Is obstructive sleep apnea a risk factor for lung cancer?—from pathophysiological mechanisms to clinical data
Editorial Commentary | Basic and Mechanism Sciences

Is obstructive sleep apnea a risk factor for lung cancer?—from pathophysiological mechanisms to clinical data

Miguel Ángel Martínez-García1,2,3^, Grace Oscullo1,2,3, Jose Daniel Gómez-Olivas1,2,3, Marina Inglés-Azorin1,2,3, Sergio Mompeán1,2,3

1Department of Pneumology, Hospital Universitario y Politécnico La Fe, Valencia, Spain; 2CIBERES de Enfermedades Respiratorias, ISCIII, Madrid, Spain; 3Health Research Institute La Fe, Valencia, Spain

^ORCID: 0000-0002-7321-1891.

Correspondence to: Miguel Ángel Martínez-García, MD. Department of Pneumology, Hospital Universitario y Politécnico La Fe, Avenida Fernando Abril Martorell 2026, Valencia 46015, Spain; CIBERES de Enfermedades Respiratorias, ISCIII, Madrid, Spain; Health Research Institute La Fe, Valencia, Spain. Email: mianmartinezgarcia@gmail.com.

Keywords: Lung cancer; sleep apnea; sleep-disordered breathing


Submitted May 20, 2023. Accepted for publication Oct 07, 2023. Published online Oct 20, 2023.

doi: 10.21037/atm-23-1641


Obstructive sleep apnea (OSA) and lung cancer are two major public health problems. On the one hand, more than one billion individuals worldwide suffer from OSA, considered as the presence of at least five respiratory events (apnea or hypopnea) per hour of sleep [apnea-hypopnea index (AHI) ≥5] (1,2). On the other hand, cancer is one of the leading causes of death, with lung cancer being the leading cause of cancer deaths worldwide with an estimated 1.8 million deaths. In 2020, 11.4% of new cancers diagnosed were lung cancers, placing it as the second most common site of incident cancers and the most common cancer in men (3,4).

Although the relationship between OSA and some cardiovascular (5-8) and metabolic (9,10) adverse outcomes is well known, only during the last decade various studies of a pathophysiological nature, as well as murine and clinical models (both in epidemiological and clinical series) have established a relationship between OSA (11,12) and sleep duration (13) and a higher prevalence, incidence or aggressiveness of some malignant tumors. It seems that this relationship is more pronounced in more severe OSA (greater number of sleep-disordered breathing events, such as apneas, hypopneas), as well as in certain histological lines of tumor cells such as melanoma, bladder, liver, cervix, kidney, pancreas and lung cancer (14,15). With this editorial, we have tried to briefly review the most relevant pathophysiological and clinical finding on the relationship between OSA and cancer.

A recent review of the pathophysiological pathways that could explain this phenomenon highlighted some possibilities: (I) cell deficiency or dysfunction (including macrophages, natural killer T-cell, CD8 and CD3 T cells, stem-cell-like and dendritic cells); (II) biological biomarkers [vascular endothelial growth factor (VEGF) and other pro-angiogenic molecules, tumoral growth factor (TGF)-α1, tumoral necrosis factor (TNF)-α, tryptophan, cyclooxygenase (COX)-2, cannabinoid receptors, programmed death-ligand 1 (PD-L1), endostatin, endothelin-1, oxidative stress molecules and paraspeckle protein-1]; (III) genetic factors [glucose genes, hypoxic inducible factor (HIF)-1α genes, common key genes between OSA and cancer, micro-RNA-320β and NF-kB factor genes]; (IV) exosomes; and (V) microbiome (16).

Most alterations in the function, quantity, activation, and deactivation of these cells or molecules seem to be mainly linked to two fundamental features of OSA: sleep fragmentation and, above all, intermittent hypoxia (IH) defined as the oxygenation-deoxygenation derived from the sleep-disordered breathing (whether or not in addition to sustained hypoxia derived from cardiopulmonary comorbidities or obesity) produced by an excess of sleep-disordered breathing (16).

The first mechanism to be described and that is still considered fundamental today was probably the overexpression of HIF-1α in a hypoxic environment (such as inside tumors), aggravated by superadded IH caused by OSA. HIF-1α overexpression increases the production of proangiogenic molecules (especially VEGF) and, in its turn, the neovascularization of the tumor, with a subsequent increase in the production of distant metastases and tumor growth. In the case of tumorigenesis, it has been speculated that the oxygenation-reoxygenation cycles produced by IH are a potent inducer of the redox system, which is recognized as an important carcinogenic pathway (17). However, it became apparent that things were probably not that simple, and that there were other mechanisms involved, beyond the existence of IH. These are probably genetically mediated or implicate cells and molecules from the immune system (18).

