Skip to main content

MINI REVIEW article

Front. Surg., 20 May 2022
Sec. Thoracic Surgery
This article is part of the Research Topic Perioperative Systemic Inflammation in Lung Cancer Surgery View all 7 articles

Perioperative Systemic Inflammation in Lung Cancer Surgery

\r\nJzsef Furk
József Furák1*Tibor NmethTibor Németh1Judit LantosJudit Lantos2Csongor FabCsongor Fabó3Tibor GcziTibor Géczi1Nomi Zombori-TthNoémi Zombori-Tóth4Dra ParczaiDóra Paróczai5Zaln SzntZalán Szántó6Zsolt Szab\r\nZsolt Szabó7
  • 1Department of Surgery, Faculty of Medicine, University of Szeged, Szeged, Hungary
  • 2Department of Neurology, Bács-Kiskun County Hospital, Kecskemét, Hungary
  • 3Department of Anesthesiology, Faculty of Medicine, University of Szeged, Szeged, Hungary
  • 4Department of Pathology, Faculty of Medicine, University of Szeged, Szeged, Hungary
  • 5Department of Medical Microbiology, Faculty of Medicine, University of Szeged, Szeged, Hungary
  • 6Department of Thoracic Surgery. Medical School, University of Pécs, Pécs, Hungary
  • 7Institute of Surgical Research, Faculty of Medicine, University of Szeged, Szeged, Hungary

Systemic inflammation (SI) is a response of the immune system to infectious or non-infectious injuries that defends the body homeostasis. Every surgical intervention triggers SI, the level of which depends on the extent of damage caused by the surgery. During the first few hours after the damage, the innate or natural immunity, involving neutrophils, macrophages, and natural killer cells, plays a main role in the defense mechanism, but thereafter the adaptive immune response ensues. The number of leukocytes is elevated, the levels of lymphocytes and natural killer cells are reduced, and the cytokines released after surgery correlate with surgical damage. Minimally invasive thoracic surgery procedures induce less inflammatory response and reduce the immune defense in patients to a more moderate level compared with the open surgery procedures; this immunosuppression can be further diminished in spontaneous ventilation cases. The normal functioning of the immune defense is important in controlling the perioperative circulatory tumor cells. Moreover, elevated levels of inflammatory cytokines before immune therapy have a negative impact on the response, and significantly shorten the progression-free survival. Clinically, the lower are the levels of cytokines released during lung surgery, the lesser is the postoperative morbidity, especially pneumonia and wound infection. The return to normal levels of lymphocytes and cytokines occurs faster after spontaneous ventilation surgery. The use of locoregional anesthesia can also reduce SI. Herein, we review the current knowledge on the effects of different operative factors on postoperative SI and defense mechanism in lung cancer surgery.

Introduction

Systemic inflammation (SI) is an immune response to infectious or non-infectious injury in the body, aimed at restoring the normal anatomy and function of the damaged organ and eradicating infectious agents. Although the initial step in this process is localized inflammation with local symptoms, this response can develop into a systematic reaction or overreaction, termed as systemic inflammatory response syndrome (SIRS). Although SIRS can be described as sepsis in the case of infection, it can be a non-infectious process as well, for example, in surgical stress cases (1, 2). The terminology and diagnostic criteria of the SI have changed with time (3). The diagnosis of SIRS is based on clinical signs (body temperature, heart rate, respiration rate, and level of white blood cells) and the presence of proinflammatory cytokines and mediators in the serum (1). For precise diagnosis and prognosis of the SI, currently, the sepsis-3 definition is widely used focusing on the organ functions, and the diagnosis is based on the sequential (or sepsis-related) organ failure assessment (SOFA) score (respiration-arterial oxygen pressure, coagulation-platelets level, liver-bilirubin level, cardiovascular-hypotension, central nervous system-Glasgow Coma Score, renal-creatinine) (4, 5).

With regard to the pathophysiological background of SI, in the first few hours after the damage, innate or natural immunity, involving neutrophils, macrophages, natural killer cells, plays a major role in the early defense mechanism, but it is soon followed by the adaptive immune response . In both types of adaptive immune responses (i.e., humoral and cell-mediated), lymphocytes (T-helper cells, cytotoxic T cells, regulatory T cells, and B cells) play a major role together with cytokines. The two types of immune responses interact very closely with each other.

Surgical trauma, including thoracic surgical procedures, induces SI through systemic proinflammatory and compensatory anti-inflammatory responses (6, 7). In lung cancer surgery, there are two important aspects of SI, viz., postoperative morbidity and control of tumor cell spreading (1, 6, 7). The currently used approaches in lung cancer surgery are open thoracotomy and minimally invasive thoracic surgery (MITS) procedures, such as video-assisted thoracic surgery (VATS) and robot-assisted thoracic surgery (RATS). MITS interventions induce lesser inflammatory response and reduce the immune defense of the patient compared with open interventions (8), and the postoperative period is easier to manage in MITS, with less morbidity, and shorter postoperative drainage time and hospital stay (9, 10). With the popularization of MITS, mainly VATS, spontaneous ventilation (SV) was developed to further reduce the surgical stress in thoracic surgery (11). This supports the clinical observation that postoperative results are simpler and smoother in shorter and less invasive procedures (12). With the minimization of surgical stress in the thoracic surgical procedures, the procedure of administering anesthesia in thoracic operations had to be changed. Generally, in thoracic surgical procedures, the additional effect of mechanical one-lung ventilation (mOLV), with its positive and negative impact, on the perioperative inflammatory response must be evaluated. The negative inflammatory effect of mOLV can be reduced with spontaneous one-lung ventilation (sOLV) (13). One of the most relevant pathophysiological observations is the reduced inflammatory response and immune changes in patients on SV (1416).

