancer. PLoS One. 1:e1292006. View Article : Google Scholar : PubMed/NCBI

45 

Loddenkemper C, Schernus M, Noutsias M, Stein H, Thiel E and Nagorsen D: In situ analysis of FOXP3+ regulatory T cells in human colorectal cancer. J Transl Med. 4:522006. View Article : Google Scholar : PubMed/NCBI

46 

Ling KL, Pratap SE, Bates GJ, Singh B, Mortensen NJ, George BD, Warren BF, Piris J, Roncador G, Fox SB, et al: Increased frequency of regulatory T cells in peripheral blood and tumour infiltrating lymphocytes in colorectal cancer patients. Cancer Immun. 7:72007.PubMed/NCBI

47 

Masugi Y, Nishihara R, Yang J, Mima K, da Silva A, Shi Y, Inamura K, Cao Y, Song M, Nowak JA, et al: Tumour CD274 (PD-L1) expression and T cells in colorectal cancer. Gut. 66:1463–1473. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Liston A and Gray DH: Homeostatic control of regulatory T cell diversity. Nat Rev Immunol. 14:154–165. 2014. View Article : Google Scholar : PubMed/NCBI

49 

Fabrizio DA, George TJ Jr, Dunne RF, Frampton G, Sun J, Gowen K, Kennedy M, Greenbowe J, Schrock AB, Hezel AF, et al: Beyond microsatellite testing: Assessment of tumor mutational burden identifies subsets of colorectal cancer who may respond to immune checkpoint inhibition. J Gastrointest Oncol. 9:610–617. 2018. View Article : Google Scholar : PubMed/NCBI

50 

Wang Z, Duan J, Cai S, Han M, Dong H, Zhao J, Zhu B, Wang S, Zhuo M, Sun J, et al: Assessment of blood tumor mutational burden as a potential biomarker for immunotherapy in patients with non-small cell lung cancer with use of a next-generation sequencing cancer gene panel. JAMA Oncol. 5:696–702. 2019. View Article : Google Scholar : PubMed/NCBI

51 

Chan TA, Yarchoan M, Jaffee E, Swanton C, Quezada SA, Stenzinger A and Peters S: Development of tumor mutation burden as an immunotherapy biomarker: Utility for the oncology clinic. Ann Oncol. 30:44–56. 2019. View Article : Google Scholar : PubMed/NCBI

52 

Cohen R, Hain E, Buhard O, Guilloux A, Bardier A, Kaci R, Bertheau P, Renaud F, Bibeau F, Fléjou JF, et al: Association of primary resistance to immune checkpoint inhibitors in metastatic colorectal cancer with misdiagnosis of microsatellite instability or mismatch repair deficiency status. JAMA Oncol. 5:551–555. 2019. View Article : Google Scholar : PubMed/NCBI

53 

Wang C, Gong J, Tu TY, Lee PP and Fakih M: Immune profiling of microsatellite instability-high and polymerase epsilon (POLE)-mutated metastatic colorectal tumors identifies predictors of response to anti-PD-1 therapy. J Gastrointest Oncol. 9:404–415. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2019
Volume 18 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kikuchi T, Mimura K, Okayama H, Nakayama Y, Saito K, Yamada L, Endo E, Sakamoto W, Fujita S, Endo H, Endo H, et al: A subset of patients with MSS/MSI‑low‑colorectal cancer showed increased CD8(+) TILs together with up‑regulated IFN‑γ. Oncol Lett 18: 5977-5985, 2019
APA
Kikuchi, T., Mimura, K., Okayama, H., Nakayama, Y., Saito, K., Yamada, L. ... Kono, K. (2019). A subset of patients with MSS/MSI‑low‑colorectal cancer showed increased CD8(+) TILs together with up‑regulated IFN‑γ. Oncology Letters, 18, 5977-5985. https://doi.org/10.3892/ol.2019.10953
MLA
Kikuchi, T., Mimura, K., Okayama, H., Nakayama, Y., Saito, K., Yamada, L., Endo, E., Sakamoto, W., Fujita, S., Endo, H., Saito, M., Momma, T., Saze, Z., Ohki, S., Kono, K."A subset of patients with MSS/MSI‑low‑colorectal cancer showed increased CD8(+) TILs together with up‑regulated IFN‑γ". Oncology Letters 18.6 (2019): 5977-5985.
Chicago
Kikuchi, T., Mimura, K., Okayama, H., Nakayama, Y., Saito, K., Yamada, L., Endo, E., Sakamoto, W., Fujita, S., Endo, H., Saito, M., Momma, T., Saze, Z., Ohki, S., Kono, K."A subset of patients with MSS/MSI‑low‑colorectal cancer showed increased CD8(+) TILs together with up‑regulated IFN‑γ". Oncology Letters 18, no. 6 (2019): 5977-5985. https://doi.org/10.3892/ol.2019.10953