Next Article in Journal
Role of DNA Methylation Profiles as Potential Biomarkers and Novel Therapeutic Targets in Head and Neck Cancer
Next Article in Special Issue
Implementation of Artificial Intelligence in Personalized Prognostic Assessment of Lung Cancer: A Narrative Review
Previous Article in Journal
Vessel-Guided Mesohepatectomy for Liver Partition and Staged Major Parenchyma-Sparing Hepatectomies with Super-Selective Portal Vein Embolization or Enhanced ALPPS to Achieve R0 Resection for Colorectal Liver Metastases at the Hepatocaval Confluence
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Prognostic Factors in Advanced Non-Small Cell Lung Cancer Patients Treated with Immunotherapy

1
Department of Pulmonary Oncology, AORN dei Colli Monaldi, 80131 Naples, Italy
2
Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy
3
Max-Planck Institute of Molecular Physiology, 44227 Dortmund, Germany
4
Division of Medical Oncology, “S.G. Moscati” Hospital, Contrada Amoretta, 83100 Avellino, Italy
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(19), 4684; https://doi.org/10.3390/cancers15194684
Submission received: 1 August 2023 / Revised: 13 September 2023 / Accepted: 19 September 2023 / Published: 22 September 2023

Abstract

:

Simple Summary

Immunotherapy in the form of Immune Checkpoint Inhibitors which currently represent one of the staples of advanced Non-Small Cell Lung Cancer treatment. However, as of today, no prognostic factors are taken in account to shape immunotherapy-treated patient selection and management according to the most authoritative international guidelines. Therefore, the identification and evaluation of possible prognostic factors in this subset of patients represent an extremely interesting and relevant topic.

Abstract

Taking into account the huge epidemiologic impact of lung cancer (in 2020, lung cancer accounted for 2,206,771 of the cases and for 1,796,144 of the cancer-related deaths, representing the second most common cancer in female patients, the most common cancer in male patients, and the second most common cancer in male and female patients) and the current lack of recommendations in terms of prognostic factors for patients selection and management, this article aims to provide an overview of the current landscape in terms of currently available immunotherapy treatments and the most promising assessed prognostic biomarkers, highlighting the current state-of-the-art and hinting at future challenges.

1. Advanced NSCLC: Epidemiology and Subtypes

The Global Cancer Observatory recorded 19.3 million tumor cases worldwide, as well as 9.9 million tumor-related deaths in 2020. In particular, lung cancer accounts for 2,206,771 of the cases and for 1,796,144 of the cancer-related deaths, representing the second most common cancer in female patients (after breast cancer) and the most common cancer in male patients; in addition, it represents the major cause of death in male patients while being the second most common cause of cancer death in female patients (after breast cancer) [1]. Similarly, according to the American Cancer Society projections for 2022, 1,918,030 tumor cases have occurred in the USA, alongside 609,360 tumor-related deaths. In particular, lung cancer has been responsible for 236,740 of the cases and for 130,180 of the cancer-related deaths, representing the second most common tumor in female patients (after breast cancer), the second most common tumor in male and female patients (after breast cancer), and the second most common tumor in male patients (after prostate cancer); moreover, it has represented the major cause of death in male patients, in females, and in males and females [2,3]. With respect to the histological definition, lung cancer can be classified in two major subtypes: Non-Small Cell Lung Cancer (NSCLC, 85% of all the reported lung cancer cases) and Small Cell Lung Cancer (SCLC, 15% of all the reported lung cancer cases) [4,5,6]. In turn, NSCLC itself is further divided into three subgroups: adenocarcinoma (40–50% of all the reported NSCLC cases), squamous cell carcinoma (20–30% of all the reported NSCLC cases), and large cell carcinoma/not otherwise specified (10–20% of all the reported NSCLC cases) [7,8,9]. With reference to the stage at diagnosis, approximately 50–60% of patients are diagnosed with an advanced disease (stage IV of the TNM classification), approximately 20–25% of patients are diagnosed with a locally advanced disease (stage III of the TNM classification), and approximately 20–25% of patients are diagnosed with an early disease (stage I/II of the TNM classification) [10,11,12].

2. Immunotherapy in the Form of ICIs: Mechanisms of Action

All of the current FDA (U.S. Food and Drug Administration) and/or EMA (European Medicines Agency)-approved and ASCO (American Society of Clinical Oncology) and/or ESMO (European Society for Medical Oncology)-recommended immunotherapeutic drugs for the treatment of advanced NSCLC fall under the category of immune checkpoint inhibitors (ICIs) and are represented by: nivolumab, pembrolizumab, cemiplimab (anti PD-1 mAbs); atezolizumab, and durvalumab (anti PD-L1 mAbs); and ipilimumab and tremelimumab (anti CTLA-4, i.e., Cytotoxic T-Lymphocyte Antigen 4 mAbs). Immune checkpoints (ICs) can be described as receptor–ligand pairs that, upon binding, stimulate (positive ICs) or inhibit (negative ICs) immune system activity—especially cytotoxic T-cells’ activity In a physiological setting, ICs help in maintaining and regulating the immunological tolerance and in fine-tuning the immune system responses. However, cancer cells can exploit negative ICs to escape immune surveillance. As of today, the two best studied and understood negative immune checkpoints are represented by PD-1-PD-L1 and by CTLA-4-B7. The PD-L1 (Programmed Death Ligand 1) protein can be found on the surface of the antigen-presenting cells (APCs) that inhibit T-cells’ activity upon ligation with its receptor PD-1 (Programmed Death Protein 1), which is located on the T-cells’ surface. In this vein, tumor cells can exploit this IC, expressing PD-L1 on their surface. This binding takes place primarily in peripheral tissues. Similarly, the B7.1/2 protein can be found on the surface of APCs, while its receptor CTLA-4 can be located on the surface of T cells; upon ligation, this receptor–ligand pair blocks T-cell activity. This binding takes place primarily in lymph nodes. Similarly to PD-L1, tumor cells can also express CTLA-4 on their surface. In summary, ICIs exert their activity by stopping these receptor–ligand bindings, preventing cancer cells from exploiting these negative ICs, and thus, inhibiting the cytotoxic T-cells’ activity, in turn in turn the immune surveillance and promoting cytotoxic T-cell-mediated cancer cell death [13,14,15,16,17,18,19,20,21,22,23,24,25].
Apart from the established role of the immune checkpoints targeted by currently approved and recommended ICIs, it is worth mentioning that the role of several other ICs is currently being investigated in order to better understand the interplay between ICs in the wider context of the tumor microenvironment and to find new suitable targets. However, as of today, these new targets are still assessed at an investigational level. The T-cell immunoglobulin and mucin-domain containing-3 (TIM-3), lymphocyte activation gene-3 (LAG-3), V-domain Ig suppressor of T-cell activation (VISTA), and OX40 and glucocorticoid-induced TNFR-related protein (GITR) are part of the most promising new ICs; with the exception of OX40 and GITR, they all act as negative ICs members [26,27]. TIM-3 is expressed on the surface of different kinds of T-cells (CD4+ cells, CD8+ cells [28], T-reg cells [29], and Th17 cells) [30], but also on the surface of APCs, and it exerts its activity upon binding the galectin-9 expressed on tumor cells. It is believed to play a role in the development of immune tolerance, and it also seems linked to T-cells’ exhaustion. In fact, its upregulation on CD8+ T cells reduces their activity and [31,32] its upregulation on T reg cells [33] enhances their performance [34]. In addition, TIM-3 is also expressed on Tumor-Infiltrating Lymphocytes and the higher levels of expression seem to be associated with a negative prognostic outcome [35]. The preclinical data point to the fact that a high TIM-3 expression is linked to resistance to anti PD-1 ICIs, thus highlighting a possible role for a dual PD-1/TIM-3 blockade treatment strategy [36,37]. Similarly to TIM-3, LAG3 is also expressed on the surface of T cells (especially TILs) and is also part of another negative IC exerting its activity upon binding its binding partners: the class II MHC, galectin-3, and LSECtin, which are expressed on the surface of tumor cells [38]. While in a physiological setting, the LAG3 pathway is activated in order to regulate T-cell activation [39], and to curb inflammatory responses [40], this pathway can also be exploited by cancer cells. The high LAG-3 expression levels also seem to be related to the resistance to anti PD-1 ICIs and with a negative prognostic outcome [41]; a dual PD-1/LAG-3 blockade therapy could also represent a viable option. VISTA, on the other hand, can act as a ligand when expressed on the surface of APCs and, more importantly, as a receptor when presented on the surface of T cells (TILs included); the binding partner(s) of VISTA expressed on tumor cells are still not fully identified [42,43,44,45,46]. Quite interestingly, while the higher VISTA expression levels seem to be associated with the higher TILs levels, they also seem to be linked to a positive prognostic outcome [47]. Conversely, OX 40 and its ligand OX40L represent a positive IC. OX40 is expressed on the surface of T cells and TILs, while OX40L is expressed on the surface of APCs [48,49,50]. In a physiological setting the OX-40-OX40L pathway mediates the co-stimulatory signals for T-cells activation, survival, and activity [51,52]. TILs with high OX40 expression levels seem to correlate with a positive prognostic outcome [53]. Therefore, OX40 agonizing monoclonal antibodies could represent an interesting strategy aimed at boosting the patients’ immune system against cancer cells. Lastly, in the same vein, GITR (expressed on the surface of T cells and TILs) and its ligand GITRL (expressed on the surface of APCs) represent another positive IC and promote and regulate T-cells’ activation and activity. GITR agonists could provide interesting results in future clinical trials [54,55,56,57,58,59,60,61] (Table 1).
As previously stated, early clinical trials are starting to investigate ICIs targeting these new IC. While it is too early to draw definite conclusions, thanks to the above-mentioned preclinical data, the most promising strategy is represented by a dual targeting of both the traditional and new IC. The notable amount of ongoing trials underlines the great interest towards this new approach (Table 2).

3. Advanced NSCLC: Current Role of Immunotherapy

With reference to the above-mentioned EMA and/or FDA-approved ICIs, according to the most recent ASCO and ESMO guidelines, they are granted their strongest recommendations—alone or in combination with chemotherapy—in the first-line setting for naïve non-oncogene-addicted patients. Therefore, these regimens represent, by far, the most used treatments in a real-world scenario [62,63] (Table 3).