In this paper, the effects of different operative factors, such as surgical aggressiveness, additional local anesthesia, and type of ventilation, on postoperative SI and defense mechanism in lung cancer surgery are discussed.

Pathophysiology of SI

In SI, the innate immune system and response are first activated. Primary immune cells (neutrophils, macrophages, natural killer cells, and dendritic cells) capable of phagocytosis and antigen presentation migrate to the surgically injured tissue. These primary immune cells recognize the damage-associated molecular patterns of the damaged tissue (e.g., surgical incision and preparation) via toll-like receptors and release proinflammatory cytokines, such as TNFα, IL-6, IL-8, and IL-1β, and anti-inflammatory cytokines, such as IL-4, IL-10, and IL-13. The release of the cytokines can be very quick. The level of IL-6, IL-8, and IL-10 may elevate at the time of skin closure after the lung resection, posing a risk of postoperative complications (17). Normal levels of these cytokines are necessary for physiological functions of the immune system. If inflammation can be controlled by the cellular defense and cytokines, it remains localized. SI can develop in advanced cases. The excessive production of proinflammatory cytokines has a negative effect on the normal functioning of the body, leading to the loss of organ function and possible multi-organ failure. The pathomechanism underlying the negative effect of cytokines in the advanced stages of SI can involve damage to the cell membrane, disseminated intravascular coagulation, and ischemia-reperfusion injury with capillary dysfunction, causing postoperative complications (18). A correlation between the levels of proinflammatory cytokines and the degree of postoperative SI has been proven (6, 19, 20).

During the SIRS, there is a massive release of immature neutrophils from the bone marrow into the circulation, causing an elevated leukocyte count after the surgery, and a lowering in the number of lymphocytes in the postoperative days by post-surgical apoptosis (19, 21). NK cells are activated by the high mobility group box protein 1, which is released from the damaged tissue. For specific immune responses, there must be an interaction between antigen-presenting cells (e.g., macrophages and dendritic cells) and T lymphocytes. The interaction of antigens and cytokines with CD4+ lymphocytes can result in their differentiation into Th1 (cellular immune response) or Th2 (humoral immune response) cells, both of which are regulated by cytokines (19, 20).

Anti-Tumor Immunity and SI

A special concern—intraoperative tumor dissemination and control of circulatory tumor cells—is emerging in cases of cancer surgery (22). In an animal study, circulatory tumor cells were significantly elevated after tumor punction but disappeared after resection. Unfortunately, circulatory tumor cells could be detected in 6 weeks after resection. In view of this, the normal functioning of the immune system is very important in the postoperative 6 weeks to eliminate the circulatory tumor cells (23). If the number of lymphocytes and NK cells is reduced, their capacity against tumor cells is also decreased (24).

Clinical studies have proven the superiority of VATS over thoracotomy, showing favorable sequelae on cellular immune functions. VATS is associated with a lesser postoperative decrease in the numbers of circulating CD3+, CD4+, and CD8+ T cells, which lowers the risk of imbalanced immunoregulation and preserves immunosurveillance, decreasing the risk of tumor growth and recurrence (24, 25). Unlike open thoracotomy, intubated VATS major lung resection and lobectomy do not significantly reduce the T lymphocyte populations (8, 26). Additionally, non-intubated VATS unveiled new approaches for patients with non-small cell lung cancer (NSCLC), as the absence of intubation is associated with less reduction in NK cell and lymphocyte populations (15, 27). The reduction in lymphocyte number is not fully understood but has been widely investigated. It may be due to the redistribution of lymphocytes to the surgical site, apoptosis, regulation of T cells with reduced helper-inducer T cells, and increased cytotoxic T cells (2830).

