Next Article in Journal
Small Molecule-Based Enzyme Inhibitors in the Treatment of Primary Hyperoxalurias
Previous Article in Journal
Identifying Barriers and Facilitators to Diet and Physical Activity Behaviour Change in Type 2 Diabetes Using a Design Probe Methodology
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

PD-L1 Expression Is Associated with Deficient Mismatch Repair and Poor Prognosis in Middle Eastern Colorectal Cancers

by
Abdul K. Siraj
1,†,
Sandeep Kumar Parvathareddy
1,†,
Padmanaban Annaiyappanaidu
1,
Wael Haqawi
1,
Maha Al-Rasheed
1,
Hadeel M. AlManea
2,
Hussah F. AlHussaini
2,
Fouad Al-Dayel
2 and
Khawla S. Al-Kuraya
1,*
1
Human Cancer Genomic Research, Research Center, King Faisal Specialist Hospital and Research Center, P.O. Box 3354, Riyadh 11211, Saudi Arabia
2
Department of Pathology, King Faisal Specialist Hospital and Research Centre, P.O. Box 3354, Riyadh 11211, Saudi Arabia
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Submission received: 23 December 2020 / Revised: 19 January 2021 / Accepted: 24 January 2021 / Published: 26 January 2021

Abstract

:
Several clinical trials are investigating the use of immune-targeted therapy with Programmed death ligand-1 (PD-L1) inhibitors for colorectal cancer (CRC), with promising results for patients with mismatch repair (MMR) deficiency or metastatic CRC. However, the prognostic significance of PD-L1 expression in CRC is controversial and such data are lacking in CRC from Middle Eastern ethnicity. We carried out this large retrospective study to investigate the prognostic and clinico-pathological impact of PD-L1 expression in Middle Eastern CRC using immunohistochemistry. A total of 1148 CRC were analyzed for PD-L1 expression. High PD-L1 expression was noted in 37.3% (428/1148) cases and was correlated with aggressive clinico-pathological features such as high malignancy grade (p < 0.0001), larger tumor size (p = 0.0007) and mucinous histology (p = 0.0005). Interestingly, PD-L1 expression was significantly higher in patients exhibiting MMR deficiency (p = 0.0169) and BRAF mutation (p = 0.0008). Furthermore, the expression of PD-L1 was found to be an independent marker for overall survival (HR = 1.45; 95% CI = 1.06–1.99; p = 0.0200). In conclusion, the results of this study indicate that PD-L1 expression could be a valid biomarker for poor prognosis in Middle Eastern CRC patients. This information can help in decision-making for anti-PD-L1 therapy in Middle Eastern CRC, especially for patients with MMR deficient tumors.

1. Introduction

Despite advances in cancer screening and therapy, colorectal cancer (CRC) is still a leading cause of cancer-related death worldwide [1,2]. In Saudi Arabia the incidence of CRC is increasing and it ranks as the most common cancer affecting Saudi males [3], with most patients presenting at advanced stage by the time they seek medical intervention [4]. Therefore, it is of great importance to identify new biomarkers that can predict patients’ survival and improve therapeutic options for advanced CRC patients.
One of the new emerging, promising biomarkers to evaluate patients’ outcome is programmed death ligand-1 (PD-L1). PD-L1, a transmembrane protein which binds to its inhibitory receptor PD-1 on T-cells, elicits T-cell anergy and leads to immunosuppression [5,6]. Several cancers, such as non-small cell lung cancer, melanoma, breast cancer and CRC, have been shown to upregulate surface expression of PD-L1 in order to escape the T-cell immune surveillance [7,8,9]. Monoclonal antibodies that target PD-L1 can reverse the T-cell anergy and re-sensitize tumor cells to antitumor immune surveillance [8,10]. These immunotherapies are being intensively evaluated for their efficacy in treatment of poor outcome cancers, including CRC [11,12,13]. Although the response rate to immunotherapies in unselected CRC is relatively low [14,15], PD-L1 inhibitors have shown good response in advanced CRC and some have been approved for treatment in patients with deficient mismatch repair (dMMR) refractory or metastatic CRC [16,17].
Over the past decade, expression of PD-L1 in CRC has been investigated by several studies [18,19,20,21]. The results obtained suggest that measurement of PD-L1 in CRC specimens could be useful for patients’ prognosis as well as for predicting patients that could benefit from anti-PD-L1 therapies [18,19]. However, studies analyzing PD-L1 and clinico-pathological markers and tumor prognosis are controversial. Therefore, we conducted this study using more than 1100 Middle Eastern CRCs to analyze the expression of PD-L1 and its correlation with clinico-pathological markers to explore its predictive/prognostic value in these patients.

2. Materials and Methods

2.1. Sample Selection

Archival samples from 1207 CRC patients diagnosed between 1990 to 2015 at King Faisal Specialist Hospital and Research Centre (Riyadh, Saudi Arabia) were available to be included in the study. However, immunohistochemical analysis was possible in only 1148 cases and hence these were included for further analysis. Clinico-pathological data were collected from patient medical records, which are summarized in Table 1. Institutional Review Board of King Faisal Specialist Hospital and Research Centre provided ethical approval for the current study. Research Advisory Council (RAC) granted waiver of informed consent for use of retrospective patient case data and archival tissue samples under project RAC# 2170 025.