4. Prognostic Factors in Advanced NSCLC Patients Treated with ICIs: Current State of the Art and Future Possibilities

4.1. PD-L1

As the above-mentioned clinical trials and international recommendations show, tumor tissue PD-L1 is the only sub-optimal biomarker whose predictive role is taken into account when considering ICI treatment in advanced NSCLC patients; in fact, while the higher PD-1 expression rates are linked to better responses to ICI treatment, patients that do not express PD-L1 can still experience meaningful responses [76]. The same does not apply, however, to its prognostic value. In fact, while large systematic studies are lacking, the available data seem to show no correlation between the PD-L1 expression levels and the survival outcomes [77]. On the other hand, the different levels of soluble PD-L1 detected in the plasma of NSCLC patients receiving ICI seem to be associated with different responses. While the lower levels of soluble PD-L1 seem to be associated with a favorable prognostic value (longer PFS and OS-, higher ORR—Overall Response Rates), the higher levels seem to be linked to a worse prognosis. A soluble PD-L1 assessment could offer an easily repeatable test (as opposed to tumor tissue biopsy), while also allowing us to better follow the dynamic changes associated with the response to therapy or lack thereof; these assessments, however, are still in an experimental phase and far from standardization [78,79,80,81].

4.2. Tumor Mutational Burden

The role of Tumor Mutational Burden (TMB), meaning the amount of mutations per DNA-coding regions, has also been investigated in advanced NSCLC patients receiving immunotherapy, thanks to the preclinical evidence that higher mutations rates lead to the higher levels of neoantigens (“hot tumors”), and thus, to a higher activation of the immune system, rendering these tumors virtually more susceptible to ICI treatment [82,83,84]. Similarly to PD-L1, almost all the major clinical trials have been focused on assessing the TMB predictive value, yet without definite findings. In fact, while pembrolizumab monotherapy in patients with ≥175 mutations per exome proved to be associated with a positive predictive value in the KEYNOTE-010 and 042 trials, this was not the case in the KEYNOTE-021 trial [85]. The combination of pembrolizumab + chemotherapy was similarly not associated with a positive predictive value in the KEYNOTE-189 and 407 trials [86].
In the same vein, the biggest trial investigating the TMB role in this setting was the CheckMate 227 study, assessing the nivolumab + ipilimumab association in 1189 naïve NSCLC patients with a TMB ≥ 10 mutations per megabase. When compared to standard chemotherapy, this association managed to provide superior results in terms of ORR, PFS, and OS, seemingly establishing a high TMB as a predictive factor in this setting. However, further data demonstrated that this benefit is also associated with a TMB < 10 mutations per megabase, albeit with a lower magnitude (median OS: 23.03 months vs. 16.72 months and 16.20 months vs. 12.42, respectively), thus possibly redefining TMB as a prognostic factor. Once again, the lack of standardization in terms of the cut-off represents a key problem [87,88].

4.3. Neutrophil-to-Lymphocytes Ratio

The preclinical data show that while lymphocytes (and particularly CD8+ ones) are the main agents involved in cancer cells killing both directly and through cytokines (e.g., IFN-γ, IL-2, and IL-12), neutrophiles foster a pro-cancer inflammatory microenvironment that inhibits lymphocytes activity via the production of a palette of cytokines (e.g., IL-10, TNF-α, and VEGF) [89,90,91]. In this sense, an elevated (albeit with different cut-offs, ranging from NLR ≥ 3 to NLR ≥ 5) Neutrophil-to-Lymphocyte Ratio (NLR) has been associated with a poor prognostic value in NSCLC patients receiving chemotherapy, targeted therapy, and immunotherapy. In fact, NSCLC patients with a high NLR receiving ICI consistently experience shorter PFS and OS, as the data coming both from single experiences and meta-analyses show [92,93,94,95,96]. For example, in a recent and large Italian retrospective trial, 252 advanced NSCLC patients receiving pembrolizumab in the first line setting were assessed, employing a cut-off of NLR ≥ 4.8. As a result, patients with a high NLR presented decisively lower OS results when compared to their low NLR counterparts: mOS 7.6 months vs. 34.8 months; interestingly, patients with a very high NLR (>10) were associated with a dismal prognosis: mOS: 3.8 months [97].

4.4. Tumor-Infiltrating Lymphocytes

Building on the same preclinical evidence, tumor-infiltrating lymphocytes (TILs) could represent another valid prognostic factor in advanced NSCLC patients receiving ICIs [89,90,91,98].
This hypothesis has been explored in a recent study in which 26 advanced NSCLC patients receiving immunotherapy were enrolled. Patients whose tumor tissue presented more than 886 CD8+ lymphocyte/mm2 were considered as high expressors, while patients with less than 886 CD8+ lymphocyte/mm2 were considered low expressors. Low expressors presented lower response rates to ICIs when compared to the high expressors (16.7%, vs. 60%); moreover, patients whose tumor tissue presented CD8+/CD4+ ratios lower than two showed lower response rates than patients whose tumor tissue presented a >2 ratio RR (13.3% vs. 43 to 50%) [99]. While interesting, these factors need further validation and standardization in larger trials.

4.5. Combined Scores

Apart from the NLR, other agents responsible for the pro-cancer inflammatory microenvironment are represented by high LDH and C-reactive protein levels; these markers are also evaluated in different scores in combination with other factors that are classically linked to poor prognosis in solid tumors, NSCLC included, like hypoalbuminemia and low platelet count [100,101,102,103,104,105]. As of today, several different combined scores have provided interesting results in NSCLC patients receiving ICIs. The Glasgow Prognostic Score (GPS) combines c-reactive protein levels >10 mg/L and hypoalbuminemia (<35 g/L), defining three different scores: 0: CRP < 10 mg/L and albumin > 35 g/L; 1: CRP ≥ 10 mg/L or albumin < 35 g/L; or 2: CRP > 10 mg/L and albumin < 35 g/L; higher GPS are associated with lower PFS and OS results in patients receiving ICIs [106,107]. Similarly, the Lung Immune Prognostic Index (LIPI) takes into account NLR (>3 as cutoff) and LDH (>upper limit of normal as cutoff), defining three scores: good (0 factor), intermediate (1 factor), and poor (2 factors); in several large studies, higher scores represent a poor prognostic factor [108,109,110].

4.6. Gut Microbiota

An ever-growing set of preclinical data underlines the role of gut microbiota in regulating adaptive and innate immunity, and thus, in partially positively regulating immune responses against cancer cells [111,112,113]. However, as of today, neither the precise mechanisms of action, the specific bacteria, nor the possible positive prognostic value are well understood. Amidst conflicting data and theories, however, some species seem to be more prevalent in patients benefitting from immunotherapy (Bifidobacterium breve/adolescentis/longum) and they seem to exert this favorable effect by improving the production of anti-cancer cytokines like IFN-γ and possibly by cross-reactions between bacteria antigens and tumor ones [114,115,116,117].

4.7. Concomitant Medications

Several types of drugs have been postulated to negatively interfere with the survival outcomes in solid tumors—NSCLC included—treated with ICIs, but Proton Pump Inhibitors (PPIs), corticosteroids (CCS), and antibiotics (ATBs) are the classes that present the most solid evidence and that thus seem to represent some negative prognostic factors. While the CCS’s immune-suppressive role is well documented and understood and suffices to explain their negative impact on ICIs’ activity, PPIs’ and ATBs’ interferences with immunotherapy effects are still being investigated, but their modulating effects on the gut microbiome seem to play a crucial role [118,119,120,121,122,123,124]. With specific respect to advanced NSCLC, a very recent and large retrospective trial enrolling 950 advanced naïve NSCLC receiving pembrolizumab further confirmed these findings, linking shorter OS to CCS’s, ATBs’, and PPIs’ treatment [125].

4.8. High Body Mass Index

Obesity has been shown to be associated with a chronic state of inflammation and this inflammation affects the host’s immune response, influencing ICIs’ efficacy in solid tumors—NSCLC included. In particular, contrarily to most of the factors listed up to this point, obesity (defined as patients’ BMI ≥ 30) seems to act as a positive prognostic factor (longer PFS and OS, higher ORR) in cancer patients because it boosts the host’s immune system, and thus, it ultimately enhances ICIs activity. While the exact mechanisms are still not completely understood, leptin seems to play a key role. In fact, the enhanced leptin signaling found in obese hosts leads to an increased number of exhausted T lymphocytes (exhaustion which could be at least partially mediated via immune checkpoints); however, quite paradoxically, these exhausted T lymphocytes seem to be more easily re-activated and boosted by ICI treatment [126,127,128,129,130,131,132,133,134,135]. A recent and extensive international retrospective study confirmed these findings in a large real-world cohort of 962 naïve advanced NSCLC patients receiving pembrolizumab. In fact, obese patients (BMI ≥ 30) were found to present longer PFS and OS and higher ORR rates [136]. Very interestingly, this favorable prognostic role does not seem to be associated with overweight patients (BMI ≥ 25). In this vein, a recent retrospective Japanese trial assessed 324 advanced NSCLC patients treated with PD-1 inhibitors, dividing them in a non-overweight group (BMI < 25; 86.1%, 279 patients) and in an overweight one (13.9%, 45 patients). As a result, no significant differences were found between the two groups in terms of ORR, PFS, and OS [137].

4.9. Weight Loss

While weight loss and cancer cachexia (mediated by cancer-linked pro-inflammatory cytokines) are well-established negative prognostic factors in advanced NSCLC patients receiving chemotherapy [138,139,140,141], they also seem to represent negative prognostic factors in advanced NSCLC patients receiving ICI. In fact, several prospective and retrospective international trials have consistently linked the lower ORR, PFS, and OS rates to weight loss and cachexia in ICI-receiving patients. While the exact mechanisms behind this relation are not fully understood, the pro-inflammatory microenvironment leading to cachexia seems to play a central role in inhibiting the host’s immune system; in particular, these pro-inflammatory cytokines seem to particularly affect the NLR and the gut microbiome composition [142,143,144,145,146,147] (Table 4).

5. Conclusions

Immunotherapy has represented an incredible revolution in the field of advanced NSCLC treatment; however, as of today (and as the most recent guidelines show), we lack effective biomarkers to both more effectively select and manage patients who could benefit the most from immunotherapy. As the above-mentioned data shows, several different factors have been investigated, but easily assessable ones like NLR, BMI, and the different combined scores not only present the most robust data, but also could allow us to dynamically and periodically re-evaluate patients. The data derived from these biomarkers are unlikely to lead to new strategies and to change our clinical practice; however, they could represent a valid addition to our armamentarium for a more efficient patient selection. In this sense, the comprehensive combined scores or panels could represent the most viable option for optimizing the vast amount of different factors while taking into account their complex interplay. The biggest problem regarding these factors, however, is represented by the current lack of validation and standardization, which should be addressed with larger and prospective trials [148,149].