T lymphocytes are essential for immunoregulation and tumor suppression. An imbalance of CD8+ cytotoxic and CD4+ helper/regulatory T cells in the tumor-infiltrating lymphocyte (TIL) population in surgically treated NSCLCs was assessed to be a prognostic indicator after surgery (31). T lymphocytes mediate anti-tumor responses and learn to recognize tumor-specific antigens bound to the major histocompatibility complex (MHC)-I. After stimulation by antigen-presenting cells, CD8+ T cells are licensed to kill tumor cells and stimulate the production of a wide range of pro-inflammatory cytokines, resulting in appropriate immune-mediated destruction. However, tumor cells develop several mechanisms, such as expression of inhibitory receptors and cytokines, to evade the host immunity. Under chronic inflammation or tumor growth, T lymphocytes experience a persistently high antigen load, leading to the expression of inhibitory receptors on their surface, which limits the evolving activation of inflammatory cells. Programmed-death 1 (PD-1) inhibitory receptor, which is an immune checkpoint, and its ligand programmed death ligand 1 (PDL-1) are essential for immune tolerance as T lymphocytes can upregulate PD-1 receptor; thus, they lose the ability of progressive proliferation and activation (32, 33). However, tumor cells can express PDL-1 and promote binding to the PD-1 receptor; therefore, tumors co-opt this pathway to suppress effector T cell-mediated cell killing and avoid cell death (34). Recently, PDL-1 inhibitors have become the focus of treatment in several cancers, such as Hodgkin’s lymphoma, melanoma, and advanced NSCLC. IgG4 monoclonal antibodies that block PD-1 have been proven to be effective in patients with advanced NSCLC, and they increase the progression-free survival (PFS) and overall survival (OS), compared with conventional chemotherapies (35, 36). In a study on stage IV NSCLC immune checkpoint inhibitor (ICI) treatment, it was verified that elevated inflammatory cytokines, including IL-6, and the neutrophyl-lymphocyte rate before the immune therapy had a negative impact on the response, and the PFS was significantly shorter (37). The negative impact of SI on the success of ICIs has been proven in another study (38).

Impact of the Surgical Approach on SI

Postoperative morbidity after VATS lobectomy has been proven to be significantly lower than that after open lobectomy. VATS is associated with reduced atrial fibrillation, renal failure, shorter hospital stay and chest tube drainage, and postoperative pneumonia (9, 10). Although these studies did not mention the reasons for the differences in postoperative pneumonia and the immune benefits of VATS/MITS, these were investigated later. The postoperative proinflammatory response is greater after open surgery than after VATS, which affects the innate immune response (39). IL-6 levels were significantly higher in open cases than in VATS (14), SIRS/SI has a significant correlation with elevated IL-6 levels (6), and the levels of IL-6 and IL-8 correlated with thoractomy, length of the surgery, and blood loss in esophageal surgery (7).

RATS, a type of MITS procedure, is increasingly being used in lung cancer surgery. The perioperative results of RATS are similar to those of the VATS, as noted in a meta-analysis (40), but the length of the surgery in some cases can be longer after RATS than after VATS. There were no differences in acute phase proteins and immune responses between the RATS and VATS procedures (41), showing that surgical trauma is similar in the two MITS methods.

Comparing the different types of MITS procedures, Tacconi reported that there was no difference in the SI between uniportal, multiportal, and hybrid VATS lobectomies. They reported that the level of SI markers returned to the preoperative levels after 5 days (42).

Impact of SV on SI

During lung resections, mOLV is a highly recommended method for providing the technical background of preparations in the chest cavity. With this type of anesthesia, the dependent/ventilated lung is used instead of the two lungs for gas exchange, which can have negative effects. In mOLV, to maintain physiological oxygen and carbon dioxide levels, anesthesia uses a higher tidal volume and oxygen concentration, positive end-expiratory pressure (PEEP), or positive pressure ventilation. Despite the protective ventilation method (43), mainly in patients with underlying lung diseases (fibrosis, emphysema, and obstructive pulmonary disease) and pulmonary hypertension, ventilation can cause injury to the alveoli through overdistension of the alveoli, resulting in volutrauma/barotrauma and atelectrauma (44, 45). These changes are the source of the accumulation of inflammatory cells (neutrophils, macrophages, and lymphocytes), release of cytokines (TNF-α, IL-6, IL-8, and IL-1β, and edema in the dependent lung (biotrauma), causing SI (46, 47). The abovementioned side effects of mOLV can be reduced with sOLV.

In both non-intubated and intubated sOLV thoracic surgeries (48, 49), SV can prevent or reduce volutrauma, atelectrauma, and biotrauma of the mOLV. Better immune responses and less immunosuppression after SV surgery have been widely investigated and published. The changes in NK cells and lymphocytes during SV surgery were lower than those in relaxed surgery cases, and the return to the baseline level required lesser time (16). These studies revealed that there is less immunosuppression after SV surgery (15) and demonstrated that SV has a long-term impact on survival. In malignant pleural effusion cases, survival was longer in patients with SV than in those who underwent relaxed surgery. SV not only affects the immune cells but also the cytokine release. In a lung metastasectomy study, the impact of the non-intubated procedure was compared with that in relaxed surgery cases. Non-intubated procedure had less impact on immune function and SI, with less release of IL-6 (27). As a clinical consequence of the lower perioperative SI, the postoperative morbidity was lower, and the hospital stay was shorter after SV thoracic surgery. In a major lung resection study, the advantages of non-intubated, SV thoracic surgery on cytokine release were revealed. IL-6 and TNF-α levels were significantly lower in non-intubated patients than in relaxed surgery patients (11). The change in stress hormone levels as a factor in the SI pathway was moderate in SV surgery compared with that in relaxed surgical cases (50).

In addition to less volutrauma, atelectrauma, and biotrauma, SV reduces the changes in immune cells and cytokine release through another pathway. This involves a sympatholytic effect of locoregional anesthesia. Both, epidural and paravertebral/intercostal anesthesia with vagus blockade can reduce surgical stress, and the levels of IL-6, IL-8, and TNF-α (51, 52), and epidural anesthesia reduces cytokine levels in open esophagectomy cases, as well (53).

