GET THE APP

Relationship of Metabolic Alterations and PD-L1 Expression in Cis
Cell & Developmental Biology

Cell & Developmental Biology
Open Access

ISSN: 2168-9296

+44 1478 350008

Review Article - (2017) Volume 6, Issue 2

Relationship of Metabolic Alterations and PD-L1 Expression in Cisplatin Resistant Lung Cancer

Wangpaichitr M1,2, Kandemir H3, Li YY4, Wu C1, Nguyen DJM5, Feun LG5, Kuo MT6 and Savaraj N1,4
1Miami VA Healthcare System, Research Service, Miami, Florida, USA, E-mail: mwangpaichitr@miami.edu
2Department of Surgery, Cardiothoracic Surgery, University of Miami, Miami, Florida, USA, E-mail: mwangpaichitr@miami.edu
3School of Medicine, Koc University, Istanbul, Turkey, E-mail: mwangpaichitr@miami.edu
4Department of Medicine, Hematology/Oncology, University of Miami, Miami, Florida, USA, E-mail: mwangpaichitr@miami.edu
5Department of Microbiology, University of Miami, Miami, Florida, USA, E-mail: mwangpaichitr@miami.edu
6Department of Translational Molecular Pathology, Texas MD Anderson, Houston, Texas, USA, E-mail: mwangpaichitr@miami.edu

Abstract

Despite numerous reports on immune checkpoint inhibitor for the treatment of non-small cell lung cancer (NSCLC), the response rate remains low but durable. Thus cisplatin still plays a major role in the treatment of NSCLC. While there are many mechanisms involved in cisplatin resistance, alteration in metabolic phenotypes with elevated levels of reactive oxygen species (ROS) are found in several cisplatin resistant tumors. These resistant cells become more reliant on mitochondria oxidative metabolism instead of glucose. Consequently, high ROS and metabolic alteration contributed to epithelial-mesenchymal transition (EMT). Importantly, recent findings indicated that EMT has a crucial role in upregulating PD-L1 expression in cancer cells. Thus, it is very likely that cisplatin resistance will lead to high expression of PD-L1/PD-1 which makes them vulnerable to anti PD-1 or anti PD-L1 antibody treatment. An understanding of the interactions between cancer cells metabolic reprogramming and immune checkpoints is critical for combining metabolism targeted therapies with immunotherapies.

<

Keywords: Lung cancer; PD-L1; EMT; Resistance; Cisplatin

Introduction

Treatment for early stage lung cancer is surgery but most patients already have locally advanced or metastatic disease at the time of diagnosis. Chemotherapy combined with radiation therapy or chemotherapy alone remains the primary modality of treatment for stage 3 and 4 disease. Targeted agents such as erlotinib or gefitinib (EGFR inhibitor) or crizotinib or ceritinib (ALK inhibitors) have shown activity in NSCLC (non-small cell lung cancer) which possess these putative types of mutation. However, both EGFR mutation and ALK mutation are rare (only 5-20%) and usually occur in women and nonsmokers. Immunotherapy with checkpoint inhibitors has received much attention lately. They offer a longer duration of response; however, the response rate is still very low in lung cancer. In fact, a recent report on PD1 inhibitor (programmed death-1) did not show improved efficacy over standard chemotherapy as first line treatment in lung cancer and did not receive FDA approval as first line therapy for NSCLC. Another checkpoint inhibitor pembrolizumab has received FDA approval for first line treatment but only in tumors which express PD-L1 (program death receptor ligand-1). Therefore, platinum containing regimen remains the first line treatment in patient with NSCLC. Despite a 50% initial response rate to platinum-based chemotherapy, the majority of lung cancer patients develop resistance to treatment. Thus, cisplatin resistance remains the major impediment for the treatment of lung cancer.