Author Contributions

Conceptualization, C.G., D.R. and S.N.; writing—original draft preparation L.D.G.; writing—review and editing, C.G., D.R., S.N., L.D.G., A.R. and L.S.; visualization L.D.G., A.R. and L.S.; supervision C.G., D.R. and S.N.; project administration C.G., D.R. and S.N. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

Dr. Gridelli received honoraria as speaker bureau and advisory board member from AstraZeneca, BMS, MSD and Roche.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Oliver, A.L. Lung Cancer: Epidemiology and Screening. Surg. Clin. N. Am. 2022, 102, 335–344. [Google Scholar] [CrossRef] [PubMed]
  3. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics, 2022. CA Cancer J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef] [PubMed]
  4. Sher, T.; Dy, G.K.; Adjei, A.A. Small cell lung cancer. Mayo Clin. Proc. 2008, 83, 355–367. [Google Scholar] [CrossRef] [PubMed]
  5. Kenfield, S.A.; Wei, E.K.; Stampfer, M.J.; Rosner, B.A.; Colditz, G.A. Comparison of aspects of smoking among the four histological types of lung cancer. Tob. Control 2008, 17, 198–204. [Google Scholar] [CrossRef]
  6. Socinski, M.A.; Obasaju, C.; Gandara, D.; Hirsch, F.R.; Bonomi, P.; Bunn, P.; Kim, E.S.; Langer, C.J.; Natale, R.B.; Novello, S.; et al. Clinicopathologic Features of Advanced Squamous NSCLC. J. Thorac. Oncol. 2016, 11, 1411–1422. [Google Scholar] [CrossRef]
  7. Molina, J.R.; Yang, P.; Cassivi, S.D.; Schild, S.E.; Adjei, A.A. Non-small cell lung cancer: Epidemiology, risk factors, treatment, and survivorship. Mayo Clin. Proc. 2008, 83, 584–594. [Google Scholar] [CrossRef]
  8. Ganti, A.K.; Klein, A.B.; Cotarla, I.; Seal, B.; Chou, E. Update of Incidence, Prevalence, Survival, and Initial Treatment in Patients with Non-Small Cell Lung Cancer in the US. JAMA Oncol. 2021, 7, 1824–1832. [Google Scholar] [CrossRef]
  9. Inamura, K. Lung Cancer: Understanding Its Molecular Pathology and the 2015 WHO Classification. Front. Oncol. 2017, 7, 193. [Google Scholar] [CrossRef]
  10. Thai, A.A.; Solomon, B.J.; Sequist, L.V.; Gainor, J.F.; Heist, R.S. Lung cancer. Lancet 2021, 398, 535–554. [Google Scholar] [CrossRef]
  11. Thandra, K.C.; Barsouk, A.; Saginala, K.; Aluru, J.S.; Barsouk, A. Epidemiology of lung cancer. Contemp. Oncol. 2021, 25, 45–52. [Google Scholar] [CrossRef]
  12. Zappa, C.; Mousa, S.A. Non-small cell lung cancer: Current treatment and future advances. Transl. Lung Cancer Res. 2016, 5, 288–300. [Google Scholar] [CrossRef] [PubMed]
  13. Wei, S.C.; Duffy, C.R.; Allison, J.P. Fundamental Mechanisms of Immune Checkpoint Blockade Therapy. Cancer Discov. 2018, 8, 1069–1086. [Google Scholar] [CrossRef] [PubMed]
  14. Granier, C.; De Guillebon, E.; Blanc, C.; Roussel, H.; Badoual, C.; Colin, E.; Saldmann, A.; Gey, A.; Oudard, S.; Tartour, E. Mechanisms of action and rationale for the use of checkpoint inhibitors in cancer. ESMO Open 2017, 2, e000213. [Google Scholar] [CrossRef]
  15. Lee, L.; Gupta, M.; Sahasranaman, S. Immune Checkpoint inhibitors: An introduction to the next-generation cancer immunotherapy. J. Clin. Pharmacol. 2016, 56, 157–169. [Google Scholar] [CrossRef]
  16. Li, B.; Chan, H.L.; Chen, P. Immune Checkpoint Inhibitors: Basics and Challenges. Curr. Med. Chem. 2019, 26, 3009–3025. [Google Scholar] [CrossRef]
  17. Bagchi, S.; Yuan, R.; Engleman, E.G. Immune Checkpoint Inhibitors for the Treatment of Cancer: Clinical Impact and Mechanisms of Response and Resistance. Annu. Rev. Pathol. 2021, 16, 223–249. [Google Scholar] [CrossRef]
  18. Pesce, S.; Trabanelli, S.; Di Vito, C.; Greppi, M.; Obino, V.; Guolo, F.; Minetto, P.; Bozzo, M.; Calvi, M.; Zaghi, E.; et al. Cancer Immunotherapy by Blocking Immune Checkpoints on Innate Lymphocytes. Cancers 2020, 12, 3504. [Google Scholar] [CrossRef]
  19. Alsaab, H.O.; Sau, S. PD-1 and PD-L1 Checkpoint Signaling Inhibition for Cancer Immunotherapy: Mechanism, Combinations, and Clinical Outcome. Front. Pharmacol. 2017, 8, 561. [Google Scholar] [CrossRef]
  20. Xu-Monette, Z.Y.; Zhang, M. PD-1/PD-L1 Blockade: Have We Found the Key to Unleash the Antitumor Immune Response? Front. Immunol. 2017, 8, 1597. [Google Scholar] [CrossRef]
  21. Hui, Y.; Boyle, T.A. PD-L1 Expression in Lung Cancer. J. Thorac. Oncol. 2016, 11, 964–975. [Google Scholar] [CrossRef]
  22. Meyers, D.E.; Bryan, P.M. Targeting the PD-1/PD-L1 axis for the treatment of non-small-cell lung cancer. Curr. Oncol. 2018, 25, e324–e334. [Google Scholar] [CrossRef] [PubMed]
  23. Buchbinder, E.I.; Desai, A. CTLA-4 and PD-1 Pathways: Similarities, Differences, and Implications of Their Inhibition. Am. J. Clin. Oncol. 2016, 39, 98. [Google Scholar] [CrossRef] [PubMed]
  24. Chae, Y.K.; Arya, A. Current landscape and future of dual anti-CTLA4 and PD-1/PD-L1 blockade immunotherapy in cancer; lessons learned from clinical trials with melanoma and non-small cell lung cancer (NSCLC). J. Immunother. Cancer 2018, 6, 39. [Google Scholar] [CrossRef]
  25. Engelhardt, J.J.; Sullivan, T.J. CTLA-4 overexpression inhibits T cell responses through a CD28-B7-dependent mechanism. J. Immunol. 2006, 177, 1052–1061. [Google Scholar] [CrossRef]
  26. Liu, Y.; Chen, P.; Wang, H.; Wu, S.; Zhao, S.; He, Y.; Zhou, C.; Hirsch, F.R. The landscape of immune checkpoints expression in non-small cell lung cancer: A narrative review. Transl. Lung Cancer Res. 2021, 10, 1029–1038. [Google Scholar] [CrossRef]
  27. Krzyżanowska, N.; Wojas-Krawczyk, K.; Milanowski, J.; Krawczyk, P. Future Prospects of Immunotherapy in Non-Small-Cell Lung Cancer Patients: Is There Hope in Other Immune Checkpoints Targeting Molecules? Int. J. Mol. Sci. 2022, 23, 3087. [Google Scholar] [CrossRef]
  28. Monney, L.; Sabatos, C.A.; Gaglia, J.L.; Ryu, A.; Waldner, H.; Chernova, T.; Manning, S.; Greenfield, E.A.; Coyle, A.J.; Sobel, R.A.; et al. Th1-specific cell surface protein Tim-3 regulates macrophage activation and severity of an autoimmune disease. Nature 2002, 415, 536–541. [Google Scholar] [CrossRef]
  29. Gao, X.; Zhu, Y.; Li, G.; Huang, H.; Zhang, G.; Wang, F.; Sun, J.; Yang, Q.; Zhang, X.; Lu, B. TIM-3 expression characterizes regulatory T cells in tumor tissues and is associated with lung cancer progression. PLoS ONE 2012, 7, e30676. [Google Scholar] [CrossRef]
  30. Hastings, W.D.; Anderson, D.E.; Kassam, N.; Koguchi, K.; Greenfield, E.A.; Kent, S.C.; Zheng, X.X.; Strom, T.B.; Hafler, D.A.; Kuchroo, V.K. TIM-3 is expressed on activated human CD4+ T cells and regulates Th1 and Th17 cytokines. Eur. J. Immunol. 2009, 39, 2492–2501. [Google Scholar] [CrossRef]
  31. Zhou, Q.; Munger, M.E.; Veenstra, R.G.; Weigel, B.J.; Hirashima, M.; Munn, D.H.; Murphy, W.J.; Azuma, M.; Anderson, A.C.; Kuchroo, V.K.; et al. Coexpression of Tim-3 and PD-1 identifies a CD8+ T-cell exhaustion phenotype in mice with disseminated acute myelogenous leukemia. Blood 2011, 117, 4501–4510. [Google Scholar] [CrossRef] [PubMed]
  32. Li, Z.; Liu, X.; Guo, R. TIM-3 plays a more important role than PD-1 in the functional impairments of cytotoxic T cells of malignant Schwannomas. Tumour Biol. 2017, 39, 1010428317698352. [Google Scholar] [CrossRef] [PubMed]
  33. Sakuishi, K.; Ngiow, S.F.; Sullivan, J.M.; Teng, M.W.; Kuchroo, V.K.; Smyth, M.J.; Anderson, A.C. TIM3FOXP3 regulatory T cells are tissue-specific promoters of T-cell dysfunction in cancer. Oncoimmunology 2013, 2, e23849. [Google Scholar] [CrossRef]
  34. Gautron, A.S.; Dominguez-Villar, M.; de Marcken, M.; Hafler, D.A. Enhanced suppressor function of TIM-3+ FoxP3+ regulatory T cells. Eur. J. Immunol. 2014, 44, 2703–2711. [Google Scholar] [CrossRef]
  35. Jia, K.; He, Y.; Dziadziuszko, R.; Zhao, S.; Zhang, X.; Deng, J.; Wang, H.; Hirsch, F.R.; Zhou, C.; Yu, H.; et al. T cell immunoglobulin and mucin-domain containing-3 in non-small cell lung cancer. TLCR Transl. Lung Cancer Res. 2019, 8, 895. [Google Scholar] [CrossRef] [PubMed]
  36. Koyama, S.; Akbay, E.A.; Li, Y.Y.; Herter-Sprie, G.S.; Buczkowski, K.A.; Richards, W.G.; Gandhi, L.; Redig, A.J.; Rodig, S.J.; Asahina, H.; et al. Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat. Commun. 2016, 7, 10501. [Google Scholar] [CrossRef] [PubMed]