Treatment of SI

Although many studies have reported the reduction of cytokine levels as a possible treatment for postoperative SI, prevention still remains the best treatment for it. Regarding the method of anesthesia administration, protective ventilation is important to prevent volutrauma (43), but the type of drugs used during narcosis can have a role in SI prevention (54). Although it is stated that sevoflurane can reduce the level of cytokines during mOLV (55), the beneficial effect of propofol over isoflurane is verified (56). Another approach to treat the pathophysiological manifestation of the SI could be to eliminate or at least reduce cytokine levels. There are many methods (filtration, dialysis [diffusion], adsorption) to overcome the challenge of reducing cytokine levels, but a real breakthrough has not been achieved yet (57). Currently, one of the most promising methods is the CytoSorb hemoadsorption, which has a positive effect in the advanced phase of the SI, like sepsis and pneumonia (58). However, the prevention of SI by eliminating cytokines in the early perioperative period or during the lung surgery has not been investigated yet, by our knowledge. In a cardiac surgery study of cardiopulmonary by-pass, there was no difference between the intraoperative and early postoperative cytokine levels and clinical results of the CytoSorb adsorption and control group (59).

Discussion

The main issue in postoperative SI is how the pathological background of pro- and anti-inflammatory responses can affect the clinical picture. Patients who have undergone lung cancer surgery expect a quick recovery from the operation and long-term survival, and the surgical treatment should be adapted to this expectation. Because every surgical intervention causes an SI response, the best results can be achieved with less harmful surgery; in other words, the more minimally invasive is the thoracic surgery, the more preserved is the immune function. Based on the changes in the cellular defense and cytokine levels, this kind of postoperative SI period can take for around 3–12 days, but it affects not only on the early postoperative morbidity, but also the 30-day mortality (15, 20, 27).

Regarding the early postoperative morbidity, the reduced levels and functions of lymphocytes and NK cells can cause diminished cellular defense. In clinical practice, this can manifest as postoperative pneumonia, wound infection, or other inflammation. Postoperative pneumonia rates were 5% and 10%, and wound infection rates were 0.4% and 1% after VATS and open lobectomies, respectively (10).

Because of the less pro-inflammatory response, some postoperative morbidities can be reduced further with the use of SV (49). In SV surgery, postoperative morbidity is lower (5%) than in relaxed surgery cases (23%) (27). Generally, after SV VATS lung resection, the postoperative fasting time, drainage time, and hospital stay were shorter than those in relaxed VATS cases (60, 61).

The long-term oncological effects of SI are discussed based on different approaches for lung cancer resection. A meta-analysis showed the advantage of relaxed VATS in long-term survival after lung cancer resection, and the reason was suspected to be the lower levels of cytokines released after VATS compared with that in open cases (62). In another review article comparing long-term survival after VATS and open surgery, no significant difference in survival was found between the two approaches (63). The same uncertain advantage exists in the long-term survival of patients undergoing SV VATS procedures. Although immune function is less reduced after SV surgery, there are very few reports about the benefits of this approach in terms of long-term survival. One of the basic publications about the immune effect and impact on survival after SV showed better survival in malignant pleural fluid surgery (15), as well as significantly better survival and disease-free survival after SV for lung cancer resections compared with that in relaxed surgery cases (64). In contrast, there was no advantage in survival or recurrence was found after SV (65).

There are some encouraging results regarding the short-term oncological effect of SI. Better compliance with adjuvant chemotherapy was found after non-intubated VATS lobectomies than after relaxed VATS lobectomies, with less toxicity, and more patients (92%) could receive the adjuvant chemotherapy protocol, compared with 72% in relaxed surgery cases (66). Less toxic and more adjuvant treatment should provide better oncological outcomes. The same thought process can be seen in the case of ICI treatment, although in the current studies on the application of ICIs in non-operated patients, the results and the message are clear: more SI before the beginning of immunotherapy is associated with less successful outcomes. Surgery for lung cancer should provide the lowest postoperative SI before the planned ICIs. Lymphocytes are crucial in anti-tumor response; thus, the impact of intubated and non-intubated VATS on the prognosis of advanced NSCLC patients receiving immunotherapies remains to be elucidated (37, 38).

In conclusion, to reduce postoperative SI, thoracic surgeons should reduce the damage caused by the surgical procedures for lung cancer while adhering to the oncological principles. The more minimally invasive the procedures used, the less immunosuppression is required by the patient. The positive effect of VATS on SI (less diminished lymphocyte function, less pro-inflammatory cytokine release) is mirrored in the better postoperative results, and to continue this beneficial minimalization of surgical aggressivity, SV surgery could be an option. Although SV is not a widely accepted procedure, it has a very simple and useful message: the positive effect of locoregional anesthesia on SI, which can be used properly in relaxed VATS and open cases.

Author Contributions

JF: writing the paper, collecting data, and conception of the manuscript. TN: writing the paper, collecting data. JL: writing the paper and collecting data, CSF: conception of the anesthesiologic aspect of the topic TG: collecting data, NZT: collecting data, DP: conception of the immunological aspect of the topic, ZSZ conception and finalizing of the manuscript. ZSSZ: conception of the anesthesiologic aspect of the topic. All authors contributed to the article and approved the submitted version.

