412
Views
6
CrossRef citations to date
0
Altmetric
Editorial

Pathogenic link between chronic obstructive pulmonary disease and squamous cell lung cancer

&
Pages 171-175 | Published online: 09 Jan 2014

Worldwide, lung cancer is the leading cause of death from cancer and tobacco smoking is associated with more than 90% of cases of lung cancer Citation[1]. Cigarette smoking is also the most important risk factor for chronic obstructive pulmonary disease (COPD) Citation[101]. Smoking-related lung diseases, such as COPD and lung cancer, are growing epidemics in women in the USA and elsewhere Citation[2]. Interestingly, COPD, a chronic inflammation of the lower airways, is also a major independent risk factor for lung carcinoma among long-term smokers Citation[3]. Lung cancer is also a leading cause of morbidity and mortality in patients with COPD Citation[4]. This evidence emerges clearly from a large longitudinal study of asymptomatic patients with mild-to-moderate COPD, where after 14.5 years follow-up, 33% of these subjects died of lung cancer Citation[4]. It is also known that smokers with COPD have a higher risk of developing a specific histological subtype of non-small-cell lung cancer (NSCLC) termed squamous cell carcinoma Citation[5,6]. Importantly, squamous cell carcinoma still represents the most common lung cancer histological subtype in European men Citation[7]. Despite significant advances in diagnostic techniques and understanding of the molecular biology, even the most recent therapeutic innovations for NSCLC have yielded little improvement in prognosis, with overall 5-year survival rates still less than 15% Citation[1,8]. Lung cancer and COPD therefore share a common risk factor – tobacco smoking – through which they may also share similar pathogenic mechanisms. However, urban and house air pollution is also recognized as a strong risk factor for both COPD and lung cancer Citation[9], and self-reported physician-diagnosed pulmonary emphysema represents a risk factor for lung cancer, even in lifelong nonsmokers Citation[10].

Since more than 90% of all cases of COPD and lung cancer are related to tobacco smoking and only a proportion of lifelong smokers will develop COPD and/or squamous cell carcinoma, we focus here on the potential pathogenic molecular link between tobacco smoking-related COPD and squamous cell carcinoma.

A well-defined stepwise progression of smoking-associated pathological premalignant changes (from basal cell hyperplasia to dysplasia and in situ squamous cell carcinoma) occurs in the normal bronchial epithelium before the development of an invasive squamous cell lung carcinoma Citation[11]. However, despite current data supporting a direct relationship between proximal airway basal progenitors and cells associated with carcinogenesis in murine models for human squamous cell lung carcinoma, the precise stem cell target of this process of carcinogenesis remains unknown Citation[12].

Molecular genetic studies of squamous cell lung carcinoma have revealed that clinically evident squamous cell lung cancers have multiple genetic and epigenetic abnormalities, including DNA sequence alterations, copy number changes and aberrant promoter hypermethylation Citation[13]. Together, these abnormalities result in the activation of oncogenes and inactivation of tumor-suppressor genes Citation[13]. In many cases these abnormalities can be found in premalignant lesions and histologically normal lung bronchial epithelial cells Citation[13].

It is conceivable that such a stepwise progression of the process of carcinogenesis could be facilitated in smokers who develop COPD owing to their impaired clearance of carcinogenic substances resulting from chronic airflow obstruction. Indeed, increased deposition of particulate matter in the major bronchi has been described recently in a computer model of the lungs of patients with COPD Citation[14].

On the basis of the recent novel molecular findings in this field, we believe that the explanation of the link between COPD and squamous cell lung carcinoma is probably more complex than a simple mechanistic one. In fact, there are also many possible molecular links between tobacco smoking-related COPD and squamous cell lung carcinoma.

The human lung is a major target organ for all inhaled carcinogens contained in tobacco smoke. Many of these compounds require enzymatic activation to exert their deleterious effects on pulmonary cells Citation[15]. Interindividual differences in in situ activation and inactivation of xenobiotics may contribute to the risk of developing both COPD and squamous cell lung carcinoma associated with these compounds. The major xenobiotic metabolizing enzymes, including both phase I cytochrome P450s (CYPs), epoxidehydrolases (EPHX), flavinmonooxygenases, such as hemeoxygenase (HMOX)1 and phase II enzymes (conjugation enzymes, including several transferases, such as glutathione S-transferases [GSTs]) are expressed in human lung tissues Citation[15]. The combination of several genetic polymorphisms in the enzymes that activate or detoxify the tobacco smoke carcinogens, such as EPHX, GST and HMOX1, might modulate the risk of chronic smokers developing both COPD and squamous cell lung carcinoma Citation[16].