2.2. DNA Isolation

DNAs were extracted from CRC tumor tissues utilizing Gentra DNA isolation kit (Gentra, Minneapolis, MN, USA) as per the manufacturer’s protocols, which were elaborated in our previous study [22].

2.3. Sanger Sequencing Analysis

Entire coding and splicing regions of exon 15 in BRAF gene among 1207 CRC samples were sequenced using Sanger sequencing technology. Primer 3 online software was utilized to design the primers (available upon request). PCR and Sanger sequencing analysis were carried out as described previously [23]. Reference sequence was downloaded from NCBI GenBank. Sequencing results were compared with the reference sequence by Mutation Surveyor V4.04 (Soft Genetics, LLC, State College, PA, USA).

2.4. Tissue Microarray Construction and Immunohistochemistry

Tissue microarray (TMA) format was utilized for immunohistochemical (IHC) analysis of samples. Construction of TMA was done as described previously [24]. Briefly, representative tumor regions from each donor tissue block were chosen and tissue cylinders with a diameter of 0.6 mm were punched and brought into recipient paraffin block with the help of a modified semiautomatic robotic precision instrument (Beecher Instruments, Woodland, WI, USA). Two cores of CRC were arrayed from each case.
Tissue microarray slides were processed and stained manually as described previously [25]. Primary antibody against PD-L1 (E1L3N, 1:50 dilution, pH 9.0, Cell Signaling Technology, Danvers, MA, USA) was used. A normal colon tissue microarray was also stained to validate the antibody. Normal tissues of different organ systems were also included in the TMA to serve as positive controls. Negative control was performed by omission of the primary antibody. A membranous and/or cytoplasmic staining was observed. Only the membrane staining was considered for scoring. PD-L1 was scored as described previously [26]. Briefly, the proportion of tumor cell membrane showing positive staining (brown color) was calculated as a percentage for each core. Scores from the two tissue cores of each tumor were averaged to yield a single percent staining score. For statistical analysis, the scores were dichotomized. Cases showing expression level of ≥5% were classified as over-expression and those with less than 5% as low expression.
Evaluation of mismatch repair (MMR) protein staining was performed as described previously [27]. Briefly, MMR protein expression was evaluated using MSH2, MSH6, MLH1 and PMS2 proteins. Details of the primary antibodies used are provided in Supplementary Table S1. Tumor was classified as deficient MMR (dMMR) if any of the four proteins showed loss of staining in cancer with concurrent positive staining in the nuclei of normal epithelial cells. Otherwise, they were classified as proficient MMR (pMMR).
Immunohistochemical scoring was done by two pathologists, blinded to the clinico-pathological characteristics. Discordant scores were reviewed together to achieve agreement.

2.5. Statistical Analysis

Associations between clinico-pathological variables and protein expression were analyzed using contingency table analysis and Chi square test. Kaplan–Meier method was used to generate overall survival curves and Mantel–Cox log rank test was used to evaluate significance. Multivariate analysis was performed using Cox proportional hazards regression model, after adjusting for clinico-pathological variables like age, gender, stage, grade, site of tumor and mismatch repair status. Two-sided tests were used for the calculations and limit of significance was defined as p value of < 0.05 for all analyses. Data analyses were performed using JMP11.0 (SAS Institute, Inc., Cary, NC, USA) software package.

3. Results

3.1. Patient Characteristics

The clinico-pathological characteristics of the 1148 CRC patients are summarized in Table 1. The median age of the study cohort was 56.3 years (inter quartile range (IQR), 47.5–67.9 years) with a male: female ratio of 1.1. Most of the tumors were located in the left colon (79.7%); 78.8% (905/1148) of patients had a moderately differentiated tumor and 69.9% (803/1148) were either stage II or stage III; 9.2% (106/1148) of tumors were MMR deficient by immunohistochemistry; 3.0% (34/1148) of tumors in our cohort were BRAF mutant. 34.6% (447/1048) patients received neoadjuvant therapy.

3.2. PD-L1 Expression in CRC Tissue Samples and Its Clinico-Pathological Associations

PD-L1 protein expression was assessed immunohistochemically in 1207 CRC tissue samples. However, immunohistochemical data were interpretable in 1148 samples and hence were included for further analysis. PD-L1 membrane staining was considered for scoring (Figure 1, Supplementary Table S2). PD-L1 over-expression was noted in 37.3% (428/1148) of cases. PD-L1 expression showed significant association with high-grade (p < 0.0001), larger tumor size (p = 0.0007) and mucinous histology (p = 0.0005). Importantly, expression of PD-L1 was found to be significantly associated with deficient mismatch repair protein expression (p = 0.0169). We also found a significant association of PD-L1 expression with BRAF mutation (p = 0.0008), despite the low frequency of BRAF mutations in our cohort (Table 2).

3.3. Prognostic Significance of PD-L1 Expression in CRC

PD-L1 expression was found to be associated with poor 5-year overall survival (OS) on univariate analysis (p = 0.0017) (Table 2, Figure 2). Next, we sought to analyze if PD-L1 was an independent prognostic predictor in CRC using Cox proportional hazards regression model, after adjusting for possible confounding factors. Multivariate analysis showed that PD-L1 expression was indeed an independent predictor of poor overall survival in our cohort (HR = 1.45; 95% CI = 1.06–1.99; p = 0.0200) (Table 3).

