eISSN: 2084-9869
ISSN: 1233-9687
Polish Journal of Pathology
Current issue Archive Manuscripts accepted About the journal Supplements Editorial board Abstracting and indexing Subscription Contact Instructions for authors Publication charge Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
Share:
Share:
Original paper

Association of PD-1 and PD-L1 protein expression with selected clinical and morphological parameters in colorectal cancers

Paulina Poter
1, 2, 3
,
Sylwia Jankowska-Szabłowska
3
,
Tomasz Kolenda
4, 5
,
Ewa Dobak
3
,
Elżbieta Urasińska
3
,
Katarzyna Karpińska-Łukaszewicz
3

  1. Department of Tumor Pathology, Greater Poland Cancer Centre, Poznań, Poland
  2. Department of Oncologic Pathology and Prophylaxis, University of Medical Sciences, Poznań, Poland
  3. Department of Pathology, Pomeranian Medical University, Szczecin, Poland
  4. Laboratory of Cancer Genetics, Greater Poland Cancer Centre, Poznań, Poland
  5. Research and Implementation Unit, Greater Poland Cancer Centre, Poznań, Poland
Pol J Pathol 2024; 75 (2)
Online publish date: 2024/05/27
Article file
- Association of PD-1.pdf  [0.40 MB]
Get citation
 
PlumX metrics:
 

Introduction

Colorectal cancer is the third most common cancer worldwide and the second cause of death from malignant tumors [1–3]. Colorectal cancers, depending on their clinical stage, are treated with surgery, chemotherapy and personalized therapy – gene therapy and immunotherapy [4–6]. Cancer cells have developed mechanisms that allow them to avoid the body’s defense reaction [7, 8]. It is possible through a number of mechanisms, including the use of inhibitory pathways at key immune points. One such point is the PD-1/PD-L1 pathway [9, 10]. In the treatment of many types of cancer, the use of the PD-1/PDL1 pathway is a therapeutic breakthrough [6]. PD-1 (programmed cell death protein 1), also known as CD279 [11], is a membrane protein found on the surface of T lymphocytes, involved in the process of cell apoptosis [12, 13]. PD-L1 is a ligand of the PD-1 protein – a key immunoregulatory molecule that suppresses the cytotoxic immune response of CD8-positive lymphocytes [14, 15]. Cancer cells and antigen-presenting cells express the PD-L1 ligand, as well as the second ligand of the PD-1 receptor, which is the PD-L2 protein [9, 16, 17]. The PD-1 and PD-L1 proteins are critical immune checkpoint proteins that are responsible for the negative regulation of the integrity and stability of the immune function of T lymphocytes [18–20].
Expression of PD-L1 ligand leads to the escape of tumor cells from immune control systems [21–23]. Anti-PD-1 and anti-PD-L1 monoclonal antibodies block the binding of PD-1 protein to its ligand (Fig. 1). Studies have shown that blockade of the PD-1/PD-L1 checkpoint using specific antibodies restores the anti-cancer activity of cytotoxic T-cells specific for the cancer antigen, which allows for therapeutic actions in patients with colorectal cancer [24]. The aim of this study was to evaluate the association of immunohistochemical expression of PD-1 and PD-L1 proteins with selected clinical and morphological parameters in patients with colorectal cancer and their survival.

Material and methods

The study included a group of 98 patients with a histopathologically confirmed diagnosis of colorectal cancer. Histological examinations of patients operated on in 2017 at the Department of General and Oncological Surgery and the Department of General and Gastroenterological Surgery of the Pomeranian Medical University in Szczecin were performed at the Department of Pathomorphology of the Pomeranian Medical University in Szczecin. The study design was approved by the local ethics committee of the university hospital. The survival data were obtained from the death register by the Systems Management Department of the Ministry of Digitization of the Republic of Poland. The clinical and morphological characteristics of the study group are presented in Table 1.