  37. Sakuishi, K.; Apetoh, L.; Sullivan, J.M.; Blazar, B.R.; Kuchroo, V.K.; Anderson, A.C. Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J. Exp. Med. 2010, 207, 2187–2194. [Google Scholar] [CrossRef]
  38. Turnis, M.E.; Andrews, L.P.; Vignali, D.A. Inhibitory receptors as targets for cancer immunotherapy. Eur. J. Immunol. 2015, 45, 1892–1905. [Google Scholar] [CrossRef] [PubMed]
  39. Workman, C.J.; Rice, D.S.; Dugger, K.J.; Kurschner, C.; Vignali, D.A. Phenotypic analysis of the murine CD4-related glycoprotein, CD223 (LAG-3). Eur. J. Immunol. 2002, 32, 2255–2263. [Google Scholar] [CrossRef]
  40. Xu, F.; Liu, J.; Liu, D.; Liu, B.; Wang, M.; Hu, Z.; Du, X.; Tang, L.; He, F. LSECtin expressed on melanoma cells promotes tumor progression by inhibiting antitumor T-cell responses. Cancer Res. 2014, 74, 3418–3428. [Google Scholar] [CrossRef]
  41. He, Y.; Yu, H.; Rozeboom, L.; Rivard, C.J.; Ellison, K.; Dziadziuszko, R.; Suda, K.; Ren, S.; Wu, C.; Hou, L.; et al. LAG-3 Protein Expression in Non–Small Cell Lung Cancer and Its Relationship with PD-1/PD-L1 and Tumor-Infiltrating Lymphocytes. JTO J. Thorac. Oncol. 2017, 12, 814–823. [Google Scholar] [CrossRef] [PubMed]
  42. Kakavand, H.; Jackett, L.A.; Menzies, A.M.; Gide, T.N.; Carlino, M.S.; Saw, R.P.M.; Thompson, J.F.; Wilmott, J.S.; Long, G.V.; Scolyer, R.A. Negative immune checkpoint regulation by VISTA: A mechanism of acquired resistance to anti-PD-1 therapy in metastatic melanoma patients. Mod. Pathol. 2017, 30, 1666–1676. [Google Scholar] [CrossRef] [PubMed]
  43. Wang, L.; Rubinstein, R.; Lines, J.L.; Wasiuk, A.; Ahonen, C.; Guo, Y.; Lu, L.F.; Gondek, D.; Wang, Y.; Fava, R.A.; et al. VISTA, a novel mouse Ig superfamily ligand that negatively regulates T cell responses. J. Exp. Med. 2011, 208, 577–592. [Google Scholar] [CrossRef] [PubMed]
  44. Nowak, E.C.; Lines, J.L.; Varn, F.S.; Deng, J.; Sarde, A.; Mabaera, R.; Kuta, A.; Le Mercier, I.; Cheng, C.; Noelle, R.J. Immunoregulatory functions of VISTA. Immunol. Rev. 2017, 276, 66–79. [Google Scholar] [CrossRef]
  45. Lines, J.L.; Pantazi, E.; Mak, J.; Sempere, L.F.; Wang, L.; O’Connell, S.; Ceeraz, S.; Suriawinata, A.A.; Yan, S.; Ernstoff, M.S.; et al. VISTA is an immune checkpoint molecule for human T cells. Cancer Res. 2014, 74, 1924–1932. [Google Scholar] [CrossRef]
  46. Le Mercier, I.; Chen, W.; Lines, J.L.; Day, M.; Li, J.; Sergent, P.; Noelle, R.J.; Wang, L. VISTA Regulates the Development of Protective Antitumor Immunity. Cancer Res. 2014, 74, 1933–1944. [Google Scholar] [CrossRef]
  47. Villarroel-Espindola, F.; Yu, X.; Datar, I.; Mani, N.; Sanmamed, M.; Velcheti, V.; Syrigos, K.; Toki, M.; Zhao, H.; Chen, L.; et al. Spatially Resolved and Quantitative Analysis of VISTA/PD-1H as a Novel Immunotherapy Target in Human Non-Small Cell Lung Cancer. Clin. Cancer Res. 2018, 24, 1562–1573. [Google Scholar] [CrossRef]
  48. Baumann, R.; Yousefi, S.; Simon, D.; Russmann, S.; Mueller, C.; Simon, H.U. Functional expression of CD134 by neutrophils. Eur. J. Immunol. 2004, 34, 2268–2275. [Google Scholar] [CrossRef]
  49. Valzasina, B.; Guiducci, C.; Dislich, H.; Killeen, N.; Weinberg, A.D.; Colombo, M.P. Triggering of OX40 (CD134) on CD4+CD25+ T cells blocks their inhibitory activity: A novel regulatory role for OX40 and its comparison with GITR. Blood 2005, 105, 2845–2851. [Google Scholar] [CrossRef]
  50. Baum, P.R.; Gayle, R.B., 3rd; Ramsdell, F.; Srinivasan, S.; Sorensen, R.A.; Watson, M.L.; Seldin, M.F.; Baker, E.; Sutherland, G.R.; Clifford, K.N.; et al. Molecular characterization of murine and human OX40/OX40 ligand systems: Identification of a human OX40 ligand as the HTLV-1-regulated protein gp34. EMBO J. 1994, 13, 3992–4001. [Google Scholar] [CrossRef]
  51. Crotty, S. T follicular helper cell differentiation, function, and roles in disease. Immunity 2014, 41, 529–542. [Google Scholar] [CrossRef] [PubMed]
  52. Rogers, P.R.; Song, J.; Gramaglia, I.; Killeen, N.; Croft, M. OX40 promotes Bcl-xL and Bcl-2 expression and is essential for long-term survival of CD4 T cells. Immunity 2001, 15, 445–455. [Google Scholar] [CrossRef] [PubMed]
  53. He, Y.; Zhang, X.; Jia, K.; Dziadziuszko, R.; Zhao, S.; Deng, J.; Wang, H.; Hirsch, F.R.; Zhou, C. OX40 and OX40L protein expression of tumor infiltrating lymphocytes in non-small cell lung cancer and its role in clinical outcome and relationships with other immune biomarkers. Transl. Lung Cancer Res. 2019, 8, 352–366. [Google Scholar] [CrossRef] [PubMed]
  54. Snell, L.M.; McPherson, A.J.; Lin, G.H.; Sakaguchi, S.; Pandolfi, P.P.; Riccardi, C.; Watts, T.H. CD8 T cell-intrinsic GITR is required for T cell clonal expansion and mouse survival following severe influenza infection. J. Immunol. 2010, 185, 7223–7234. [Google Scholar] [CrossRef]
  55. Nocentini, G.; Riccardi, C. GITR: A modulator of immune response and inflammation. Adv. Exp. Med. Biol. 2009, 647, 156–173. [Google Scholar]
  56. Snell, L.M.; Lin, G.H.; McPherson, A.J.; Moraes, T.J.; Watts, T.H. T-cell intrinsic effects of GITR and 4-1BB during viral infection and cancer immunotherapy. Immunol. Rev. 2011, 244, 197–217. [Google Scholar] [CrossRef]
  57. Kanamaru, F.; Youngnak, P.; Hashiguchi, M.; Nishioka, T.; Takahashi, T.; Sakaguchi, S.; Ishikawa, I.; Azuma, M. Costimulation via glucocorticoid-induced TNF receptor in both conventional and CD25+ regulatory CD4+ T cells. J. Immunol. 2004, 172, 7306–7314. [Google Scholar] [CrossRef]
  58. Ko, K.; Yamazaki, S.; Nakamura, K.; Nishioka, T.; Hirota, K.; Yamaguchi, T.; Shimizu, J.; Nomura, T.; Chiba, T.; Sakaguchi, S. Treatment of advanced tumors with agonistic anti-GITR mAb and its effects on tumor-infiltrating Foxp3+CD25+CD4+ regulatory T cells. J. Exp. Med. 2005, 202, 885–891. [Google Scholar] [CrossRef]
  59. Clouthier, D.L.; Watts, T.H. Cell-specific and context-dependent effects of GITR in cancer, autoimmunity, and infection. Cytokine Growth Factor Rev. 2014, 25, 91–106. [Google Scholar] [CrossRef]
  60. Aida, K.; Miyakawa, R.; Suzuki, K.; Narumi, K.; Udagawa, T.; Yamamoto, Y.; Chikaraishi, T.; Yoshida, T.; Aoki, K. Suppression of Tregs by anti-glucocorticoid induced TNF receptor antibody enhances the antitumor immunity of interferon-alpha gene therapy for pancreatic cancer. Cancer Sci. 2014, 105, 159–167. [Google Scholar] [CrossRef]
  61. Zhou, P.; L’italien, L.; Hodges, D.; Schebye, X.M. Pivotal roles of CD4+ effector T cells in mediating agonistic anti-GITR mAb-induced-immune activation and tumor immunity in CT26 tumors. J. Immunol. 2007, 179, 7365–7375. [Google Scholar] [CrossRef]
  62. Hendriks, L.E.; Kerr, K.M.; Menis, J.; Mok, T.S.; Nestle, U.; Passaro, A.; Peters, S.; Planchard, D.; Smit, E.F.; Solomon, B.J.; et al. Non-oncogene-addicted metastatic non-small-cell lung cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann. Oncol. 2023, 34, 358–376. [Google Scholar] [CrossRef] [PubMed]
  63. Singh, N.; Temin, S.; Baker, S., Jr.; Blanchard, E.; Brahmer, J.R.; Celano, P.; Duma, N.; Ellis, P.M.; Elkins, I.B.; Haddad, R.Y.; et al. Therapy for Stage IV Non-Small-Cell Lung Cancer without Driver Alterations: ASCO Living Guideline. J. Clin. Oncol. 2022, 40, 3323–3343. [Google Scholar] [CrossRef] [PubMed]
  64. Reck, M.; Rodríguez-Abreu, D.; Robinson, A.G.; Hui, R.; Csőszi, T.; Fülöp, A.; Gottfried, M.; Peled, N.; Tafreshi, A.; Cuffe, S.; et al. Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2016, 375, 1823–1833. [Google Scholar] [CrossRef] [PubMed]
  65. Herbst, R.S.; Giaccone, G.; de Marinis, F.; Reinmuth, N.; Vergnenegre, A.; Barrios, C.H.; Morise, M.; Felip, E.; Andric, Z.; Geater, S.; et al. Atezolizumab for First-Line Treatment of PD-L1-Selected Patients with NSCLC. N. Engl. J. Med. 2020, 383, 1328–1339. [Google Scholar] [CrossRef]
  66. Sezer, A.; Kilickap, S.; Gümüş, M.; Bondarenko, I.; Özgüroğlu, M.; Gogishvili, M.; Turk, H.M.; Cicin, I.; Bentsion, D.; Gladkov, O.; et al. Cemiplimab monotherapy for first-line treatment of advanced non-small-cell lung cancer with PD-L1 of at least 50%: A multicentre, open-label, global, phase 3, randomised, controlled trial. Lancet 2021, 397, 592–604. [Google Scholar] [CrossRef]