4. Discussion

Several studies have shown that PD-1/PD-L1 pathway has become a potential therapeutic target for a wide range of human malignancies [28,29,30,31,32,33]. However, the associations between expression of PD-L1 and clinico-pathological parameters are not consistent [18,34,35]. Tumor heterogeneity, antibody used for PD-L1 expression analysis, variation in cutoff point and sample size are among the factors which could explain the inconsistency among these studies. Therefore, in this study we comprehensively analyzed a large cohort of Middle Eastern CRC from a single institute in order to reach a better conclusion.
We found PD-L1 to be expressed in 37.3% (428/1148) of CRC tumors. There was a significant correlation between PD-L1 expression and higher tumor grade, larger tumor size and mucinous histology. Previous studies have found that PD-L1 expression correlated with advanced stage and grade [19,20,36] while other studies failed to confirm this correlation [18,37,38]. Interestingly, multivariate analysis showed that PD-L1 expression was a predictive marker for OS in the entire cohort. Analogously, a recent large meta-analysis by Wang et al. [39] in a large cohort of 8823 CRC patients from 32 studies also showed that PD-L1 expression was an independent predictor for poor OS. Another meta-analysis comprising 13 studies with 3905 CRC patients also showed a significant association between expression of PD-L1 and poor OS [40]. In contrast, a study by Li et al. [18] found PD-L1 expression in CRC tumor cells to be associated with favorable OS and disease-free survival on univariate analysis. However, this significance was lost on multivariate analysis.
Recent evidence suggests that MMR deficient tumors could provide a robust marker for anti-PD-1/PD-L1 response [16,41,42]. A strong association between PD-L1 tumor expression and MMR deficiency has been documented in endometrial and colorectal carcinoma [43,44] and the efficacy of anti-PD-1/PD-L1 therapy has been shown to increase in MMR deficient tumors [16,45,46]. MMR deficiency allows for rapid accumulation of neoantigens [47], which could explain the positive relationship between PD-L1 expression and better immunotherapeutic response in dMMR tumors. In this study, we showed significantly higher PD-L1 expression in dMMR tumors (p = 0.0169), suggesting that this subset of Middle Eastern CRCs may benefit from PD-L1 targeted therapy. Another mechanism of immune evasion commonly relies on mutation in BRAF [48]. In contrast to MMR deficiency, BRAF mutation in Middle Eastern CRC is not highly prevalent, with only 3% of our entire cohort being positive for this mutation (Table 1). Notably, PD-L1 expression was significantly higher in BRAF mutant tumors, which is in concordance with a previous study by Rosenbaum et al. [43].
However, recent evidence has shown that IHC staining of PD-L1 in CRC may not identify all patients who might respond to anti-PD-L1 agents. A phase II clinical trial (CheckMate 142) has shown similar overall response rates to immunotherapy in patients with and without PD-L1 expression [17,49]. A possible explanation could be the effect of tumor heterogeneity on the predictive value of PD-L1 expression. Indeed, this has led to the use of non-invasive molecular imaging methods (PET-CT) for PD-1/PD-L1, which are more informative, as they provide an image of the entire tumor area, both primary and metastatic, thus accounting for tumor heterogeneity [50,51,52]. However, IHC has the advantage of providing information on the cell type expressing PD-L1 (tumor cells vs. immune cells) and also being cost-effective. Hence, IHC continues to be the method of choice to determine the expression of PD-L1.

5. Conclusions

In conclusion, this large study from a single institute and unique ethnicity provided evidence on the importance of evaluation for PD-L1 expression and its correlation with clinico-pathological markers and prognostic outcomes. Furthermore, the information available from this report suggests that PD-L1 could be utilized as a prognostic marker in Middle Eastern CRC patients for risk stratification and that PD-1/PD-L1 inhibitors might be a valid therapeutic option for patients with advanced CRCs harboring BRAF mutation or showing deficient MMR. Thus, our study provides scientific rationale for clinicians to select this subset of CRC patients for anti-PD-1/PD-L1 immunotherapy.

Supplementary Materials

The following are available online at https://www.mdpi.com/2075-4426/11/2/73/s1, Table S1: Antibodies used for the mismatch repair immunohistochemistry assay, Table S2: PD-L1 expression stratified by proportion of immunohistochemical staining.

Author Contributions

Conceptualization, K.S.A.-K. and A.K.S.; methodology, A.K.S. and S.K.P.; software, S.K.P.; validation, S.K.P., P.A. and W.H.; formal analysis, S.K.P. and A.K.S.; investigation, A.K.S., S.K.P., P.A., W.H. and M.A.-R.; resources, H.M.A., H.F.A. and F.A.-D.; data curation, A.K.S. and S.K.P.; writing—original draft preparation, K.S.A.-K.; writing—review and editing, A.K.S. and S.K.P.; visualization, S.K.P. and A.K.S.; supervision, A.K.S. and K.S.A.-K.; project administration, A.K.S. and K.S.A.-K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Institutional Review Board of King Faisal Specialist Hospital and Research Centre provided ethical approval for the current study. Research Advisory Council (RAC) granted waiver of informed consent for use of retrospective patient case data and archival tissue samples under project RAC# 2170 025.

Informed Consent Statement

Patient consent was waived since only archival tissue blocks were used.

Data Availability Statement

The data presented in this study are available in the article.