The technique of tissue microarray (TMA) made of paraffin blocks containing material collected for routine histopathological examination from a postoperative preparation fixed in 10% formalin and embedded in paraffin was used in the study [25–27]. Representative regions from the primary histopathological specimen stained with hematoxylin and eosin were taken. Then the manual tissue arrayer (Beecher Instruments, Silver Spring, MD, USA) was used to prepare TMA. The histological preparations of intestinal tumors were also evaluated for the presence of tumor budding and the presence of lymphocytes penetrating the tumor tissue and infiltrating the tumor area (TIL) in 1 mm2. In the case of TIL, “+” was defined as present (> 30%) and “–” as absence of TIL (< 30%) [28], while the presence of tumor budding was defined as “+” when there were single tumor cells or small clusters of up to 5 cells in the stroma at the periphery of the tumor and “–” as the absence of this phenomenon [29]. In addition, in 28 patients whose KRAS gene status was assessed for diagnostic purposes, the relationship between KRAS gene status and the expression of PD-1 and PD-L1 proteins in cancer cells and in TIL was examined. To evaluate the expression of PD-1 and PD-L1 proteins, immunohistochemical staining was performed using monoclonal antibodies against the mentioned proteins. A mouse monoclonal antibody (NAT105; Roche Diagnostics Poland; catalog no. 07099029001) was used to assess PD-1 receptor expression. For the evaluation of PD-L1 ligand expression, a rabbit monoclonal antibody against PD-L1 (VENTANA SP263; Roche Diagnostics Polska; catalog no. 07419821001) was used. Those antibodies were optimally diluted to a form compatible with VENTANA detection kits and BenchMark devices (Roche Diagnostics Polska).
The presence of PD-1 protein expression in lymphocytes and PD-L1 in cancer cells and/or lymphocytes in stained immunohistochemical slides was first evaluated by an experienced pathomorphologist, and then virtual image analysis was used to standardize and unify the results. The slides were scanned using an APERIO CS scanner (Aperio Technologies Inc. California, USA). For image analysis, software that applies staining evaluation algorithms directly to the scanned image (Image Scope Version 11.2.0.780; Aperio Technologies, Inc. 2003–2012) was used.
In the case of a cytoplasmic reaction, the cytoplasmic v2 algorithm allows for assessment of the percentage of cells expressing a given protein, as well as for assessing the intensity of staining of these cells (lack of reaction (0), weak reaction (1+), medium intensity reaction (2+), strong reaction (3+)). In the case of the PD-1 protein, the reaction was evaluated in lymphocytes infiltrating the tumor tissue (Fig. 2). The expression of a given protein was evaluated according to the H-score [30, 31]: H-score = 1  (% of 1+ cells) + 2  (% of 2+ cells) + 3  (% of 3+ cells).
The score has a range of 0–300.
For the membrane reaction, we used the membrane v9 algorithm designed to assess HER2 receptor status, which is also recommended to assess the expression of other proteins in the cell membrane. This algorithm, like the previous one, allows for assessment of the percentage of cells expressing a given protein, as well as for assessing the intensity of staining of these cells (0, 1+, 2+, 3+). In the case of PD-L1 protein, expression was assessed in the membrane of cancer cells (Fig. 3A) and lymphocytes infiltrating the tumor tissue (Fig. 3B). For further analysis, the H-score formula described earlier was used.
The normality of the distributions of all variables was checked by the Shapiro-Wilk test. These variables were described by means, standard deviations, medians, and minimum and maximum values. The statistical differences between the two groups were checked with the Mann-Whitney U test. The Kruskal-Wallis test was used to compare more than two groups. Spearman’s rank correlation was used to test the correlation between variables. The results were described by the correlation coefficient r and the probability of p. Survival analysis was prepared using the log-rank (Mantel-Cox) test (pa) and Gehan-Breslow-Wilcoxon test (pb). All tests were performed using GraphPad Prism 8 software. The statistically significant differences in all the tests performed were those for which the probability was lower than 0.05.

Results

Immunohistochemical expression of PD-1 and PD-L1 proteins
Expression of PD-1 protein, which was studied in lymphocytes accompanying tumor infiltration in the intestinal wall, was found in 29 (29.59%) of the cases studied. PD-L1 ligand expression was studied in lymphocytes, as well as in colon cancer cells. PD-L1 protein expression was found in lymphocytes in 43 (43.88%) cases, and in cancer cells in 7 (7.14%) cases (Table 2).
Immunohistochemical expression of PD-1 and PD-L1 proteins and selected clinical and morphological parameters
The results are shown in Tables 3 and 4.