Acknowledgments

We would like to thank Editage (www.editage.com) for English language editing.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher's Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Zotova NV, Chereshnev VA, Gusev EY. Systemic inflammation: methodological approaches to identification of the common pathological process. PLoS ONE. (2016) 11:e0155138. doi: 10.1371/journal.pone.0155138

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Bone RC, Balk RA, Cerra FB, Dellinger RP, Fein AM, Knaus WA, et al. Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. The ACCP/SCCM Consensus Conference Committee. American College of Chest Physicians/Society of Critical Care Medicine. Chest. (1992) 101:1644–55. doi: 10.1378/chest.101.6.1644

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Marik PE, Taeb AM. SIRS, qSOFA and new sepsis definition. J Thorac Dis. (2017) 9:943–5. doi: 10.21037/jtd.2017.03.125

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Fernando SM, Rochwerg B, Seely AJE. Clinical implications of the Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). CMAJ. (2018) 190:E1058–9. doi: 10.1503/cmaj.170149

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Vincent JL, Moreno R, Takala J, Willatts S, De Mendonça A, Bruining H, et al. The SOFA (Sepsis-related Organ Failure Assessment) score to describe organ dysfunction/failure. On behalf of the Working Group on Sepsis-Related Problems of the European Society of Intensive Care Medicine. Intensive Care Med. (1996) 22:707–10. doi: 10.1007/BF01709751

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Takenaka K, Ogawa E, Wada H, Hirata T. Systemic inflammatory response syndrome and surgical stress in thoracic surgery. J Crit Care. (2006) 21:48–53. doi: 10.1016/j.jcrc.2005.07.001

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Sakamoto K, Arakawa H, Mita S, Ishiko T, Ikei S, Egami H, et al. Elevation of circulating interleukin 6 after surgery: factors influencing the serum level. Cytokine. (1994) 6:181–6. doi: 10.1016/1043-4666(94)90040-x

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Leaver HA, Craig SR, Yap PL, Walker WS. Lymphocyte responses following open and minimally invasive thoracic surgery. Eur J Clin Investig. (2000) 30:230–8. doi: 10.1046/j.1365-2362.2000.00622.x

CrossRef Full Text | Google Scholar

9. Scott WJ, Allen MS, Darling G, Meyers B, Decker PA, Putnam JB, et al. Video-assisted thoracic surgery versus open lobectomy for lung cancer: a secondary analysis of data from the American College of Surgeons Oncology Group Z0030 randomized clinical trial. J Thorac Cardiovasc Surg. (2010) 139:976–81. doi: 10.1016/j.jtcvs.2009.11.059

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Villamizar NR, Darrabie MD, Burfeind WR, Petersen RP, Onaitis MW, Toloza E, et al. Thoracoscopic lobectomy is associated with lower morbidity compared with thoracotomy. J Thorac Cardiovasc Surg. (2009) 138:419–25. doi: 10.1016/j.jtcvs.2009.04.026

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Jeon J, Sung S, Moon Y, Koo J, Hyun K, Han K, et al. Comparison of early postoperative cytokine changes in patients undergoing intubated and non-intubated thoracic surgery: a randomized controlled trial. Interact Cardiovasc Thorac Surg. (2021) 32:343–50. doi: 10.1093/icvts/ivaa265

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Misthos P, Katsaragakis S, Theodorou D, Milingos N, Skottis I. The degree of oxidative stress is associated with major adverse effects after lung resection: a prospective study. Eur J Cardiothorac Surg. (2006) 29:591–5. doi: 10.1016/j.ejcts.2005.12.027

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Kiss G, Castillo M. Nonintubated anesthesia in thoracic surgery: general issues. Ann Transl Med. (2015) 3:110. doi: 10.3978/j.issn.2305-5839.2015.04.21

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Nagahiro I, Andou A, Aoe M, Sano Y, Date H, Shimizu N. Pulmonary function, postoperative pain, and serum cytokine level after lobectomy: a comparison of VATS and conventional procedure. Ann Thorac Surg. (2001) 72:362–5. doi: 10.1016/s0003-4975(01)02804-1

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Mineo TC, Ambrogi V. Immune effects after uniportal nonintubated video-thoracoscopic operations. Video-assist. J Thorac Surg. (2018) 3:4–10. doi: 10.21037/vats.2018.01.02

CrossRef Full Text | Google Scholar

16. Vanni G, Tacconi F, Sellitri F, Ambrogi V, Mineo TC, Pompeo E. Impact of awake videothoracoscopic surgery on postoperative lymphocyte responses. Ann Thorac Surg. (2010) 90:973–8. doi: 10.1016/j.athoracsur.2010.04.070

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Kaufmann KB, Heinrich S, Staehle HF, Bogatyreva L, Buerkle H, Goebel U. Perioperative cytokine profile during lung surgery predicts patients at risk for postoperative complications—a prospective, clinical study. PLoS One. (2018) 13:e0199807. doi: 10.1371/journal.pone.0199807

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Jaffer U, Wade RG, Gourlay T. Cytokines in the systemic inflammatory response syndrome: a review. HSR Proc Intensive Care Cardiovasc Anesth. (2010) 2:161–75.