Acknowledgments

The authors acknowledge the technical assistance provided by Felisa DeVera.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA A Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [Green Version]
  2. Keum, N.; Giovannucci, E. Global burden of colorectal cancer: Emerging trends, risk factors and prevention strategies. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 713–732. [Google Scholar] [CrossRef]
  3. Alrawaji, A.; Alzahrani, A.; Almadouj, A.; Alzahrani, B.; Alhashmi, A.; Sharaheli. Cancer Incidence Report Saudi Arabia 2015; Saudi Cancer Registry: Riyadh, Saudi Arabia, 2018. [Google Scholar]
  4. Alsanea, N.; Abduljabbar, A.S.; Alhomoud, S.; Ashari, L.H.; Hibbert, D.; Bazarbashi, S. Colorectal cancer in Saudi Arabia: Incidence, survival, demographics and implications for national policies. Ann. Saudi Med. 2015, 35, 196–202. [Google Scholar] [CrossRef]
  5. Qin, W.; Hu, L.; Zhang, X.; Jiang, S.; Li, J.; Zhang, Z.; Wang, X. The diverse function of PD-1/PD-L pathway beyond cancer. Front. Immunol. 2019, 10, 2298. [Google Scholar] [CrossRef]
  6. Zheng, Y.; Fang, Y.C.; Li, J. PD-L1 expression levels on tumor cells affect their immunosuppressive activity. Oncol. Lett. 2019, 18, 5399–5407. [Google Scholar] [CrossRef] [Green Version]
  7. Chen, N.; Fang, W.; Zhan, J.; Hong, S.; Tang, Y.; Kang, S.; Zhang, Y.; He, X.; Zhou, T.; Qin, T. Upregulation of PD-L1 by EGFR activation mediates the immune escape in EGFR-driven NSCLC: Implication for optional immune targeted therapy for NSCLC patients with EGFR mutation. J. Thorac. Oncol. 2015, 10, 910–923. [Google Scholar] [CrossRef] [Green Version]
  8. Smyth, M.J.; Ngiow, S.F.; Ribas, A.; Teng, M.W. Combination cancer immunotherapies tailored to the tumour microenvironment. Nat. Rev. Clin. Oncol. 2016, 13, 143. [Google Scholar] [CrossRef]
  9. Han, Y.; Liu, D.; Li, L. PD-1/PD-L1 pathway: Current researches in cancer. Am. J. Cancer Res. 2020, 10, 727. [Google Scholar]
  10. Xu-Monette, Z.Y.; Zhang, M.; Li, J.; Young, K.H. PD-1/PD-L1 blockade: Have we found the key to unleash the antitumor immune response? Front. Immunol. 2017, 8, 1597. [Google Scholar] [CrossRef] [Green Version]
  11. Dang, T.O.; Ogunniyi, A.; Barbee, M.S.; Drilon, A. Pembrolizumab for the treatment of PD-L1 positive advanced or metastatic non-small cell lung cancer. Expert Rev. Anticancer Ther. 2016, 16, 13–20. [Google Scholar] [CrossRef] [Green Version]
  12. Zou, Y.; Zou, X.; Zheng, S.; Tang, H.; Zhang, L.; Liu, P.; Xie, X. Efficacy and predictive factors of immune checkpoint inhibitors in metastatic breast cancer: A systematic review and meta-analysis. Ther. Adv. Med Oncol. 2020, 12. [Google Scholar] [CrossRef]
  13. Oliveira, A.F.; Bretes, L.; Furtado, I. Review of PD-1/PD-L1 inhibitors in metastatic dMMR/MSI-H colorectal cancer. Front. Oncol. 2019, 9, 396. [Google Scholar] [CrossRef] [Green Version]
  14. Cogdill, A.P.; Andrews, M.C.; Wargo, J.A. Hallmarks of response to immune checkpoint blockade. Br. J. Cancer 2017, 117, 1–7. [Google Scholar] [CrossRef] [Green Version]
  15. Lichtenstern, C.R.; Ngu, R.K.; Shalapour, S.; Karin, M. Immunotherapy, Inflammation and Colorectal Cancer. Cells 2020, 9, 618. [Google Scholar] [CrossRef] [Green Version]
  16. Le, D.T.; Uram, J.N.; Wang, H.; Bartlett, B.R.; Kemberling, H.; Eyring, A.D.; Skora, A.D.; Luber, B.S.; Azad, N.S.; Laheru, D. PD-1 blockade in tumors with mismatch-repair deficiency. N. Engl. J. Med. 2015, 372, 2509–2520. [Google Scholar] [CrossRef]
  17. Overman, M.J.; McDermott, R.; Leach, J.L.; Lonardi, S.; Lenz, H.-J.; Morse, M.A.; Desai, J.; Hill, A.; Axelson, M.; Moss, R.A. 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. 2017, 18, 1182–1191. [Google Scholar] [CrossRef]
  18. Li, Y.; Liang, L.; Dai, W.; Cai, G.; Xu, Y.; Li, X.; Li, Q.; Cai, S. Prognostic impact of programed cell death-1 (PD-1) and PD-ligand 1 (PD-L1) expression in cancer cells and tumor infiltrating lymphocytes in colorectal cancer. Mol. Cancer 2016, 15, 55. [Google Scholar] [CrossRef] [Green Version]