  67. Mok, T.S.K.; Wu, Y.L.; Kudaba, I.; Kowalski, D.M.; Cho, B.C.; Turna, H.Z.; Castro, G., Jr.; Srimuninnimit, V.; Laktionov, K.K.; Bondarenko, I.; et al. Pembrolizumab versus chemotherapy for previously untreated, PD-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (KEYNOTE-042): A randomised, open-label, controlled, phase 3 trial. Lancet 2019, 393, 1819–1830. [Google Scholar] [CrossRef]
  68. Paz-Ares, L.; Ciuleanu, T.E.; Cobo, M.; Schenker, M.; Zurawski, B.; Menezes, J.; Richardet, E.; Bennouna, J.; Felip, E.; Juan-Vidal, O.; et al. First-line nivolumab plus ipilimumab combined with two cycles of chemotherapy in patients with non-small-cell lung cancer (CheckMate 9LA): An international, randomised, open-label, phase 3 trial. Lancet Oncol. 2021, 22, 198–211, Erratum in Lancet Oncol. 2021, 22, e92. [Google Scholar] [CrossRef]
  69. Gogishvili, M.; Melkadze, T.; Makharadze, T.; Giorgadze, D.; Dvorkin, M.; Penkov, K.; Laktionov, K.; Nemsadze, G.; Nechaeva, M.; Rozhkova, I.; et al. Cemiplimab plus chemotherapy versus chemotherapy alone in non-small cell lung cancer: A randomized, controlled, double-blind phase 3 trial. Nat. Med. 2022, 28, 2374–2380. [Google Scholar] [CrossRef]
  70. Johnson, M.L.; Cho, B.C.; Luft, A.; Alatorre-Alexander, J.; Geater, S.L.; Laktionov, K.; Kim, S.W.; Ursol, G.; Hussein, M.; Lim, F.L.; et al. Durvalumab with or without Tremelimumab in Combination with Chemotherapy as First-Line Therapy for Metastatic Non-Small-Cell Lung Cancer: The Phase III POSEIDON Study. J. Clin. Oncol. 2023, 41, 1213–1227. [Google Scholar] [CrossRef]
  71. Hellmann, M.D.; Paz-Ares, L.; Bernabe Caro, R.; Zurawski, B.; Kim, S.W.; Carcereny Costa, E.; Park, K.; Alexandru, A.; Lupinacci, L.; de la Mora Jimenez, E.; et al. Nivolumab plus Ipilimumab in Advanced Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2019, 381, 2020–2031. [Google Scholar] [CrossRef] [PubMed]
  72. Gandhi, L.; Rodríguez-Abreu, D.; Gadgeel, S.; Esteban, E.; Felip, E.; De Angelis, F.; Domine, M.; Clingan, P.; Hochmair, M.J.; Powell, S.F.; et al. Pembrolizumab plus Chemotherapy in Metastatic Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 378, 2078–2092. [Google Scholar] [CrossRef] [PubMed]
  73. West, H.; McCleod, M.; Hussein, M.; Morabito, A.; Rittmeyer, A.; Conter, H.J.; Kopp, H.G.; Daniel, D.; McCune, S.; Mekhail, T.; et al. Atezolizumab in combination with carboplatin plus nab-paclitaxel chemotherapy compared with chemotherapy alone as first-line treatment for metastatic non-squamous non-small-cell lung cancer (IMpower130): A multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2019, 20, 924–937. [Google Scholar] [CrossRef] [PubMed]
  74. Socinski, M.A.; Jotte, R.M.; Cappuzzo, F.; Orlandi, F.; Stroyakovskiy, D.; Nogami, N.; Rodríguez-Abreu, D.; Moro-Sibilot, D.; Thomas, C.A.; Barlesi, F.; et al. Atezolizumab for First-Line Treatment of Metastatic Nonsquamous NSCLC. N. Engl. J. Med. 2018, 378, 2288–2301. [Google Scholar] [CrossRef]
  75. Paz-Ares, L.; Luft, A.; Vicente, D.; Tafreshi, A.; Gümüş, M.; Mazières, J.; Hermes, B.; Çay Şenler, F.; Csőszi, T.; Fülöp, A.; et al. Pembrolizumab plus Chemotherapy for Squamous Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 379, 2040–2051. [Google Scholar] [CrossRef]
  76. Davis, A.A.; Patel, V.G. The role of PD-L1 expression as a predictive biomarker: An analysis of all US Food and Drug Administration (FDA) approvals of immune checkpoint inhibitors. J. Immunother. Cancer 2019, 7, 278. [Google Scholar] [CrossRef]
  77. Fumet, J.D.; Richard, C.; Ledys, F.; Klopfenstein, Q.; Joubert, P.; Routy, B.; Truntzer, C.; Gagné, A.; Hamel, M.A.; Guimaraes, C.F.; et al. Prognostic and predictive role of CD8 and PD-L1 determination in lung tumor tissue of patients under anti-PD-1 therapy. Br. J. Cancer 2018, 119, 950–960, Erratum in Br. J. Cancer 2019, 121, 283. [Google Scholar] [CrossRef]
  78. Oh, S.Y.; Kim, S.; Keam, B.; Kim, T.M.; Kim, D.W.; Heo, D.S. Soluble PD-L1 is a predictive and prognostic biomarker in advanced cancer patients who receive immune checkpoint blockade treatment. Sci. Rep. 2021, 11, 19712. [Google Scholar] [CrossRef]
  79. Okuma, Y.; Wakui, H.; Utsumi, H.; Sagawa, Y.; Hosomi, Y.; Kuwano, K.; Homma, S. Soluble Programmed Cell Death Ligand 1 as a Novel Biomarker for Nivolumab Therapy for Non-Small-cell Lung Cancer. Clin. Lung Cancer 2018, 19, 410–417. [Google Scholar] [CrossRef]
  80. Costantini, A.; Julie, C.; Dumenil, C.; Hélias-Rodzewicz, Z.; Tisserand, J.; Dumoulin, J.; Giraud, V.; Labrune, S.; Chinet, T.; Emile, J.F.; et al. Predictive role of plasmatic biomarkers in advanced non-small cell lung cancer treated by nivolumab. Oncoimmunology 2018, 7, e1452581. [Google Scholar] [CrossRef]
  81. Costantini, A.; Takam Kamga, P.; Dumenil, C.; Chinet, T.; Emile, J.F.; Giroux Leprieur, E. Plasma Biomarkers and Immune Checkpoint Inhibitors in Non-Small Cell Lung Cancer: New Tools for Better Patient Selection? Cancers 2019, 11, 1269. [Google Scholar] [CrossRef] [PubMed]
  82. Bravaccini, S.; Bronte, G.; Ulivi, P. TMB in NSCLC: A Broken Dream? Int. J. Mol. Sci. 2021, 22, 6536. [Google Scholar] [CrossRef] [PubMed]
  83. Wang, P.; Chen, Y.; Wang, C. Beyond Tumor Mutation Burden: Tumor Neoantigen Burden as a Biomarker for Immunotherapy and Other Types of Therapy. Front. Oncol. 2021, 11, 672677. [Google Scholar] [CrossRef] [PubMed]
  84. Maleki Vareki, S. High and low mutational burden tumors versus immunologically hot and cold tumors and response to immune checkpoint inhibitors. J. Immunother. Cancer 2018, 6, 157. [Google Scholar] [CrossRef] [PubMed]
  85. Herbst, R.S.L.; Kowalski, G.; Nishio, M.; Tsk, M. Association between tissue TMB (tTMB) and clinical outcomes with pembrolizumab monotherapy (pembro) in PD-L1 positive advanced NSCLC in the KEYNOTE-010 and -042 trials. Ann. Oncol. 2019, 30 (Suppl. S5), v851–v934. [Google Scholar] [CrossRef]
  86. Paz-Ares, L.; Vicente, D.; Tafreshi, A.; Robinson, A.; Parra, H.S.; Mazières, J.; Hermes, B.; Cicin, I.; Medgyasszay, B.; Beatrix, B.; et al. Pembrolizumab (pembro) + chemotherapy (chemo) in metastatic squamous NSCLC: Final analysis and progression after the next line of therapy (PFS2) in KEYNOTE-407. Ann. Oncol. 2019, 30, v918–v919. [Google Scholar] [CrossRef]
  87. Hellmann, M.D.; Ciuleanu, T.E.; Pluzanski, A.; Lee, J.S.; Otterson, G.A.; Audigier-Valette, C.; Minenza, E.; Linardou, H.; Burgers, S.; Salman, P.; et al. Nivolumab plus Ipilimumab in Lung Cancer with a High Tumor Mutational Burden. N. Engl. J. Med. 2018, 378, 2093–2104. [Google Scholar] [CrossRef]
  88. BMS Press Release [Internet] Bristol-Myers Squibb Provides Update on the Ongoing Regulatory Review of Opdivo Plus Low-Dose Yervoy in First-Line Lung Cancer Patients with Tumor Mutational Burden ≥10 mut/Mb. Available online: https://news.bms.com/press-release/corporatefinancial-news/bristol-myers-squibb-provides-update-ongoing-regulatory-review (accessed on 10 July 2023).
  89. Ramesh, P.; Shivde, R.; Jaishankar, D.; Saleiro, D.; Le Poole, I.C. A Palette of Cytokines to Measure Anti-Tumor Efficacy of T Cell-Based Therapeutics. Cancers 2021, 13, 821. [Google Scholar] [CrossRef]
  90. Balkwill, F.; Mantovani, A. Inflammation and cancer: Back to Virchow? Lancet 2001, 357, 539–545. [Google Scholar] [CrossRef]
  91. Brummel, K.; Eerkens, A.L.; de Bruyn, M.; Nijman, H.W. Tumour-infiltrating lymphocytes: From prognosis to treatment selection. Br. J. Cancer 2022, 128, 451–458. [Google Scholar] [CrossRef]
  92. Cao, D.; Xu, H.; Xu, X.; Guo, T.; Ge, W. A reliable and feasible way to predict the benefits of Nivolumab in patients with non-small cell lung cancer: A pooled analysis of 14 retrospective studies. Oncoimmunology 2018, 7, e1507262. [Google Scholar] [CrossRef] [PubMed]
  93. Pavan, A.; Calvetti, L.; Dal Maso, A.; Attili, I.; Del Bianco, P.; Pasello, G.; Guarneri, V.; Aprile, G.; Conte, P.; Bonanno, L. Peripheral blood markers identify risk of immune-related toxicity in advanced non-small cell lung Cancer treated with immune-checkpoint inhibitors. Oncologist 2019, 24, 1128–1136. [Google Scholar] [CrossRef] [PubMed]
  94. Ren, F.; Zhao, T.; Liu, B.; Pan, L. Neutrophil-lymphocyte ratio (NLR) predicted prognosis for advanced non-small-cell lung cancer (NSCLC) patients who received immune checkpoint blockade (ICB). Onco. Targets Ther. 2019, 12, 4235–4244. [Google Scholar] [CrossRef]