PubMed Abstract | Google Scholar

19. Dąbrowska AM, Słotwiński R. The immune response to surgery and infection. Cent Eur J Immunol. (2014) 39:532–7. doi: 10.5114/ceji.2014.47741

CrossRef Full Text | Google Scholar

20. Breunig A, Gambazzi F, Beck-Schimmer B, Tamm M, Lardinois D, Oertli D, et al. Cytokine & chemokine response in the lungs, pleural fluid and serum in thoracic surgery using one-lung ventilation. J Inflamm (Lond). (2011) 8:32. doi: 10.1186/1476-9255-8-32

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Chen L, Deng H, Cui H, Fang J, Zuo Z, Deng J, et al. Inflammatory responses and inflammation-associated diseases in organs. Oncotarget. (2018) 9:7204–18. doi: 10.18632/oncotarget.23208

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Alieva M, van Rheenen J, Broekman MLD. Potential impact of invasive surgical procedures on primary tumor growth and metastasis. Clin Exp Metastasis. (2018) 35:319–31. doi: 10.1007/s10585-018-9896-8

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Juratli MA, Siegel ER, Nedosekin DA, Sarimollaoglu M, Jamshidi-Parsian A, Cai C, et al. In vivo long-term monitoring of circulating tumor cells fluctuation during medical interventions. PLoS ONE. (2015) 10:e0137613. doi: 10.1371/journal.pone.0137613

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Zhang LB, Wang B, Wang XY, Zhang L. Influence of video-assisted thoracoscopic lobectomy on immunological functions in non-small cell lung cancer patients. Med Oncol. (2015) 32:201. doi: 10.1007/s12032-015-0639-2

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Ng CSH, Whelan RL, Lacy AM, Yim APC. Is minimal access surgery for cancer associated with immunologic benefits? World J Surg. (2005) 29:975–81. doi: 10.1007/s00268-005-0029-6

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Ng CSH, Lee TW, Wan S, Wan IYP, Sihoe ADL, Arifi AA, et al. Thoracotomy is associated with significantly more profound suppression in lymphocytes and natural killer cells than video-assisted thoracic surgery following major lung resections for cancer. J Invest Surg. (2005) 18:81–8. doi: 10.1080/08941930590926320

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Mineo TC, Sellitri F, Vanni G, Gallina FT, Ambrogi V. . Immunological and inflammatory impact of non-intubated lung metastasectomy. Int J Mol Sci. (2017) 18:1466. doi: 10.3390/ijms18071466

CrossRef Full Text | Google Scholar

28. Dupont G, Flory L, Morel J, Lukaszewicz AC, Patoir A, Presles E, et al. Postoperative lymphopenia: an independent risk factor for postoperative pneumonia after lung cancer surgery, results of a case-control study. PLoS ONE. (2018) 15(13):e0205237. doi: 10.1371/journal.pone.0205237

CrossRef Full Text | Google Scholar

29. Hogan BV, Peter MB, Shenoy HG, Horgan K, Hughes TA. Surgery induced immunosuppression. Surgeon. (2011) 9:38–43. doi: 10.1016/j.surge.2010.07.011

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Ogawa K, Hirai M, Katsube T, Murayama M, Hamaguchi K, Shimakawa T, et al. Suppression of cellular immunity by surgical stress. Surgery. (2000) 127:329–36. doi: 10.1067/msy.2000.103498

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Giatromanolaki A, Anestopoulos I, Panayiotidis MI, Mitrakas A, Pappa A, Koukourakis MI. Prognostic relevance of the relative presence of CD4, CD8 and CD20 expressing tumor infiltrating lymphocytes in operable non-small cell lung cancer patients. Anticancer Res. (2021) 41:3989–95. doi: 10.21873/anticanres.15196

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Wherry EJ. T cell exhaustion. Nat Immunol. (2011) 12:492–9. doi: 10.1038/ni.2035

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Speiser DE, Utzschneider DT, Oberle SG, Münz C, Romero P, Zehn D. T cell differentiation in chronic infection and cancer: functional adaptation or exhaustion? Nat Rev Immunol. (2014) 14:768–74. doi: 10.1038/nri3740

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Seidel JA, Otsuka A, Kabashima K. Anti-PD-1 and anti-CTLA-4 therapies in cancer: mechanisms of action, efficacy, and limitations. Front Oncol. (2018) 8:86. doi: 10.3389/fonc.2018.00086

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Reck M, Rodríguez-Abreu D, Robinson AG, Hui R, Csőszi T, Fülöp A, et al. Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer. N Engl J Med. (2016) 375:1823–33. doi: 10.1056/NEJMoa1606774

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Herbst RS, Garon EB, Kim D-W, Cho BC, Gervais R, Perez-Gracia JL, et al. Five year survival update from KEYNOTE-010: pembrolizumab versus docetaxel for previously treated, programmed death-ligand 1-positive advanced NSCLC. J Thorac Oncol. (2021) 16:1718–32. doi: 10.1016/j.jtho.2021.05.001