In any case, one prominent characteristic of this relationship is that the impact of OSA does not seem to be related equally to all cancer sites. Furthermore, it can vary even with respect to the same cancer site, depending on the line of tumor cell involved—probably due to the different sensitivity of cancer cells to the action of IH and its consequences (for example, the different densities of active VEGF receptors in tumor cells) (19).

Table 1 shows the different pathophysiological mechanisms described to establish a relationship between OSA and lung cancer. As can be seen, and as occurs with most other tumors, many of the intimate molecular mechanisms that associate OSA with a greater progression or incidence of lung cancer are linked to the overexpression of HIF-1α and the various mechanisms that this sets in motion (20-25). A genetic component cannot be entirely ruled out, however, since OSA and lung cancer share some common genes (26).

Table 1

Factors hypothesized to promote lung cancer in OSA patients

Induction of HIF-1α as an expression acting as transcriptional activator of ATAD2
Common genes in OSA and lung cancer (MOAP1, CBX7, PDGFB and MAP2K3)
Elevation of PD-L1 monocytes/macrophages induced by exosomes that correlate with HIF-1α expression
Down-regulation of microRNA-320β with increasing of CDT1 via USP37
Promotion of ESM1 via HIF-1α pathway
Promotion of lung CSC-like properties by activation of mtROS mediated by Bach-1
Up-regulation of TGF-β1, VEGF and Foxp3 + Tregs expression
Increased expression of beta-catenin and Nrf2 as tumor growth factors

OSA, obstructive sleep apnea; HIF-1α, hypoxic inducible factor-1α; ATAD2, ATPase family AAA domain-containing protein 2; PD-L1, programmed death-ligand 1; CDT1, chromatin licensing and DNA replication factor 1; USP37, deubiquitinating enzyme family member; ESM1, endothelial cell-specific molecule-1; CSC, cancer stem cells; mtROS, mitochondrial reactive oxygen species; TGF, tumor growth factor; VEGF, vascular endothelial growth factor; Foxp3, forkhead box P3; Nrf2, nuclear factor erythroid-2-related factor-2.

It is worth noting that most of the studies were conducted in non-small cell lung cancer (NSCLC), and the evidence in small cell tumors is therefore very limited. Moreover, it is also striking that not all NSCLC cell lines responded in the same way to IH. In an interesting study of a mouse model, Marhuenda et al. (27) observed how cell proliferation “in vitro” responded differently to varying intensities of IH or sustained hypoxia, according to the cell lines exposed. The cell lines used were: H522, H1437 [human adenocarcinoma; p53 mutant and epidermal growth factor receptor (EGFR) wild-type], H1975 (human adenocarcinoma; p53 mutant, EGFR mutant) and H520 (human squamous cell lung cancer; p53 mutant, EGFR wild-type). It was observed that the H520 line (squamous cell lung cancer) proliferated faster in the presence of IH than in that of sustained hypoxia, and also faster than the adenocarcinoma lines. Within the three different lines of adenocarcinoma, IH did not seem to have any effect, although sustained hypoxia did produce a significant increase in cell proliferation. Evidence to extrapolate the previously described finding to human populations is desirable, as IH is a hallmark of OSA and sustained hypoxia is characteristic of other cardiopulmonary diseases or obesity that can coexist with OSA, and therefore, depending on these circumstances, the type of hypoxia created (intermittent, sustained or mixed) could produce different responses in the proliferation of the various lung cancer cell lines.

From a clinical point of view, two recent meta-analyses agree that the presence of OSA is a risk factor, independent of other confounding factors, for a higher incidence of lung cancer although there is much less scientific evidence for any relationship with higher mortality. Cheong et al. (28) found that patients with OSA were approximately 30% more likely to develop lung cancer than patients with no OSA. Ma et al. (29), for their part, included seven studies to reanalyze the relationship between OSA and cancer incidence, observing results similar to those of Cheong et al. (28). Furthermore, this higher incidence seemed to be independent of smoking, as it was replicated in a sensitivity analysis that used three studies without smokers [risk ratio (RR) 1.34; 95% CI: 1.22 to 1.48]. Moreover, subgroup analysis suggested that the association between OSA and a higher risk of lung cancer was not significantly affected by study characteristics such as design, source of population, sample size, evaluation methods for OSA, follow-up duration, methods for validation of lung cancer or study quality scores (28). Finally, Chen et al. (30) concluded in a third meta-analysis that OSA was not a risk factor for higher lung cancer mortality, although their analysis only included three small studies including 67 patients with lung cancer and comorbid OSA and 45 patients with lung cancer and no OSA. However, the odds ratio (OR) showed double the mortality for the group of lung cancer and OSA, although this was not statistically significant (probably due to the small sample size and the lack of analysis of the main confounders).