Immunohistochemical expression of PD-1 and PD-L1 proteins and patient survival
It was found that the expression of the PD-1 protein had a statistically significant impact on the patient survival in the first year after the diagnosis – 100% survival in patients with expression of the PD-1 protein (p = 0.018). However, there was no statistically significant relationship between PD-1 protein expression and patient survival at 5 years after diagnosis. There was no statistically significant relationship between patient survival and PD-L1 protein expression in either cancer cells or lymphocytes. These data are presented in Figures 4 and 5.

Discussion

The interactions of PD-1 and PD-L1 involve immunosuppressive processes in the tumor growth environment. In the last decade, monoclonal antibody therapy (anti-PD-1/anti-PD-L1) has been introduced to block the pro-tumor activity of this checkpoint. The use of PD-1/PD-L1 inhibitors is expected to block the anti-tumor activity of this complex [32, 33]. The expression of PD-L1 protein on the surface of cancer cells and PD-1 protein in lymphocytes accompanying the cancer infiltrate is an important predictive indicator for anti-PD-1 and anti-PD-L1 antibody therapy [34]. However, a clinical response to anti-PD-1/PD-L1 therapy is observed in only a minority of patients. Targeting DNA synthesis and replication through chemotherapy results in the elimination of cancer cells, while blocking the PD-1/PD-L1 checkpoint stimulates tumor-specific T-cells. This is because cell death is followed by an increase in the presence of tumor antigens, which stimulates lymphocytes [35]. Thus, taking this process into account, it seems that an appropriate combination of chemotherapeutic treatment with PD-1/PD-L1 inhibitors may lead to an increase in treatment efficacy especially in patients with less immunogenic and chemotherapy- sensitive tumors [18]. In addition, studies indicate that the use of PD-1 and PD-L1 inhibitors combined with radiotherapy in patients with advanced non-small cell lung cancer (NSCLC) improves both overall survival and recurrence-free survival. Geng et al. found that the use of PD-1/PD-L1 inhibitors after radiotherapy was more beneficial than concurrent PD-1/PD-L1 inhibitor therapy with radiotherapy or the use of radiotherapy after PD-1/PD-L1 inhibitors [36]. This result is in line with the theory that radiotherapy causes double-stranded DNA breaks and increases CD8+ T-cell infiltration, which in turn increases PD-L1 protein expression [36–39].
The aim of the present study was to compare the expression of PD-1 and PD-L1 proteins in patients with colorectal cancer with clinical and morphological parameters, KRAS gene status, tumor budding, and patient’s survival. An immunohistochemical method was used to evaluate the expression of PD-1 and PD-L1 proteins.
A similar study related to PD-L1 protein expression in colorectal cancer was conducted by Shan et al., demonstrating immunohistochemically the presence of expression in 42.5% of colorectal cancer cases studied. The authors found a positive correlation of PD-L1 expression with the occurrence of lymph node metastasis, distant metastasis, and the depth of tumor infiltration. However, there was no correlation with gender or the degree of histological differentiation of the tumor [40], which in turn were found in the present study. On the other hand, Masugi and his team analyzed the occurrence of PD-L1 protein expression in both tumor cells and the stroma, where 89% and 5% of cases, respectively, showed expression [41]. That is the opposite of the present study, where PD-L1 protein expression in cancer cells was observed in a lower percentage of cases than in lymphocytes, i.e. 7.1% of cases with PD-L1 expression in cancer cells and 43.9% of cases with expression in lymphocytes. Masugi studied the correlation of PD-L1 protein expression with the density of FOXP3 cells [41], which are regulatory T-cells that play an important role in inhibiting the anti-tumor response [42]. This relationship was found to be inversely related to PD-L1 protein expression in colorectal cancers, which may suggest an effect of PD-L1-expressing cancer cells on regulatory T-cells in the tumor microenvironment [41]. Zhao and his team also found a statistically significant correlation between regulatory T-cell infiltration and PD-L1 protein expression in cancer cells (expression was observed in 48.21% of cases, while PD-L1 expression was not observed in tumor infiltrating cells). An association of PD-L1 protein expression in cancer cells with the depth of tumor infiltration and the presence of lymph node metastasis was also found [42]. In the present study, PD-L1 protein expression in lymphocytes was demonstrated in 43.9% of cases. PD-L1 protein expression in lymphocytes was found to be related to clinical stage and the presence of lymph node metastasis. The higher the clinical stage was, the less frequent was the expression of PD-L1 protein in lymphocytes. Moreover, PD-L1 protein expression in lymphocytes was observed more frequently in patients who did not have lymph node metastases. In his study of PD-L1 protein expression, Masugi observed in colorectal cancers both a membranous and cytoplasmic reaction in the histochemical reaction [41], which is confirmed by other researchers [42–47]. The authors emphasized that evaluation of PD-L1 expression in tissue was challenging because there is no universally accepted immunohistochemical method to demonstrate this expression, and PD-L1 expression in the cell membrane is likely to be associated with the possibility of binding to the PD-1 receptor. Membrane expression may be masked in cells with strong cytoplasmic expression, so both cytoplasmic and membrane expression levels were used for calculations, and were evaluated independently by two specialists [41]. Also in the present study, a two-level evaluation was used – slides were evaluated by an experienced pathomorphologist, and then calculations were made using image analysis software to determine PD-L1 protein expression in cell membranes.