Although cigarette smoke may be directly mutagenic, polymorphisms in the genes controlling acquired somatic mutations may also contribute, at least to some extent, to the observed differing susceptibilities to COPD and squamous cell lung carcinoma. Alterations in cell-cycle regulation and apoptosis leading to malignant transformation could be caused by common genetic variants in tumor-suppressor genes. A critical downstream mediator of the protein p53 is the tumor-suppressor gene p21, which is transcriptionally activated by p53 to induce cell-cycle arrest and DNA repair. One of the single-nucleotide polymorphisms in p21 results in an amino acid change in codon 31 from Ser to Arg. This polymorphism is located in a highly conserved region of p21 and is believed to affect its molecular function. The p21 Ser31Arg polymorphism is a moderate-risk allele for squamous cell carcinoma Citation[17]. Lee and colleagues, in a case–control study involving 206 Taiwanese subjects with COPD and 210 healthy smokers as control subjects, reported that the distribution frequencies of genotypes of p21WAP/CIP1 codon 31 were significantly different between the COPD and control groups Citation[18]. Furthermore, higher odds ratios (ORs) for COPD were seen for individuals with p21WAP/CIP1 R/R and R/S genotypes against S/S genotype (OR = 2.07) Citation[18]. p21WAP/CIP1 protein expression is increased in alveolar macrophages (a key cell in the pathogenesis of COPD Citation[19]) and bronchial epithelial cells of patients with COPD Citation[20] and, in vitro, hydrogen peroxide (an oxidative stress) induces cytoplasmic expression of p21WAP/CIP1 in airway epithelial cells and fails to induce their apoptosis Citation[20]. These data suggest that an increased expression of p21WAP/CIP1 in the lower airways caused by chronic exposure to tobacco smoking may represent another potential molecular link between COPD and squamous cell lung carcinoma.

COPD is recognized as a chronic inflammation of the lower airways Citation[101]. A causal relationship between inflammation and cancer had previously been proposed by Galen and later by Wirchow, who noticed the infiltration of leukocytes in malignant tissues and suggested that cancers arise at regions of chronic inflammation Citation[21]. How exactly chronic airway inflammation is linked to lung carcinogenesis is still not completely understood Citation[21,22]. However the transcription factor NF-κB could well be an important player in this process since it is activated in bronchial epithelium and in the inflammatory cells in the lower airways of COPD and in the premalignant lesions of the bronchial epithelium and neoplastic cells of squamous cell lung carcinoma Citation[23–25].

NF-κB activation and subsequent transactivation of inflammation-related genes may play a central role in both COPD and squamous cell lung carcinoma. Indeed, in addition to its tumor-promoting role, which depends on the stimulation of cell proliferation and inhibition of cell death, NF-κB may also participate in tumor initiation Citation[26]. For instance, NF-κB-activated macrophages in the bronchial tissues of COPD patients can release oxidants in the proximity of the basal bronchial epithelial cells to cause their DNA damage. Interestingly, in vitro inhibition of NF-κB using bortezomib or the compound BAY-11–7085 sensitizes squamous cell lung carcinoma cell lines to death induced by histone deacetylases inhibitors Citation[27,28].

Interestingly, in vitro, nicotine acting on nicotinic receptors may stimulate MAPK (p44/42) and Akt-dependent proliferation and NF-κB-dependent survival of squamous cell lung cancer cell lines, conferring a survival advantage to these cells Citation[29,30]. It also induces lung fibroblasts to produce fibronectin, a major component of the extracellular matrix Citation[31].

Normal human bronchial epithelium and squamous cell lung cancer cell lines are able to produce acetylcholine Citation[32,33], which has been shown to promote the proliferation of neoplastic cells acting on nicotinic receptors in vitroCitation[33] and the proliferation of lung fibroblasts and myofibroblasts acting on muscarinic receptors Citation[34]. There is a complete absence of studies on the potential effect of smoking or its components (such as nicotine) to modulate the release of acetylcholine from the non-neuronal cholinergic system in the airways. Notably, acetylcholinesterase activity in squamous cell carcinoma is 80% less than in its adjacent noncancerous tissue Citation[35].