Despite inconsistencies in available data on the relationship between OSA and lung cancer in terms of the quality and generalizability of the evidence and therefore it is reasonable to approach some findings with caution, there is an interesting body of scientific evidence on the biological plausibility of this relationship. It seems that the overexpression of HIF-1α as a consequence of IH is the most important pathophysiological factor, although not all lung cancer cell lines respond in the same way. The existing data from clinical series, as gathered in meta-analyses, suggest the presence of a relationship between OSA and lung cancer in humans, although the following limitations of these studies need to be addressed in future research: (I) most of the studies were retrospective, included few patients and had short follow-ups; (II) small cell lung cancer was very little studied; (III) some confounders (especially smoking, obesity and other comorbidities) could be determining factors and should be studied in-depth; (IV) the methodology of the studies was quite heterogeneous; (V) it is necessary to carry out studies balanced by gender, as these could both be determining factors in the relationship; (VI) mortality studies in lung cancer are needed to shed more light on the effect of OSA on the aggressiveness of cancer; (VII) finally, and probably most importantly, it is necessary to carry out clinical studies on the effect of continuous positive airway pressure (CPAP)—which eliminates IH—on the incidence and aggressiveness of lung cancer, as well as its interaction with other anti-cancer treatments.


Acknowledgments

Funding: None.


Footnote

Provenance and Peer Review: This article was a standard submission to the journal. The article has undergone external peer review.