As reported previously, good therapeutic results with the drugs nivolumab and pembrolizumab, as well as atezolizumab, have been obtained in the treatment of NSCLC. Mansour et al. studied by immunohistochemistry both formalin-fixed and paraffin-embedded histologic and cytologic samples of lung cancer. In both variants, they detected PD-L1 protein expression in 55% of cases, but high PD-L1 expression in the tumor in histological specimens correlated with better response to treatment. Squamous cell carcinomas showed higher PD-L1 expression than adenocarcinomas.
There are reports on the association of KRAS and epidermal growth factor receptor (EGFR) gene status with PD-L1 expression. The highest PD-L1 expression occurred in cases with KRAS mutations, and the lowest for cases with EGFR mutations and in cases without mutations in both KRAS and EGFR [48]. In the case of colorectal cancer, KRAS gene status was one of the first biomarkers to be studied, and is now the primary criterion that qualifies colorectal cancer patients for treatment with the monoclonal antibodies cetuximab and panitumumab. These drugs target the EGFR receptor [49–52]. The efficacy of therapy directed against EGFR in patients without mutations in the KRAS gene is 8–11%. Mutations in the KRAS gene are detected in about 40% of patients with colorectal cancer [53–55]. It is therefore necessary to find new predictive biomarkers for patients who, despite having the wild-type KRAS gene, are resistant to treatment directed against the EGFR. In the present study, 28 patients with defined KRAS gene status were examined. The KRAS mutation was detected in half of the cases. However, none of the 28 patients, either with or without mutations in KRAS, had PD-L1 protein expression in colorectal cancer cells. The association of PD-L1 protein expression in lymphocytes with KRAS gene status was tested, but no statistical significance was found. A significant correlation was obtained by analyzing the association of KRAS gene status with PD-1 protein expression in lymphocytes – none of the cases with KRAS mutation showed PD-1 protein expression. However, the low abundance of samples tested for KRAS gene status should be kept in mind here.
Studies have reported that the clinical response rate in various types of cancer treated with PD-1/PD-L1 inhibitors is 30–50% [56]. Therefore, it seems that identifying predictive biomarkers to select patients and improve treatment efficacy without additional and unwarranted side effects is necessary to create targeted therapies for the individual patient. Immunotherapy, based on the use of immune checkpoint inhibitors, has revolutionized the treatment of various types of cancer in recent years. However, when it comes to colorectal cancer, currently only patients with either a stable DNA repair system or a high degree of microsatellite instability can benefit from such therapy, and they represent only 5% of patients with advanced colorectal cancer. Clinical trials are underway to evaluate treatment efficacy when PD-1/PD-L1 inhibitors are combined with other immune checkpoint inhibitors, as well as with chemotherapy, radiotherapy and molecularly targeted therapies [57–59]. The role of PD-L1 expression in colorectal cancer is not well defined, and published studies show inconsistent results regarding the association of PD-L1 expression with prognosis [60, 61]. In the present study, the low percentage of patients showing PD-L1 protein expression in cancer cells shows that the study should have been conducted on a larger number of patients, which would have provided more reliable results. Conclusions Despite a slight improvement in the survival of patients with colorectal cancer, about half of patients need new therapies. Unlike other malignancies, such as kidney cancer, lung cancer, or melanoma, colorectal cancer shows a very low response rate to PD-1 or PD-L1 protein blockade [62–64], but studies conducted on the PD-1/PD-L1 pathway indicate that it may be an important mechanism for cancer cells to escape from immune surveillance [65]. This leads us to believe that in the future, the therapeutic use of this pathway will become increasingly applicable in the treatment of colorectal cancer as well. However, the study of the association of PD-1 and PD-L1 protein expression with clinical, morphological, genetic and molecular parameters or prognosis of colorectal cancer requires further research.