Accumulating evidence suggests that tumor metabolism is in fact interconnected to drug resistance and it has proven to be one of the most important challenges in cancer treatment [1-3]. The observations of metabolic differences in cancer cells were first reported by Otto Warburg [4,5]. He showed that cancer cells prefer to utilize glucose even in the presence of oxygen; hence this led to the term “aerobic glycolysis”. This difference in energy metabolism between tumor and normal tissue has been utilized successfully in the development of a diagnostic imaging technique, fluoro-deoxy-glucose positron emission tomography (FDGPET) for cancer detection. However, what is not known is why certain tumors are PET-negative (not taking up FDG), and why PET negativity does not always correlate with tumor response. Thus, it is conceivable that PET negative’s tumors have undergone metabolic reprogramming after chemotherapy and are no longer addicted to glucose. To further support this notion, it has been shown that therapy-resistant tumors have altered metabolic phenotypes relative to treatment-naive tumors, with increased reliance on mitochondrial metabolism in the resistant cancers [6-9]. Increased mitochondrial metabolic activity can lead to high levels of reactive oxygen species (ROS) [10]. In fact, many have discovered that elevated reactive oxygen species (ROS) are found in cisplatin resistant (CR) cell lines including those derived from patients who failed cisplatin [11-14].

ROS, a harmful by-product of metabolism played an important role in signaling pathways. ROS is known to facilitate the activation of receptor tyrosine kinase signaling as well as PI3K/ AKT which plays a vital role in cell growth/proliferation, survival, and motility [15,16]. Moreover, during the past decade, elevate ROS level in tumor cells have been implicated in epithelial-mesenchymal transition (EMT) [17-19]. Importantly, recent reports have shown that EMT played an essential role in upregulating PD-L1 (programmed death ligand-1) expression [20].

In this review, we provide a possible link between metabolic alteration and PD-L1 expression in cisplatin resistant lung cancer (Figure 1). Understanding these complex interrelationships will provide a new approach in overcoming the cisplatin resistance in lung cancer.

cell-developmental-cisplatin-results

Figure 1: Acquired resistance to cisplatin results in accumulation of cellular ROS. Increased ROS levels involved in metabolic reprogramming by switching cisplatin resistant cells from glycolysis toward oxidative metabolism and triggers epithelial-mesenchymal transition (EMT). Furthermore, induction of EMT may lead to increase in PD-L1 expression in tumor cells.

Cancer cells and their carbon sources

It is known that most if not all tumors utilize glycolysis instead of oxidative phosphorylation (OXPHOS) (4, 5). This is due to upregulation of glycolytic enzymes and glucose transporters [21,22]. In fact, increased glucose uptake is one of the hallmarks for malignant transformation [23,24]. Recently, it has been shown that up-regulation of pyruvate kinase-M2 (PKM2), an enzyme in the glycolytic pathway which converts phosphoenolpyruvate (PEP) to pyruvate, could be an answer for the aerobic glycolysis observed in Warburg’s theory. PKM2 is a key protein in directing tumor cells toward glycolysis [25]. PKM2 increases the DNA binding of HIF1α. Consequently, increases in HIF1α target gene expression. Cells expressing high levels of PKM2 are known to consume less oxygen and produce more lactate [25]. On the other hand, reduced PKM2 activity allows accumulation of glucose-6-phosphate and thus shifts glucose flux toward the pentose phosphate pathway (PPP) to generate reduced NADPH. Consistent with this notion, acute increases in intracellular concentrations of ROS caused inhibition of the glycolytic enzyme pyruvate kinase M2 (PKM2) through oxidation of Cys358 in lung cancer cells [26].

Although increased glucose metabolism in cancer cell has been recognized as the main carbon skeleton source of energy, we and others [27] have shown that cisplatin resistant (CR) cells are no longer addicted to the glycolytic pathway [11,28]. CR cells use other carbon sources to replenish TCA cycle intermediates (anaplerosis) for their energy demand and biosynthesis. In this regard, reports have shown that certain tumor types are highly addicted to glutamine [29,30]. The cellular demand in these tumors outstrips its glutamine supply; hence glutamine becomes the conditionally essential amino acid. Moreover, studies have shown that reduction in lactate dehydrogenase A (LDHA) expression in cancer cells either by genetic knock down (shRNA) or inhibitor (FX11) resulted in the shift to oxidative phosphorylation (OXPHOS) and increased intracellular ROS [31,32]. Therefore, there is no doubt that alteration in metabolism has gained its status as a core hallmark of cancer.