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Kauffmann-Guerrero D, Kahnert K, Kiefl R, Sellmer L, Walter J, Behr J, et al. Systemic inflammation and pro-inflammatory cytokine profile predict response to checkpoint inhibitor treatment in NSCLC: a prospective study [Sci. rep.]. Sci Rep. (2021) 11:10919. doi: 10.1038/s41598-021-90397-y

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Keegan A, Ricciuti B, Garden P, Cohen L, Nishihara R, Adeni A, et al. Plasma IL-6 changes correlate to PD-1 inhibitor responses in NSCLC. J Immunother Cancer. (2020) 8:e000678. doi: 10.1136/jitc-2020-000678

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Jones RO, Anderson NH, Murchison JT, Brittan M, Simon EJ, Casali G, et al. Innate immune responses after resection for lung cancer via video-assisted thoracoscopic surgery and thoracotomy. Innovations (Phila). (2014) 9:93–103. doi: 10.1097/IMI.0000000000000061

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Guo FMD, Ma DMD, Li SMD. Compare the prognosis of Da Vinci robot-assisted thoracic surgery (RATS) with video-assisted thoracic surgery (VATS) for non-small cell lung cancer. Medicine. (2019) 98:e17089. doi: 10.1097/MD.0000000000017089

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Erus S, Öztürk AB, Albayrak Ö, İncir S, Kapdağlı MH, Cesur EE, et al. Immune profiling after minimally invasive lobectomy. Interact Cardiovasc Thorac Surg. (2021) 32:291–7. doi: 10.1093/icvts/ivaa296

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Tacconi F, Carlea F, La Rocca E, Vanni G, Ambrogi V. Systemic inflammation after uniport, multiport, or hybrid VATS lobectomy for lung cancer. Thorac Cardiovasc Surg. (2021) 17:10.1055/s-0041-1731824. doi: 10.1055/s-0041-1731824

CrossRef Full Text | Google Scholar

43. Kozian A, Schilling T, Schütze H, Senturk M, Hachenberg T, Hedenstierna G. Ventilatory protective strategies during thoracic surgery: effects of alveolar recruitment maneuver and low-tidal volume ventilation on lung density distribution. Anesthesiology. (2011) 114:1025–35. doi: 10.1097/ALN.0b013e3182164356

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Lohser J. Chapter IS 5. Physiology of the lateral decubitus position, open chest and one-lung ventilation. In Principles and practice of anesthesia for thoracic surgery, Vol. 71, Slinger P, editor, p. 71–80. doi: 10.1007/978-1-4419-0184-2_5.

45. Kozian A, Schilling T, Röcken C, Breitling C, Hachenberg T, Hedenstierna G. Increased alveolar damage after mechanical ventilation in a porcine model of thoracic surgery. J Cardiothorac Vasc Anesth. (2010) 24:617–23. doi: 10.1053/j.jvca.2009.09.016

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Lohser J, Slinger P. Lung injury after one-lung ventilation: a review of the pathophysiologic mechanisms affecting the ventilated and the collapsed lung. Anesth Analg. (2015) 121:302–18. doi: 10.1213/ANE.0000000000000808

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Kozian A, Schilling T, Fredén F, Maripuu E, Röcken C, Strang C, et al. One-lung ventilation induces hyperperfusion and alveolar damage in the ventilated lung: an experimental study. Br J Anaesth. (2008) 100:549–59. doi: 10.1093/bja/aen021

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Gonzalez-Rivas D, Fernandez R, de la Torre M, Rodriguez JL, Fontan L, Molina F. Single-port thoracoscopic lobectomy in a nonintubated patient: the least invasive procedure for major lung resection? Interact Cardiovasc Thorac Surg. (2014) 19:552–5. doi: 10.1093/icvts/ivu209

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Furák J, Barta Z, Lantos J, Ottlakán A, Németh T, Pécsy B, et al. Better intraoperative cardiopulmonary stability and similar postoperative results of spontaneous ventilation combined with intubation than non-intubated thoracic surgery. Gen Thorac Cardiovasc Surg. (2022) 5:10.1007/s11748-021-01768-1. doi: 10.1007/s11748-021-01768-1

CrossRef Full Text | Google Scholar

50. Tacconi F, Pompeo E, Sellitri F, Mineo TC. Surgical stress hormones response is reduced after awake videothoracoscopy. Interact Cardiovasc Thorac Surg. (2010) 10:666–71. doi: 10.1510/icvts.2009.224139

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Mineo TC. Epidural anesthesia in awake thoracic surgery. Eur J Cardiothorac Surg. (2007) 32:13–9. doi: 10.1016/j.ejcts.2007.04.004

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Zhan Y, Chen G, Huang J, Hou B, Liu W, Chen S. Effect of intercostal nerve block combined with general anesthesia on the stress response in patients undergoing minimally invasive mitral valve surgery. Exp Ther Med. (2017) 14:3259–64. doi: 10.3892/etm.2017.4868

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Fares KM, Mohamed SA, Hamza HM, Sayed DM, Hetta DF. Effect of thoracic epidural analgesia on pro-inflammatory cytokines in patients subjected to protective lung ventilation during Ivor Lewis esophagectomy. Pain Phys. (2014) 17:305–15. Erratum in: Pain Phys (2014) 17:475. PMID: 25054390

