Next Article in Journal
Plant-Growth-Promoting Rhizobacteria Emerging as an Effective Bioinoculant to Improve the Growth, Production, and Stress Tolerance of Vegetable Crops
Next Article in Special Issue
Cell Behavior of Non-Small Cell Lung Cancer Is at EGFR and MicroRNAs Hands
Previous Article in Journal
Polycistronic Artificial microRNA-Mediated Resistance to Cucumber Green Mottle Mosaic Virus in Cucumber
Previous Article in Special Issue
L-Type Amino Acid Transporter 1 Regulates Cancer Stemness and the Expression of Programmed Cell Death 1 Ligand 1 in Lung Cancer Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Safety of Surgery after Neoadjuvant Targeted Therapies in Non-Small Cell Lung Cancer: A Narrative Review

1
Thoracic Surgery Department, Medical University of Gdansk, Sklodowskiej-Curie 3A, 80-211 Gdansk, Poland
2
Department of Oncology and Radiotherapy, Medical University of Gdansk, Sklodowskiej-Curie 3A, 80-211 Gdansk, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(22), 12244; https://doi.org/10.3390/ijms222212244
Submission received: 30 September 2021 / Revised: 7 November 2021 / Accepted: 10 November 2021 / Published: 12 November 2021

Abstract

:
New drugs, including immune checkpoint inhibitors and targeted therapy, have changed the prognosis in a subset of patients with advanced lung cancer, and are now actively investigated in a number of trials with neoadjuvant and adjuvant regimens. However, no phase III randomized studies were published yet. The current narrative review proves that targeted therapies are safe in neoadjuvant approach. Unsurprisingly, administration of therapy is related to an acceptable toxicity profile. Severe adverse events’ rate that rarely compromises outcomes of patients with advanced lung cancer is not that commonly accepted in early lung cancer as it may lead to missing the chance of curative surgery. Among those complications, the most important factors that may limit the use of targeted therapies are severe respiratory adverse events precluding the resection occurring after treatment with some anaplastic lymphoma kinase and rarely after epidermal growth factor receptor tyrosine kinase inhibitors. At this point, in the presented literature assessing the feasibility of neoadjuvant therapies with anaplastic lymphoma kinase and epidermal growth factor receptor tyrosine kinase inhibitors, we did not find any unexpected intraoperative events that would be of special interest to a thoracic surgeon. Moreover, the postoperative course was associated with typical rate of complications.

1. Introduction

Targeted therapies and immunotherapy are the cornerstones of progress in the treatment of advanced lung cancer [1,2,3]. In the era of lung cancer screening, there is a potential for increased incidence of early disease [4,5]. Adverse events of novel neoadjuvant protocols as well as their implications on surgical outcomes are crucial. Surgery is a gold standard in the treatment of non-small-cell lung cancer (NSCLC) being twice as effective in terms of overall survival (OS) when comparing to stereotactic body radiation therapy [6]. Nevertheless, all kinds of ablative therapies should still undergo assessment within clinical trials.
Surgical resection leads to 80–90% of 5-year OS in the IA1-IA3 stages of NSCLC. Poorer results in patients treated in stages IB (73%), IIA (65%), IIB (56%), IIIA (41%), and IIIB (24%) stimulate further efforts for the implementation of complex treatment protocols [7]. In a LACE meta-analysis of individual data of 4584 patients, an adjuvant chemotherapy led to improvement of OS by 5.4% (hazard ratio (HR) 0.89, 95% confidence interval (CI) 0.82–0.96) and disease-free survival (DFS) by 5.8% (HR 0.84 95%, CI 0.78–0.91) [8]. A systematic review of 32 randomized trials involving more than 10,000 participants compared the timing of chemotherapy (neoadjuvant versus adjuvant) and did not demonstrated an advantage of either strategy (OS HR 0.99, 95% CI 0.81–1.21; DFS HR 0.96, 95% CI 0.77–1.20) [9]. Another systematic review of 15 randomized controlled trials including 2385 patients who underwent neoadjuvant chemotherapy disclosed a benefit in terms of improved survival by 5% (HR 0.87, 95% CI 0.78–0.96) [10]. Although equivalent efficacy of preoperative and postoperative chemotherapy strategies are suggested, postoperative chemotherapy remains the standard treatment. Authors suggest that perioperative chemotherapy regimens reached a plateau of their efficacy [8,9,10].
New drugs, including immune checkpoint inhibitors and targeted therapy, have changed the prognosis in a subset of patients with advanced NSCLC [1,2,3], and are now actively investigated in a number of trials with neoadjuvant and adjuvant regimens [11,12,13]. Immunotherapy with or without chemotherapy significantly improves survival of patients with advanced NSCLC depending on the programmed cell death ligand-1 (PD-L1) expression [14]. However, molecular subtypes of NSCLC with driver mutations may derive less benefit from immune checkpoint inhibitors [15,16]. Due to progress in molecular diagnostic technologies, a number of actionable genetic abnormalities can be found in NSCLC and a number of targeted agents are being used in advanced stages of the disease [17]. In NSCLC patients, epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) alterations are most frequently found, especially in no or light smokers.
Activating mutations of epidermal growth factor receptor (EGFR)—exon 19 deletion and exon 21 substitution are present in 10–36% of Caucasian patients with adenocarcinoma of the lung [18,19,20]. Chromosomal rearrangements of ALK are diagnosed less commonly in 4–5% of patients and are not exclusive for adenocarcinoma [21,22]. The frequency of driver mutations in Asian patients is higher [23,24], while the data in other ethnicities are conflicting [25]. Diagnosis of these two groups of common molecular aberrations allows effective use of targeted therapies.
There are attempts to include targeted therapies into complex strategies of treatment of early NSCLC [12]. Toxicity profiles in the treatment of patients with advanced NSCLC are well documented [26,27]. Implementing novel agents not previously used preoperatively leads to new patient safety questions [28]. Safety issues reported in trials with neoadjuvant immunotherapy are monitored and are a matter of interdisciplinary discussion [29,30]. While offering targeted therapies in neoadjuvant settings, their influence on the perioperative period should raise the interest of thoracic surgeons. The key issue of the neoadjuvant approach is safety. Benefits of previously used neoadjuvant chemotherapy are limited [8]. If an unacceptably high complication rate or perioperative mortality is recorded during the trial, it reduces the benefit in OS, which may explain the absence of neoadjuvant therapies in the guidelines [31,32]. The new neoadjuvant therapies have a potential for increased rate of postoperative complications and escalated intraoperative complexity. Before involving the patient in a multidisciplinary treatment approach, other issues must be addressed, e.g., what would be the effect of the neoadjuvant treatment on the delay of the resection? How many patients would not qualify for resection due to progression and/or complications? Additional issues: thoracoscopy-open conversion rate, the extent of surgery, resectability rate, and quality of life will be answered as the body of evidence accumulates.
In clinical trials, the indications for EGFR and ALK tyrosine kinase inhibitors (TKIs) inhibitors are being extended beyond stage IV (Figure 1). Improved DFS in most of the trials with adjuvant EGFR-TKIs in EGFR mutant patients in stage IB-IIIA adenocarcinoma (for example ADAURA trial) promises the change of current practice in these patients [12,33,34,35,36]. Equal effect of neoadjuvant versus adjuvant chemotherapy on OS [8,9,10] is not observed in recently published studies with neoadjuvant and adjuvant targeted therapies [12,33,34,35,36,37,38,39,40,41,42,43,44,45]. However, different treatment protocols and foremost different EGFR mutation status of the patients treated in these trials precludes an exact comparison of the results. Patient’s adherence to the treatment, low neoadjuvant treatment discontinuation rate, low rate of grade 3/4 toxicities leading to significant delay of surgery, and low rate of progression on the neoadjuvant therapy are essential to eventually include targeted therapies in future studies assessing efficacy. Results of ongoing trials with third-generation EGFR-TKI (NeoADAURA NCT04351555, LAURA NCT03521154, NCT03433469) are awaited [46].
In this narrative review, we have analyzed and presented all available scientific papers on the topic of safety of surgery after neoadjuvant targeted therapies. The papers were extracted after searching electronic databases and scanning reference lists from relevant studies. The literature search was not restricted by the time of publication.

2. Typical Complications of EGFR-TKIs

The most common adverse events of administration of EGFR-TKIs in advanced NSCLC are skin toxicity, gastrointestinal toxicity, pulmonary toxicity, hepatic toxicity, and ocular toxicity [26,47,48,49]. EGFR-TKIs are generally well tolerated. However, administration of the drugs may be related to a few toxicities leading to treatment discontinuation and/or dose reduction due to poor patients’ adherence [47,48]. Grade 3/4 toxicities according to the Common Toxicity Criteria for Adverse Events (CTCAE) are reported to occur in 29% of patients treated with gefitinib, 54% of patients treated with erlotinib, and 42% of patients treated with afatinib. Toxicities lead to discontinuation of treatment in <10% of patients and are lower when comparing to chemotherapy [49,50]. Pulmonary toxicity is important from the point of view of eventual lung surgery because development of interstitial lung disease (ILD) being a consequence of administration of EGFR-TKIs significantly impairs physiological fitness [51]. Inadequate pulmonary reserve may result in resignation from surgery [52,53].

2.1. Skin Toxicity

High expression of EGFR in the basal layer of the skin leads to common skin toxicity due to EGFR-TKIs. The incidence of this complication is reported in 66–100% of patients [54,55]. Grade 3 skin toxicity is reported in 2–20% of patients [56]. The most common presentation are: rash, xerosis, erythema, fissures, telangiectasia, pruritus [56]. Most of the recommendations concerning the prevention and treatment of skin toxicity of EGFR-TKIs are based on clinical routine [26]. Topical and systemic use of corticosteroids and antibiotics, topical use of retinoids, vitamin K and emollients are the treatment options [26,54]. The adequate supportive treatment usually allows for relief of symptoms and continuation of treatment.

2.2. Gastrointestinal Toxicity

Impairment of the function of the basal layer of the epithelium of the tongue, esophagus, and intestines during treatment with EGFR-TKIs leads to a second common group of complications [57]. The most significant gastrointestinal adverse event, diarrhea is reported in 21–95% of patients [58]. The onset of diarrhea is usually present in the first few weeks of therapy. Diarrhea is more severe in patients treated with afatinib [59]. Usually loperamide and adequate hydration are needed. In some patients, dose reduction is indicated [60]. Other complications related to the mucosa of the gastrointestinal tract are stomatitis and mucositis [57]. The adverse events are usually manageable with adequate supportive care and eventually dose reduction of the drugs [26,58].

2.3. Pulmonary Toxicity

Pulmonary toxicity is a very rare complication of treatment with EGFR-TKIs; however, it may have important implications from the perspective of physicians planning multidisciplinary treatment. The most common presentations are interstitial lung disease (ILD), pneumonitis, pneumonia, and hemoptysis [61,62,63]. The incidence of any grade pulmonary toxicity is low and occurs in 0.7–4.0% of patients treated with various generation EGFR-TKIs for advanced NSCLC [62,64]. However, if this complication occurs, the mortality in EGFR-TKI-induced ILD or pneumonitis is as high as 36% [65]. The mechanism of pulmonary injury is debated and there are is no causative treatment [26,66]. Risk factors for the development of ILD are preexisting ILD, male sex, older age, history of tobacco smoking, worse performance status [67]. The symptoms characteristic for rapid onset of ILD comprise of worsening dry cough and dyspnea within days to weeks [68,69]. The diagnosis is made on the basis of chest X-ray or high-resolution computerized tomography of the chest after exclusion of pneumonia and cancer progression [70]. Negative cultures obtained in bronchoalveolar lavage obviously do not preclude infectious origin of pulmonary infiltrates, which may require molecular testing for various pathogens responsible for interstitial pneumonia. Treatment discontinuation and steroids are the only interventions offered when ILD after EGFR-TKIs occurs [26,69].

2.4. Cardiac Complications

Even 10% of patients treated with osimertinib and 5% of patients treated with first- and second-generations EGFR-TKIs may be diagnosed with QT interval prolongation [71]. QT prolongation is usually labeled as grade 1 or 2 toxicity and usually does not lead to life-threatening cardiac arrhythmias [64].

2.5. Treatment-Related Mortality

Lethal toxicity of targeted therapies is rare. In advanced-stage NSCLC, the administration of gefitinib, erlotinib, and afatinib is related to deaths in 2.3%, 0.8%, and 1.1%, respectively. The most common cause of death is pneumonitis (65%) [48,49].

3. Neoadjuvant EGFR-TKIs in Early NSCLC

There is a limited experience with neoadjuvant administration of EGFR-TKIs in early NSCLC. No phase III randomized studies were published. Most of the studies deliver a low level of evidence from single-arm trials [37,38,43,44] and retrospective observations [41,42]. We refer only to two small randomized controlled trials directly comparing the neoadjuvant therapies with the use of erlotinib vs. gemcitabine/cisplatin chemotherapy [39,40]. The study protocols evolved as the body of evidence for the efficacy of EGFR-TKIs grew. Primarily, the patients were enrolled in the trials regardless of the presence of driver mutations [40,44]. Further, only patients with tumors harboring sensitizing ex19del/L858R mutations were included in observational or interventional studies [37,39,40,42].
The rate of pathological complete response (pCR) after neoadjuvant use of EGFR-TKIs is low (0–12%) [37,38,39]. Major pathological response (MPR) is noted in 8–24% of patients [38,43,47]. The results of MPR and pCR rates are worse in the case of EGFR-TKIs than with immune checkpoint inhibitors reported recently [72]. Relatively low pathological response rates after neoadjuvant EGFR-TKIs may be a result of wild-type tumors in some studies and a high rate of not radical resections (5–50%) [40,42]. These shortcomings may be corrected by meticulous preoperative qualification of patients and elaborating the study protocol. Neoadjuvant treatment should not aim to downstage tumor characterized by marginal resectability. The primary aim should be improving the OS and DFS by reducing distant relapse instead of reducing the number of local recurrences. On the other hand, limited duration of neoadjuvant therapy may result in insufficient exposition to the EGFR-TKIs. Whether this factor is responsible for better results of adjuvant [12] versus neoadjuvant trials will be answered by future research.
The safety of the EGFR-TKIs in the preoperative setting is high. The toxicity profile of neoadjuvant EGFR-TKIs is the same as in the case of therapy in an advanced stage. Most of the studies report the number of perioperative complications (Table 1). No intraoperative events were mentioned in the analyzed studies. Postoperative mortality was 0%. The complications after surgery were typical and their level was low. Deterioration of pulmonary function after neoadjuvant administration of erlotinib was reported in 2 studies where it was observed in 3–5% of patients [39,44]. In one retrospective study with the use of different agents, 7% of the resections were complicated by chylothorax, which is higher than commonly reported in surgical databases [73,74]. This high rate of complications may be a result of a small study group.

4. Typical Complications in ALK Inhibitors

Treatment with ALK TKIs is usually well tolerated and grade 3/4 toxicities are rarely observed [76]. The most commonly reported adverse events affect the digestive tract, lungs, and heart [77]. Fatigue, visual disorders, peripheral oedema, and neurological disturbances are also common [76,77]. Observed toxicities lead to discontinuation of treatment in 5–12% of patients with NSCLC [78,79]. There are differences in the toxicity profiles between different ALK inhibitors. Gastrointestinal and hepatic toxicities are common during treatment with ceritinib, visual disorders with crizotinib, edema, myalgias and dysgeusia with alectinib, and respiratory complications probably with brigatinib [77].

4.1. Gastrointestinal and Hepatic Complications

Toxicities related to the alimentary tract are very common but usually mild. Diarrhea, nausea, vomiting, and constipation are reported in up to 55% of patients. Grade ≥3 toxicities are observed in less than 3% [79,80,81]. Nausea, vomiting, and diarrhea rates are highest with ceritinib and lowest with alectinib. Constipation occurs most commonly with crizotinib and alectinib comparing to brigatinib and ceritinib. Liver toxicity is usually limited to biochemical changes and acute liver injury with clinical implications is extremely rare [27]. All grades transaminase elevation is reported in 19–33% of patients treated with ALK-TKIs [79,80,81,82] with grade ≥3 occurring in less than 5% of patients [27,78]. Liver toxicity is more frequent in patients treated with ceritinib and crizotinib than with alectinib or brigatinib.

4.2. Pulmonary Complications

Pulmonary toxicity is of significant concern when neoadjuvant therapy is considered, especially in the case of lung cancer surgery. Upper respiratory infections are observed in patients receiving crizotinib and alectinib. Cough is noted in 25% patients receiving brigatinib and 16% of patients receiving crizotinib [80]. However, in the case of underlying advanced NSCLC, it may be difficult to differentiate drug-related adverse events from symptoms of the disease itself. ILD occurs in 0–3% of patients [27,80]. In the review of the four PROFILE trials with crizotinib, the incidence rate of drug-related ILD was assessed as 1.2% [83]. Typical pulmonary complications of brigatinib and ceritinib are pneumonia and respiratory failure with the serious adverse events (SAE) rate up to 14% [78]. Oppositely pulmonary adverse events are rarely observed in patients treated with other ALK-TKIs [84]. Interestingly, brigatinib-related ILD may occur rapidly, even within the first seven days of therapy [80,81].

4.3. Cardiac Complications

Cardiac arrhythmias are the most common complications of ALK-TKIs [85]. Bradycardia and QTc prolongation are transient and rarely lead to cardiac insufficiency [85]. Disfunction of the left ventricle is not reported [86]. Death as a result of arrhythmias has rarely been recorded [27].

4.4. Treatment-Related Mortality

Treatment-related mortality is uncommon and was reported in 0.9% of patients [27]. The most important reasons for treatment-related mortality are pulmonary complications, e.g., ILD, pneumonitis, and pneumonia [80].

5. Neoadjuvant Treatment with ALK Inhibitors in Early NSCLC

The PROFILE 1014 study demonstrated improved objective response rate and progression-free survival in ALK-positive non-squamous NSCLC patients treated with crizotinib comparing to chemotherapy in first-line therapy [87]. Since then, ALK-TKIs became a standard of care in ALK-positive advanced NSCLC. Expanding the indications beyond advanced NSCLC led to the introduction of this therapy to neoadjuvant, induction, and adjuvant protocols in NSCLC. Positive results of trials comparing the second-generation ALK-TKIs–alectinib and brigatinib [80,88] are expected to lead to introduction these agents in individualized treatment protocols.
Neoadjuvant therapy aims to improve the OS by reducing the rate of distant relapse. The patients accepted for neoadjuvant treatment should be upfront resectable, as the local control is not the aim of this approach. In contrast, the downstaging-induction treatment is an approach which may be accepted only in a highly selected population of patients with locally advanced NSCLC. A number of published case studies present a downstaging rather than a neoadjuvant approach due to locally advanced disease [89,90]. It should be underlined that only carefully planned trials, where only patients with objectively resectable tumors are accepted, will aim to prove survival benefit. Currently, high-quality evidence for the neoadjuvant use of ALK inhibitors is lacking. In the largest series of patients operated on after neoadjuvant crizotinib pCR occurred in 18% of patients, pathological response in 91%, pathological nodal downstaging in 27%, and R0 resection rate in 91%. These results together are rationales for an ongoing phase II trial (NCT03088930).
Currently available, heterogeneous data demonstrate that neoadjuvant therapy with ALK-inhibitors is safe (Table 2). A low rate of treatment-related SAE and low rate of postoperative complications were reported. There was no perioperative mortality.

6. Final Remarks

Trials with neoadjuvant administration of immunotherapy or chemo-immunotherapy report much higher rates of obtained pathological responses than trials with neoadjuvant EGFR and ALK-TKIs (21–45% MPR and 7–15% pCR) [94]. Immunotherapy and chemotherapy combinations may lead to even higher rates of pathological response: MPR 57–83% and pCR 33–63% [73,95]. It is hypothesized that the immune checkpoint inhibitors are more efficient in the neoadjuvant approach comparing to adjuvant due to intact immune landscape of the primary tumor and regional lymph nodes. The presence of tumor accompanied by T-cell infiltration may promote an adaptive antitumor immune response. By inducing immune response before surgery, effective immune memory, targeting primary tumor and potential micrometastatic disease, can be generated to spread durable protection against cancer recurrence and distant metastases development [96]. This effect is not utilized in the mechanisms of action, neither of molecularly driven therapies nor chemotherapy. Molecularly driven therapies are limited by timedelaying the surgery. EGFR and ALK mutated tumors are characterized by a lower tumor mutation burden, they present lower numbers of antigens that generate less prominent immunological responses. Those tumors are less susceptible to the immune checkpoint inhibitors, which require constant development of effective therapies also in neoadjuvant protocols [95].
The current narrative review proves that molecularly driven therapies are safe in a neoadjuvant approach. Unsurprisingly, administration of therapy is related to an acceptable toxicity profile. However, the SAEs’ rate that rarely compromises outcomes of patients with advanced NSCLC is not that commonly accepted in early NSCLC, as it may lead to missing the chance of curative surgery. Among those complications, the most important factors that may limit the use of targeted therapies are the grade ≥3 respiratory adverse events precluding the resection [37], occurring after treatment with some ALK inhibitors and rarely after EGFR-TKIs.
Additionally, novel agents involved in neoadjuvant protocols raise concerns of unpredicted intraoperative difficulties (for example perihilar fibrosis). At this point, in the presented literature assessing the feasibility of neoadjuvant therapies with EGFR and ALK inhibitors, we did not find any unexpected intraoperative events that would be of special interest to a thoracic surgeon. Moreover, the postoperative course was associated with the typical rate of complications.

Author Contributions

Conceptualization, T.M., R.D., A.K. and W.R.; methodology, T.M., R.D., A.K. and W.R.; software, R.D.; validation, T.M., R.D., A.K. and W.R.; formal analysis, T.M., R.D., A.K. and W.R.; investigation, T.M., R.D., A.K. and W.R.; resources, T.M., R.D., A.K. and W.R.; data curation, T.M., R.D., A.K. and W.R.; writing—original draft preparation, T.M., R.D., A.K. and W.R.; writing—review and editing, T.M., R.D., A.K. and W.R.; visualization, T.M. and R.D.; supervision, W.R.; project administration, T.M.; funding acquisition, T.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding. Article Processing Charge has been supported by the Medical University of Gdansk.

Conflicts of Interest

Tomasz Marjanski reports having an advisory/consultancy and speaker bureau/expert testimony role with Roche/Genentech. Robert Dziedzic, Anna Kowalczyk, and Witold Rzyman declare noconflict of interest.

Abbreviations

ALKanaplastic lymphoma kinase
ALTAlanine aminotransferase
ASTAspartate aminotransferase
CTCAECommon Toxicity Criteria for Adverse Events
DFSdisease-free survival
EGFRepidermal growth factor receptor
HRhazard ratio
GIgastrointestinal
ILDinterstitial lung disease
MPRmajor pathological response
NRnot reported
NSCLCnon-smallcell lung cancer
OSoverall survival
PALpersistent air leak
pCRpathological complete response
PD-L1programmed cell death ligand-1
SAEserious adverse event
TKItyrosine kinase inhibitors

References

  1. Horn, L.; Pao, W. EML4-ALK: Honing in on a New Target in Non-Small-Cell Lung Cancer. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2009, 27, 4232–4235. [Google Scholar] [CrossRef]
  2. Wang, C.; Thudium, K.B.; Han, M.; Wang, X.T.; Huang, H.; Feingersh, D.; Garcia, C.; Wu, Y.; Kuhne, M.; Srinivasan, M.; et al. In Vitro Characterization of the Anti-PD-1 Antibody Nivolumab, BMS-936558, and in Vivo Toxicology in Non-Human Primates. Cancer Immunol. Res. 2014, 2, 846–856. [Google Scholar] [CrossRef] [Green Version]
  3. Rosell, R.; Carcereny, E.; Gervais, R.; Vergnenegre, A.; Massuti, B.; Felip, E.; Palmero, R.; Garcia-Gomez, R.; Pallares, C.; Sanchez, J.M.; et al. Erlotinib versus Standard Chemotherapy as First-Line Treatment for European Patients with Advanced EGFR Mutation-Positive Non-Small-Cell Lung Cancer (EURTAC): A Multicentre, Open-Label, Randomised Phase 3 Trial. Lancet Oncol. 2012, 13, 239–246. [Google Scholar] [CrossRef]
  4. De Koning, H.J.; van der Aalst, C.M.; de Jong, P.A.; Scholten, E.T.; Nackaerts, K.; Heuvelmans, M.A.; Lammers, J.-W.J.; Weenink, C.; Yousaf-Khan, U.; Horeweg, N.; et al. Reduced Lung-Cancer Mortality with Volume CT Screening in a Randomized Trial. N. Engl. J. Med. 2020, 382, 503–513. [Google Scholar] [CrossRef] [PubMed]
  5. Aberle, D.R.; Adams, A.M.; Berg, C.D.; Black, W.C.; Clapp, J.D.; Fagerstrom, R.M.; Gareen, I.F.; Gatsonis, C.; Marcus, P.M.; Sicks, J.D. Reduced Lung-Cancer Mortality with Low-Dose Computed Tomographic Screening. N. Engl. J. Med. 2011, 365, 395–409. [Google Scholar] [CrossRef] [Green Version]
  6. Cao, C.; Wang, D.; Chung, C.; Tian, D.; Rimner, A.; Huang, J.; Jones, D.R. A Systematic Review and Meta-Analysis of Stereotactic Body Radiation Therapy versus Surgery for Patients with Non–Small Cell Lung Cancer. J. Thorac. Cardiovasc. Surg. 2019, 157, 362–373. [Google Scholar] [CrossRef] [PubMed]
  7. Goldstraw, P.; Chansky, K.; Crowley, J.; Rami-Porta, R.; Asamura, H.; Eberhardt, W.E.E.; Nicholson, A.G.; Groome, P.; Mitchell, A.; Bolejack, V.; et al. The IASLC Lung Cancer Staging Project: Proposals for Revision of the TNM Stage Groupings in the Forthcoming (Eighth) Edition of the TNM Classification for Lung Cancer. J. Thorac. Oncol. 2016, 11, 39–51. [Google Scholar] [CrossRef] [Green Version]
  8. Pignon, J.P.; Tribodet, H.; Scagliotti, G.V.; Douillard, J.Y.; Shepherd, F.A.; Stephens, R.J.; Dunant, A.; Torri, V.; Rosell, R.; Seymour, L.; et al. Lung Adjuvant Cisplatin Evaluation: A Pooled Analysis by the LACE Collaborative Group. J. Clin. Oncol. 2008, 26, 3552–3559. [Google Scholar] [CrossRef]
  9. Lim, E.; Harris, G.; Patel, A.; Adachi, I.; Edmonds, L.; Song, F. Preoperative versus Postoperative Chemotherapy in Patients with Resectable Non-Small Cell Lung Cancer: Systematic Review and Indirect Comparison Meta-Analysis of Randomized Trials. J. Thorac. Oncol. 2009, 4, 1380–1388. [Google Scholar] [CrossRef]
  10. Burdett, S. Preoperative Chemotherapy for Non-Small-Cell Lung Cancer: A Systematic Review and Meta-Analysis of Individual Participant Data. Lancet 2014, 383, 1561–1571. [Google Scholar] [CrossRef] [Green Version]
  11. Felip, E.; Altorki, N.; Zhou, C.; Csőszi, T.; Vynnychenko, I.; Goloborodko, O.; Luft, A.; Akopov, A.; Martinez-Marti, A.; Kenmotsu, H.; et al. Adjuvant Atezolizumab after Adjuvant Chemotherapy in Resected Stage IB-IIIA Non-Small-Cell Lung Cancer (IMpower010): A Randomised, Multicentre, Open-Label, Phase 3 Trial. Lancet 2021, 398, 1344–1357. [Google Scholar] [CrossRef]
  12. Wu, Y.-L.; Tsuboi, M.; He, J.; John, T.; Grohe, C.; Majem, M.; Goldman, J.W.; Laktionov, K.; Kim, S.-W.; Kato, T.; et al. Osimertinib in Resected EGFR -Mutated Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2020, 383, 1711–1723. [Google Scholar] [CrossRef]
  13. Chmielewska, I.; Stencel, K.; Kalinka, E.; Ramlau, R.; Krawczyk, P. Neoadjuvant and Adjuvant Immunotherapy in Non-Small Cell Lung Cancer-Clinical Trials Experience. Cancers 2021, 13, 5048. [Google Scholar] [CrossRef]
  14. Yu, Y.; Zeng, D.; Ou, Q.; Liu, S.; Li, A.; Chen, Y.; Lin, D.; Gao, Q.; Zhou, H.; Liao, W.; et al. Association of Survival and Immune-Related Biomarkers with Immunotherapy in Patients with Non-Small Cell Lung Cancer: A Meta-Analysis and Individual Patient-Level Analysis. JAMA Netw. Open 2019, 2, 1–17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Calles, A.; Riess, J.W.; Brahmer, J.R. Checkpoint Blockade in Lung Cancer With Driver Mutation: Choose the Road Wisely. Am. Soc. Clin. Oncol. Educ. Book Am. Soc. Clin. Oncol Annu. Meet 2020, 40, 372–384. [Google Scholar] [CrossRef]
  16. Yang, H.; Zhu, J.; Xiao, R.; Liu, Y.; Yu, F.; Cai, L.; Qiu, M.; He, F. EGFR Mutation Status in Non-Small Cell Lung Cancer Receiving PD-1/PD-L1 Inhibitors and Its Correlation with PD-L1 Expression: A Meta-Analysis. Cancer Immunol. Immunother. 2021. [Google Scholar] [CrossRef]
  17. Stencel, K.; Chmielewska, I.; Milanowski, J.; Ramlau, R. Non-Small-Cell Lung Cancer: New Rare Targets-New Targeted Therapies-State of The Art and Future Directions. Cancers 2021, 13, 1829. [Google Scholar] [CrossRef] [PubMed]
  18. Krawczyk, P.; Ramlau, R.; Chorostowska-Wynimko, J.; Powrózek, T.; Lewandowska, M.A.; Limon, J.; Wasąg, B.; Pankowski, J.; Kozielski, J.; Kalinka-Warzocha, E.; et al. The Efficacy of EGFR Gene Mutation Testing in Various Samples from Non-Small Cell Lung Cancer Patients: A Multicenter Retrospective Study. J. Cancer Res. Clin. Oncol. 2015, 141, 61–68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Cardenal, F.; Camps, C.; Majem, M.; Lopez-vivanco, G.; Isla, D.; Provencio, M.; Insa, A.; Massuti, B.; Gonzalez-larriba, J.L.; Paz-ares, L.; et al. Screening for epidermal growth factor receptor mutations in lung cancer. N. Engl. J. Med. 2019, 361, 958–967. [Google Scholar]
  20. Lindeman, N.I.; Cagle, P.T.; Beasley, M.B.; Chitale, D.A.; Dacic, S.; Giaccone, G.; Jenkins, R.B.; Kwiatkowski, D.J.; Saldivar, J.S.; Squire, J.; et al. Molecular Testing Guideline for Selection of Lung Cancer Patients for EGFR and ALK Tyrosine Kinase Inhibitors: Guideline from the College of American Pathologists, International Association for the Study of Lung Cancer, and Association for Molecular Patho. J. Thorac. Oncol. 2013, 8, 823–859. [Google Scholar] [CrossRef] [Green Version]
  21. Letovanec, I.; Finn, S.; Zygoura, P.; Smyth, P.; Soltermann, A.; Bubendorf, L.; Speel, E.J.M.; Marchetti, A.; Nonaka, D.; Monkhorst, K.; et al. Evaluation of NGS and RT-PCR Methods for ALK Rearrangement in European NSCLC Patients: Results from the European Thoracic Oncology Platform Lungscape Project. J. Thorac. Oncol. 2018, 13, 413–425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Blackhall, F.H.; Peters, S.; Bubendorf, L.; Dafni, U.; Kerr, K.M.; Hager, H.; Soltermann, A.; O’Byrne, K.J.; Dooms, C.; Sejda, A.; et al. Prevalence and Clinical Outcomes for Patients with ALK-Positive Resected Stage I to III Adenocarcinoma: Results from the European Thoracic Oncology Platform Lungscape Project. J. Clin. Oncol. 2014, 32, 2780–2787. [Google Scholar] [CrossRef]
  23. Shi, Y.; Au, J.S.K.; Thongprasert, S.; Srinivasan, S.; Tsai, C.M.; Khoa, M.T.; Heeroma, K.; Itoh, Y.; Cornelio, G.; Yang, P.C. A Prospective, Molecular Epidemiology Study of EGFR Mutations in Asian Patients with Advanced Non-Small-Cell Lung Cancer of Adenocarcinoma Histology (PIONEER). J. Thorac. Oncol. 2014, 9, 154–162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Lee, H.; Han, J.; Choi, Y.-L. Real-World Analysis of the EGFR Mutation Test in Tissue and Plasma Samples from Non-Small Cell Lung Cancer. Diagnostics 2021, 11, 1695. [Google Scholar] [CrossRef]
  25. Schabath, M.B.; Cress, W.D.; Muñoz-Antonia, T. Racial and Ethnic Differences in the Epidemiology and Genomics of Lung Cancer. Cancer Control. 2016, 23, 338–346. [Google Scholar] [CrossRef] [Green Version]
  26. Płużański, A.; Piórek, A. Side Effects of Tyrosine Kinase Inhibitors—Management Guidelines. Oncol. Clin. Pract. 2016, 12, 113–118. [Google Scholar] [CrossRef]
  27. Zhu, Q.; Hu, H.; Weng, D.S.; Zhang, X.F.; Chen, C.L.; Zhou, Z.Q.; Tang, Y.; Xia, J.C. Pooled Safety Analyses of ALK-TKI Inhibitor in ALK-Positive NSCLC. BMC Cancer 2017, 17, 412. [Google Scholar] [CrossRef] [Green Version]
  28. Yang, C.F.J.; McSherry, F.; Mayne, N.R.; Wang, X.; Berry, M.F.; Tong, B.; Harpole, D.H.; D’Amico, T.A.; Christensen, J.D.; Ready, N.E.; et al. Surgical Outcomes After Neoadjuvant Chemotherapy and Ipilimumab for Non-Small Cell Lung Cancer. Ann. Thorac. Surg. 2018, 105, 924–929. [Google Scholar] [CrossRef] [Green Version]
  29. Stiles, B.M.; Sepesi, B.; Broderick, S.R.; Bott, M.J. Perioperative Considerations for Neoadjuvant Immunotherapy in Non–Small Cell Lung Cancer. J. Thorac. Cardiovasc. Surg. 2020, 160, 1376–1382. [Google Scholar] [CrossRef] [PubMed]
  30. Lee, J.M.; Kim, A.W.; Marjanski, T.; Falcoz, P.-E.; Tsuboi, M.; Wu, Y.-L.; Sun, S.W.; Gitlitz, B.J. Important Surgical and Clinical Endpoints in Neoadjuvant Immunotherapy Trials in Resectable Non-Small Cell Lung Cancer. JTO Clin. Res. Rep. 2021, 2, 100221. [Google Scholar] [CrossRef] [PubMed]
  31. Albain, K.S.; Swann, R.S.; Rusch, V.R.; Turrisi, A.T.; Shepherd, F.A.; Smith, C.; Chen, Y.; Robert, B.; Feins, R.; Gandara, D.R.; et al. Radiotherapy plus chemotherapy with or without surgical resection for stage III non-small-cell lung cancer: A phase III randomised controlled trial. Lancet 2009, 374, 379–386. [Google Scholar] [CrossRef] [Green Version]
  32. Ettinger, D.S.; Wood, D.E.; Aisner, D.L.; Akerley, W.; Bauman, J.; Chirieac, L.R.; D’Amico, T.A.; DeCamp, M.M.; Dilling, T.J.; Dobelbower, M.; et al. Non–Small Cell Lung Cancer, Version 5.2017, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. J. Natl. Compr. Canc. Netw. 2017, 15, 504–535. [Google Scholar] [CrossRef]
  33. Kelly, K.; Altorki, N.K.; Eberhardt, W.E.E.; OBrien, M.E.R.; Spigel, D.R.; Crinò, L.; Tsai, C.M.; Kim, J.H.; Cho, E.K.; Hoffman, P.C.; et al. Adjuvant Erlotinib versus Placebo in Patients with Stage IB-IIIA Nonsmall-Cell Lung Cancer (RADIANT): A Randomized, Double-Blind, Phase III Trial. J. Clin. Oncol. 2015, 33, 4007–4014. [Google Scholar] [CrossRef] [PubMed]
  34. Pennell, N.A.; Neal, J.W.; Chaft, J.E.; Azzoli, C.G.; Jänne, P.A.; Govindan, R.; Evans, T.L.; Costa, D.B.; Wakelee, H.A.; Heist, R.S.; et al. Select: A Phase II Trial of Adjuvant Erlotinib in Patients with Resected Epidermal Growth Factor Receptor-Mutant Non-Small-Cell Lung Cancer. J. Clin. Oncol. 2019, 37, 97–104. [Google Scholar] [CrossRef]
  35. Yue, D.; Xu, S.; Wang, Q.; Li, X.; Shen, Y.; Zhao, H.; Chen, C.; Mao, W.; Liu, W.; Liu, J.; et al. Erlotinib versus Vinorelbine plus Cisplatin as Adjuvant Therapy in Chinese Patients with Stage IIIA EGFR Mutation-Positive Non-Small-Cell Lung Cancer (EVAN): A Randomised, Open-Label, Phase 2 Trial. Lancet Respir. Med. 2018, 6, 863–873. [Google Scholar] [CrossRef]
  36. Zhong, W.Z.; Wang, Q.; Mao, W.M.; Xu, S.T.; Wu, L.; Shen, Y.; Liu, Y.Y.; Chen, C.; Cheng, Y.; Xu, L.; et al. Gefitinib versus Vinorelbine plus Cisplatin as Adjuvant Treatment for Stage II–IIIA (N1–N2) EGFR-Mutant NSCLC (ADJUVANT/CTONG1104): A Randomised, Open-Label, Phase 3 Study. Lancet Oncol. 2018, 19, 139–148. [Google Scholar] [CrossRef]
  37. Xiong, L.; Li, R.; Sun, J.; Lou, Y.; Zhang, W.; Bai, H.; Wang, H.; Shen, J.; Jing, B.; Shi, C.; et al. Erlotinib as Neoadjuvant Therapy in Stage IIIA (N2) EGFR Mutation-Positive Non-Small Cell Lung Cancer: A Prospective, Single-Arm, Phase II Study. Oncologist 2019, 24, 157. [Google Scholar] [CrossRef] [Green Version]
  38. Zhang, Y.; Fu, F.; Hu, H.; Wang, S.; Li, Y.; Hu, H.; Chen, H. Gefitinib as Neoadjuvant Therapy for Resectable Stage II-IIIA Non–Small Cell Lung Cancer: A Phase II Study. J. Thorac. Cardiovasc. Surg. 2021, 161, 434–442. [Google Scholar] [CrossRef] [PubMed]
  39. Zhong, W.Z.; Chen, K.N.; Chen, C.; Gu, C.D.; Wang, J.; Yang, X.N.; Mao, W.M.; Wang, Q.; Qiao, G.B.; Cheng, Y.; et al. Erlotinib versus Gemcitabine plus Cisplatin as Neoadjuvant Treatment of Stage IIIA-N2 EGFR-Mutant Non-Small-Cell Lung Cancer (EMERGING-CTONG 1103): A Randomized Phase II Study. J. Clin. Oncol. 2019, 37, 2235–2245. [Google Scholar] [CrossRef]
  40. Zhong, W.; Yang, X.; Yan, H.; Zhang, X.; Su, J.; Chen, Z.; Liao, R.; Nie, Q.; Dong, S.; Zhou, Q.; et al. Phase II Study of Biomarker-Guided Neoadjuvant Treatment Strategy for IIIA-N2 Non-Small Cell Lung Cancer Based on Epidermal Growth Factor Receptor Mutation Status. J. Hematol. Oncol. 2015, 8, 54. [Google Scholar] [CrossRef] [Green Version]
  41. Bao, Y.; Gu, C.; Xie, H.; Zhao, S.; Xie, D.; Chen, C.; Jiang, G.; Dai, C.; Zhu, Y. Comprehensive Study of Neoadjuvant Targeted Therapy for Resectable Non-Small Cell Lung Cancer. Ann. Transl. Med. 2021, 9, 493. [Google Scholar] [CrossRef] [PubMed]