Peer Review File: Available at https://atm.amegroups.com/article/view/10.21037/atm-23-1641/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://atm.amegroups.com/article/view/10.21037/atm-23-1641/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Benjafield AV, Ayas NT, Eastwood PR, et al. Estimation of the global prevalence and burden of obstructive sleep apnoea: a literature-based analysis. Lancet Respir Med 2019;7:687-98. [Crossref] [PubMed]
  2. Mediano O, González Mangado N, Montserrat JM, et al. International Consensus Document on Obstructive Sleep Apnea. Arch Bronconeumol 2022;58:52-68. [Crossref] [PubMed]
  3. Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin 2021;71:209-49. [Crossref] [PubMed]
  4. Cayuela L, López-Campos JL, Otero R, et al. The Beginning of the Trend Change in Lung Cancer Mortality Trends in Spain, 1980-2018. Arch Bronconeumol (Engl Ed) 2021;57:115-21. [Crossref] [PubMed]
  5. Velásquez-Rodríguez J, Ortiz-Maraima T, Rodríguez-Viñoles MP, et al. Serum Leptin and Ultrasound Markers of Early Atherosclerosis in Patients with Sleep Apnea Hypopnea Syndrome. Arch Bronconeumol (Engl Ed) 2021;57:230-1. [Crossref] [PubMed]
  6. Navarro-Soriano C, Martínez-García MA, Torres G, et al. Long-term Effect of CPAP Treatment on Cardiovascular Events in Patients With Resistant Hypertension and Sleep Apnea. Data From the HIPARCO-2 Study. Arch Bronconeumol (Engl Ed) 2021;57:165-71. [Crossref] [PubMed]
  7. Javaheri S, Barbe F, Campos-Rodriguez F, et al. Sleep Apnea: Types, Mechanisms, and Clinical Cardiovascular Consequences. J Am Coll Cardiol 2017;69:841-58. [Crossref] [PubMed]
  8. Fernández-Bello I, Monzón Manzano E, García Río F, et al. Procoagulant State of Sleep Apnea Depends on Systemic Inflammation and Endothelial Damage. Arch Bronconeumol 2022;58:117-24. [Crossref] [PubMed]
  9. Alonso-Fernández A, Cerdá Moncadas M, Álvarez Ruiz De Larrinaga A, et al. Impact of Obstructive Sleep Apnea on Gestational Diabetes Mellitus. Arch Bronconeumol 2022;58:219-27. [Crossref] [PubMed]
  10. Labarca G, Reyes T, Jorquera J, et al. CPAP in patients with OSA and type-2 diabtes. A systematic review and meta-analysis. Clin Resp J 2018;12:2316-8. [Crossref] [PubMed]
  11. Martínez-García MÁ, Campos-Rodriguez F, Barbé F. Cancer and OSA: Current Evidence From Human Studies. Chest 2016;150:451-63. [Crossref] [PubMed]
  12. Almendros I, Martinez-Garcia MA, Farré R, et al. Obesity, sleep apnea, and cancer. Int J Obes (Lond) 2020;44:1653-67. [Crossref] [PubMed]
  13. Gómez Olivas JD, Campos-Rodriguez F, Nagore E, et al. Sleep Duration and Cutaneous Melanoma Aggressiveness. A Prospective Observational Study in 443 Patients. Arch Bronconeumol 2021;57:776-8. [Crossref] [PubMed]
  14. Cao Y, Ning P, Li Q, et al. Cancer and obstructive sleep apnea: An updated meta-analysis. Medicine (Baltimore) 2022;101:e28930. [Crossref] [PubMed]
  15. Gozal D, Ham SA, Mokhlesi B. Sleep Apnea and Cancer: Analysis of a Nationwide Population Sample. Sleep 2016;39:1493-500. [Crossref] [PubMed]
  16. Sánchez-de-la-Torre M, Cubillos C, Veatch OJ, et al. Potential Pathophysiological Pathways in the Complex Relationships between OSA and Cancer. Cancers (Basel) 2023;15:1061. [Crossref] [PubMed]
  17. García-Río F, Alcázar-Navarrete B, Castillo-Villegas D, et al. Biological Biomarkers in Respiratory Diseases. Arch Bronconeumol 2022;58:323-33. [Crossref] [PubMed]
  18. Gozal D, Almendros I, Hakim F. Sleep apnea awakens cancer: A unifying immunological hypothesis. Oncoimmunology 2014;3:e28326. [Crossref] [PubMed]
  19. Martinez-Garcia MA, Campos-Rodriguez F, Almendros I, et al. Cancer and Sleep Apnea: Cutaneous Melanoma as a Case Study. Am J Respir Crit Care Med 2019;200:1345-53. [Crossref] [PubMed]
  20. Hao S, Li F, Jiang P, et al. Effect of chronic intermittent hypoxia-induced HIF-1α/ATAD2 expression on lung cancer stemness. Cell Mol Biol Lett 2022;27:44. [Crossref] [PubMed]
  21. Liu Y, Lu M, Chen J, et al. Extracellular vesicles derived from lung cancer cells exposed to intermittent hypoxia upregulate programmed death ligand 1 expression in macrophages. Sleep Breath 2022;26:893-906. [Crossref] [PubMed]
  22. Gu X, Zhang J, Shi Y, et al. ESM1/HIF-1α pathway modulates chronic intermittent hypoxia-induced non-small-cell lung cancer proliferation, stemness and epithelial-mesenchymal transition. Oncol Rep 2021;45:1226-34. [Crossref] [PubMed]
  23. Hao S, Zhu X, Liu Z, et al. Chronic intermittent hypoxia promoted lung cancer stem cell-like properties via enhancing Bach1 expression. Respir Res 2021;22:58. [Crossref] [PubMed]
  24. Liu Y, Lao M, Chen J, et al. Short-term prognostic effects of circulating regulatory T-Cell suppressive function and vascular endothelial growth factor level in patients with non-small cell lung cancer and obstructive sleep apnea. Sleep Med 2020;70:88-96. [Crossref] [PubMed]
  25. Liu Y, Song X, Wang X, et al. Effect of chronic intermittent hypoxia on biological behavior and hypoxia-associated gene expression in lung cancer cells. J Cell Biochem 2010;111:554-63. [Crossref] [PubMed]
  26. Wang W, He L, Ouyang C, et al. Key Common Genes in Obstructive Sleep Apnea and Lung Cancer are Associated with Prognosis of Lung Cancer Patients. Int J Gen Med 2021;14:5381-96. [Crossref] [PubMed]
  27. Marhuenda E, Campillo N, Gabasa M, et al. Effects of Sustained and Intermittent Hypoxia on Human Lung Cancer Cells. Am J Respir Cell Mol Biol 2019;61:540-4. [Crossref] [PubMed]
  28. Cheong AJY, Tan BKJ, Teo YH, et al. Obstructive Sleep Apnea and Lung Cancer: A Systematic Review and Meta-Analysis. Ann Am Thorac Soc 2022;19:469-75. [Crossref] [PubMed]
  29. Ma H, Zhang X, Han J, et al. Sleep-disordered breathing and risk of lung cancer: a meta-analysis longitudinal follow-up studies. Eur J Cancer Prev 2022;31:245-52. [Crossref] [PubMed]
  30. Chen MX, Chen LD, Zeng AM, et al. Obstructive sleep apnea and the risk of mortality in patients with lung cancer: a meta-analysis. Sleep Breath 2022;26:559-66. [Crossref] [PubMed]
Cite this article as: Martínez-García MÁ, Oscullo G, Gómez-Olivas JD, Inglés-Azorin M, Mompeán S. Is obstructive sleep apnea a risk factor for lung cancer?—from pathophysiological mechanisms to clinical data. Ann Transl Med 2023;11(12):422. doi: 10.21037/atm-23-1641

Download Citation