PubMed Abstract | Google Scholar

54. Alhayyan A, McSorley S, Roxburgh C, Kearns R, Horgan P, McMillan D. The effect of anesthesia on the postoperative systemic inflammatory response in patients undergoing surgery: a systematic review and meta-analysis. Surg Open Sci. (2020) 2(1):1–21. doi: 10.1016/j.sopen.2019.06.001

PubMed Abstract | CrossRef Full Text | Google Scholar

55. De Conno E, Steurer MP, Wittlinger M, Zalunardo MP, Weder W, Schneiter D, et al. Anesthetic-induced improvement of the inflammatory response to one-lung ventilation. Anesthesiology. (2009) 110:1316–26. doi: 10.1097/ALN.0b013e3181a10731

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Kotani N, Hashimoto H, Sessler DI, Yasuda T, Ebina T, Muraoka M, et al. Expression of genes for proinflammatory cytokines in alveolar macrophages during propofol and isoflurane anesthesia. Anesth Analg. (1999) 89:1250–6. doi: 10.1213/00000539-199911000-00032

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Bonavia A, Groff A, Karamchandani K, Singbartl K. Clinical utility of extracorporeal cytokine hemoadsorption therapy: a literature review. Blood Purif. (2018) 46:337–49. doi: 10.1159/000492379

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Akil A, Ziegeler S, Reichelt J, Rehers S, Abdalla O, Semik M, et al. Combined use of CytoSorb and ECMO in patients with severe pneumogenic sepsis. Thorac Cardiovasc Surg. (2021) 69:246–51. doi: 10.1055/s-0040-1708479

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Poli EC, Alberio L, Bauer-Doerries A, Marcucci C, Roumy A, Kirsch M, et al. Cytokine clearance with CytoSorb® during cardiac surgery: a pilot randomized controlled trial. Crit Care. (2019) 23:108. doi: 10.1186/s13054-019-2399-4

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Liu J, Cui F, Pompeo E, Gonzalez-Rivas D, Chen H, Yin W, et al. The impact of non-intubated versus intubated anaesthesia on early outcomes of video-assisted thoracoscopic anatomical resection in non-small-cell lung cancer: a propensity score matching analysis. Eur J Cardiothorac Surg. (2016) 50:920–5. doi: 10.1093/ejcts/ezw160

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Deng HY, Zhu ZJ, Wang YC, Wang WP, Ni PZ, Chen LQ. Non-intubated video-assisted thoracoscopic surgery under loco-regional anaesthesia for thoracic surgery: a meta-analysis. Interact Cardiovasc Thorac Surg. (2016) 23:31–40. doi: 10.1093/icvts/ivw055

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Taioli E, Lee DS, Lesser M, Flores R. Long-term survival in video-assisted thoracoscopic lobectomy vs open lobectomy in lung-cancer patients: a meta-analysis. Eur J Cardiothorac Surg. (2013) 44:591–7. doi: 10.1093/ejcts/ezt051

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Detterbeck F. Thoracoscopic versus open lobectomy debate: the pro argument. Thorac Surg Sci. (2009) 6:Doc04. doi: 10.3205/tss000018

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Zheng J, Liang H, Wang R, Zhong R, Jiang S, Wang W, et al. Perioperative and long-term outcomes of spontaneous ventilation video-assisted thoracoscopic surgery for non-small cell lung cancer. Transl Lung Cancer Res. (2021) 10:3875–87. doi: 10.21037/tlcr-21-629

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Wang ML, How CH, Hung MH, Huang HH, Hsu HH, Cheng YJ, et al. Long-term outcomes after nonintubated versus intubated thoracoscopic lobectomy for clinical stage I non-small cell lung cancer: a propensity-matched analysis. J Formos Med Assoc. (2021) 120:1949–56. doi: 10.1016/j.jfma.2021.04.018

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Furák J, Paróczai D, Burián K, Szabó Z, Zombori T. Oncological advantage of nonintubated thoracic surgery: better compliance of adjuvant treatment after lung lobectomy. Thorac Cancer. (2020) 11:3309–16. doi: 10.1111/1759-7714.13672

CrossRef Full Text | Google Scholar

Keywords: thoracotomy, video-assisted thoracic surgery, non-intubated, one-lung ventilation, systemic inflammation, immune cells, cytokines

Citation: Furák J, Németh T, Lantos J, Fabó C, Géczi T, Zombori-Tóth N, Paróczai D, Szántó Z and Szabó Z (2022) Perioperative Systemic Inflammation in Lung Cancer Surgery. Front. Surg. 9:883322. doi: 10.3389/fsurg.2022.883322

Received: 24 February 2022; Accepted: 2 May 2022;
Published: 20 May 2022.

Edited by:

Federico Tacconi, University of Rome Tor Vergata, Italy

Reviewed by:

Alessandro Palleschi, IRCCS Ca ‘Granda Foundation Maggiore Policlinico Hospital, Italy

Copyright © 2022 Furák, Németh, Lantos, Fabó, Géczi, Zombori-Tóth, Paróczai, Szántó and Szabó. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: József Furák jfurak@gmail.com

Specialty section: This article was submitted to Thoracic Surgery, a section of the journal Frontiers in Surgery

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.