  19. Yang, L.; Xue, R.; Pan, C. Prognostic and clinicopathological value of PD-L1 in colorectal cancer: A systematic review and meta-analysis. Oncotargets Ther. 2019, 12, 3671. [Google Scholar] [CrossRef]
  20. Lee, L.H.; Cavalcanti, M.S.; Segal, N.H.; Hechtman, J.F.; Weiser, M.R.; Smith, J.J.; Garcia-Aguilar, J.; Sadot, E.; Ntiamoah, P.; Markowitz, A.J. Patterns and prognostic relevance of PD-1 and PD-L1 expression in colorectal carcinoma. Mod. Pathol. 2016, 29, 1433–1442. [Google Scholar] [CrossRef] [Green Version]
  21. Droeser, R.A.; Hirt, C.; Viehl, C.T.; Frey, D.M.; Nebiker, C.; Huber, X.; Zlobec, I.; Eppenberger-Castori, S.; Tzankov, A.; Rosso, R. Clinical impact of programmed cell death ligand 1 expression in colorectal cancer. Eur. J. Cancer 2013, 49, 2233–2242. [Google Scholar] [CrossRef]
  22. Abubaker, J.; Jehan, Z.; Bavi, P.; Sultana, M.; Al-Harbi, S.; Ibrahim, M.; Al-Nuaim, A.; Ahmed, M.; Amin, T.; Al-Fehaily, M. Clinicopathological analysis of papillary thyroid cancer with PIK3CA alterations in a Middle Eastern population. J. Clin. Endocrinol. Metab. 2008, 93, 611–618. [Google Scholar] [CrossRef] [Green Version]
  23. Bu, R.; Siraj, A.K.; Al-Obaisi, K.A.; Beg, S.; Al Hazmi, M.; Ajarim, D.; Tulbah, A.; Al-Dayel, F.; Al-Kuraya, K.S. Identification of novel BRCA founder mutations in Middle Eastern breast cancer patients using capture and Sanger sequencing analysis. Int. J. Cancer 2016, 139, 1091–1097. [Google Scholar] [CrossRef] [Green Version]
  24. Siraj, A.; Bavi, P.; Abubaker, J.; Jehan, Z.; Sultana, M.; Al-Dayel, F.; Al-Nuaim, A.; Alzahrani, A.; Ahmed, M.; Al-Sanea, O. Genome-wide expression analysis of Middle Eastern papillary thyroid cancer reveals c-MET as a novel target for cancer therapy. J. Pathol. 2007, 213, 190–199. [Google Scholar] [CrossRef]
  25. Bavi, P.; Jehan, Z.; Atizado, V.; Al-Dossari, H.; Al-Dayel, F.; Tulbah, A.; Amr, S.S.; Sheikh, S.S.; Ezzat, A.; El-Solh, H. Prevalence of fragile histidine triad expression in tumors from Saudi Arabia: A tissue microarray analysis. Cancer Epidemiol. Prev. Biomark. 2006, 15, 1708–1718. [Google Scholar] [CrossRef] [Green Version]
  26. Mesnage, S.; Auguste, A.; Genestie, C.; Dunant, A.; Pain, E.; Drusch, F.; Gouy, S.; Morice, P.; Bentivegna, E.; Lhomme, C. Neoadjuvant chemotherapy (NACT) increases immune infiltration and programmed death-ligand 1 (PD-L1) expression in epithelial ovarian cancer (EOC). Ann. Oncol. 2017, 28, 651–657. [Google Scholar] [CrossRef]
  27. Siraj, A.K.; Prabhakaran, S.; Bavi, P.; Bu, R.; Beg, S.; Hazmi, M.A.; Al-Rasheed, M.; Al-Assiri, M.; Sairafi, R.; Al-Dayel, F. Prevalence of Lynch syndrome in a Middle Eastern population with colorectal cancer. Cancer 2015, 121, 1762–1771. [Google Scholar] [CrossRef]
  28. Nguyen, H.D.; Liao, Y.-C.; Ho, Y.-S.; Chen, L.-C.; Chang, H.-W.; Cheng, T.-C.; Liu, D.; Lee, W.-R.; Shen, S.-C.; Wu, C.-H. The α9 nicotinic acetylcholine receptor mediates nicotine-induced pd-l1 expression and regulates melanoma cell proliferation and migration. Cancers 2019, 11, 1991. [Google Scholar] [CrossRef] [Green Version]
  29. Aguiar Jr, P.N.; Santoro, I.L.; Tadokoro, H.; de Lima Lopes, G.; Filardi, B.A.; Oliveira, P.; Mountzios, G.; De Mello, R.A. The role of PD-L1 expression as a predictive biomarker in advanced non-small-cell lung cancer: A network meta-analysis. Immunotherapy 2016, 8, 479–488. [Google Scholar] [CrossRef]
  30. Qin, T.; Zeng, Y.-D.; Qin, G.; Xu, F.; Lu, J.-B.; Fang, W.-F.; Xue, C.; Zhan, J.-H.; Zhang, X.-K.; Zheng, Q.-F.; et al. High PD-L1 expression was associated with poor prognosis in 870 Chinese patients with breast cancer. Oncotarget 2015, 6, 33972. [Google Scholar] [CrossRef] [Green Version]
  31. Tamura, T.; Ohira, M.; Tanaka, H.; Muguruma, K.; Toyokawa, T.; Kubo, N.; Sakurai, K.; Amano, R.; Kimura, K.; Shibutani, M. Programmed death-1 ligand-1 (PDL1) expression is associated with the prognosis of patients with stage II/III gastric cancer. Anticancer Res. 2015, 35, 5369–5376. [Google Scholar]