  95. Ren, F.; Zhao, T.; Liu, B.; Pan, L. Prognostic role of pretreatment neutrophil-to-lymphocyte ratio in non-small cell lung cancer patients treated with systemic therapy: A meta-analysis. Transl. Lung Cancer Res. 2019, 8, 214–226. [Google Scholar] [CrossRef]
  96. Templeton, A.J.; McNamara, M.G.; Šeruga, B.; Vera-Badillo, F.E.; Aneja, P.; Ocaña, A.; Leibowitz-Amit, R.; Sonpavde, G.; Knox, J.J.; Tran, B.; et al. Prognostic Role of Neutrophil-to-Lymphocyte Ratio in Solid Tumors: A Systematic Review and Meta-Analysis. JNCI J. Natl. Cancer Inst. 2014, 106, dju124. [Google Scholar] [CrossRef]
  97. Romano, F.J.; Ronga, R.; Ambrosio, F.; Arundine, D.; Longo, V.; Galetta, D.; Gridelli, C.; Maione, P.; Palma, V.; Damiano, V.; et al. Neutrophil-to-Lymphocyte Ratio Is a Major Prognostic Factor in Non-small Cell Lung Carcinoma Patients Undergoing First Line Immunotherapy With Pembrolizumab. Cancer Diagn. Progn. 2023, 3, 44–52. [Google Scholar] [CrossRef] [PubMed]
  98. Schalper, K.A.; Brown, J.; Carvajal-Hausdorf, D.; McLaughlin, J.; Velcheti, V.; Syrigos, K.N.; Herbst, R.S.; Rimm, D.L. Objective measurement and clinical significance of TILs in non-small cell lung cancer. J. Natl. Cancer Inst. 2015, 107, dju435. [Google Scholar] [CrossRef]
  99. Uryvaev, A.; Passhak, M.; Hershkovits, D.; Sabo, E.; Bar-Sela, G. The role of tumor-infiltrating lymphocytes (TILs) as a predictive biomarker of response to anti-PD1 therapy in patients with metastatic non-small cell lung cancer or metastatic melanoma. Med. Oncol. 2018, 35, 25. [Google Scholar] [CrossRef]
  100. Feng, Y.; Xiong, Y.; Qiao, T.; Li, X.; Jia, L.; Han, Y. Lactate dehydrogenase A: A key player in carcinogenesis and potential target in cancer therapy. Cancer Med. 2018, 7, 6124–6136. [Google Scholar] [CrossRef]
  101. Liu, R.; Cao, J.; Gao, X.; Zhang, J.; Wang, L.; Wang, B.; Guo, L.; Hu, X.; Wang, Z. Overall survival of cancer patients with serum lactate dehydrogenase greater than 1000 IU/L. Tumour Biol. 2016, 37, 14083–14088. [Google Scholar] [CrossRef]
  102. Claps, G.; Faouzi, S.; Quidville, V.; Chehade, F.; Shen, S.; Vagner, S.; Robert, C. The multiple roles of LDH in cancer. Nat. Rev. Clin. Oncol. 2022, 19, 749–762. [Google Scholar] [CrossRef]
  103. Lee, D.S.; Park, K.R.; Kim, S.J.; Chung, M.J.; Lee, Y.H.; Chang, J.H.; Kang, J.H.; Hong, S.H.; Kim, M.S.; Kim, Y.S. Serum lactate dehydrogenase levels at presentation in stage IV non-small cell lung cancer: Predictive value of metastases and relation to survival outcomes. Tumour Biol. 2016, 37, 619–625. [Google Scholar] [CrossRef]
  104. Hart, P.C.; Rajab, I.M.; Alebraheem, M.; Potempa, L.A. C-Reactive Protein and Cancer-Diagnostic and Therapeutic Insights. Front. Immunol. 2020, 11, 595835. [Google Scholar] [CrossRef]
  105. Xiao, X.; Wang, S.; Long, G. C-reactive protein is a significant predictor of improved survival in patients with advanced non-small cell lung cancer. Medicine 2019, 98, e16238. [Google Scholar] [CrossRef] [PubMed]
  106. Kasahara, N.; Sunaga, N.; Tsukagoshi, Y.; Miura, Y.; Sakurai, R.; Kitahara, S.; Yokobori, T.; Kaira, K.; Mogi, A.; Maeno, T.; et al. Post-treatment Glasgow Prognostic Score Predicts Efficacy in Advanced Non-small-cell Lung Cancer Treated with Anti-PD1. Anticancer Res. 2019, 39, 1455–1461. [Google Scholar] [CrossRef] [PubMed]
  107. Kang, H.S.; Shin, A.Y.; Yeo, C.D.; Kim, S.K.; Park, C.K.; Kim, J.S.; Kim, S.J.; Lee, S.H.; Kim, J.W. Significance of Glasgow Prognostic Scores in NSCLC Patients Treated with Immunotherapy after Platinum-based Cytotoxic Chemotherapy. Vivo 2021, 35, 3423–3430. [Google Scholar] [CrossRef] [PubMed]
  108. Aldea, M.; Benitez, J.C.; Mezquita, L. The Lung Immune Prognostic Index (LIPI) stratifies prognostic groups in advanced non-small cell lung cancer (NSCLC) patients. Transl. Lung Cancer Res. 2020, 9, 967–970. [Google Scholar] [CrossRef]
  109. Ruiz-Bañobre, J.; Areses-Manrique, M.C.; Mosquera-Martínez, J.; Cortegoso, A.; Afonso-Afonso, F.J.; de Dios-Álvarez, N.; Fernández-Núñez, N.; Azpitarte-Raposeiras, C.; Amenedo, M.; Santomé, L.; et al. Evaluation of the lung immune prognostic index in advanced non-small cell lung cancer patients under nivolumab monotherapy. Transl. Lung Cancer Res. 2019, 8, 1078–1085. [Google Scholar] [CrossRef]
  110. Mezquita, L.; Auclin, E.; Ferrara, R.; Charrier, M.; Remon, J.; Planchard, D.; Ponce, S.; Ares, L.P.; Leroy, L.; Audigier-Valette, C.; et al. Association of the Lung Immune Prognostic Index with Immune Checkpoint Inhibitor Outcomes in Patients with Advanced Non-Small Cell Lung Cancer. JAMA Oncol. 2018, 4, 351–357. [Google Scholar] [CrossRef]
  111. Fernandes, M.R.; Aggarwal, P.; Costa, R.G.F.; Cole, A.M.; Trinchieri, G. Targeting the gut microbiota for cancer therapy. Nat. Rev. Cancer 2022, 22, 703–722. [Google Scholar] [CrossRef]
  112. Ağagündüz, D.; Cocozza, E.; Cemali, Ö.; Bayazıt, A.D.; Nanì, M.F.; Cerqua, I.; Morgillo, F.; Saygılı, S.K.; Berni Canani, R.; Amero, P.; et al. Understanding the role of the gut microbiome in gastrointestinal cancer: A review. Front. Pharmacol. 2023, 14, 1130562. [Google Scholar] [CrossRef] [PubMed]
  113. Cheng, W.Y.; Wu, C.Y.; Yu, J. The role of gut microbiota in cancer treatment: Friend or foe? Gut 2020, 69, 1867–1876. [Google Scholar] [CrossRef] [PubMed]
  114. Rahman, M.M.; Islam, M.R.; Shohag, S.; Ahasan, M.T.; Sarkar, N.; Khan, H.; Hasan, A.M.; Cavalu, S.; Rauf, A. Microbiome in cancer: Role in carcinogenesis and impact in therapeutic strategies. Biomed. Pharmacother. 2022, 149, 112898. [Google Scholar] [CrossRef] [PubMed]
  115. Sivan, A.; Corrales, L.; Hubert, N.; Williams, J.B.; Aquino-Michaels, K.; Earley, Z.M.; Benyamin, F.W.; Lei, Y.M.; Jabri, B.; Alegre, M.L.; et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 2015, 350, 1084–1089. [Google Scholar] [CrossRef]
  116. Stingele, F.; Corthésy, B.; Kusy, N.; Porcelli, S.A.; Kasper, D.L.; Tzianabos, A.O. Zwitterionic polysaccharides stimulate T cells with no preferential V beta usage and promote anergy, resulting in protection against experimental abscess formation. J. Immunol. 2004, 172, 1483–1490. [Google Scholar] [CrossRef]
  117. Routy, B.; Le Chatelier, E.; Derosa, L.; Duong, C.P.M.; Alou, M.T.; Daillère, R.; Fluckiger, A.; Messaoudene, M.; Rauber, C.; Roberti, M.P. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 2018, 359, 91–97. [Google Scholar] [CrossRef]
  118. Viaud, S.; Saccheri, F.; Mignot, G.; Yamazaki, T.; Daillère, R.; Hannani, D.; Enot, D.P.; Pfirschke, C.; Engblom, C.; Pittet, M.J.; et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science 2013, 342, 971–976. [Google Scholar] [CrossRef]
  119. Tsikala-Vafea, M.; Belani, N.; Vieira, K.; Khan, H.; Farmakiotis, D. Use of antibiotics is associated with worse clinical outcomes in patients with cancer treated with immune checkpoint inhibitors: A systematic review and meta-analysis. Int. J. Infect. Dis. 2021, 106, 142–154, Erratum in Int. J. Infect. Dis. 2021, 113, 34–35. [Google Scholar] [CrossRef]
  120. Chang, Y.; Lin, W.Y.; Chang, Y.C.; Huang, C.H.; Tzeng, H.E.; Abdul-Lattif, E.; Wang, T.H.; Tseng, T.H.; Kang, Y.N.; Chi, K.Y. The Association between Baseline Proton Pump Inhibitors, Immune Checkpoint Inhibitors, and Chemotherapy: A Systematic Review with Network Meta-Analysis. Cancers 2022, 15, 284. [Google Scholar] [CrossRef]
  121. Chen, B.; Yang, C.; Dragomir, M.P.; Chi, D.; Chen, W.; Horst, D.; Calin, G.A.; Li, Q. Association of proton pump inhibitor use with survival outcomes in cancer patients treated with immune checkpoint inhibitors: A systematic review and meta-analysis. Ther. Adv. Med. Oncol. 2022, 14, 17588359221111703. [Google Scholar] [CrossRef]
  122. Dar, S.; Merza, N.; Qatani, A.; Rahim, M.; Varughese, T.; Mohammad, A.; Masood, F.; Reza, F.Z.; Wan, S.; Almas, T. Impact of proton-pump inhibitors on the efficacy of immune checkpoint inhibitors in non-small cell lung cancer: A systematic review and meta-analysis. Ann. Med. Surg. 2022, 78, 103752. [Google Scholar] [CrossRef]