Redox and oxidative metabolism (OXMET)

A number of investigators have shown that cisplatin can inhibit thioredoxin reductase (TrxR) which leads to increased ROS. As a result, DNA damage occurs which can lead to cell death [33,34]. In order to adapt and survive at higher ROS levels and to evade cell death caused by cisplatin, CR cells use less thioredoxin-1(TRX1) and employ other antioxidant systems to compensate for the lack of TRX1 [35,37]. In fact, many reports showed that CR cells have higher level of glutathione (GSH) proteins [12,38,39]. Lower intracellular TRX1 also was not due to the protein degradation caused by cathepsin-D [40], but as a consequence of increased TRX1 secretion. TRX1 is secreted via special secretory pathway called “leaderless pathway”. This pathway is known to secrete low molecular weight proteins which lack signal peptide [41-43]. The mechanism of how this pathway functions remains poorly understood. Nevertheless, increased TRX1 secretion usually occurs when cells are under stress [44-46] and is found in patients who received cisplatin treatment [47,48]. In fact, many investigators have reported that higher serum TRX1 resulted in bad prognosis and drug resistance [49,51]. Decreased intracellular TRX1 has also been shown to reprogram lung cancer cells to become more reliant on oxidative metabolism (OXMET) [11] and overexpression of Txnip, an inhibitor of TRX1, can lead to adipogensis [52]. Thus, these findings could have future implication for drug development to selectively kill CR cells that have high ROS and low TRX1 levels.

ROS triggers epithelial-mesenchymal transition (EMT) in cancer cells

One of the first studies that established a direct connection between ROS and EMT was reported in the cross-talk signaling between ROS and TGF-β [53]. TGF-β stimulated ROS production was responsible for E-cadherin repression [53]. EMT related molecular events can also be stimulated by H2O2 treatment. It was noteworthy that the crucial event in EMT is represented by the disassembly of the epithelial structure and thus, E-cadherin down regulation was the most relevant step [19,54]. Several transcription factors contributed to this event for allowing E-cadherin repression. One of the important factors was the two zinc fingers E-box binding homeobox transcription factor ZEB1 and ZEB2 [55]. While ZEB1 is known to repress T lymphocytes IL2 gene expression , ZEB2 (previously known as SMADIP1, SIP1) can activate TGFβ. These repressors collaborate with histone deacetylases and histone demethylases, ensuring the maintenance of the silenced state of the E-cadherin gene [56].

The relationship between EMT and PD-L1 regulations

EMT is known as a driving force for metastasis and drug resistance. The presence of EMT signifies poor prognosis in many tumors including NSCLC [57]. Another factor which dictates tumor cells behavior is the immune cells in the tumor microenvironment. Studies have been carried out in lung adenocarcinoma which showed an increase in inflammatory signal cytokines such as IFN gamma and immune checkpoints markers including PD-L1/2, PD1, TIM3, B7H3, BTLA, LAG3 and CTLA4 [57]. Correlation between EMT and PD-L1 expression [58-60] is illustrated in Table 1. Furthermore, induction of EMT increased immunosuppressive cytokines and increased immunosuppressive CD8+ tumor infiltrating lymphocytes in preclinical models of lung, melanoma, pancreatic cancer, and breast cancer [61- 63]. Importantly, microRNA200 and ZEB1 axis, which is known to control cancer cell migration/invasion and EMT, can also regulate PD-L1 expression. Decrease in PD-L1 expressions was reported as a consequence of ectopic microRNA 200 expression or ZEB1 knockdown models [20]. In fact, low microRNA200 with high ZEB1 and PD-L1 expressions in mesenchymal tumors created a microenvironment of decreased CD8+ T-cells populations [20].