  32. Yu, D.; Liu, X.; Han, G.; Liu, Y.; Zhao, X.; Wang, D.; Bian, X.; Gu, T.; Wen, L. The let-7 family of microRNAs suppresses immune evasion in head and neck squamous cell carcinoma by promoting PD-L1 degradation. Cell Commun. Signal. 2019, 17, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Uccini, S.; Al-Jadiry, M.F.; Pepe, G.; Scarpino, S.; Al-Hadad, S.A.; Ruco, L. PD-L1 expression in pediatric Epstein-Barr virus positive classic Hodgkin lymphoma is not associated with 9p24. 1 amplification. Pediatr. Blood Cancer 2019, 66, e27757. [Google Scholar]
  34. Koganemaru, S.; Inoshita, N.; Miura, Y.; Miyama, Y.; Fukui, Y.; Ozaki, Y.; Tomizawa, K.; Hanaoka, Y.; Toda, S.; Suyama, K. Prognostic value of programmed death-ligand 1 expression in patients with stage III colorectal cancer. Cancer Sci. 2017, 108, 853–858. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Wei, X.-L.; Wu, Q.-N.; Chen, D.-L.; Zeng, Z.-L.; Lu, J.-B.; Liu, Z.-X.; Ju, H.-Q.; Ren, C.; Pan, Z.-Z.; Wang, F.-H. The clinical and biomarker association of programmed death ligand 1 and its spatial heterogeneous expression in colorectal cancer. J. Cancer 2018, 9, 4325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Valentini, A.M.; Di Pinto, F.; Cariola, F.; Guerra, V.; Giannelli, G.; Caruso, M.L.; Pirrelli, M. PD-L1 expression in colorectal cancer defines three subsets of tumor immune microenvironments. Oncotarget 2018, 9, 8584. [Google Scholar] [CrossRef]
  37. Wang, L.; Ren, F.; Wang, Q.; Baldridge, L.A.; Monn, M.F.; Fisher, K.W.; Sheng, W.; Zhou, X.; Du, X.; Cheng, L. Significance of programmed death ligand 1 (PD-L1) immunohistochemical expression in colorectal cancer. Mol. Diagn. Ther. 2016, 20, 175–181. [Google Scholar] [CrossRef]
  38. Hamada, T.; Cao, Y.; Qian, Z.R.; Masugi, Y.; Nowak, J.A.; Yang, J.; Song, M.; Mima, K.; Kosumi, K.; Liu, L. Aspirin use and colorectal cancer survival according to tumor CD274 (programmed cell death 1 ligand 1) expression status. J. Clin. Oncol. 2017, 35, 1836. [Google Scholar] [CrossRef]
  39. Wang, S.; Yuan, B.; Wang, Y.; Li, M.; Liu, X.; Cao, J.; Li, C.; Hu, J. Clinicopathological and prognostic significance of PD-L1 expression in colorectal cancer: A meta-analysis. Int. J. Colorectal Dis. 2020, 36, 117–130. [Google Scholar] [CrossRef]
  40. Ni, X.; Sun, X.; Wang, D.; Chen, Y.; Zhang, Y.; Li, W.; Wang, L.; Suo, J. The clinicopathological and prognostic value of programmed death-ligand 1 in colorectal cancer: A meta-analysis. Clin. Transl. Oncol. 2019, 21, 674–686. [Google Scholar] [CrossRef]
  41. Marcus, L.; Lemery, S.J.; Keegan, P.; Pazdur, R. FDA approval summary: Pembrolizumab for the treatment of microsatellite instability-high solid tumors. Clin. Cancer Res. 2019, 25, 3753–3758. [Google Scholar] [CrossRef] [Green Version]
  42. Sahin, I.H.; Akce, M.; Alese, O.; Shaib, W.; Lesinski, G.B.; El-Rayes, B.; Wu, C. Immune checkpoint inhibitors for the treatment of MSI-H/MMR-D colorectal cancer and a perspective on resistance mechanisms. Br. J. Cancer 2019, 121, 809–818. [Google Scholar] [CrossRef] [PubMed]
  43. Rosenbaum, M.W.; Bledsoe, J.R.; Morales-Oyarvide, V.; Huynh, T.G.; Mino-Kenudson, M. PD-L1 expression in colorectal cancer is associated with microsatellite instability, BRAF mutation, medullary morphology and cytotoxic tumor-infiltrating lymphocytes. Mod. Pathol. 2016, 29, 1104–1112. [Google Scholar] [CrossRef] [PubMed]
  44. Howitt, B.E.; Shukla, S.A.; Sholl, L.M.; Ritterhouse, L.L.; Watkins, J.C.; Rodig, S.; Stover, E.; Strickland, K.C.; D’Andrea, A.D.; Wu, C.J. Association of polymerase e–mutated and microsatellite-instable endometrial cancers with neoantigen load, number of tumor-infiltrating lymphocytes, and expression of PD-1 and PD-L1. JAMA Oncol. 2015, 1, 1319–1323. [Google Scholar] [CrossRef]
  45. Overman, M.J.; Lonardi, S.; Wong, K.Y.M.; Lenz, H.-J.; Gelsomino, F.; Aglietta, M.; Morse, M.A.; Van Cutsem, E.; McDermott, R.; Hill, A. Durable clinical benefit with nivolumab plus ipilimumab in DNA mismatch repair-deficient/microsatellite instability-high metastatic colorectal cancer. J. Clin. Oncol. 2018, 36, 773–779. [Google Scholar] [CrossRef] [PubMed]