  123. Gaucher, L.; Adda, L.; Séjourné, A.; Joachim, C.; Chaby, G.; Poulet, C.; Liabeuf, S.; Gras-Champel, V.; Masmoudi, K.; Moreira, A.; et al. Impact of the corticosteroid indication and administration route on overall survival and the tumor response after immune checkpoint inhibitor initiation. Ther. Adv. Med. Oncol. 2021, 13, 1758835921996656. [Google Scholar] [CrossRef]
  124. Goodman, R.S.; Johnson, D.B.; Balko, J.M. Corticosteroids and Cancer Immunotherapy. Clin. Cancer Res. 2023, 29, 2580–2587. [Google Scholar] [CrossRef] [PubMed]
  125. Cortellini, A.; Di Maio, M.; Nigro, O.; Leonetti, A.; Cortinovis, D.L.; Aerts, J.G.; Guaitoli, G.; Barbieri, F.; Giusti, R.; Ferrara, M.G.; et al. Differential influence of antibiotic therapy and other medications on oncological outcomes of patients with non-small cell lung cancer treated with first-line pembrolizumab versus cytotoxic chemotherapy. J. Immunother. Cancer 2021, 9, e002421. [Google Scholar] [CrossRef] [PubMed]
  126. Assumpção, J.A.F.; Pasquarelli-do-Nascimento, G.; Duarte, M.S.V.; Bonamino, M.H.; Magalhães, K.G. The ambiguous role of obesity in oncology by promoting cancer but boosting antitumor immunotherapy. J. Biomed. Sci. 2022, 29, 12. [Google Scholar] [CrossRef] [PubMed]
  127. Finelli, C. Obesity and immunotherapy: The surprisingly positive association! Immunotherapy 2020, 12, 541–544. [Google Scholar] [CrossRef] [PubMed]
  128. Woodall, M.J.; Neumann, S.; Campbell, K.; Pattison, S.T.; Young, S.L. The Effects of Obesity on Anti-Cancer Immunity and Cancer Immunotherapy. Cancers 2020, 12, 1230. [Google Scholar] [CrossRef]
  129. Wang, Z.; Aguilar, E.G.; Luna, J.I.; Dunai, C.; Khuat, L.T.; Le, C.T.; Mirsoian, A.; Minnar, C.M.; Stoffel, K.M.; Sturgill, I.R.; et al. Paradoxical effects of obesity on T cell function during tumor progression and PD-1 checkpoint blockade. Nat. Med. 2019, 25, 141–151. [Google Scholar] [CrossRef]
  130. Mullen, M.; Gonzalez-Perez, R.R. Leptin-Induced JAK/STAT signaling and cancer growth. Vaccines 2016, 4, 26. [Google Scholar] [CrossRef]
  131. Cortellini, A.; Bersanelli, M.; Buti, S.; Cannita, K.; Santini, D.; Perrone, F.; Giusti, R.; Tiseo, M.; Michiara, M.; Di Marino, P.; et al. A multicenter study of body mass index in cancer patients treated with anti-PD-1/PD-L1 immune checkpoint inhibitors: When overweight becomes favorable. J. Immunother. Cancer 2019, 7, 57. [Google Scholar] [CrossRef]
  132. McQuade, J.L.; Daniel, C.R.; Hess, K.R.; Mak, C.; Wang, D.Y.; Rai, R.R.; Park, J.J.; Haydu, L.E.; Spencer, C.; Wongchenko, M.; et al. Association of body-mass index and outcomes in patients with metastatic melanoma treated with targeted therapy, immunotherapy, or chemotherapy: A retrospective, multicohort analysis. Lancet Oncol. 2018, 19, 310–322. [Google Scholar] [CrossRef]
  133. Martini, D.J.; Kline, M.R.; Liu, Y.; Shabto, J.M.; Williams, M.A.; Khan, A.I.; Lewis, C.; Collins, H.; Akce, M.; Kissick, H.T.; et al. Adiposity may predict survival in patients with advanced stage cancer treated with immunotherapy in phase 1 clinical trials. Cancer 2020, 126, 575–582. [Google Scholar] [CrossRef]
  134. Kichenadasse, G.; Miners, J.O.; Mangoni, A.A.; Rowland, A.; Hopkins, A.M.; Sorich, M.J. Association between body mass index and overall survival with immune checkpoint inhibitor therapy for advanced non-small cell lung cancer. JAMA Oncol. 2020, 6, 512–518. [Google Scholar] [CrossRef]
  135. Petrelli, F.; Cortellini, A.; Indini, A.; Tomasello, G.; Ghidini, M.; Nigro, O.; Salati, M.; Dottorini, L.; Iaculli, A.; Varricchio, A.; et al. Obesity paradox in patients with cancer: A systematic review and meta-analysis of 6,320,365 patients. medRxiv 2020. [Google Scholar] [CrossRef]
  136. Cortellini, A.; Ricciuti, B.; Tiseo, M.; Bria, E.; Banna, G.L.; Aerts, J.G.; Barbieri, F.; Giusti, R.; Cortinovis, D.L.; Migliorino, M.R.; et al. Baseline BMI and BMI variation during first line pembrolizumab in NSCLC patients with a PD-L1 expression ≥50%: A multicenter study with external validation. J. Immunother. Cancer 2020, 8, e001403. [Google Scholar] [CrossRef] [PubMed]
  137. Tateishi, A.; Horinouchi, H.; Yoshida, T.; Masuda, K.; Jo, H.; Shinno, Y.; Okuma, Y.; Goto, Y.; Yamamoto, N.; Ohe, Y. Correlation between body mass index and efficacy of anti-PD-1 inhibitor in patients with non-small cell lung cancer. Respir. Investig. 2022, 60, 234–240. [Google Scholar] [CrossRef] [PubMed]
  138. Kimura, M.; Naito, T.; Kenmotsu, H.; Taira, T.; Wakuda, K.; Oyakawa, T.; Hisamatsu, Y.; Tokito, T.; Imai, H.; Akamatsu, H.; et al. Prognostic impact of cancer cachexia in patients with advanced non-small cell lung cancer. Support. Care Cancer 2015, 23, 1699–1708. [Google Scholar] [CrossRef]
  139. Dewys, W.D.; Begg, C.; Lavin, P.T.; Band, P.R.; Bennett, J.M.; Bertino, J.R.; Cohen, M.H.; Douglass, H.O., Jr.; Engstrom, P.F.; Ezdinli, E.Z.; et al. Prognostic effect of weight loss prior to chemotherapy in cancer patients. Eastern Cooperative Oncology Group. Am. J. Med. 1980, 69, 491–497. [Google Scholar] [CrossRef]
  140. Watte, G.; Nunes, C.H.A.; Sidney-Filho, L.A.; Zanon, M.; Altmayer, S.P.L.; Pacini, G.S.; Barros, M.; Moreira, A.L.S.; Alves, R.J.V.; Zelmanowicz, A.M.; et al. Proportional weight loss in six months as a risk factor for mortality in stage IV non-small cell lung cancer. J. Bras. Pneumol. 2018, 44, 505–509. [Google Scholar] [CrossRef]
  141. Patel, J.D.; Pereira, J.R.; Chen, J.; Liu, J.; Guba, S.C.; John, W.J.; Orlando, M.; Scagliotti, G.; Bonomi, P.D. Relationship between efficacy outcomes and weight gain during treatment of advanced, non-squamous, non-small-cell lung cancer patients. Ann. Oncol. 2016, 27, 1612–1619. [Google Scholar] [CrossRef]
  142. Lee, C.S.; Devoe, C.E.; Zhu, X.; Fishbein, J.S.; Seetharamu, N. Pretreatment nutritional status and response to checkpoint inhibitors in lung cancer. Lung Cancer Manag. 2020, 9, LMT31. [Google Scholar] [CrossRef] [PubMed]
  143. Miyawaki, T.; Naito, T.; Kodama, A.; Nishioka, N.; Miyawaki, E.; Mamesaya, N.; Kawamura, T.; Kobayashi, H.; Omori, S.; Wakuda, K.; et al. Desensitizing Effect of Cancer Cachexia on Immune Checkpoint Inhibitors in Patients with Advanced NSCLC. JTO Clin. Res. Rep. 2020, 1, 100020. [Google Scholar] [CrossRef] [PubMed]
  144. Jo, H.; Yoshida, T.; Horinouchi, H.; Yagishita, S.; Matsumoto, Y.; Shinno, Y.; Okuma, Y.; Goto, Y.; Yamamoto, N.; Takahashi, K.; et al. Prognostic significance of cachexia in advanced non-small cell lung cancer patients treated with pembrolizumab. Cancer Immunol. Immunother. 2022, 71, 387–398. [Google Scholar] [CrossRef]
  145. Shijubou, N.; Sumi, T.; Yamada, Y.; Nakata, H.; Mori, Y.; Chiba, H. Immunological and nutritional predictive factors in patients receiving pembrolizumab for the first-line treatment of non-small cell lung cancer. J. Cancer Res. Clin. Oncol. 2022, 148, 1893–1901. [Google Scholar] [CrossRef] [PubMed]
  146. Hakozaki, T.; Nolin-Lapalme, A.; Kogawa, M.; Okuma, Y.; Nakamura, S.; Moreau-Amaru, D.; Tamura, T.; Hosomi, Y.; Takeyama, H.; Richard, C.; et al. Cancer Cachexia among Patients with Advanced Non-Small-Cell Lung Cancer on Immunotherapy: An Observational Study with Exploratory Gut Microbiota Analysis. Cancers 2022, 14, 5405. [Google Scholar] [CrossRef]
  147. Degens, J.H.R.J.; Dingemans, A.C.; Willemsen, A.C.H.; Gietema, H.A.; Hurkmans, D.P.; Aerts, J.G.; Hendriks, L.E.L.; Schols, A.M.W.J. The prognostic value of weight and body composition changes in patients with non-small-cell lung cancer treated with nivolumab. J. Cachexia Sarcopenia Muscle 2021, 12, 657–664. [Google Scholar] [CrossRef]
  148. Brueckl, W.M.; Ficker, J.H.; Zeitler, G. Clinically relevant prognostic and predictive markers for immune-checkpoint-inhibitor (ICI) therapy in non-small cell lung cancer (NSCLC). BMC Cancer 2020, 20, 1185. [Google Scholar] [CrossRef]
  149. Šutić, M.; Vukić, A.; Baranašić, J.; Försti, A.; Džubur, F.; Samaržija, M.; Jakopović, M.; Brčić, L.; Knežević, J. Diagnostic, Predictive, and Prognostic Biomarkers in Non-Small Cell Lung Cancer (NSCLC) Management. J. Pers. Med. 2021, 11, 1102. [Google Scholar] [CrossRef]
Table 1. Currently clinically targetable Immune Checkpoint and future candidates.
Table 1. Currently clinically targetable Immune Checkpoint and future candidates.