Head and Neck
Cancer
N=50
PD-L1 positive
64%
Associate with EMT (Low
E-cadherin expressions
p=0.010
Ock et al.
2016
Lung
Adenocarcinoma
N=220
PD-L1 positive
42%
Associate with EMT (Low
E-cadherin expressions)
p<0.001
Shimoji
et al.
2016
Lung
Adenocarcinoma
N=477
PD-L1 positive
60%
Associate with EMT (High
vimentin expressions)
p<0.01
Kim et al.
2016

Table 1: Correlations between PD-L1 expression and EMT [58-60].

PD-L1, a ligand of PD1 is an immune regulatory protein deriving from B7 family of T-cell co-regulatory molecules [64]. Their interaction prevents T-cell activation and proliferation including cell apoptosis and creates cancer resistance. So far, PD-L1 was found in many solid neoplasms such as cancer of the breast, colon, esophagus, stomach, ovaries, pancreas and lung [64]. As a prognostic marker, PD-L1 expression is a poor prognostic factor for gastric cancer, liver cancer, esophageal cancer, ovarian cancer, bladder cancer, but served as a better prognostic factor for breast and merkel cell carcinoma [65], while remains controversial in melanoma and lung cancer. Many reports suggest that EGFR and KRAS mutations contribute to increase in PD-L1 expression; however, the molecular mechanism behind this important biochemical event remains to be elucidated [66,67].

Drug resistance and PD-L1 expression

Recent studies have shown that treatment with cisplatin, carboplatin, paclitaxel, and 5-FU contribute to acquired PD-L1 expression in many solid tumors including small cell lung cancer (SCLC) and NSCLC [64,68,69]. Knocking-down PD-L1 was able to overcome cisplatin resistance. Further investigations also supported PD-L1 as the main resistance mechanism against cisplatin in SCLC via the over expression of DNMT1 or KIT. Down-regulation of these two proteins showed less PD-L1 expression and could overcome cisplatin resistance in H60 and H82 cell lines [69]. Thus, targeting the cellular PD-L1 may hypersensitize aggressive lung cancer to standard chemotherapy.

Concluding Remarks

Immunotherapy with checkpoint inhibitors has received much attention lately. This type of therapy offers a longer duration of response; however, the response rate is still low in lung cancer. In fact, a recent report on immunotherapy did not show improved efficacy over standard chemotherapy and failed as first line treatment in lung cancer. Therefore, the majority of lung cancer patients still require the traditional chemotherapeutic agents such as cisplatin or carboplatin to control their disease. We have found that the major biochemical alterations in cisplatin resistance are increasing ROS and metabolic reprograming which can be used to kill cisplatin resistant cells. Furthermore, the tumor microenvironment may also be modified in these resistant tumors by multiple factors including immune cells such as tumor-infiltrating lymphocyte. These resistant cells undergo epithelialmesenchymal transition to enable invasion/metastasis as well as escape immune surveillance by expressing PD-L1/PD1. Combination of ROS inducing agent with immunomodulation approach may ultimately lead to cisplatin resistant cell death (Figure 2).

cell-developmental-immune-system

Figure 2: Cancers cells adapt and exploit immune system to evade immune surveillance by activating PD-L1/PD1 axis. ZEB1 and microRNA200 can regulate this axis. KRAS or EGFR mutation may also influence PD-L 1 expression. Blocking PD1 and PD-L1 interaction with checkpoint inhibitor(s) in combination with ROS inducing agent may lead to new approaches to overcome cisplatin resistant lung cancer.

Acknowledgement

We regret that we were unable to cite many excellent studies that shaped our understanding of cancer metabolism, EMT, and PD-L1. This work is supported by BLR&D Career Development Award-2 (1K2BX001289) to Wangpaichitr M. and NIH/NCI (1R01CA149260) to Kuo MT. and Savaraj N. Woman’s Cancer Association of the University of Miami award to Savaraj N.

References

  1. Zhao Y, Butler EB, Tan M (2013) Targeting cellular metabolism to improve cancer therapeutics. Cell Death Dis 4: e532.
  2. Kroemer G, Pouyssegur J (2008) Tumor cell metabolism: Cancer's Achilles' heel. Cancer Cell 13: 472-482.
  3. Munoz-Pinedo C, El Mjiyad N, and Ricci JE (2012) Cancer metabolism: Current perspectives and future directions. Cell Death Dis 3: e248.
  4. Warburg O (1956) On respiratory impairment in cancer cells. Science 124: 269-270.
  5. Ippolito L, Marini A, Cavallini L, Morandi A, Pietrovito L, et al. (2016) Metabolic shift toward oxidative phosphorylation in docetaxel resistant prostate cancer cells. Oncotarget 7: 61890-61904.
  6. Zhang G, Frederick DT, Wu L, Wei Z, Krepler C, et al. (2016) Targeting mitochondrial biogenesis to overcome drug resistance to MAPK inhibitors. J Clin Invest 126: 1834-1856.
  7. Viale A, Pettazzoni P, Lyssiotis CA, Ying H, Sanchez N, et al. (2014) 514: 628-632.
  8. Vazquez F, Lim JH, Chim H, Bhalla K, Girnun G, et al. (2013) PGC1alpha expression defines a subset of human melanoma tumors with increased mitochondrial capacity and resistance to oxidative stress. Cancer Cell 23: 287-301.
  9. Liou GY, Storz P (2010) Reactive oxygen species in cancer. Free Radic Res 44: 479-496.
  10. Wangpaichitr M, Sullivan EJ, Theodoropoulos G, Wu C, You M, et al. (2012) The relationship of thioredoxin-1 and cisplatin resistance: Its impact on ROS and oxidative metabolism in lung cancer cells. Mol Cancer Ther 11: 604-615.
  11. Wangpaichitr M, Wu C, You M, Maher JC, Dinh V, et al. (2009) N1,N3-Dimethyl-N1,N3-bis(phenylcarbonothioyl) Propanedihydrazide (Elesclomol) selectively kills cisplatin resistant lung cancer cells through reactive oxygen species (ROS). Cancers 1: 23-28.
  12. Chen J, Solomides C, Parekh H, Simpkins F, Simpkins H (2015) Cisplatin resistance in human cervical, ovarian and lung cancer cells. Cancer Chemother Pharmacol 75: 1217-1227.
  13. Marullo R, Werner E, Degtyareva N, Moore B, Altavilla G, et al. (2013) Cisplatin induces a mitochondrial-ROS response that contributes to cytotoxicity depending on mitochondrial redox status and bioenergetic functions. PLoS ONE 8: e81162.
  14. Wang Y, Zeigler MM, Lam GK, Hunter MG, Eubank TD, et al. (2007) The role of the NADPH oxidase complex, p38 MAPK and Akt in regulating human monocyte/macrophage survival. Am J Respir Cell Mol Biol 36: 68-77.
  15. Pan MH, Ho CT (2008) Chemopreventive effects of natural dietary compounds on cancer development. Chem Soc Rev 37: 2558-2574.
  16. Cichon MA, Radisky DC (2014) ROS-induced epithelial-mesenchymal transition in mammary epithelial cells is mediated by NF-kB-dependent activation of snail. Oncotarget 5: 2827-2838.
  17. Giannoni E, Parri M, Chiarugi P (2012) EMT and oxidative stress: A bidirectional interplay affecting tumor malignancy. Antioxid Redox Signal 16: 1248-1263.
  18. Cannito S, Novo E, di Bonzo LV, Busletta C, Colombatto S, et al. (2010) Epithelial-mesenchymal transition: from molecular mechanisms, redox regulation to implications in human health and disease. Antioxid Redox Signal 12: 1383-1430.
  19. Chen L, Gibbons DL, Goswami S, Cortez MA, Ahn YH, et al. (2014) Metastasis is regulated via microRNA-200/ZEB1 axis control of tumour cell PD-L1 expression and intratumoral immunosuppression. Nat Commun 5: 5241.
  20. Kurtoglu M, Maher JC, Lampidis TJ (2007) Differential toxic mechanisms of 2-deoxy-D-glucose versus 2-fluorodeoxy-D-glucose in hypoxic and normoxic tumor cells. Antioxid Redox Signal 9: 1383-1390.
  21. Geschwind JF, Georgiades CS, Ko YH, Pedersen PL, (2014) Recently elucidated energy catabolism pathways provide opportunities for novel treatments in hepatocellular carcinoma. Expert Rev Anticancer Ther 4: 449-457.
  22. Garber K (2006) Energy deregulation: Licensing tumors to grow. Science 312: 1158-1159.
  23. Hanahan D, Weinberg RA (2011) Hallmarks of cancer: The next generation. Cell 144: 646-674.
  24. Vander Heiden MG, Locasale JW, Swanson KD, Sharfi H, Heffron GJ, et al. (2010) Evidence for an alternative glycolytic pathway in rapidly proliferating cells. Science 329: 1492-1499.
  25. Anastasiou D, Poulogiannis G, Asara JM, Boxer MB, Jiang JK, et al. (2011) Inhibition of pyruvate kinase M2 by reactive oxygen species contributes to cellular antioxidant responses. Science 334: 1278-1283.
  26. Sullivan EJ, Kurtoglu M, Brenneman R, Liu H, Lampidis TJ (2014) Targeting cisplatin-resistant human tumor cells with metabolic inhibitors. Cancer Chemother Pharmacol 73: 417-427.
  27. Wangpaichitr M, Wu CJ, Li Y, Nguyen DJM, Shah S, et al. (2017) Exploiting ROS and metabolic differences to kill cisplatin resistant lung cancer oncotarget.
  28. Wise DR, Thompson CB (2010) Glutamine addiction: A new therapeutic target in cancer. Trends Biochem Sci 35: 427-433.
  29. DeBerardinis RJ, Mancuso A, Daikhin E, Nissim I, Yudkoff M, et al. (2007) Beyond aerobic glycolysis: transformed cells can engage in glutamine metabolism that exceeds the requirement for protein and nucleotide synthesis. Proc Natl Acad Sci USA 104: 19345-19350.
  30. Le A, Cooper CR, Gouw AM, Dinavahi R, Maitra A, et al. (2010) Inhibition of lactate dehydrogenase A induces oxidative stress and inhibits tumor progression. Proc Natl Acad Sci USA 107: 2037-2042.
  31. Arseneault R, Chien A, Newington JT, Rappon T, Harris R, et al. (2013) Attenuation of LDHA expression in cancer cells leads to redox-dependent alterations in cytoskeletal structure and cell migration. Cancer Lett 338: 255-266.
  32. Saitoh M, Nishitoh H, Fujii M, Takeda K, Tobiume K, et al. (1998) Mammalian thioredoxin is a direct inhibitor of apoptosis signal-regulating kinase (ASK) 1. Embo J 17: 2596-2606.
  33. Sun Y, Rigas B (2008) The thioredoxin system mediates redox-induced cell death in human colon cancer cells: implications for the mechanism of action of anticancer agents. Cancer Res 68: 8269-8277.
  34. Witte AB, Anestal K, Jerremalm E, Ehrsson H, Arner ES (2005) Inhibition of thioredoxin reductase but not of glutathione reductase by the major classes of alkylating and platinum-containing anticancer compounds. Free Radic Biol Med 39: 696-703.
  35. Mostert V, Hill KE, Burk RF (2003) Loss of activity of the selenoenzyme thioredoxin reductase causes induction of hepatic heme oxygenase-1. FEBS Lett 541: 85-88.
  36. Lu Y, Cederbaum A (2007) The mode of cisplatin-induced cell death in CYP2E1-overexpressing HepG2 cells: modulation by ERK, ROS, glutathione and thioredoxin. Free Radic Biol Med 43: 1061-1075.
  37. Okuno S, Sato H, Kuriyama-Matsumura K, Tamba M, Wang H, et al. (2003) Role of cystine transport in intracellular glutathione level and cisplatin resistance in human ovarian cancer cell lines. Br J Cancer 88: 951-956.
  38. Godwin AK, Meister A, O'Dwyer PJ, Huang CS, Hamilton TC, et al. (1992) High resistance to cisplatin in human ovarian cancer cell lines is associated with marked increase of glutathione synthesis. Proc Natl Acad Sci USA 89: 3070-3074.
  39. Haendeler J, Popp R, Goy C, Tischler V, Zeiher AM, et al. (2005) Cathepsin D and H2O2 stimulate degradation of thioredoxin-1: implication for endothelial cell apoptosis. J Biol Chem 280: 42945-42951.
  40. Rubartelli A, Bajetto A, Allavena G, Wollman E, Sitia R (1992) Secretion of thioredoxin by normal and neoplastic cells through a leaderless secretory pathway. J Biol Chem 267: 24161-24164.
  41. Rubartelli A, Bajetto A, Bonifaci N, Di Blas E, Solito E, et al. (1993) A novel way to get out of the cell. Cytotechnology 11 Suppl 1: S37-40.
  42. Rubartelli A, Bonifaci N, Sitia R (1995) High rates of thioredoxin secretion correlates with growth arrest in hepatoma cells. Cancer Res 55: 675-680.
  43. Miyamoto S, Kawano H, Sakamoto T, Soejima H, Kajiwara I, et al. (2004) Increased plasma levels of thioredoxin in patients with coronary spastic angina. Antioxid Redox Signal 6: 75-80.
  44. Nakamura H, De Rosa S, Roederer M, Anderson MT, Dubs JG, et al. (1996) Elevation of plasma thioredoxin levels in HIV-infected individuals. Int Immunol 8: 603-611.
  45. Nakamura H, Vaage J, Valen G, Padilla CA, Bjornstedt M, et al. (1998) Measurements of plasma glutaredoxin and thioredoxin in healthy volunteers and during open-heart surgery. Free Radic Biol Med 24: 1176-1186.
  46. Sasada T, Nakamura H, Ueda S, Iwata S, Ueno M, et al. (2008) Secretion of thioredoxin enhances cellular resistance to cis-diamminedichloroplatinum (II). Antioxid Redox Signal 2: 695-705.
  47. Sasada T, Nakamura H, Ueda S, Sato N, Kitaoka Y, et al. (1999) Possible involvement of thioredoxin reductase as well as thioredoxin in cellular sensitivity to cis-diamminedichloroplatinum (II). Free Radic Biol Med 27: 504-514.
  48. Park BJ, Cha MK, Kim IH (2014) Thioredoxin 1 as a serum marker for ovarian cancer and its use in combination with CA125 for improving the sensitivity of ovarian cancer diagnoses. Biomarkers 19: 604-610.
  49. Yokomizo A, Ono M, Nanri H, Makino Y, Ohga T, et al. (1995) Cellular levels of thioredoxin associated with drug sensitivity to cisplatin, mitomycin C, doxorubicin and etoposide. Cancer Res 55: 4293-4296.
  50. Kim SJ, Miyoshi Y, Taguchi T, Tamaki Y, Nakamura H, et al. (2005) High thioredoxin expression is associated with resistance to docetaxel in primary breast cancer. Clin Cancer Res 11: 8425-8430
  51. Chutkow WA, Lee RT (2011) Thioredoxin regulates adipogenesis through thioredoxin interacting protein (Txnip) protein stability. J Biol Chem.
  52. Rhyu DY, Yang Y, Ha H, Lee GT, Song JS, et al. (2005) Role of reactive oxygen species in TGF-beta1-induced mitogen-activated protein kinase activation and epithelial-mesenchymal transition in renal tubular epithelial cells. J Am Soc Nephrol 16: 667-675.
  53. Kalluri R, Weinberg RA (2009) The basics of epithelial-mesenchymal transition. J Clin Invest 119: 1420-1428.
  54. Gregory PA, Bert AG, Paterson EL, Barry SC, Tsykin A, et al. (2008) The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat Cell Biol 10: 593-601.
  55. Peinado H, Olmeda D, Cano A (2007) Snail, Zeb and bHLH factors in tumour progression: an alliance against the epithelial phenotype? Nature reviews 7: 415-428.
  56. Lou Y, Diao L, Cuentas ER, Denning WL, Chen L, et al. (2016) Epithelial-mesenchymal transition is associated with a distinct tumor microenvironment including elevation of inflammatory signals and multiple immune checkpoints in lung adenocarcinoma. Clin Cancer Res 22: 3630-3642.
  57. Ock CY, Kim S, Keam B, Kim M, Kim TM, et al. (2016) PD-L1 expression is associated with epithelial-mesenchymal transition in head and neck squamous cell carcinoma. Oncotarget 7: 15901-15914.
  58. Shimoji M, Shimizu S, Sato K, Suda K, Kobayashi Y, et al. (2016) Clinical and pathologic features of lung cancer expressing programmed cell death ligand 1 (PD-L1). Lung cancer (Amsterdam, Netherlands) 98: 69-75.
  59. Kim S, Koh J, Kim MY, Kwon D, Go H, et al. (2016) PD-L1 expression is associated with epithelial-to-mesenchymal transition in adenocarcinoma of the lung. Hum Pathol 58: 7-14.
  60. Kudo-Saito C, Shirako H, Takeuchi T, Kawakami Y (2009) Cancer metastasis is accelerated through immunosuppression during Snail-induced EMT of cancer cells. Cancer Cell 15: 195-206.
  61. Lu H, Knutson KL, Gad E, Disis ML (2006) The tumor antigen repertoire identified in tumor-bearing neu transgenic mice predicts human tumor antigens. Cancer Res 66: 9754-9761.
  62. Kudo-Saito C, Shirako H, Ohike M, Tsukamoto N, Kawakami Y (2013) CCL2 is critical for immunosuppression to promote cancer metastasis. Clin Exp Metastasis 30: 393-405.
  63. Van Der Kraak L, Goel G, Ramanan K, Kaltenmeier C, Zhang L, et al. (2016) 5-Fluorouracil upregulates cell surface B7-H1 (PD-L1) expression in gastrointestinal cancers. J Immunother Cancer 4: 65.
  64. Wang X, Teng F, Kong L, Yu J (2016) PD-L1 expression in human cancers and its association with clinical outcomes. Onco Targets Ther 9: 5023-5039.
  65. Ji M, Liu Y, Li Q, Li XD, Zhao WQ, et al. (2015) PD-1/PD-L1 pathway in non-small-cell lung cancer and its relation with EGFR mutation. J Transl Med 13: 5.
  66. Gatalica Z, Snyder C, Maney T, Ghazalpour A, Holterman DA, et al. (2014) Programmed cell death 1 (PD-1) and its ligand (PD-L1) in common cancers and their correlation with molecular cancer type. Cancer Epidemiol Biomarkers Prev 23: 2965-2970.
  67. Gong W, Song Q, Lu X, Gong W, Zhao J, et al. (2011) Paclitaxel induced B7-H1 expression in cancer cells via the MAPK pathway. J Chemother 23: 295-299.
  68. Yan F, Pang J, Peng Y, Molina JR, Yang P, et al. (2016) Elevated cellular PD1/PD-L1 expression confers acquired resistance to cisplatin in small cell lung cancer cells. PLoS ONE 11: e0162925.
Citation: Wangpaichitr M, Kandemir H, Li YY, Wu C, Nguyen DJM, et al. (2017) Relationship of Metabolic Alterations and PD-L1 Expression in Cisplatin Resistant Lung Cancer. Cell Dev Biol 6:183.

Copyright: © 2017 Wangpaichitr M, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top