The authors declare no conflict of interest.
References
1. Phipps O, Brookes MJ, Al-Hassi HO. Iron deficiency, immunology, and colorectal cancer. Nutr Rev 2021; 79: 88-97.
2. Dekker E, Tanis PJ, Vleugels JLA, et al. Colorectal cancer. Lancet 2019; 394: 1467-1480.
3. Keum N, Giovannucci E. Global burden of colorectal cancer: emerging trends, risk factors and prevention strategies. Nat Rev Gastroenterol Hepatol 2019; 16: 713-732.
4. Bosman FT, Bujko K, Chmielik E, et al. Wybrane zagadnienia z patomorfologii i patokliniki jelita grubego i odbytu. Pol J Pathol 2014; 65: 32-36.
5. Li J, Zhen L, Zhang Y, et al. Circ-104916 is downregulated in gas‑ tric cancer and suppresses migration and invasion of gastric cancer cells. Onco Targets Ther 2017; 10: 3521-3529.
6. Gao Y, Li SU, Xu D, et al. Prognostic value of programmed death-1, programmed death-ligand 1, programmed death- ligand 2 expression, and CD8 (+) T cell density in primary tumors and met‑ astatic lymph nodes from patients with stage T1-4N+ M0 gastric adenocarcinoma. Chin J Cancer 2017; 36: 61-74.
7. Wu C. Systemic therapy for colon cancer. Surg Oncol Clin N Am 2018; 27: 235-242.
8. Markowska A, Sajdak S, Lubin J, et. al. Znaczenie PD-1 – receptora programowanej śmierci-1 – i jego ligandów w immunoterapii raka jajnika. Curr Gynecol Oncol 2016; 14: 117-120.
9. Postow MA, Callahan MK, Wolchok JD. Immune checkpoint blockade in cancer therapy. J Clin Oncol 2015; 33: 1974-1982.
10. Welz JA. The promise of PD-1 pathway blockade in the treatment of metastatic cancer. MOP Magnifier 2014; 1: 1-7.
11. Lai CY, Tseng PC, Chen CL, et al. Different induction of PD-L1 (CD274) and PD-1 (CD279) expression in THP-1-differentiated types 1 and 2 macrophages. J Inflamm Res 2021; 14: 5241-5249.
12. Ishida Y, Agata Y, Shibahara K, et al. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J 1992; 11: 3887-3895.
13. Dong H, Zhu G, Tamada K, et al. B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion. Nat Med 1999; 5: 1365-1369.
14. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012; 12: 252-264.
15. Kalim M, Iqbal Khan MS, Zhan J. Programmed cell death ligand-1: a dynamic immune checkpoint in cancer therapy. Chem Biol Drug Des 2020; 95: 552-566.
16. Callahan MK, Postow MA, Wolchok JD. CTLA-4 and PD-1 pathway blockade: combinations in the clinic. Front Oncol 2015; 15: 385.
17. Cai J, Qi Q, Qian X, et al. The role of PD-1/PD-L1 axis and macrophage in the progression and treatment of cancer. J Cancer Res Clin Oncol 2019; 145: 1377-1385.
18. Ghosh C, Luong G, Sun Y. A snapshot of the PD-1/PD-L1 pathway. J Cancer 2021; 12: 2735-2746.
19. Sharpe AH, Pauken KE. The diverse functions of the PD1 inhibitory pathway. Nat Rev Immunol 2018; 18: 153-167.
20. Hui E, Cheung J, Zhu J, et al. T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science 2017; 355: 1428-1433.
21. Zou W, Chen L. Inhibitory B7-family molecules in the tumour microenvironment. Nat Rev Immunol 2008; 8: 467-477.
22. Willis BC, Sloan EA, Atkins KA, et al. Mismatch repair status and PDL1 expression in clear cell carcinomas of the ovary and endometrium. Mod Pathol 2017; 114: 106-107.
23. Gangling T, Boran C, Jingzhang L, et al. MACC1 regulates PDL1 expression and tumor immunity through the c-Met/AKT/mTOR pathway in gastric cancer cells. Cancer Med 2019; 8: 7044-7054.
24. Nardone V, Tini P, Pastina P, et al. Radiomics predicts survival of patients with advanced non‑small cell lung cancer undergoing PD‑1 blockade using nivolumab. Oncol Lett 2020; 19: 1559-1566.
25. Biunno I, Paiola E, De Blasio P. The application of the tissue microarray (TMA) technology to analyze cerebral organoids. J Histochem Cytochem 2021; 69: 451-460.
26. Koo M, Squires JM, Ying D, et al. Making a tissue microarray. Methods Mol Biol 2019; 1897: 313-323.
27. Glinsmann-Gibson B, Wisner L, Stanton M, et al. Recommendations for tissue microarray construction and quality assurance. Appl Immunohistochem Mol Morphol 2020; 28: 325-330.
28. Li Y, Liang L, Dai W, et al. 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.
29. Koelzer VH, Zlobec I, Lugli A. Tumor budding in colorectal cancer – ready for diagnostic practice? Human Pathology 2016; 47: 4-19.
30. Lewandowska M. Ekspresja epireguliny i amfireguliny w komórkach nowotworowych i w komórkach mikrośrodowiska raków jelita grubego uzyskanych za pomocą laserowej mikrodyssekcji. Pomorski Uniwersytet Medyczny, Szczecin 2016.
31. Hu WH, Chen HH, Yen SL, et al. Increased expression of interleukin-23 associated with progression of colorectal cancer. J Surg Oncol 2017; 115: 208-212. 
32. Lei Q, Wang D, Sun K, et al. Resistance mechanisms of anti- PD1/PDL1 therapy in solid tumors. Front Cell Dev Biol 2020; 8: 672.
33. Oliveira AF, Bretes L, Furtado I. Review of PD-1/PD-L1 inhibitors in metastatic dMMR/MSI-H colorectal cancer. Front Oncol 2019; 9: 396.
34. Krawczyk P, Wojas-Krawczyk K. Przeciwciała monoklonalne przeciw immunologicznym punktom kontroli w leczeniu chorych na nowotwory. Onkol Prakty Klin 2015; 11: 76-86.
35. Bracci L, Schiavoni G, Sistigu A, et al. Immune-based mechanisms of cytotoxic chemotherapy: implications for the design of novel and rationale-based combined treatments against cancer. Cell Death Differ 2014; 21: 15 -25.
36. Geng Y, Zhang Q, Feng S, et al. Safety and efficacy of PD-1/PD-L1 inhibitors combined with radiotherapy in patients with non-small-cell lung cancer: a systematic review and meta-analysis. Cancer Med 2021; 10: 1222-1239.
37. Sato H, Niimi A, Yasuhara T, et al. DNA double-strand break repair pathway regulates PD-L1 expression in cancer cells. Nat Commun 2017; 8: 1751.
38. Choe EA, Cha YJ, Kim JH, et al. Dynamic changes in PD-L1 expression and CD8+ T cell infiltration in non-small cell lung cancer following chemoradiation therapy. Lung Cancer 2019; 136: 30-36.
39. Chen S, Crabill GA, Pritchard TS, et al. Mechanisms regulating PD-L1 expression on tumor and immune cells. J Immunother Cancer 2019; 7: 305.
40. Shan T, Chen S, Wu T, et al. PD-L1 expression in colon cancer and its relationship with clinical prognosis. Int J Clin Exp Pathol 2019; 12: 1764-1769.
41. Masugi Y, Nishihara R, Yang J, et al. Tumour CD274 (PD-L1) expression and T cells in colorectal cancer. Gut 2017; 66: 1463-1473.
42. Zhao LW, Li C, Zhang RL, et al. B7-H1 and B7-H4 expression in colorectal carcinoma: correlation with tumor FOXP3(+) regulatory T-cell infiltration. Acta Histochem 2014; 116: 1163-1168.
43. Jeschke J, Bizet M, Desmedt C, et al. Dna methylation – based immune response signature improves patient diagnosis in multiple cancers. J Clin Invest 2017; 127: 3090-3102.
44. Le DT, Uram JN, Wang H, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med 2015; 372: 2509-2520.
45. Hua D, Sun J, Mao Y, et al. B7-H1 expression is associated with expansion of regulatory T cells in colorectal carcinoma. World J Gastroenterol 2012; 18: 971-978.
46. Liang M, Li J, Wang D, et al. T-cell infiltration and expressions of T lymphocyte co-inhibitory B7- H1 and B7-H4 molecules among colorectal cancer patients in northeast China’s Heilongjiang province. Tumour Biol 2014; 35: 55-60.
47. Song M, Chen D, Lu B, et al. PTEN loss increases PD-L1 protein expression and affects the correlation between PD-L1 expression and clinical parameters in colorectal cancer. PLoS One 2013; 8: e65821.
48. Mansour MSI, Malmros K, Mager U, et al. PD-L1 expression in non-small cell lung cancer specimens: association with clinicopathological factors and molecular alterations. Int J Mol Sci 2022; 23: 4517.
49. Lièvre A, Bachet JB, Le Corre D, et al. KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer. Cancer Res 2006; 66: 3992-3995.
50. Benvenuti S, Sartore-Bianchi A, Di Nicolantonio F, et al. Oncogenic activation of the RAS/RAF signaling pathway impairs the response of metastatic colorectal cancers to anti-epidermal growth factor receptor antibody therapies. Cancer Res 2007; 67: 2643-2648.
51. Karapetis CS, Khambata-Ford S, Jonker DJ, et al. K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N Engl J Med 2008; 359: 1757-1765.
52. Khambata-Ford S, Garrett CR, Meropol NJ, et al. Expression of epiregulin and amphiregulin and K-ras mutation status predict disease control in metastatic colorectal cancer patients treated with cetuximab. J Clin Oncol 2007; 25: 3230-3237.
53. Roth AD, Tejpar S, Delorenzi M, et al. Prognostic role of KRAS and BRAF in stage II and III resected colon cancer: results of the translational study on the PETACC-3, EORTC 40993, SAKK 60-00 trial. J Clin Oncol 2010; 28: 466-474.
54. Cunningham D, Humblet Y, Siena S, et al. Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer. N Engl J Med 2004; 351: 337-345.
55. Van Cutsem E, Peeters M, Siena S, et al. Open-label phase III trial of panitumumab plus best supportive care compared with best supportive care alone in patients with chemotherapy-refractory metastatic colorectal cancer. J Clin Oncol 2007; 25: 1658-1664.
56. Gandini S, Massi D, Mandala M. PD-L1 expression in cancer patients receiving anti PD-1/PD-L1 antibodies: a systematic review and meta-analysis. Crit Rev Oncol Hematol 2016; 100: 88-98.
57. Ooki A, Shinozaki E, Yamaguchi K. Immunotherapy in colorectal cancer: current and future strategies. J Anus Rectum Colon 2021; 5: 11-24.
58. Lee LH, Cavalcanti MS, Segal NH, et al. Patterns and prognostic relevance of PD-1 and PD-L1 expression in colorectal carcinoma. Mod Pathol 2016; 29: 1433-1442.
59. Rosenbaum MW, Bledsoe JR, Morales-Oyarvide V, et al. 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
60. Droeser RA, Hirt C, Viehl CT, et al. Clinical impact of programmed cell death ligand 1 expression in colorectal cancer. Eur J Cancer 2013; 49: 2233-2242.
61. Choueiri TK, Fay AP, Gray KP, et al. PD-L1 expression in nonclear-cell renal cell carcinoma. Ann Oncol 2014; 25: 2178-2184.
62. Topalian SL, Sznol M, McDermott DF, et al. Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab, J Clin Oncol 2014; 32: 1020-1030
63. Brahmer JR, Tykodi SS, Chow LQ, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 2012; 366: 2455-2465.
64. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012; 366: 2443-2454.
65. Grzywnowicz M, Giannopoulos K. Znaczenie receptora programowanej śmierci 1 oraz jego ligandów w układzie immunologicznym oraz nowotworach. Acta Haematol Pol 2012; 43: 132-145.
Copyright: © 2024 Polish Association of Pathologists and the Polish Branch of the International Academy of Pathology This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.