ICBinding Partners Expressed
Respectively on
RoleTargetability
PD-1—PD-L1T cells—tumor cellsNegative ICCurrently targetable
CTLA-4—B7T cells—tumor cellsNegative ICCurrently targetable
TIM-3—Galectin-9T cells—tumor cellsNegative ICStill not targetable
LAG3—class II MHC/galectin-3/LSECtinT cells—tumor cellsNegative ICStill not targetable
VISTA—Still unknownT cellsNegative ICStill not targetable
OX40—OX40LT cells—APCsPositive ICStill not targetable
GITR—GITRLT cells—APCsPositive ICStill not targetable
Table 2. Clinical trial investigating ICIs targeting new ICs.
Table 2. Clinical trial investigating ICIs targeting new ICs.
ICICIDesignIdentifier
TIM-3LY3415244
(anti TIM-3 and PD-L1)
LY3415244
in
advanced or metastatic solid neoplasms
NCT03752177
TIM-3LY3321367
(anti TIM-3)
LY3321367 ±
LY3300054
(anti PD-L1 ICI)
in
advanced or metastatic solid neoplasms
NCT03099109
TIM-3Sym023
(anti TIM-3)
Sym023
in
advanced or metastatic solid neoplasms or lymphomas
NCT03489343
TIM-3TSR-022
(anti TIM-3)
TSR-022 + TSR-042
(anti PD-L1 ICI) +
carboplatin + pemetrexed/nab-paclitaxel
in
advanced or metastatic solid neoplasms
NCT03307785
TIM-3Sym023
(anti TIM-3)
Sym023 +
Sym021
(anti PD-1 ICI)
in
advanced or metastatic solid neoplasms or lymphomas
NCT03311412
TIM-3TSR-022
(anti TIM-3)
TSR-022 ± TSR-042 (anti PD-L1 ICI)
/
TSR-022 ± TSR-033 (anti LAG3 ICI)
in
advanced or metastatic solid neoplasms
NCT02817633
TIM-3RO7121661
(anti TIM-3 and PD-1)
RO7121661
in
advanced or metastatic solid neoplasms
NCT03708328
TIM-3INCAGN02390
(anti TIM-3)
INCAGN02390
in
advanced or metastatic solid neoplasms
NCT03652077
TIM-3MBG453
(anti TIM-3)
MBG453 ± PDR001
(anti PD-1 ICI)
in
advanced or metastatic solid neoplasms
NCT02608268
TIM-3BGB-A425
(anti TIM-3)
BGB-A425 ±
tislelizumab
(anti PD-1 ICI)
in
advanced or metastatic solid neoplasms
NCT03744468
LAG 3Sym022
(anti LAG 3)
Sym022
in
advanced or metastatic solid neoplasms or lymphomas
NCT03489369
LAG 3LAG525
(anti LAG 3)
LAG525 ±
PDR001
(anti PD-1 ICI)
in
advanced or metastatic solid neoplasms
NCT02460224
LAG 3IMP321
(anti LAG 3)
IMP321
in
advanced or metastatic solid neoplasms
NCT03252938
LAG 3Relatlimab
(anti LAG 3)
Relatlimab ±
nivolumab
in
advanced or metastatic solid neoplasms
NCT02966548
LAG 3XmAb®22841
(anti LAG 3 and CTLA-4)
XmAb®22841 ±
pembrolizumab
in
advanced or metastatic solid neoplasms
NCT03849469
LAG 3Relatlimab
(anti LAG 3)
Relatlimab ±
nivolumab
in
advanced or metastatic solid neoplasms
NCT01968109
LAG 3MGD013
(anti LAG 3)
MGD013
in
advanced or metastatic solid neoplasms or lymphomas
NCT03219268
VISTACA-170
(anti VISTA and PD-L1 and PD-2)
CA-170
in
advanced or metastatic solid neoplasms or lymphomas
NCT02812875
OX40MEDI0562
(anti OX-40)
MEDI0562 +
durvalumab
/
MEDI0562 +
tremelimumab
in
advanced or metastatic solid neoplasms
NCT02705482
OX40MOXR0916
(anti OX-40)
MOXR0916 +
atezolizumab
in
advanced or metastatic solid neoplasms
NCT02410512
OX40INCAGN01949
(anti OX-40)
INCAGN01949 +
nivolumab
/
INCAGN01949 +
ipilimumab
/
INCAGN01949 +
nivolumab
+
ipilimumab
in
advanced or metastatic solid neoplasms
NCT03241173
OX40PF-04518600
(anti OX-40)
PF-04518600 ±
05082566
(4-1BB ICI)
in
advanced or metastatic solid neoplasms
NCT02315066
OX40ATOR-1015
(anti OX-40)
ATOR-1015
in
advanced or metastatic solid neoplasms
NCT03782467
OX40SL-279252
(anti OX-40)
SL-279252
in
advanced or metastatic solid neoplasms or lymphomas
NCT03894618
OX40ATOR-1015
(anti OX-40)
ATOR-1015
in
advanced or metastatic solid neoplasms
NCT03782467
OX40SL-279252
(anti OX-40)
SL-279252
in
advanced or metastatic solid neoplasms or lymphomas
NCT03894618
GITRINCAGN01876
(anti GITR)
INCAGN01876
in
advanced or metastatic solid neoplasms
NCT02697591
GITRTRX518
(anti GITR)
TRX518
in
advanced or metastatic solid neoplasms
NCT01239134
GITRGWN323
(anti GITR)
GWN323 ± PDR001
(anti PD-1 ICI)
in
advanced or metastatic solid neoplasms or lymphomas
NCT02740270
GITROMP-336B11
(anti GITR)
OMP-336B11
in
advanced or metastatic solid neoplasms
NCT03295942
Table 3. Current ESMO and/or ASCO-recommended ICIs or ICI-containing combinations for the treatment of naïve advanced NSCLC without driver mutations.
Table 3. Current ESMO and/or ASCO-recommended ICIs or ICI-containing combinations for the treatment of naïve advanced NSCLC without driver mutations.
ICI or ICI-Containing CombinationSubset of PatientsRegulatory ApprovalPivotal TrialUpdated
Survival Data (mPFS/OS) *
PembrolizumabSquamous and nonsquamous with PD-L1 TPS ≥ 50%ASCO-recommended
ESMO-recommended
KEYNOTE-024
[64]
PFS: 7.7 months
OS: 26.3 months
AtezolizumabSquamous and nonsquamous with PD-L1 TPS ≥ 50%ASCO-recommended
ESMO-recommended
IMpower110
[65]
PFS: 8.2 months
OS: 18.9 months
CemiplimabSquamous and nonsquamous with PD-L1 TPS ≥ 50%ASCO-recommended
ESMO-recommended
Empower-Lung 1
[66]
PFS: 6.3 months
OS: 23.4 months
PembrolizumabSquamous and nonsquamous with any PD-L1 TPSASCO-recommendedKEYNOTE-042
[67]
PFS: 5.6 months
OS: 16.4 months
Nivolumab + Ipilimumab + short course of platinum-based histology-specific doublet chemotherapy followed by Nivolumab + IpilimumabSquamous and nonsquamous with any PD-L1 TPSASCO-recommended
ESMO-recommended
CheckMate 9LA
[68]
PFS: 6.4 months
OS: 15.8 months
Cemiplimab + platinum-based histology-specific doublet chemotherapy followed by Cemiplimab (+Pemetrexed if nonsquamous)Squamous and nonsquamous with any PD-L1 TPSESMO-recommendedEmpower-Lung 3
[69]
PFS: 8.2 months
OS: 21.1 months
Durvalumab + Tremelimumab + platinum-based histology-specific doublet chemotherapy followed by Durvalumab + one additional Tremelimumab dose (+Pemetrexed if nonsquamous)Squamous and nonsquamous with any PD-L1 TPSESMO-recommendedPOSEIDON
[70]
PFS: 6.2 months
OS: 14.0 months
Nivolumab + IpilimumabSquamous and nonsquamous with a PD-L1 TPS ≥ 1%ESMO-recommendedCheckMate 227
[71]
PFS: 5.1 months
OS: 17.1 months
Pembrolizumab + Platinum + Pemetrexed followed by Pembrolizumab + PemetrexedNonsquamous with any PD-L1 TPSASCO-recommended
ESMO-recommended
KEYNOTE-189
[72]
PFS: 9.0 months
OS: 22.0 months
Atezolizumab + Carboplatin + Nab-Paclitaxel followed by AtezolizumabNonsquamous with any PD-L1 TPSASCO-recommended
ESMO-recommended
IMpower130 [73]PFS: 7.0 months
OS: 18.6 months
Atezolizumab + Carboplatin + Paclitaxel + Bevacizumab followed by Atezolizumab + BevacizumabNonsquamous with any PD-L1 TPSASCO-recommended
ESMO-recommended
IMpower150
[74]
PFS: 8.4 months
OS: 19.5 months
Pembrolizumab + Carboplatin + (Nab)Paclitaxel followed by PembrolizumabSquamous with any PD-L1 TPSASCO-recommended
ESMO-recommended
KEYNOTE-407
[75]
PFS: 8.0 months
OS: 17.2 months
* mPFS: Median Progression Free Survival; mOS: Median Overall Survival.
Table 4. Most promising prognostic factors under investigation in advanced NSCLC patients treated with ICIs.
Table 4. Most promising prognostic factors under investigation in advanced NSCLC patients treated with ICIs.
FactorPrognostic Value
High soluble PD-L1 levelsNegative
High Tumor Mutational BurdenPositive
High Neutrophil-to-lymphocytes ratioNegative
High levels of Tumor-infiltrating lymphocytesPositive
High Glasgow Prognostic ScoreNegative
High Lung Immune Prognostic IndexNegative
Gut microbiotaDebated
Corticosteroids, antibiotics, and PPI useNegative
High Body Mass IndexPositive (BMI ≥ 30)
Weight loss and cachexiaNegative
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Rocco, D.; Della Gravara, L.; Ragone, A.; Sapio, L.; Naviglio, S.; Gridelli, C. Prognostic Factors in Advanced Non-Small Cell Lung Cancer Patients Treated with Immunotherapy. Cancers 2023, 15, 4684. https://doi.org/10.3390/cancers15194684

AMA Style

Rocco D, Della Gravara L, Ragone A, Sapio L, Naviglio S, Gridelli C. Prognostic Factors in Advanced Non-Small Cell Lung Cancer Patients Treated with Immunotherapy. Cancers. 2023; 15(19):4684. https://doi.org/10.3390/cancers15194684

Chicago/Turabian Style

Rocco, Danilo, Luigi Della Gravara, Angela Ragone, Luigi Sapio, Silvio Naviglio, and Cesare Gridelli. 2023. "Prognostic Factors in Advanced Non-Small Cell Lung Cancer Patients Treated with Immunotherapy" Cancers 15, no. 19: 4684. https://doi.org/10.3390/cancers15194684

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