  46. Azad, N.S.; Gray, R.J.; Overman, M.J.; Schoenfeld, J.D.; Mitchell, E.P.; Zwiebel, J.A.; Sharon, E.; Streicher, H.; Li, S.; McShane, L.M. Nivolumab is effective in mismatch repair–deficient noncolorectal cancers: Results from arm Z1D—A subprotocol of the NCI-MATCH (EAY131) study. J. Clin. Oncol. 2020, 38, 214–222. [Google Scholar] [CrossRef]
  47. Mardis, E.R. Neoantigens and genome instability: Impact on immunogenomic phenotypes and immunotherapy response. Genome Med. 2019, 11, 71. [Google Scholar] [CrossRef] [PubMed]
  48. Ilieva, K.M.; Correa, I.; Josephs, D.H.; Karagiannis, P.; Egbuniwe, I.U.; Cafferkey, M.J.; Spicer, J.F.; Harries, M.; Nestle, F.O.; Lacy, K.E. Effects of BRAF mutations and BRAF inhibition on immune responses to melanoma. Mol. Cancer Ther. 2014, 13, 2769–2783. [Google Scholar] [CrossRef] [Green Version]
  49. André, T.; Lonardi, S.; Wong, M.; Leone, F.; McDermott, R.S.; Morse, M.A.; Neyns, B.; Leach, J.L.; Alfonso, P.G.; Lee, J.J.; et al. Nivolumab + ipilimumab combination in patients with DNA mismatch repair-deficient/microsatellite instability-high metastatic colorectal cancer: First report of the full cohort from CheckMate-142. Paper Presented at 2018 Gastrointestinal Cancers Symposium, San Francisco, CA, USA,, 20 January 2018. Abstract 553. [Google Scholar]
  50. Broos, K.; Keyaerts, M.; Lecocq, Q.; Renmans, D.; Nguyen, T.; Escors, D.; Liston, A.; Raes, G.; Breckpot, K.; Devoogdt, N. Non-invasive assessment of murine PD-L1 levels in syngeneic tumor models by nuclear imaging with nanobody tracers. Oncotarget 2017, 8, 41932. [Google Scholar] [CrossRef] [Green Version]
  51. Broos, K.; Lecocq, Q.; Raes, G.; Devoogdt, N.; Keyaerts, M.; Breckpot, K. Noninvasive imaging of the PD-1: PD-L1 immune checkpoint: Embracing nuclear medicine for the benefit of personalized immunotherapy. Theranostics 2018, 8, 3559. [Google Scholar] [CrossRef]
  52. Niemeijer, A.; Leung, D.; Huisman, M.; Bahce, I.; Hoekstra, O.; Van Dongen, G.; Boellaard, R.; Du, S.; Hayes, W.; Smith, R. Whole body PD-1 and PD-L1 positron emission tomography in patients with non-small-cell lung cancer. Nat. Commun. 2018, 9, 1–5. [Google Scholar] [CrossRef]
Figure 1. Programmed death ligand-1 (PD-L1) immunohistochemical staining in colorectal cancer (CRC) tissue microarray (TMA). Representative examples of tumors showing (A) positive membranous expression and (B) negative membranous expression of PD-L1 in CRC tissues. (20×/0.70 objective on an Olympus BX 51 microscope. (Olympus America Inc., Center Valley, PA, USA) with the inset showing a 40× 0.85 aperture magnified view of the same TMA spot). Brown color represents PD-L1 staining and blue color represents absence of PD-L1 staining.
Figure 1. Programmed death ligand-1 (PD-L1) immunohistochemical staining in colorectal cancer (CRC) tissue microarray (TMA). Representative examples of tumors showing (A) positive membranous expression and (B) negative membranous expression of PD-L1 in CRC tissues. (20×/0.70 objective on an Olympus BX 51 microscope. (Olympus America Inc., Center Valley, PA, USA) with the inset showing a 40× 0.85 aperture magnified view of the same TMA spot). Brown color represents PD-L1 staining and blue color represents absence of PD-L1 staining.
Jpm 11 00073 g001
Figure 2. Survival analysis of PD-L1 protein expression. Kaplan–Meier survival plot showing statistically significant poor overall survival in PD-L1 positive cases compared to PD-L1 low expression (p = 0.0017).
Figure 2. Survival analysis of PD-L1 protein expression. Kaplan–Meier survival plot showing statistically significant poor overall survival in PD-L1 positive cases compared to PD-L1 low expression (p = 0.0017).
Jpm 11 00073 g002
Table 1. Clinico-pathological variables for the patient cohort (n = 1148).
Table 1. Clinico-pathological variables for the patient cohort (n = 1148).
Clinico-Pathological Parametern (%)
Age
Median56.3
Range (IQR)47.5–67.9
Gender
Male599 (52.2)
Female549 (47.8)
Histological subtype
Adenocarcinoma1019 (88.8)
Mucinous carcinoma128 (11.2)
Histological grade
Well differentiated109 (9.5)
Moderately differentiated905 (78.8)
Poorly differentiated109 (9.5)
Unknown25 (2.2)
Tumor site
Left914 (79.7)
Right214 (18.6)
Unknown20 (1.7)
TNM Stage
I148 (12.9)
II370 (32.2)
III433 (37.7)
IV143 (12.5)
Unknown54 (4.7)
MMR protein expression status
dMMR106 (9.2)
pMMR1042 (90.8)
BRAF mutation
Present34 (3.0)
Absent1092 (95.1)
Unknown22 (1.9)
Neoadjuvant therapy
Yes447 (34.6)
No701 (65.4)
IQR: inter quartile range, dMMR: deficient mismatch repair, pMMR: proficient mismatch repair.
Table 2. Correlation of PD-L1 IHC expression with clinico-pathological parameters in colorectal carcinoma.
Table 2. Correlation of PD-L1 IHC expression with clinico-pathological parameters in colorectal carcinoma.
Clinico-Pathological ParametersTotalPD-L1 PositivePD-L1 Negativep Value
n%n%n%
Total Number of Cases1148 42837.372062.7
Age (years)
≤5037332.515441.321958.70.0523
>5077567.527435.450164.6
Sex
Male59952.221736.238263.80.4400
Female54947.821138.433861.6
Tumor Site
Left colon91481.033837.057663.00.6238
Right colon21419.08338.813161.2
Histological Type
Adenocarcinoma101988.836135.465864.60.0005
Mucinous Carcinoma12811.26651.66248.4
pT
T1353.21131.42468.60.0007
T216915.55733.711266.3
T376570.326434.550165.5
T411911.06453.85546.2
pN
N055250.819234.836065.20.2790
N133230.611935.821364.2
N220218.68341.111958.9
pM
M095587.034135.761464.30.1804
M114313.05941.38458.7
Tumor Stage
I14813.55134.59765.50.4309
II37033.812634.124465.9
III43339.616237.427162.6
IV14313.15941.38458.7
Differentiation
Well differentiated1099.62321.18678.9<0.0001
Moderate differentiated90580.632435.858164.2
Poor differentiated1099.76559.64440.4
MMR IHC
dMMR1069.25148.15551.90.0169
pMMR104290.837736.266563.8
BRAF mutation
Present343.02264.71235.30.0008
Absent109297.039135.870164.2
Overall Survival
5 Years 66.1 74.90.0017
dMMR: deficient mismatch repair, pMMR: proficient mismatch repair, pT: pathologic tumor size, pN: pathologic lymph node metastasis, pM: pathologic distant metastasis.
Table 3. Univariate and Multivariate analysis of PD-L1 expression using Cox Proportional Hazard Model.
Table 3. Univariate and Multivariate analysis of PD-L1 expression using Cox Proportional Hazard Model.
Clinico-Pathological ParametersUnivariateMultivariate
Hazard Ratio (95 % CI)p ValueHazard Ratio (95% CI)p Value
Age (years)
>50 (vs. ≤50)
1.06 (0.81–1.41)0.66480.99 (0.72–1.38)0.9801
Sex
Male (vs Female)
1.00 (0.77–1.31)0.97231.11 (0.81–1.50)0.5183
Stage
IV (vs. I–III)
5.10 (3.81–6.80)<0.00015.72 (4.17–7.84)<0.0001
Grade
3 (vs. 1–2)
1.68 (1.15–2.38)0.00841.76 (1.16–2.68)0.0083
Site
Left (vs. Right)
0.77 (0.49–1.16)0.22370.79 (0.50–1.25)0.3147
MMR IHC
dMMR (vs. pMMR)
0.87 (0.53–1.35)0.55140.58 (0.31–0.99)0.0483
PD-L1 expression
Positive (vs. negative)
1.53 (1.17–1.99)0.00211.45 (1.06–1.99)0.0200
dMMR: deficient mismatch repair, pMMR: proficient mismatch repair.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Siraj, A.K.; Parvathareddy, S.K.; Annaiyappanaidu, P.; Haqawi, W.; Al-Rasheed, M.; AlManea, H.M.; AlHussaini, H.F.; Al-Dayel, F.; Al-Kuraya, K.S. PD-L1 Expression Is Associated with Deficient Mismatch Repair and Poor Prognosis in Middle Eastern Colorectal Cancers. J. Pers. Med. 2021, 11, 73. https://0-doi-org.brum.beds.ac.uk/10.3390/jpm11020073

AMA Style

Siraj AK, Parvathareddy SK, Annaiyappanaidu P, Haqawi W, Al-Rasheed M, AlManea HM, AlHussaini HF, Al-Dayel F, Al-Kuraya KS. PD-L1 Expression Is Associated with Deficient Mismatch Repair and Poor Prognosis in Middle Eastern Colorectal Cancers. Journal of Personalized Medicine. 2021; 11(2):73. https://0-doi-org.brum.beds.ac.uk/10.3390/jpm11020073

Chicago/Turabian Style

Siraj, Abdul K., Sandeep Kumar Parvathareddy, Padmanaban Annaiyappanaidu, Wael Haqawi, Maha Al-Rasheed, Hadeel M. AlManea, Hussah F. AlHussaini, Fouad Al-Dayel, and Khawla S. Al-Kuraya. 2021. "PD-L1 Expression Is Associated with Deficient Mismatch Repair and Poor Prognosis in Middle Eastern Colorectal Cancers" Journal of Personalized Medicine 11, no. 2: 73. https://0-doi-org.brum.beds.ac.uk/10.3390/jpm11020073

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop