2. Materials and Methods
We performed a retrospective cohort study, approved by the ethical institutional review board, of adult locally advanced or metastatic non-small cell lung cancer (NSCLC) patients treated with immune checkpoint inhibitors (ICI) at Coltea Clinical Hospital, Bucharest, Romania, for a period of 5 years from 1 November 2017 (first PD-1 inhibitor used at our institution, Nivolumab) until 30 November 2022. We assessed the baseline characteristics of NSCLC patients, type of immunotherapy treatment and type of response using registered data. For endocrine toxicity evaluation, we integrated exclusion criteria to avoid bias, such as patients with a history of cervical radiotherapy due to head and neck cancer or lymph node metastasis, patients with just one ICI administration and those lost in follow-up. Patients must have been evaluated for thyroid functional tests (TFTs) at baseline and a minimum of one follow-up value. We did not intend to evaluate diabetes mellitus as endocrine immune-related adverse event. Oncologist specialist requested endocrinology evaluation based on modified TFTs or high clinical suspicion of endocrine dysfunction, such as sudden onset of headache of inexplicable cause, digestive intolerance (nausea, vomiting and diarrhea), inexplicable weight loss, orthostatic hypotension with vertigo, hyponatremia.
Endocrine irAEs were characterized as any occurring autoimmune endocrinopathy during treatment with ICIs and related to immunotherapy. Endocrine adverse events not related to immunotherapy were evaluated separately. Patients’ evaluation, diagnostics and treatment were performed by the endocrinologist specialist from our institution. Thyroid laboratory testing was performed at the Coltea Clinical Hospital Laboratory, Bucharest, Romania and included screening for autoimmune thyroiditis i.e., TSH, fT4, T3, and ATPO. Adrenal function reserve was evaluated using basal cortisol and ACTH as well as low dose (1 ug) ACTH stimulation test with cortisol measurements at 30 and 60 min. Our laboratory’s reference ranges for adults for thyroid-stimulating hormone (TSH) are 0.465–4.68 mIU/L (VITROS_ECI), 0.55–4.78 mIU/L (ATTELICA) and 0.465–4.68 mIU/L (VITROS 7600), respectively, and for free thyroxine (FT4) are 10–28.2 pmol/L (VITROS_ECI), 11.5–22.7 pmol/L (ATTELICA) and 10–28.2 pmol/L (VITROS 7600). Cortisol laboratory reference ranges are 123–626 nmol/L (blood sampling must be performed before 10:00 a.m.) and 46.2–389 nmol/L (blood sampling must be performed after 5:00 p.m.) (VITROS 7600). Results of other endocrine tests, if needed and especially requested by endocrinologist, such as thyroid peroxidase (TPO) antibodies, adrenocorticotropic hormone (ACTH), follicle stimulating hormone (FSH), luteinizing hormone (LH) and prolactin, were assessed in external clinic and interpreted based on validated reference ranges.
The main outcomes of the study were descriptive analyses of real-world NSCLC patients treated with ICIs according to NIH protocols in Romania, medical reasons of oncologist to request endocrinology assessment, patients’ access to comprehensive endocrine evaluation, and natural history and frequency of endocrine irAEs in locally advanced or metastatic NSCLC patients treated with ICIs. The secondary endpoint was to take into consideration the probability of any correlation between endocrine irAEs and time of ICI treatment.
4. Discussion
The tremendous effective success of immunotherapy in many malignancies over the last decade has become a routine medical practice in oncology. Nevertheless, discrepancies in different countries may occur. Starting with one major predicament, in Romania, the first ICI reimbursed by the National Insurance House (NIH) and used in NSCLC patients was nivolumab [
14] in November 2017 [
15], and only approved indication was as second-line monotherapy; this is the only used indication of Nivolumab till today. During the next five years, other ICIs entered into clinical practice, such as pembrolizumab in September 2018 [
16], just in first-line monotherapy indication [
17]; atezolizumab in July 2021 [
18] in second-line monotherapy indication [
19]; and finally, durvalumab [
20], in February 2022 [
21]. This reality explains the small proportion of NSCLC patients treated with atezolizumab and durvalumab in our institution in the selected period of this retrospective analysis. Cemiplimab is not reimbursed by NIH for use in NSCLC patients in Romania [
22]. Nowadays, to simplify the process of searching for the reimbursed indication of ICIs throughout the abundant approved European Medicines Agency (EMA) market authorizations, one should check the Romanian Medical Oncology Society’s site [
23] and follow legal steps to correctly fill in an NIH file. Limitations in treating NSCLC patients with all the extensive and effective new antibodies narrow the horizon of patient’s clinical benefits. First-line nivolumab plus ipilimumab combined with two cycles of chemotherapy indication in NSCLC patients [
24,
25] was finally revised and actually during process of approval for reimbursement by NIH since April 2023. Nivolumab was not available in this indication in our study.
Secondly, but equally as important for therapeutic decision making, molecular tests should be carried out; however, in Romania, they are not reimbursed by NIH. European Society of Medical Oncology (ESMO) guidelines recommend molecular tests in advanced non-squamous cell carcinoma and in unusual cases of squamous cell carcinoma [
26]. Standard molecular tests include EGFR mutation status and ALK rearrangements (level of evidence I, A). Testing should also include ROS1 rearrangements, BRAF V600 mutation status, NTRK rearrangements, MET exon 14 skipping mutations, MET amplifications, RET rearrangements, KRAS G12C mutations and HER2 mutations (level of evidence II, A). PD-L1 expression should be systematically determined in advanced NSCLC, and PD-L1 testing is required for first-line pembrolizumab and atezolizumab monotherapy, as well as second-line pembrolizumab (level of evidence I, A) [
27]. In Romania, provided in the form of sponsored vouchers by pharmaceutical companies, minimal biomarker testing comprises EGFR mutation, ALK rearrangements and PD-L1 expression, according to NIH-supported medicines. Limited access to diagnosis and comprehensive biomarker panels in Romania were recently reported by The Swedish Institute for Health Economics, which analyzed access to cancer drugs in Europe [
28].
Furthermore, NSCLC patients treated with ICIs should be evaluated for response. Measurements following response evaluation criteria in solid tumors (RECIST) v1.1 should be used [
29]. In real-world clinical practice, RECIST evaluation is not always detailed in imaging reports (CT scans in the majority of cases). Consequently, the oncologist must judge the response to ICI based on radiologist conclusions, especially to properly assess maintenance criteria for continuing ICI treatment in NIH records. The difficulty for cancer patients to access imaging centers, low-income affordability and time-consuming schedules reduce the opportunity for consistent radiological evaluation. Thus, sporadic evaluation may happen, and may elucidate the proportion of NSCLC patients from our retrospective study who were not assessed for ICI response, which was 30%.
In our retrospective study, we decided to perform several exclusion criteria for the NSCLC cohort to be appraised for endocrine dysfunctions. The rationale behind these constrictive exclusion criteria was to better identify patients to evaluate for endocrine toxicities and avoid bias. Thyroid dysfunction assessment consists of baseline and dynamic TFT, so patients without baseline TFT were excluded. Cervical radiotherapy for head and neck cancers is well known as a bias in thyroid function abnormalities [
30,
31,
32], so it was a pertinent omission judgement. In daily clinical practice, referral to endocrinology evaluation is elective. As endocrine irAEs have been reported in randomized clinical trials [
33,
34,
35,
36,
37,
38] and in updated systematic reviews and meta-analyses [
39,
40], ESMO guidelines recommend TFT at the baseline every 4–6 weeks during treatment, as well as 4–6 weeks after the last cycle [
9].
Endocrine toxicities were detected in all ICI regimens in a large FDA adverse-event-reporting system [
41]. We do not have experience in our retrospective study with anti-CTLA4/PD-L1 antibodies or combinations, so all endocrine irAEs reported in this analysis are related to anti-PD-1 antibody treatment.
Thyroid dysfunction is one of the most common endocrine irAEs in NSCLC patients treated with anti-PD-1 antibodies [
42]. Moreover, in our study, thyroid toxicity was the most frequently reported endocrine irAE (6 out of 13 patients). Brilli et al. testified a significant association between the development of overt thyroid dysfunction and both TSH and positive antithyroid antibody (ATAb) levels at the baseline [
43]. In our study, 97% of patients had normal baseline TSH levels, and ATAb were not performed, as it is not a routine clinical practice. On the other hand, early hypothyroidism during ICI could be predicted by higher baseline TSH levels [
44]. We reported five patients with hypothyroidism, and all of them presented normal TSH baseline values and needed lifelong replacement therapy. Another study, which reported a high incidence of thyroid disorders (21%) with a PD-L1 inhibitor, concluded that elevated thyroid peroxidase (TPO) antibodies at the time of thyroid irAEs might impact the gravity of thyroid dysfunction, thus helping to identify patients who will progress to overt hypothyroidism and require thyroid hormone replacement [
45]. We did not assess TPO antibodies at the baseline as it is not possible in routine oncological practice. The same study outlined that diffusely increased thyroids on
18-fluorodeoxyglucose–positron emission tomography–computed tomography (
18FDG-PET/CT) may predict the occurrence of thyroid dysfunction. This observation is supported by another review that acknowledged the ability of
18FDG-PET/CT to detect autoimmune thyroiditis [
46]. Although the normal thyroid gland usually does not express high 18F-FDG uptake, there is an increased incidental finding of thyroiditis detected by PET/CT. The mechanism of underlying uptake in autoimmune thyroiditis is not well understood, but could possibly be clarified by the activated lymphocyte microenvironment in the infiltrated thyroid. The author also concluded that the detection of thyroiditis by
18FDG-PET/CT may become a prognostic marker. Supplementarily, the probability of immune-mediated thyroiditis associated with a better response to immunotherapy is even stronger if radiologic manifestations are present (evaluated through Doppler ultrasound or conventional thyroid scintigraphy with technetium Tc-99 m pertechnetate imaging methods) [
47]. Wrapping up, clinical research suggests that thyroiditis may be a biomarker for antitumor immune response, emphasizing the need to further characterize its underlying mechanism. In Romania, there are distinctive eligibility conditions for reimbursed
18FDG-PET/CT in the National Oncology Programme [
48], which was prohibitive to our study setting.
Finally, higher TSH levels and the presence of antithyroglobulin autoantibodies (TgAbs) and/or antithyroid peroxidase autoantibodies (TPOAbs) might be used as pre-treatment biomarkers and TgAbs and/or TPOAbs increase and thyroglobulin (Tg) elevation might be applied as during-treatment predictive biomarkers [
49]. The clinical practice value of these specified biomarkers might hypothetically predict thyroid irAEs and manage them appropriately. As mentioned above, in daily clinical practice, we did not assess TgAbs or TPOAbs as pretreatment biomarkers, and we assessed them as during-treatment biomarkers only if indicated by the endocrinologist.
Primary adrenal insufficiency (PAI) is a rare endocrine irAE [
50], and adrenal crisis can be life-threatening, so increased awareness of this endocrine toxicity should be a main apprehension in oncological routine clinical practice for patients treated with ICIs. Nonspecific symptoms such as fatigue, nausea, hypotension, anorexia and dyselectrolytemia in laboratory testing (i.e., hyponatremia, hyperkalemia and hypoglycemia) are usually common in cancer patients in a metastatic setting. So, regular measurements of serum cortisol and adrenocorticotropin (ACTH) could be recommended for cancer patients treated with immunotherapy to smooth an early diagnosis [
51]. In our institution, ACTH laboratory testing is not routinely rated, so it was inaccessible as part of the baseline, and exclusively evaluated on the endocrinologist’s request during the irAE diagnostic algorithm. If necessary, the ACTH test was provided externally. Given the special conditions of transport and storage for ACTH evaluation, no samples were collected in our institution. Another imperative argument in interpreting adrenal disorder in metastatic NSCLC patients is the high tumor burden in adrenal metastasis. In our retrospective study, we reported 22 patients with adrenal metastatic sites and 1 patient with PAI related to this. In randomized clinical trials [
33,
34,
35,
36,
37,
38], there was no description of adrenal metastasis. Subsequently, in real-life medical practice, it is vital to properly identify the etiology of PAI. Even though steroids are formally not indicated during ICI treatment (with concessions for a small dose of prednisone) in everyday oncological practice, urgent situations to use steroids are common. In our report, we define two cases of PAI due to steroid treatment. In this perspective, we emphasize the magnitude of ruling out steroid treatment and adrenal metastasis in cancer patients treated with ICI for accurately recognizing immune-related PAI.
A meta-analysis from 2018 reported hypophysitis more often as endocrine irAE related to anti-CTLA-4 antibodies and a low incidence of 0.4% of patients treated with anti-PD-1 antibodies [
52]. As mentioned before, we do not report experience with anti-CTLA-4 antibodies in this retrospective analysis. In the selected NSCLC population revised for endocrine irAEs, we identified five cases of hypophysitis (4.5%) that could be explained by the superior doubt of a more experienced endocrinologist in our institution. A recent overview of pituitary disorder proposed insightful recommendations for magnetic resonance imaging (MRI) to confirm hypophysitis and to disregard differential diagnoses, especially pituitary metastasis [
53]. Although moderate pituitary enlargement can be rapidly reversible, ICI-induced hypophysitis should not be ruled out due simply to normal imaging. For patients treated with anti-PD-1/anti-PD-L1 agents, no abnormalities on MRI may be common, and this was one potential reason why hypophisitis in patients treated with PD-1/PD-L1 agents might have been underestimated [
41]. In our study, we did not report MRI imaging for hypophisitis, as this imaging method is not a routine-based clinical practice. A retrospective study from Greece that included thyroid and pituitary gland irAEs proved statistical significance of progression-free survival and overall survival for cancer patients treated with immunotherapy, concluding that endocrine toxicity may be a positive predictor of ICI response [
54]. In that context, predictive biomarkers of pituitary gland irAEs are of great interest in the medical community [
55], but are not always of clinical utility, and no biomarker has been proven to effectively foresee the likelihood of evolving a specific endocrine irAE after ICI [
49]. The pathophysiological pattern and underlying mechanism of anti-PD-1/anti-PD-L1 antibody-related hypophysitis were projected, and incriminated type IV hypersensitivity as the main pathway [
56]. Currently, there is an innovative focus of medical research and an emerging concept of onco-immuno-endocrinology. Immune-related hypophysitis is described as paraneoplastic autoimmune hypophysitis, a novel clinical entity [
57].
Immunotherapy in metastatic NSCLC patients substantially improved the prognosis and survival benefit of these patients. The existing reported correlation between immune-related adverse events and improved response to immune checkpoint inhibitors was corroborated with higher survival rates [
58]. In conjunction with the fact that most irAEs can be successfully managed, it became crucially important to identify the patients at risk of adverse events in a timely manner. Predictive and sensitive biomarkers might be useful to adequately stratify the risks in these patients and to monitor them closely for early detection and treatment. Several cytokines, and chemokines such as IL-2, IL-8, granulocyte colony-stimulating factor (G-CSF), IFN-γ, TNF-α, granulocyte–macrophage colony-stimulating factor (GM-CSF), monocyte chemoattractant protein (MCP-1), antithyroglobulin autoantibody (TgAb) and anti-TPO autoantibody (TPOAb), have been identified to be significantly interrelated with thyroid irAEs [
6]. In a recently informed review, Shalit et al. defined pretreatment and during treatment biomarkers for each endocrine dysfunction, such as specific human leukocyte antigen (HLA) alleles, antipituitary antibodies (APAs) and anti-GNAL abs (anti-guanine-nucleotide-binding protein G(olf) subunit alpha antibodies) for pituitary dysfunction [
49]. Remarkably, higher levels of absolute eosinophilic count might have predictive values for endocrine irAEs. We illustrated the biomarkers that we used as baselines and for monitoring, such as TSH and FT4, which are common knowledge in routine clinical practice. Regrettably, we did not have data to publish for TPOAb, ACTH, FSH and LH values to all the patients and not in a monitoring setting by all means. Clinical judgment and vast experience in the oncological background of the specialist endocrinologist in our institution led the management of these patients.
All endocrine irAEs that occurred and were described in our retrospective study were guideline-based treated and follow-up supervised [
59,
60]. In a real-life setting, it is essential to perfectly diagnose endocrine adverse events, as not all of them are immune-mediated, as our study entirely revealed.
In contrast with all the data presented above, we must stress that an ESMO open systematic review and meta-analysis of randomized trials found no statistically significant correlations between the ICI therapy effects on specific irAEs (i.e., endocrine) [
61]. Further data is needed to integrate these results.
The main limitations of our study stem from its retrospective nature, which cannot exclude potential confounders and is also not as accurate regarding the estimation of PFS and other parameters as prospective clinical trials. The retrospective design narrowed the opportunity of extended research and was restrained just to medical information filled up in the archive. It is a well known potential selection bias in a retrospective study but we used this method for database evaluation. As some patients were lost in the follow-up, essential data might have been missed (such as imaging data, laboratory tests, quality of life during ICI). We used several pertinent exclusion criteria to select a homogenous population of NSCLC patients, but this selection could also add potential selection bias. It was a relatively small sample size population evaluated for irAEs, and we did not provide (as they were not available) MRI or imaging data for irAE diagnostics or laboratory analysis of ACTH, ATPO or other biomarkers, as recommended in guidelines. It should also be noted that our study only enrolled patients with NSCLC from Romania, which limits generalizability of the results to other cancer types and/or other countries with potentially different patterns of clinical practice. This single tertiary-level hospital retrospective study might not be in line with medical experience from larger oncological Institutes. In-depth statistical analysis should be provided from this retrospective study to progress the understanding of the prognostic and predictive values of endocrine irAEs in immunotherapy-treated NSCLC patients in Romania.