Theoretically, an increased release of acetylcholine in the lung of smokers, via an autocrine or paracrine loop, may cause both small airway fibrosis (a characteristic of the COPD lung Citation[101]), thus contributing to the development of COPD, and enhanced cholinergic signaling on bronchial epithelium, contributing to carcinogenesis.

In this way, in long-term smokers, chronic nicotine inhalation and/or endogenous acetylcholine released locally in the lung might both play a role in COPD and squamous cell lung carcinoma development.

Tobacco smoking-related COPD is also associated with increased oxidative stress in the lower airways, which can cause DNA damage and carcinogenesis Citation[36]. Damage to DNA induces several cellular responses that enable cells either to eliminate or cope with the damage or to activate a programmed cell death process, presumably to eliminate cells with potentially catastrophic mutations Citation[37]. DNA repair mechanisms include direct, base excision, nucleotide excision, double-strand break and crosslink repair Citation[37]. A lack of DNA repair may be another common mechanism linking both COPD and squamous cell lung carcinoma Citation[38]. For example, one possibility is that heritable genetic polymorphisms influence the efficiency of both DNA repair damage in the bronchial epithelium and connective tissue damage repair. It is also likely that genetic factors modulating the quality of DNA repair explain the absence of both COPD and squamous cell lung cancer in most smokers Citation[38]. Several polymorphisms in DNA repair genes have already been reported to be associated with squamous cell lung carcinoma risk Citation[39]. Clearly, owing to the complexity and the inter-relationships of the many different pathways involved in the process of DNA repair in the lung, this area deserves to be fully explored in large ad hoc studies.

Recent molecular studies investigating gene expression in the bronchial epithelial cells of a control group of smokers with normal lung function compared with smokers with COPD using microarrays Citation[40] are very useful and provide novel clues on the possible molecular links between COPD and squamous cell carcinoma of the lung. Among others, the protein AKR1B10 appears to be of particular interest since it is overexpressed in the bronchial epithelium of COPD patients and in the neoplastic cells of squamous cell carcinoma of the lung Citation[40,41], suggesting its involvement in the multistep carcinogenesis of squamous cell carcinoma of the lung. Aldo–keto reductases are NADPH-dependent oxidoreductases that catalyze the reduction of a variety of carbonyl compounds. AKR1B10 is a member of this superfamily and reduces aromatic and aliphatic aldehyde substrates and has putative physiologic roles in steroid metabolism or detoxification of reactive aldehydes Citation[41]. Data suggest that AKR1B10 translocates to the nucleus during the cell cycle and is involved in the regulation of the cell cycle in a fashion yet to be identified. Another possibility is that AKR1B10 promotes carcinogenesis of squamous cell lung carcinoma through its enzymatic activity, which counteracts the conversion of β-carotene to retinoic acid Citation[41].

Many new compounds that target the molecular pathology of advanced squamous cell lung carcinoma are now undergoing clinical trials Citation[42]; however, we believe that we need to promote more studies on the molecular biology of smokers with premalignant bronchial lesions of the squamous cell lung carcinoma compared with a control group of smokers with and without COPD to unravel the complex molecular interactions between COPD and early squamous cell lung carcinoma. These studies may allow the discovery of new molecular targets of the early carcinogenesis process that, in the foreseeable future, may render the prevention of invasive squamous cell lung carcinoma a reality.

Financial & competing interests disclosure

Supported by teh Associazione per la Ricerca e la Cura dell’Asma (ARCA, Padova, Italy), Fondo per Ricerca Scientifica di Interesse Locale 2007 of the University of Ferrara (ex60%), GlaxoSmithKline (UK) and Novartis (UK). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

No writing assistance was utilized in the production of this manuscript.

References

  • Parkin DM, Bray F, Ferlay J et al. Global cancer statistics, 2002. CA. Cancer J. Clin.55(2), 74–108 (2005).
  • Ben-Zaken O, Cohen S, Parè D et al. The growing burden of chronic obstructive pulmonary disease and lung cancer in women: examining sex differences in cigarette smoke metabolism. Am. J. Respir. Crit. Care Med.176(2), 113–120 (2007).
  • Mannino DM, Aguayo SM, Petty TL et al. Low lung function and incident lung cancer in the United States: data from the First National Health and Nutrition Examination Survey follow-up. Arch. Intern. Med.163(12), 1475–1480 (2003).
  • Anthonisen NR, Skeans MA, Wise RA et al. The effects of a smoking cessation intervention on 14.5-year mortality: a randomized clinical trial. Ann. Intern. Med.142(4), 233–239 (2005).
  • Malhotra S, Lam S, Man SF et al. The relationship between stage 1 and 2 non-small cell lung cancer and lung function in men and women. BMC Pulm. Med.6(2), 1–6 (2006).
  • Papi A, Casoni G, Caramori G et al. COPD increases the risk of squamous histological subtype in smokers who develop non-small-cell lung cancer. Thorax59(8), 679–681 (2004).
  • Janssen-Heijnen ML, Coebergh JW. Trends in incidence and prognosis of the histological subtypes of lung cancer in North America, Australia, New Zealand and Europe. Lung Cancer31(2–3), 123–137 (2001).
  • Carney DN. Lung cancer – time to move on from chemotherapy. N. Engl. J. Med.346(2), 126–128 (2002).
  • Zhang JJ, Smith KR. Household air pollution from coal and biomass fuels in china: measurements, health impacts, and interventions. Environ. Health Perspect.115(6), 848–855 (2007).
  • Turner MC, Chen Y, Krewski D et al. COPD associated with lung cancer mortality in a prospective study of never smokers. Am. J. Respir. Crit. Care Med.176(3), 285–290 (2007)
  • Auerbach O, Hammond EC, Garfinkel L. Changes in bronchial epithelium in relation to cigarette smoking, 1955–1960 vs. 1970–1977. N. Engl. J. Med.300(24), 381–385 (1979).
  • Giangreco A, Groot KR, Janes SM. Lung cancer and lung stem cells: strange bedfellows? Am. J. Respir. Crit. Care Med.175(6), 547–553 (2007).
  • Sato A, Shames DS, Gazdar AF et al. A translational view of the molecular pathogenesis of lung cancer. J. Thorac. Oncol.2(4), 327–343 (2007).
  • Segal RA, Martonen TB, Kim CS et al. Computer simulations of particle deposition in the lungs of chronic obstructive pulmonary disease patients. Inhal. Toxicol.14(7), 705–720 (2002).
  • Zhang JY, Wang Y, Prakash C. Xenobiotic-metabolizing enzymes in human lung. Curr. Drug Metab.7(8), 939–948 (2006).
  • Caramori G, Adcock IM. Gene–environment interactions in the development of chronic obstructive pulmonary disease. Curr. Opin. Allergy Clin. Immunol.6(5), 323–338 (2006).
  • Popanda O, Edler L, Waas P et al. Elevated risk of squamous-cell carcinoma of the lung in heavy smokers carrying the variant alleles of the TP53 Arg72Pro and p21 Ser31Arg polymorphisms. Lung Cancer55(1), 25–34 (2007).
  • Lee YL, Chen W, Tsai WK et al. Polymorphisms of p53 and p21 genes in chronic obstructive pulmonary disease. J. Lab. Clin. Med.147(5), 228–233 (2006).
  • Barnes PJ. Alveolar macrophages as orchestrators of COPD. COPD1(1), 59–70 (2004).
  • Tomita K, Caramori G, Lim S et al. Increased p21CIP1/WAF1 and bcl-xL expression and reduced apoptosis in alveolar macrophages from smokers. Am. J. Respir. Crit. Care Med.166(3), 724–731 (2002).
  • Balkwill F, Mantovani A. Inflammation and cancer: back to Virchow? Lancet357(9255), 539–545 (2001).
  • Ballaz S, Mulshine JL. The potential contributions of chronic inflammation to lung carcinogenesis. Clin. Lung Cancer5(1), 46–62 (2003).
  • Di Stefano A, Caramori G, Oates T et al. Increased expression of NF-κB in bronchial biopsies from smokers and patients with chronic obstructive pulmonary disease (COPD). Eur. Resp. J.20(3), 556–563 (2002).
  • Caramori G, Adcock IM, Ito K. Anti-inflammatory inhibitors of IκB kinase in asthma and COPD. Curr. Opin. Investig. Drugs5(11), 1141–1147 (2004).
  • Tang X, Liu D, Shishodia S et al. Nuclear factor-κB (NF-κB) is frequently expressed in lung cancer and preneoplastic lesions. Cancer107(11), 2637–2646 (2006).
  • Greten FR, Karin M. The IKK/NF-κB activation pathway-a target for prevention and treatment of cancer. Cancer Lett.206(2), 193–199 (2004).
  • Denlinger CE, Keller MD, Mayo MW et al. Combined proteasome and histone deacetylase inhibition in non-small cell lung cancer. J. Thorac. Cardiovasc. Surg.127(4), 1078–1086 (2004).
  • Rundall BK, Denlinger CE, Jones DR. Combined histone deacetylase and NF-κB inhibition sensitizes non-small cell lung cancer to cell death. Surgery136(2), 416–425 (2004).
  • Carlisle DL, Liu X, Hopkins TM et al. Nicotine activates cell-signaling pathways through muscle-type and neuronal nicotinic acetylcholine receptors in non-small cell lung cancer cells. Pulm. Pharmacol. Ther. (2006) (Epub ahead of print).
  • Tsurutani J, Castillo SS, Brognard J et al. Tobacco components stimulate Akt-dependent proliferation and NFκB-dependent survival in lung cancer cells. Carcinogenesis26(7), 1182–1195 (2005).
  • Roman J, Ritzenthaler JD, Gil-Acosta A et al. Nicotine and fibronectin expression in lung fibroblasts: implications for tobacco-related lung tissue remodeling. FASEB J.18(12), 1436–1438 (2004).
  • Racke K, Matthiesen S. The airway cholinergic system: physiology and pharmacology. Pulm. Pharmacol. Ther.17(4), 181–198 (2004).
  • Song P, Sekhon HS, Proskocil B et al. Synthesis of acetylcholine by lung cancer. Life Sci.72(18–19), 2159–2168 (2003).
  • Pieper MP, Chaudhary NI, Park JE. Acetylcholine-induced proliferation of fibroblasts and myofibroblasts in vitro is inhibited by tiotropium bromide. Life Sci.80(24–25), 2270–2273 (2007).
  • Martinez-Moreno P, Nieto-Ceron S, Torres-Lanzas J et al. Cholinesterase activity of human lung tumours varies according to their histological classification. Carcinogenesis27(3), 429–436 (2006).
  • Psarras S, Caramori G, Contoli M et al. Oxidants in asthma and chronic obstructive pulmonary disease (COPD). Curr. Pharm. Des.11(6), 2053–2062 (2005).
  • Sancar A, Lindsey-Boltz LA, Unsal-Kacmaz K et al. Molecular mechanisms of mammalian DNA repair and the DNA damage checkpoints. Annu. Rev. Biochem.73, 39–85 (2004).
  • Brody JS, Spira A. State of the art. Chronic obstructive pulmonary disease, inflammation, and lung cancer. Proc. Am. Thorac. Soc.3(6), 535–537 (2006).
  • Popanda O, Schattenberg T, Phong T et al. Specific combinations of DNA repair gene variants and increased risk for non-small cell lung cancer. Carcinogenesis25(12), 2433–2441 (2004).
  • Pierrou S, Broberg P, O’Donnell RA et al. Expression of genes involved in oxidative stress responses in airway epithelial cells of smokers with chronic obstructive pulmonary disease Am. J. Respir. Crit. Care Med.175(6), 577–586 (2007).
  • Fukumoto S, Yamauchi N, Moriguchi H et al. Overexpression of the aldo–keto reductase family protein AKR1B10 is highly correlated with smokers’ non-small cell lung carcinomas. Clin. Cancer Res.11(5), 1776–1785 (2005).
  • Ramalingam S, Belani CP. Recent advances in targeted therapy for non-small cell lung cancer. Expert Opin. Ther. Targets11(2), 245–257 (2007).

Website

  • Global Initiative for Chronic Obstructive Lung Disease. Global Strategy for the diagnosis, management and prevention of chronic obstructive pulmonary disease. NHLBI/WHO workshop report. Bethesda, National Heart, Lung and Blood Institute, April 2001; NIH Publication No 2701:1–100. Last update 2006. www.goldcopd.com

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.