  42. Lv, C.; Ma, Y.; Feng, Q.; Lu, F.; Chi, Y.; Wu, N.; Fang, J.; Yang, Y. Does Neoadjuvant Targeted Therapy Provide an Opportunity for Resectable EGFR-Mutant Lung Cancer: A Real-World Retrospective Study. J. Thorac. Dis. 2020, 12, 5324–5335. [Google Scholar] [CrossRef] [PubMed]
  43. Tan, A.; Chua, K.P.; Takano, A.; Alvarez, J.; Ong, B.H.; Koh, T.; Aung, Z.W.; Jain, A.; Lai, G.; Tan, W.L.; et al. P1.17-07 Neoadjuvant Gefitinib in Resectable Early Stage EGFR Mutant Non-Small Cell Lung Cancer (NSCLC): A Window-of-Opportunity Study. J. Thorac. Oncol. 2019, 14, S609–S610. [Google Scholar] [CrossRef]
  44. Schaake, E.E.; Kappers, I.; Codrington, H.E.; Valdés Olmos, R.A.; Teertstra, H.J.; Van Pel, R.; Burgers, J.A.; Van Tinteren, H.; Klomp, H.M. Tumor Response and Toxicity of Neoadjuvant Erlotinib in Patients with Early-Stage Non-Small-Cell Lung Cancer. J. Clin. Oncol. 2012, 30, 2731–2738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Wang, T.; Liu, Y.; Zhou, B.; Wang, Z.; Liang, N.; Zhang, Y.; Dong, Z.; Li, J. Effects of Icotinib on Early-Stage Non-Small-Cell Lung Cancer as Neoadjuvant Treatment with Different Epidermal Growth Factor Receptor Phenotypes. Onco. Targets. Ther. 2016, 9, 1735–1741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Tsuboi, M.; Weder, W.; Escriu, C.; Blakely, C.; He, J.; Dacic, S.; Yatabe, Y.; Zeng, L.; Walding, A.; Chaft, J. P03.02 Neoadjuvant Osimertinib with/without Chemotherapy vs Chemotherapy for EGFR Mutated Resectable NSCLC: NeoADAURA. J. Thorac. Oncol. 2021, 16, S258. [Google Scholar] [CrossRef]
  47. Zhao, Y.; Cheng, B.; Chen, Z.; Li, J.; Liang, H.; Chen, Y.; Zhu, F.; Li, C.; Xu, K.; Xiong, S.; et al. Toxicity Profile of Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors for Patients with Lung Cancer: A Systematic Review and Network Meta-Analysis. Crit. Rev. Oncol. Hematol. 2021, 160, 103305. [Google Scholar] [CrossRef]
  48. Yin, X.; Zhao, Z.; Yin, Y.; Shen, C.; Chen, X.; Cai, Z.; Wang, J.; Chen, Z.; Yin, Y.; Zhang, B. Adverse Event Profiles of Epidermal Growth Factor Receptor-Tyrosine Kinase Inhibitors in Cancer Patients: A Systematic Review and Meta-Analysis. Clin. Transl. Sci. 2021, 14, 919–933. [Google Scholar] [CrossRef]
  49. Ding, P.N.; Lord, S.J.; Gebski, V.; Links, M.; Bray, V.; Gralla, R.J.; Yang, J.C.H.; Lee, C.K. Risk of Treatment-Related Toxicities from EGFR Tyrosine Kinase Inhibitors: A Meta-Analysis of Clinical Trials of Gefitinib, Erlotinib, and Afatinib in Advanced EGFR-Mutated Non–Small Cell Lung Cancer. J. Thorac. Oncol. 2017, 12, 633–643. [Google Scholar] [CrossRef] [Green Version]
  50. Mok, T.S.; Wu, Y.; Thongprasert, S.; Yang, C.; Saijo, N.; Sunpaweravong, P.; Han, B.; Margono, B.; Ichinose, Y.; Nishiwaki, Y.; et al. Gefitinib or carboplatin–paclitaxel in pulmonary adenocarcinoma. N. Engl. J. 2009, 361, 947–957. [Google Scholar] [CrossRef]
  51. Margaritopoulos, G.A.; Antoniou, K.M.; Wells, A.U. Comorbidities in Interstitial Lung Diseases. Eur. Respir. Rev. 2017, 26, 1–15. [Google Scholar] [CrossRef]
  52. Brunelli, A.; Charloux, A.; Bolliger, C.T.; Rocco, G.; Sculier, J.-P.; Varela, G.; Licker, M.; Ferguson, M.K.; Faivre-Finn, C.; Huber, R.M.; et al. ERS/ESTS Clinical Guidelines on Fitness for Radical Therapy in Lung Cancer Patients (Surgery and Chemo-Radiotherapy). Eur. Respir. J. 2009, 34, 17–41. [Google Scholar] [CrossRef] [Green Version]
  53. Brunelli, A.; Kim, A.W.; Berger, K.I.; Addrizzo-Harris, D.J. Physiologic Evaluation of the Patient with Lung Cancer Being Considered for Resectional Surgery: Diagnosis and Management of Lung Cancer, 3rd Ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest 2013, 143, e166S–e190S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Melosky, B.; Leighl, N.B.; Rothenstein, J.; Sangha, R.; Stewart, D.; Papp, K. Management of Egfr Tki-Induced Dermatologic Adverse Events. Curr. Oncol. 2015, 22, 123–132. [Google Scholar] [CrossRef] [Green Version]
  55. Giovannini, M.; Gregorc, V.; Belli, C.; Roca, E.; Lazzari, C.; Viganó, M.G.; Serafico, A.; Villa, E. Clinical Significance of Skin Toxicity Due to EGFR-Targeted Therapies. J. Oncol. 2009, 2009, 849051. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Kozuki, T. Skin Problems and EGFR-Tyrosine Kinase Inhibitor. Jpn. J. Clin. Oncol. 2016, 46, 291–298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Holcmann, M.; Sibilia, M. Mechanisms Underlying Skin Disorders Induced by EGFR Inhibitors. Mol. Cell Oncol. 2015, 2, e1004969. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Aw, D.C.W.; Tan, E.H.; Chin, T.M.; Lim, H.L.; Lee, H.Y.; Soo, R.A. Management of Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor-Related Cutaneous and Gastrointestinal Toxicities. Asia Pac. J. Clin. Oncol. 2018, 14, 23–31. [Google Scholar] [CrossRef] [PubMed]
  59. Yang, J.C.H.; Reguart, N.; Barinoff, J.; Köhler, J.; Uttenreuther-Fischer, M.; Stammberger, U.; O’Brien, D.; Wolf, J.; Cohen, E.E. Diarrhea Associated with Afatinib: An Oral ErbB Family Blocker. Expert. Rev. Anticancer Ther. 2013, 13, 729–736. [Google Scholar] [CrossRef]
  60. Hirsh, V.; Blais, N.; Burkes, R.; Verma, S.; Croitoru, K. Management of Diarrhea Induced by Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors. Curr. Oncol. 2014, 21, 329–336. [Google Scholar] [CrossRef] [Green Version]
  61. Peerzada, M.M.; Spiro, T.P.; Daw, H.A. Pip Tyrosine Kinase Pneumonitis. Clin. Adv. Hematol. Oncol. 2011, 9, 824–836. [Google Scholar] [PubMed]
  62. Qi, W.X.; Sun, Y.J.; Shen, Z.; Yao, Y. Risk of Interstitial Lung Disease Associated with EGFR-TKIs in Advanced Non-Small-Cell Lung Cancer: A Meta-Analysis of 24 Phase III Clinical Trials. J. Chemother. 2015, 27, 40–51. [Google Scholar] [CrossRef] [PubMed]
  63. Gong, J.; Zhang, L. Tyrosine Kinase Inhibitors as Induction Therapy in Nonsmall-Cell Lung Cancer. Curr. Opin. Oncol. 2021, 33, 55–58. [Google Scholar] [CrossRef] [PubMed]
  64. Soria, J.-C.; Ohe, Y.; Vansteenkiste, J.; Reungwetwattana, T.; Chewaskulyong, B.; Lee, K.H.; Dechaphunkul, A.; Imamura, F.; Nogami, N.; Kurata, T.; et al. Osimertinib in Untreated EGFR-Mutated Advanced Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 378, 113–125. [Google Scholar] [CrossRef]
  65. Gemma, A.; Kudoh, S.; Ando, M.; Ohe, Y.; Nakagawa, K.; Johkoh, T.; Yamazaki, N.; Arakawa, H.; Inoue, Y.; Ebina, M.; et al. Final Safety and Efficacy of Erlotinib in the Phase 4 POLARSTAR Surveillance Study of 10,708 Japanese Patients with Non-Small-Cell Lung Cancer. Cancer Sci. 2014, 105, 1584–1590. [Google Scholar] [CrossRef] [PubMed]
  66. Hong, D.; Zhang, G.; Zhang, X.; Lian, X. Pulmonary Toxicities of Gefitinib in Patients with Advanced Non-Small-Cell Lung Cancer: A Meta-Analysis of Randomized Controlled Trials. Medicine 2016, 95, e3008. [Google Scholar] [CrossRef]
  67. Shi, L.; Tang, J.; Tong, L.; Liu, Z. Risk of Interstitial Lung Disease with Gefitinib and Erlotinib in Advanced Non-Small Cell Lung Cancer: A Systematic Review and Meta-Analysis of Clinical Trials. Lung Cancer 2014, 83, 231–239. [Google Scholar] [CrossRef]
  68. Akamatsu, H.; Inoue, A.; Mitsudomi, T.; Kobayashi, K.; Nakagawa, K.; Mori, K.; Nukiwa, T.; Nakanishi, Y.; Yamamoto, N. Interstitial Lung Disease Associated with Gefitinib in Japanese Patients with EGFR-Mutated Non-Small-Cell Lung Cancer: Combined Analysis of Two Phase III Trials (NEJ 002 and WJTOG 3405). Jpn J. Clin. Oncol. 2013, 43, 664–668. [Google Scholar] [CrossRef] [PubMed]
  69. Gupta, K.; Uchel, T.; Karamian, G.; Loschner, A. Pulmonary Complications of Tyrosine Kinase Inhibitors and Immune Checkpoint Inhibitors in Patients with Non-Small Cell Lung Cancer. Cancer Treat. Res. Commun. 2021, 28, 100439. [Google Scholar] [CrossRef]
  70. Johkoh, T.; Lee, K.S.; Nishino, M.; Travis, W.D.; Ryu, J.H.; Lee, H.Y.; Ryerson, C.J.; Franquet, T.; Bankier, A.A.; Brown, K.K.; et al. Chest CT Diagnosis and Clinical Management of Drug-Related Pneumonitis in Patients Receiving Molecular Targeting Agents and Immune Checkpoint Inhibitors: A Position Paper from the Fleischner Society. Radiology 2021, 298, 550–566. [Google Scholar] [CrossRef] [PubMed]
  71. Schiefer, M.; Hendriks, L.E.L.; Dinh, T.; Lalji, U.; Dingemans, A.-M.C. Current Perspective: Osimertinib-Induced QT Prolongation: New Drugs with New Side-Effects Need Careful Patient Monitoring. Eur. J. Cancer 2018, 91, 92–98. [Google Scholar] [CrossRef]
  72. Provencio, M.; Nadal, E.; Insa, A.; García-Campelo, M.R.; Casal-Rubio, J.; Dómine, M.; Majem, M.; Rodríguez-Abreu, D.; Martínez-Martí, A.; De Castro Carpeño, J.; et al. Neoadjuvant Chemotherapy and Nivolumab in Resectable Non-Small-Cell Lung Cancer (NADIM): An Open-Label, Multicentre, Single-Arm, Phase 2 Trial. Lancet Oncol. 2020, 21, 1413–1422. [Google Scholar] [CrossRef]
  73. Chen, C.; Wang, Z.; Hao, J.; Hao, X.; Zhou, J.; Chen, N.; Liu, L.; Pu, Q. Chylothorax after Lung Cancer Surgery: A Key Factor Influencing Prognosis and Quality of Life. Ann. Thorac. Cardiovasc. Surg. 2020, 26, 303–310. [Google Scholar] [CrossRef] [PubMed]
  74. Martucci, N.; Tracey, M.; Rocco, G. Postoperative Chylothorax. Thorac. Surg. Clin. 2015, 25, 523–528. [Google Scholar] [CrossRef] [PubMed]
  75. Han, B.; Xiong, L.; Li, R.; Sun, J.; Lou, Y.; Zhang, Y. Erlotinib as Neoadjuvant Treatment in Patients with IIIA-N2 Non-Small Cell Lung Cancer(NSCLC) With Activating Epidermal Growth Factor Receptor (EGFR) Mutation (NCT01217619, ESTERN). Ann. Oncol. 2012, 23, ix413. [Google Scholar] [CrossRef]
  76. Wang, L.; Wang, W. Safety and Efficacy of Anaplastic Lymphoma Kinase Tyrosine Kinase Inhibitors in Non-small Cell Lung Cancer (Review). Oncol. Rep. 2021, 45, 13–28. [Google Scholar] [CrossRef]
  77. Kassem, L.; Shohdy, K.S.; Lasheen, S.; Abdel-Rahman, O.; Ali, A.; Abdel-Malek, R.R. Safety Issues with the ALK Inhibitors in the Treatment of NSCLC: A Systematic Review. Crit. Rev. Oncol. Hematol. 2019, 134, 56–64. [Google Scholar] [CrossRef]
  78. Hou, H.; Sun, D.; Liu, K.; Jiang, M.; Liu, D.; Zhu, J.; Zhou, N.; Cong, J.; Zhang, X. The Safety and Serious Adverse Events of Approved ALK Inhibitors in Malignancies: A Meta-Analysis. Cancer Manag. Res. 2019, 11, 4109–4118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Gristina, V.; Mantia, M.L.; Iacono, F.; Galvano, A.; Russo, A.; Bazan, V. The Emerging Therapeutic Landscape of Alk Inhibitors in Non-Small Cell Lung Cancer. Pharmaceuticals 2020, 13, 474. [Google Scholar] [CrossRef]
  80. Camidge, D.R.; Kim, H.R.; Ahn, M.-J.; Yang, J.C.-H.; Han, J.-Y.; Lee, J.-S.; Hochmair, M.J.; Li, J.Y.-C.; Chang, G.-C.; Lee, K.H.; et al. Brigatinib versus Crizotinib in ALK-Positive Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 379, 2027–2039. [Google Scholar] [CrossRef] [PubMed]
  81. Gettinger, S.N.; Bazhenova, L.A.; Langer, C.J.; Salgia, R.; Gold, K.A.; Rosell, R.; Shaw, A.T.; Weiss, G.J.; Tugnait, M.; Narasimhan, N.I.; et al. Activity and Safety of Brigatinib in ALK-Rearranged Non-Small-Cell Lung Cancer and Other Malignancies: A Single-Arm, Open-Label, Phase 1/2 Trial. Lancet Oncol. 2016, 17, 1683–1696. [Google Scholar] [CrossRef]
  82. Liu, B.; Yuan, M.; Sun, Y.; Cheng, Z.; Zhang, Z.; Hou, S.; Wang, X.; Liu, J. Incidence and Risk of Hepatic Toxicities Associated with Anaplastic Lymphoma Kinase Inhibitors in the Treatment of Non-Small-Cell Lung Cancer: A Systematic Review and Meta-Analysis. Oncotarget 2018, 9, 9480–9488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Yoneda, K.Y.; Scranton, J.R.; Cadogan, M.A.; Tassell, V.; Nadanaciva, S.; Wilner, K.D.; Stollenwerk, N.S. Interstitial Lung Disease Associated With Crizotinib in Patients With Advanced Non–Small Cell Lung Cancer: Independent Review of Four PROFILE Trials. Clin. Lung Cancer 2017, 18, 472–479. [Google Scholar] [CrossRef]
  84. Pellegrino, B.; Facchinetti, F.; Bordi, P.; Silva, M.; Gnetti, L.; Tiseo, M. Lung Toxicity in Non-Small-Cell Lung Cancer Patients Exposed to ALK Inhibitors: Report of a Peculiar Case and Systematic Review of the Literature. Clin. Lung Cancer 2018, 19, e151–e161. [Google Scholar] [CrossRef]
  85. Zaborowska-Szmit, M.; Krzakowski, M.; Kowalski, D.M.; Szmit, S. Cardiovascular Complications of Systemic Therapy in Non-Small-Cell Lung Cancer. J. Clin. Med. 2020, 9, 1268. [Google Scholar] [CrossRef] [PubMed]
  86. Jin, Y.; Xu, Z.; Yan, H.; He, Q.; Yang, X.; Luo, P. A Comprehensive Review of Clinical Cardiotoxicity Incidence of FDA-Approved Small-Molecule Kinase Inhibitors. Front. Pharmacol. 2020, 11, 891. [Google Scholar] [CrossRef]
  87. Solomon, B.J.; Mok, T.; Kim, D.-W.; Wu, Y.-L.; Nakagawa, K.; Mekhail, T.; Felip, E.; Cappuzzo, F.; Paolini, J.; Usari, T.; et al. First-Line Crizotinib versus Chemotherapy in ALK-Positive Lung Cancer. N. Engl. J. Med. 2014, 371, 2167–2177. [Google Scholar] [CrossRef] [Green Version]
  88. Hida, T.; Nokihara, H.; Kondo, M.; Kim, Y.H.; Azuma, K.; Seto, T.; Takiguchi, Y.; Nishio, M.; Yoshioka, H.; Imamura, F.; et al. Alectinib versus Crizotinib in Patients with ALK-Positive Non-Small-Cell Lung Cancer (J-ALEX): An Open-Label, Randomised Phase 3 Trial. Lancet 2017, 390, 29–39. [Google Scholar] [CrossRef]
  89. Tian, Y.; Huang, J.; Li, C.; Jiang, L.; Lin, H.; Lu, P.; Luo, Q.; Yang, G. Perioperative Crizotinib in a Patient with Stage IIIB ALK-Positive Non-Small Cell Lung Cancer: A Case Report. Ann. Transl. Med. 2020, 8, 770. [Google Scholar] [CrossRef]
  90. Xie, X.H.; Zhan, Z.J.; Qin, Y.Y.; Jiang, J.H.; Yin, W.Q.; Zheng, R.H.; Li, S.Y.; Zhou, C.Z. Case Report: Neoadjuvant and Adjuvant Crizotinib Targeted Therapy in Stage IIIA-N2 ALK-Positive Non-Small-Cell Lung Cancer. Front. Oncol. 2021, 11, 785. [Google Scholar] [CrossRef]
  91. Kilickap, S.; Onder, S.; Dizdar, O.; Erman, M.; Uner, A. Short-Time Use of Crizotinib as Neoadjuvant in ALK-Positive Non-Small Cell Lung Carcinoma Can Be a Chance for Resectability. Cancer Chemother. Pharmacol. 2019, 83, 1195–1196. [Google Scholar] [CrossRef] [PubMed]
  92. Zhang, C.; Li, S.L.; Nie, Q.; Dong, S.; Shao, Y.; Yang, X.N.; Wu, Y.L.; Yang, Y.; Zhong, W.Z. Neoadjuvant Crizotinib in Resectable Locally Advanced Non–Small Cell Lung Cancer with ALK Rearrangement. J. Thorac. Oncol. 2019, 14, 726–731. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Imanishi, N.; Yoneda, K.; Taira, A.; Ichiki, Y.; Sato, N.; Hisaoka, M.; Tanaka, F. Major Pathologic Response to Alectinib in ALK-Rearranged Adenocarcinoma of the Lung. Surg. Case Rep. 2018, 4, 19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Lee, J.; Chaft, J.; Nicholas, A.; Patterson, A.; Waqar, S.; Toloza, E.; Haura, E.; Raz, D.; Reckamp, K.; Merritt, R.; et al. PS01.05 Surgical and Clinical Outcomes With Neoadjuvant Atezolizumab in Resectable Stage IB–IIIB NSCLC: LCMC3 Trial Primary Analysis. J. Thorac. Oncol. 2021, 16, S59–S61. [Google Scholar] [CrossRef]
  95. Gavralidis, A.; Justin, F. Immunotherapy in EGFR-Mutant and ALK-Positive Lung Cancer. Cancer J. 2020, 26, 517–524. [Google Scholar] [CrossRef] [PubMed]
  96. Liu, J.; Blake, S.J.; Yong, M.C.R.; Harjunpää, H.; Ngiow, S.F.; Takeda, K.; Young, A.; O’Donnell, J.S.; Allen, S.; Smyth, M.J.; et al. Improved Efficacy of Neoadjuvant Compared to Adjuvant Immunotherapy to Eradicate Metastatic Disease. Cancer Discov. 2016, 6, 1382–1399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. General study protocols of neoadjuvant approach in clinical trials. ALK-TKI anaplastic lymphoma kinase tyrosine kinase inhibitors, EGFR-TKI epidermal growth factor receptor tyrosine kinase inhibitors, NSCLC non-small cell lung cancer.
Figure 1. General study protocols of neoadjuvant approach in clinical trials. ALK-TKI anaplastic lymphoma kinase tyrosine kinase inhibitors, EGFR-TKI epidermal growth factor receptor tyrosine kinase inhibitors, NSCLC non-small cell lung cancer.
Ijms 22 12244 g001
Table 1. Toxicities of neoadjuvant EGFR-TKIs described in the literature. SAE, serious adverse event; EGFR-TKI, epidermal growth factor receptor tyrosine kinase inhibitor; NR, not reported; PAL, persistent air leak; GI, gastrointestinal; ALT, Alanine aminotransferase; AST, Aspartate aminotransferase.
Table 1. Toxicities of neoadjuvant EGFR-TKIs described in the literature. SAE, serious adverse event; EGFR-TKI, epidermal growth factor receptor tyrosine kinase inhibitor; NR, not reported; PAL, persistent air leak; GI, gastrointestinal; ALT, Alanine aminotransferase; AST, Aspartate aminotransferase.
StudyEGFR-TKI in Study GroupNumber of Patients in Study GroupDelay of SurgeryEGFR-TKI ComplicationsIntraoperative Complications in EGFR-TKI GroupR0 Resection Rate in EGFR-TKI GroupPostoperative Complications in EGFR-TKI Group
Schaake et al., 2012 [44]Erlotinib6024 daysRash 61%
Diarrhea 35%
Pneumonitis 5%
0%7% of patients found out to be unresectablePneumonia 2%
PAL 2%
Blood transfusion 3%
Han et al., 2012 [75]Erlotinib756 daysNR0%20% of resected patientsNR
Zhong et al., 2015 [40]Erlotinib246 weeks of treatmentRash 100%
Diarrhea 42%
0%50%0%
Tan et al., 2019 [43]Gefitinib14At least 4 weeks of treatmentAST/ALT elevation 8%NRNRNR
W.Z. Zhong et al., 2019 [39]Erlotinib376 weeks of treatmentRash 76%
Diarrhea 68%
Deterioration of pulmonary function precluding surgery 3%
0%73%Arrhytmia 6%
Lung infection 6%
Poor wound healing 6%
PAL 3%
Pneumothorax 3%
Xiong et al., 2019 [37]Erlotinib1956 daysRash 26%
13% were not operated due to SAE
NR68% of resected patients NR
Lv et al., 2020 [42]Different agents438 weeks of treatmentNR0%95%Chylothorax 7%
Atelectasis 5%
Arrhytmia 2%
Y. Zhang et al., 2021 [38]Gefitinib3561 daysSkin toxicity 69%
GI symptoms 49%
0%12% of patients found out to be stage IV at surgeryChylothorax 12%
Bao et al., 2021 [41]Different agents42NRNRNRNRNR
Table 2. Toxicities of neoadjuvant ALK inhibitors reported in the literature. ALK, anaplastic lymphoma kinase; NR, not reported.
Table 2. Toxicities of neoadjuvant ALK inhibitors reported in the literature. ALK, anaplastic lymphoma kinase; NR, not reported.
StudyALK Inhibitor in Study GroupNumber of Patients Delay of SurgeryALK Inhibitor ComplicationsIntraoperative ComplicationsPostoperative Complications
Tian et al., 2020 [89]Crizotinib112 weeksGrade 1 hepatic damageMild edemaNoneNone
Kilickap et al., 2019 [90]Crizotinib16 weeksNRNRNR
Xie et al., 2021 [91]Gemcitabine, cisplatine, crizotinib12 monthsNoneNRNR
C. Zhang et al., 2019 [92]Crizotinib11Median 41 daysGrade 4 hepatitis 9%NonePneumonia 9%
Dyspnoe 9%
Imanishi et al., 2018 [93]Alectinib13 monthsNoneNoneNone
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Marjanski, T.; Dziedzic, R.; Kowalczyk, A.; Rzyman, W. Safety of Surgery after Neoadjuvant Targeted Therapies in Non-Small Cell Lung Cancer: A Narrative Review. Int. J. Mol. Sci. 2021, 22, 12244. https://doi.org/10.3390/ijms222212244

AMA Style

Marjanski T, Dziedzic R, Kowalczyk A, Rzyman W. Safety of Surgery after Neoadjuvant Targeted Therapies in Non-Small Cell Lung Cancer: A Narrative Review. International Journal of Molecular Sciences. 2021; 22(22):12244. https://doi.org/10.3390/ijms222212244

Chicago/Turabian Style

Marjanski, Tomasz, Robert Dziedzic, Anna Kowalczyk, and Witold Rzyman. 2021. "Safety of Surgery after Neoadjuvant Targeted Therapies in Non-Small Cell Lung Cancer: A Narrative Review" International Journal of Molecular Sciences 22, no. 22: 12244. https://doi.org/10.3390/ijms222212244

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop