Back to Journals » Lung Cancer: Targets and Therapy » Volume 12

Immunotherapy for Stage III NSCLC: Durvalumab and Beyond

Authors Fitzpatrick O, Naidoo J

Received 18 August 2021

Accepted for publication 21 October 2021

Published 2 November 2021 Volume 2021:12 Pages 123—131

DOI https://doi.org/10.2147/LCTT.S305466

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Dr Justinn Cochran



Orla Fitzpatrick,1 Jarushka Naidoo1,2

1Department of Oncology, Beaumont Hospital, RCSI University of Health Sciences, Dublin, Ireland; 2Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, 21231, USA

Correspondence: Jarushka Naidoo
Beaumont Hospital, Beaumont Road, Dublin, 9, Ireland
, Tel +353 1 809-3000
Email [email protected]

Abstract: Immunocheckpoint inhibitors (ICIs) have altered the treatment landscape of a wide range of malignancies, including non–small cell lung cancer (NSCLC). This class of agents inhibits the interaction between PD1 and PDL1, and was shown to be efficacious in the landmark PACIFIC trial with 1 year of maintenance durvalumab (anti-PDL1 antibody). This trial demonstrated that its use as a consolidation treatment given after definitive chemoradiotherapy improved progression free survival and overall survival compared to standard-of-care treatment. In this review, we discuss both clinical trial and real-world data that have been published since PACIFIC that support the use of durvalumab for stage III unresectable NSCLC. In addition, we highlight specific populations that may require special considerations for the use of durvalumab in this setting, such as oncogene-addicted NSCLC, the toxicity of immunotherapy, and future directions in ICI research in stage III NSCLC.

Keywords: lung cancer, immunotherapy, durvalumab

Background

Non–small cell lung cancer (NSCLC) is the leading cause of cancer-related deaths worldwide, and approximately a third of patients have locally advanced disease at diagnosis.1,2 Treatment options for NSCLC vary depending on the stage at diagnosis, but recent advances in genomic profiling, targeted therapy, and cancer immunotherapy have resulted in an expansion of treatment options and improved disease outcomes.3

One area that has revolutionized the treatment of advanced NSCLC is the use of immunocheckpoint inhibitors (ICIs), highlighting the dynamic relationship between immunology and oncology. Tumor cells often downregulate expression of proteins involved in immunosurveillance, shielding cancer cells from protective immunoresponses.4 Multiple targets of immunosuppressive pathways have been developed, facilitating the activation of immunomediated destruction of tumor cells.5 One pathway includes PD1 and PDL1 targets. When PDL1 binds to PD1, it induces T-cell signaling that results in apoptosis and anergy, and it is this interaction that has been utilized to reinvigorate an antitumor T-cell response.6–8 In recent years, a number of monoclonal antibodies targeting this interaction have demonstrated efficacy, including pembrolizumab, nivolumab, and cemiplimab targeting PD1, and atezolizumab, avelumab, and durvalumab targeting PDL1. These drugs have been shown to be efficacious in a wide range of malignancies, both in early and advanced disease, including lung cancer, melanoma, renal-cell carcinoma, Hodgkin’s lymphoma, cutaneous squamous-cell carcinoma, urothelial cancer, and metastatic Merkel-cell carcinoma.9–17 The incorporation of ICIs into the treatment landscape for advanced NSCLC has changed how we treat this disease in the last 5–8 years.18,19 In the US, there are now more than five standard treatment options that incorporate immunotherapy in the first-line treatment of advanced NSCLC,19–24 and their use has expanded to earlier-stage NSCLC.

Stage III NSCLC is a heterogeneous disease that is treated with a multimodality approach, incorporating chemotherapy, radiotherapy, and surgical resection in selected cases.25 In patients with unresectable locally advanced (stage III) NSCLC, the standard of care has been definitive chemoradiotherapy since the early 2000s, with a platinum-based doublet-chemotherapy regimen in combination with concurrent radiotherapy (RT).26 A phase III clinical trial demonstrated that a combination of concurrent cisplatin, etoposide, and chest radiotherapy resulted in a benefit in median OS of 15 months, with 3- and 5-year survival of 17% and 15%, respectively.27 This approach of combination chemoradiotherapy with carboplatin, pemetrexed, and thoracic radiotherapy consolidated the data available on cisplatin and etoposide, increasing chemotherapy treatment options.27,28 The incidence of neutropenia was similar in both studies (35% vs 42%), with fatigue, dehydration, and anemia the most common adverse events (AEs) noted.28 The delivery of concurrent versus sequential chemoradiotherapy has been associated with a higher incidence of both hematological and nonhematological toxicity, notably esophagitis, but it has proven significantly effective in sustaining survival outcomes and became a standard of care >10 years ago.29 With concurrent treatment in one study, median progression-free survival (PFS) was 8 months and 5-year survival <20%.30

More recently, several studies have explored the use of immunotherapy after definitive chemoradiotherapy for stage III NSCLC. This “consolidation treatment” is delivered with the intent of reducing recurrence, and is administered after completion of chemoradiotherapy.31 In this narrative review, we highlight clinical trial and real-world data that support the use of immunotherapy for stage III NSCLC, in particular durvalumab, populations that may require special consideration such as oncogene-addicted NSCLC, the toxicity of immunotherapy for stage III NSCLC, and future directions.

Immunotherapy for Stage III NSCLC

A landmark study, the PACIFIC trial, published in 2017, assessed the potential benefit of durvalumab as consolidation treatment after completion of concurrent chemoradiotherapy in unresectable stage III NSCLC.26 This trial included 713 patients, randomly assigned to receive either durvalumab or placebo, commencing 1–42 days after combined chemoradiotherapy at a 2:1 ratio. Durvalumab was administered as an intravenous infusion every 2 weeks at 10 mg/kg. This phase III study demonstrated a benefit in both PFS (16.8 months vs 5.6 months, P<0.001) and overall survival (OS) (23.2 months vs 14.6 months, P<0.001) compared with placebo.26 Patients who received maintenance durvalumab also demonstrated prolonged median time to death or distant metastases in favour of durvalumab (28.3 months versus 16.2 months) and lower incidence of brain metastases (6.3% vs 11.8%) compared with placebo.32

Recently, a 4-year update on survival outcomes in the PACIFIC trial demonstrated that 49.6% of patients who received durvalumab remained alive at 4 years compared with 36.3% who received a placebo, with 35.5% of patients who received durvalumab alive with no disease progression versus 19.5% who received a placebo.33 Five-year survival outcomes were published this year at ASCO, showing 42.9% of patients who received durvalumab remained alive at 5 years and approximately a third remained alive and free of disease progression34

Real-World Data on Durvalumab for Stage III NSCLC

While clinical trial data demonstrate an impressive benefit for durvalumab in stage III NSCLC in the maintenance setting, other studies have examined whether this approach yields favorable outcomes in a real-world setting. Three retrospective studies in this area have shown patients with increased expression of PDL1 were more likely to receive durvalumab and showed improved PFS and OS in more heterogeneous patient cohorts compared to the PACIFIC trial.35–37

In the first of these three studies, a retrospective analysis conducted in Germany including 437 patients with stage III NSCLC, patients were treated with platinum-based chemoradiotherapy followed by durvalumab as per the PACIFIC regimen, and 50.3% were eligible to receive durvalumab.35,38 Reasons for inability to receive durvalumab included insufficient response to chemoradiotherapy based on RECIST38 (32.4% of patients), PDL1 positivity <1% (22.3% of patients), grade 2 or higher radiation pneumonitis (12.6%), and 3.5% due to prior autoimmune disease, mainly rheumatoid arthritis.35 Those with higher PDL1 expression (≥50%) were more likely to receive durvalumab (OR 2.4, P=0.006).35

Other retrospective studies have assessed survival outcomes in patients who received chemoradiotherapy alone compared with those who also received durvalumab consolidation treatment. A multicenter retrospective analysis of 147 patients found that PFS and 12-month OS was higher in patients who received durvalumab consolidation, but was specific to patients with PDL1 expression ≥50%.36 Notably, pneumonitis incidence was similar in both groups, yet those who developed grade 2+ pneumonitis had lower 12-month OS, regardless of treatment group.36

A retrospective study in Germany covering 56 centers and 126 patients assessed PFS, OS, and safety outcomes for durvalumab in patients with stage III NSCLC, including those with poorer performance status and autoimmune disease.37 In this study, 71.2% of patients had PDL1 expression >1%, slightly higher than the PACIFIC trial,37 and 54% completed 12 months of durvalumab consolidation treatment. Patients assessed in this analysis demonstrated a median PFS of 20.1 months with durvalumab, while 12- and 24-month OS was 78.6% and 66.0%, respectively.37 The incidence of both intrathoracic and extrathoracic metastases, including brain metastases, was lower in the durvalumab group. With further subgroup analysis, younger patients, female patients, and those with a good performance status showed improved PFS, again demonstrating the efficacy of durvalumab consolidation treatment.37

Separately, a prospective study of 26 patients with PDL1-positive unresectable stage III NSCLC showed impressive efficacy for durvalumab consolidation treatment, with 12-month PFS of 62% and 12-month OS of 100%.39 This study differed in terms of including patients that had received concurrent or sequential chemoradiotherapy, and it included one patient with stage IV disease. At the time of publication, a number of participants in this study were still receiving durvalumab, with a median follow-up of 20.6 months.39 Taken together, these data support the PACIFIC data, but also identify that in real-world settings there are particular considerations, including tumor PDL1 expression, comorbidities, and autoimmune disease history, that may limit or curtail the use of durvalumab.

Other Immunotherapeutic Approaches for Stage III NSCLC

The efficacy of consolidation treatment with ICIs, specifically PD1 or PDL1 blockade, has been assessed when given sequentially after a platinum-based doublet in stage III resectable NSCLC, and due to significant improvements in both PFS and OS, further research into optimal timing of this blockade has taken place. A pilot study of 21 patients with resectable NSCLC in 2018 investigated administering a PD1 blockade with nivolumab prior to surgery, and showed a major pathological response of 45%.40 Delivery of two doses of nivolumab prior to surgery did not delay surgery, with few AEs.40 The NADIM trial also investigated the use of neoadjuvant nivolumab in stage IIIa resectable NSCLC, and showed 77.1% 24-month PFS and 90% OS.41 The addition of nivolumab to chemotherapy did not result in a significantly higher incidence of AEs, and there was no delay to planned surgery.41 The phase II NEOSTAR trial of 44 patients investigated the use of neoadjuvant PD1 blockade with nivolumab alone or in combination with ipilimumab prior to surgery in resectable NSCLC. The use of combination ICIs resulted in a higher pathological complete-response rate (38% vs 10%), indicating neoadjuvant ICIs, alone or in combination, can improve survival in advanced NSCLC.42

The use of concurrent ICIs with chemoradiation in stage III unresectable NSCLC in the KEYNOTE-799 trial evaluated response rate as per RECIST criteria and incidence of grade 3 or higher pneumonitis with the addition of pembrolizumab in comparison to standard-of-care treatment.38 Although median OS and PFS were not reached after 1 year of follow-up, there was an objective response rate of 70% in standard-of-care treatment with and without pembrolizumab, with an incidence of 8% of grade 3 or higher pneumonitis demonstrating tolerable side effects of combination treatment.43 An additional phase II trial assessed the use of consolidation pembrolizumab in unresectable stage III NSCLC in 93 patients.44 Although the primary end point of time to metastatic disease or death was not reached, OS estimates of 80.5% at 12 months and 68.7% at 24 months indicated promising results with consolidation PD1 blockade.44

Atezolizumab is currently in a phase II trial (AFT-16, NCT03102242) assessing PDL1 blockage both before and after definitive chemoradiotherapy in stage III unresectable NSCLC, with four cycles being administered prior to platinum doublet chemotherapy, followed by 1 year of adjuvant atezolizumab therapy.45 Although secondary end points are awaited, initial results show it has been well tolerated, with a disease-control rate of 82.4% for patients with PDL1-negative tumors and 90.9% for PDL1-positive tumors.45 These studies emphasize the tolerability of combined neoadjuvant treatment in resectable NSCLC, and current data suggest improved disease-response rates.

The use of consolidation durvalumab has been assessed in resectable NSCLC. The SAKK 16/14 trial on 67 patients with resectable stage IIIA NSCLC investigated the role of preoperative chemotherapy and durvalumab, followed by consolidation treatment for up to 1 year.46 This single-arm phase II trial did not reach median event-free survival or OS at 28 months, but did demonstrate safety or presurgical ICI use and 1-year event-free survival of 73%.46

Immunotherapy in Stage III Oncogene-Addicted NSCLC

Since the PACIFIC trial, a number of studies have focused on investigating the efficacy of durvalumab in particular subsets of patients with NSCLC who may have differential responses to immunotherapy, such as EGFR-mutant NSCLC. A multicenter retrospective analysis of 37 patients investigating its use in EGFR-mutated stage III NSCLC, failed to show PFS or OS benefit in this subgroup (10.3 vs 6.9 months, P=0.993).47 When durvalumab consolidation therapy was compared to consolidation treatment with EGFR tyrosine-kinase inhibitors (TKI), the survival benefit was significantly longer in the EGFR tyrosine-kinase inhibitor–treatment subgroup (P=0.023) highlighting that it may not offer survival benefits in tumors with different genomic features.47 This was confirmed in a study evaluating durvalumab efficacy in ERBB2/EGFR-mutant stage III NSCLC, again demonstrating shorter PFS than the ERBB2/EGFR wild-type cohort.48

Toxicity of Immunotherapy in Stage III NSCLC

Durvalumab consolidation treatment has demonstrated improved PFS and OS compared to placebo; however, it has not come without immunorelated toxicities. ICIs can affect multiple organ systems.49 The most common AEs of any grade in those receiving anti-PDL1 treatment are fatigue, gastrointestinal (bloody diarrhea, abdominal pain, hepatitis, and jaundice), endocrine (altered thyroid function and hypocalcemia), peripheral neuropathy, and dermatological irAEs.50–53 The most common AE is skin rash, reported by up to 40% of patients, but severe dermatological AEs are rarely reported, even in those receiving combined immunotherapy.54,55 Respiratory AEs, such as pneumonitis, are the most common cause of immunorelated deaths, and have been reported 7–23 months after commencing treatment.56 The incidence of pneumonitis in clinical trials compared to real-world settings appears to vary quite significantly (3%–5% versus 19%, respectively) and may be multifactorial, related to patient selection, pharmacovigilance, and increased awareness of AEs.57

Pneumonitis is of particular interest in stage III NSCLC, as these patients are also at high risk of developing radiation pneumonitis due to the temporal proximity of chemotherapy, radiation treatment, and consolidation with durvalumab.58,59 Differentiating between radiation pneumonitis and immunorelated pneumonitis can be difficult clinically due to timing of onset and overlapping symptoms, and thus the comparison of morphology on CT imaging has become increasingly important. The distribution of changes, extent of lung involvement, presence of ground-glass opacity, consolidation, fibrosis, acute respiratory distress syndrome, and the presence of sharp borders around the edge of the affected areas are all taken into consideration.60 Radiation pneumonitis classically displays unilateral involvement, smaller areas confined to the radiation field, and sharp borders, whereas immunorelated pneumonitis tends to be bilateral with a larger area involved, and is less likely to display sharp borders.60 Immunomediated pneumonitis is considered a diagnosis of exclusion, and workup to rule out other etiologies, including infection, should take place, with input from respiratory physicians as needed.61

The management of immunomediated pneumonitis includes corticosteroid therapy, holding or delaying treatment, or permanently discontinuing treatment in severe cases, according to the Common Terminology Criteria for Adverse Events grade of pneumonitis.56,62 Patients who develop pneumonitis that does not clinically improve with corticosteroids may need rescue therapy with additional immunosuppressive agents, including infliximab, mycophenolate mofetil, or intravenous immunoglobulin.62

There is an increased incidence of severe pneumonitis, both radiation- and immunorelated, in patients with poorer performance status, worse lung function, prior respiratory disease, and smoking history.59,63 Pulmonary function testing is not routinely performed prior to commencement of ICI treatment, but is used as part of the diagnosis of pneumonitis, commonly demonstrating a restrictive pattern and significantly decreased diffusion capacity of the lungs for carbon monoxide.64 The development of a standardized patient assessment prior to commencement of ICIs in patients with preexisting lung disease would identify those with poor lung function who may need heightened monitoring for AEs during their treatment course.65–67

The PACIFIC trial reported that 29.9% of patients who received durvalumab and 26.1% of those in the placebo group had grade 3 or 4 AEs, most commonly pneumonia.26 Overall, 15.4% of patients who received durvalumab discontinued treatment secondary to AEs compared to 9.8% of patients in the placebo group.26 The most frequent AE resulting in cessation of consolidation treatment was pneumonitis. Other commonly reported AEs included diarrhea, rash, and pruritus. Immunomediated AEs of any grade in the durvalumab group were approximately triple those in the placebo group (24.2% vs 8.1%).26 Similar AE incidence was reported in follow-up studies.

Follow-up analysis of the PACIFIC trial data reported grade 3 or 4 AEs in 30.5% of the durvalumab group versus 26.1% of the placebo group, again reporting pneumonitis as the most common AE, with 4.8% of patients in the durvalumab group discontinuing treatment due to this compared to 2.6% of the placebo group.32 Respiratory AEs remained the most common AE in other studies, but some reported a lower incidence of pneumonitis — as low as 15% compared to 23% in the PACIFIC trial.37 The rate of AEs were higher in patients with a history of autoimmune disease, with one study reporting 78% of patients with a known diagnosis of autoimmune disease having an AE of any grade. Despite a higher incidence of AEs in this subgroup, it did not significantly affect PFS or OS.37

Cost of Durvalumab for Stage III NSCLC

Since publication of the PACIFIC trial, the importance of access to durvalumab for stage III NSCLC has been increasingly emphasized. Regulatory agencies in the US, Canada, Australia, Japan, Singapore, and India have approved durvalumab consolidation treatment in unresectable stage III NSCLC. Controversially, the European Medicines Agency (EMA) approved its use only in patients whose NSCLCs have PDL1 expression ≥1%. Multiple studies have demonstrated the cost-effectiveness of durvalumab, and although funding varies across different countries, the overall conclusion is that durvalumab consolidation treatment is cost-effective compared to combined chemoradiotherapy alone, accentuating the importance of increased access.68–70 Cost-effectiveness analysis has shown no consolidation therapy resulted in a mean cost of US$185,944 and mean quality-adjusted life-years of 2.34. Durvalumab consolidation increased quality-adjusted life-years to 2.57, with an increase in mean cost to $201,563.68 A retrospective study carried out in Japan in 2019 of 81 patients with unresectable stage III NSCLC found 70% would be eligible to receive durvalumab consolidation therapy based on PACIFIC criteria.71 Since approval by the EMA in 2018, its administration has risen annually, and now approximately 50% of eligible patients with stage III NSCLC are gaining access to durvalumab.72 A retrospective review of 82 patients in Japan with unresectable stage III NSCLC highlighted that although some patients may meet eligibility criteria for durvalumab therapy initially, this status may change after they undergo chemoradiotherapy, so initial eligibility for treatment may not coincide with real-world administration rates.73

Discussion

The use of ICIs has altered the landscape of anticancer treatment in diagnoses with previously poor outcomes. Anti-PDL1 maintenance treatment with durvalumab after combined chemoradiotherapy improves both PFS and OS in unresectable stage III NSCLC. This was first demonstrated in the PACIFIC trial, but multiple studies published after this have consolidated these results in the last 4 years in real-world settings. In this review, we have highlighted the prospective and real-world data in support of durvalumab maintenance in stage III NSCLC, other immunotherapeutic approaches for stage III NSCLC, such as neoadjuvant immunotherapy, immunotherapy for those with oncogene-addicted NSCLCs, and immunotherapy toxicity that is of particular interest in stage III NSCLC.

Our review has also highlighted several controversial issues in the use of immunotherapy for stage III NSCLC. Although the PACIFIC trial did not select for PDL1 status in its inclusion criteria and its access in the US and other countries did not select for it, a division developed when the EMA approval process required a PDL1 status ≥1%. PACIFIC trial criteria also specified a World Health Organization performance status of 0 or 1 and excluded patients with a history of any autoimmune disease.26 However, stage III NSCLC can be considered a broad spectrum of disease, and subsequent studies have noted that a large proportion of patients with this disease have a performance status >1 when commencing treatment, highlighting that the patients enrolled in the original study may not fully represent a real-world population.37 Subsequent real-world studies have included a broader range of patients, including those with autoimmune disease and a performance status >1, confirming durvalumab benefits in more heterogeneous and real-world patient cohorts.37 While the PACIFIC trial did not specify tumor analysis, follow-up studies have also demonstrated reduced efficacy of durvalumab in patients with EGFR-mutant NSCLCs.47

An open question in stage III NSCLC that remains unaddressed is the optimal duration of ICI therapy. Multiple trials have assessed continuing treatment for varying durations: until disease progression, unacceptable toxicity, or fixed duration of 1 or 2 years. Three trials assessing nivolumab — CA-209-003, Checkmate-153, and Checkmate-017/057 — varied from up to 2 years’ treatment, 1 year versus continuous, and continuous treatment, respectively.74–76 Current evidence suggests that continuing ICIs beyond 1 year can improve outcomes, including PFS and OS, up to 5 years compared to continuing docetaxel.76 Pembrolizumab-treatment duration in Keynote-001 and Keynote-010 was either continuous or up to 2 years, respectively. In Keynote-001, continuous treatment showed an OS of 25% at 5 years in both treatment-naïve and previously treated advanced NSCLC.77 Keynote-010 showed similar data: patients treated with 2 years of pembrolizumab showed increased OS, and a second course of pembrolizumab after disease progression provided significant disease control.78 Atezolizumab duration in the OAK trial was continuous, and follow-up data after 2 years demonstrated a survival benefit compared to docetaxel.79 All three ICIs here — nivolumab, pembrolizumab, and atezolizumab — work via the PD1–PDL1 pathway, and with the addition of durvalumab as a new consolidation therapy, further research into the optimum duration of treatment is required.

Lastly, as more patients survive from stage III NSCLC after durvalumab and live beyond 5 years, patients may enter an era of “survivorship” after NSCLC. Current issues of survivorship in lung cancer include but are not limited to physical symptoms, psychological distress and socioeconomic issues secondary to cost of treatment, and time spent out of work.80 There may be unique issues related to survivorship after immunotherapy, such as the management of long-term immunotoxiciy, eg, hypothyroidism and pneumonitis.81 A retrospective review of 159 patients who received nivolumab, pembrolizumab, or atezolizumab identified almost 40% of patients developed new or worsening AEs after 6 months of treatment.81 This emphasizes the need to ascertain optimal treatment duration to increase PFS and OS, but limit the incidence of long-term toxicity. The development of a multidisciplinary immunorelated toxicity team has been shown to be beneficial and feasible in the acute management of ICI toxicity, and perhaps this approach with long-term survival is also required.82 A study encompassing the importance of a multidisciplinary-team approach to survivorship issues post–ICI treatment used a 1-hour webcast available on demand for physicians to increase awareness of AEs, a resource that encompasses both medical and psychological long-term effects, including quality-of-life parameters.83,84

Advanced NSCLC has long been a diagnosis associated with poor outcomes, high symptom burden, and limited treatment options. The advent of immunotherapy, and in this case durvalumab consolidation treatment in stage III unresectable NSCLC, has been shown to improve survival significantly, providing hope to both clinicians and patients about the changing landscape of lung cancer management.

Disclosure

Dr Jarushka Naidoo reports grants and personal fees from AstraZeneca, Bristol Myers Squibb, Merck, Roche/Genentech, Takeda, Pfizer, and Daiichi Sankyo during the conduct of the study. The authors report no other conflicts of interest in this work.

References

1. de Groot PM, Wu CC, Carter BW, Munden RF. The epidemiology of lung cancer. Transl Lung Cancer Res. 2018;7(3):220. doi:10.21037/tlcr.2018.05.06

2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin. 2015;65(1):5–29. doi:10.3322/caac.21254

3. Barta JA, Powell CA, Wisnivesky JP. Global epidemiology of lung cancer. Ann Global Health. 2019;85. doi:10.5334/aogh.2419

4. Yang H, Shen K, Zhu C, Li Q, Zhao Y, Ma X. Safety and efficacy of durvalumab (MEDI4736) in various solid tumors. Drug Des Devel Ther. 2018;12:2085–2096. doi:10.2147/DDDT.S162214

5. Darvin P, Toor SM, Sasidharan Nair V, Elkord E. Immune checkpoint inhibitors: recent progress and potential biomarkers. Exp Mol Med. 2018;50(12):1–11. doi:10.1038/s12276-018-0191-1

6. Butte MJ, Keir ME, Phamduy TB, Sharpe AH, Freeman GJ. Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses. Immunity. 2007;27(1):111–122. doi:10.1016/j.immuni.2007.05.016

7. Shi L, Chen S, Yang L, Li Y. The role of PD-1 and PD-L1 in T-cell immune suppression in patients with hematological malignancies. J Hematol Oncol. 2013;6(1):74. doi:10.1186/1756-8722-6-74

8. Naidoo J, Page DB, Wolchok JD. Immune checkpoint blockade. Hematol Oncol Clin North Am. 2014;28(3):585–600. doi:10.1016/j.hoc.2014.02.002

9. Mahoney KM, Freeman GJ, McDermott DF. The next immune-checkpoint inhibitors: PD-1/PD-L1 blockade in melanoma. Clin Ther. 2015;37(4):764–782. doi:10.1016/j.clinthera.2015.02.018

10. Akinleye A, Rasool Z. Immune checkpoint inhibitors of PD-L1 as cancer therapeutics. J Hematol Oncol. 2019;12(1):92. doi:10.1186/s13045-019-0779-5

11. Flippot R, Escudier B, Albiges L. Immune checkpoint inhibitors: toward new paradigms in renal cell carcinoma. Drugs. 2018;78(14):1443–1457. doi:10.1007/s40265-018-0970-y

12. Iwai Y, Hamanishi J, Chamoto K, Honjo T. Cancer immunotherapies targeting the PD-1 signaling pathway. J Biomed Sci. 2017;24. doi:10.1186/s12929-017-0329-9

13. Kleffel S, Posch C, Barthel SR, et al. Melanoma cell-intrinsic PD-1 receptor functions promote tumor growth. Cell. 2015;162(6):1242–1256. doi:10.1016/j.cell.2015.08.052

14. Weinstock M, McDermott D. Targeting PD-1/PD-L1 in the treatment of metastatic renal cell carcinoma. Ther Adv Urol. 2015;7(6):365–377. doi:10.1177/1756287215597647

15. Xu-Monette ZY, Zhou J, Young KH. PD-1 expression and clinical PD-1 blockade in B-cell lymphomas. Blood. 2018;131:68–83. doi:10.1182/blood-2017-07-740993

16. Stenehjem DD, Tran D, Nkrumah MA, Gupta S. PD1/PDL1 inhibitors for the treatment of advanced urothelial bladder cancer. Onco Targets Ther. 2018;11:5973–5989. doi:10.2147/OTT.S135157

17. Chan IS, Bhatia S, Kaufman HL, Lipson EJ. Immunotherapy for Merkel cell carcinoma: a turning point in patient care. J Immunother Cancer. 2018;6(1):23. doi:10.1186/s40425-018-0335-9

18. Reck M, Rodríguez-Abreu D, Robinson AG, et al. Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer. N Engl J Med. 2016;375(19):1823–1833. doi:10.1056/NEJMoa1606774

19. Gandhi L, Rodríguez-Abreu D, Gadgeel S, et al. Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N Engl J Med. 2018;378(22):2078–2092. doi:10.1056/NEJMoa1801005

20. Paz-Ares L, Luft A, Vicente D, et al. Pembrolizumab plus chemotherapy for squamous non-small-cell lung cancer. N Engl J Med. 2018;379(21):2040–2051. doi:10.1056/NEJMoa1810865

21. Mok TSK, Wu YL, Kudaba 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(10183):1819–1830. doi:10.1016/S0140-6736(18)32409-7

22. Hellmann Md, Paz-Ares L, Bernabe Caro R, et al. Nivolumab plus ipilimumab in advanced non-small-cell lung cancer. N Engl J Med. 2019;381(21):2020–2031. doi:10.1056/NEJMoa1910231

23. Paz-Ares L, Ciuleanu TE, Cobo M, 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(2):198–211. doi:10.1016/S1470-2045(20)30641-0

24. Socinski MA, Jotte RM, Cappuzzo F, et al. Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC. N Engl J Med. 2018;378(24):2288–2301. doi:10.1056/NEJMoa1716948

25. Evison M. The current treatment landscape in the UK for stage III NSCLC. Br J Cancer. 2020;123(Suppl 1):3–9. doi:10.1038/s41416-020-01069-z

26. Antonia SJ, Villegas A, Daniel D, et al. Durvalumab after chemoradiotherapy in stage III non-small-cell lung cancer. N Engl J Med. 2017;377(20):1919–1929. doi:10.1056/NEJMoa1709937

27. Albain KS, Crowley JJ, Turrisi AT, et al. Concurrent cisplatin, etoposide, and chest radiotherapy in pathologic stage IIIB non-small-cell lung cancer: a Southwest Oncology Group phase II study, SWOG 9019. J Clin Oncol. 2002;20(16):3454–3460. doi:10.1200/JCO.2002.03.055

28. Govindan R, Bogart J, Stinchcombe T, et al. Randomized phase II study of pemetrexed, carboplatin, and thoracic radiation with or without cetuximab in patients with locally advanced unresectable non-small-cell lung cancer: cancer and Leukemia Group B trial 30407. J Clin Oncol. 2011;29(23):3120–3125. doi:10.1200/JCO.2010.33.4979

29. Curran WJ, Paulus R, Langer CJ, et al. Sequential vs. concurrent chemoradiation for stage III non-small cell lung cancer: randomized phase III trial RTOG 9410. J Natl Cancer Inst. 2011;103(19):1452–1460. doi:10.1093/jnci/djr325

30. Hu C, Machtay M, Dignam JJ, Paulus R, Bradley JD. Progression-free survival (PFS) and cardiac-toxicity-adjusted-PFS (CTA-PFS) as predictors of overall survival (OS) in locally advanced non-small cell lung cancers (LA-NSCLC) treated with concurrent chemoradiation (CCRT): a secondary analysis of NRG Oncology RTOG 0617. J Clin Oncol. 2018;36:8539. doi:10.1200/jco.2018.36.15_suppl.8539

31. Skrzypski M, Jassem J. Consolidation systemic treatment after radiochemotherapy for unresectable stage III non-small cell lung cancer. Cancer Treat Rev. 2018;66:114–121. doi:10.1016/j.ctrv.2018.04.001

32. Antonia SJ, Villegas A, Daniel D, et al. Overall survival with durvalumab after chemoradiotherapy in stage III NSCLC. N Engl J Med. 2018;379(24):2342–2350. doi:10.1056/NEJMoa1809697

33. Faivre-Finn C, Vicente D, Kurata T, et al. Four-year survival with durvalumab after chemoradiotherapy in stage III NSCLC-an update from the PACIFIC trial. J Thorac Oncol. 2021;16(5):860–867. doi:10.1016/j.jtho.2020.12.015

34. Spigel DR, Faivre-Finn C, Gray JE, et al. Five-year survival outcomes with durvalumab after chemoradiotherapy in unresectable stage III NSCLC: an update from the PACIFIC trial. J Clin Oncol. 2021;39:8511. doi:10.1200/jco.2021.39.15_suppl.8511

35. Eichkorn T, Bozorgmehr F, Regnery S, et al. Consolidation immunotherapy after platinum-based chemoradiotherapy in patients with unresectable stage III non-small cell lung cancer-cross-sectional study of eligibility and administration rates. Front Oncol. 2020;10:586449. doi:10.3389/fonc.2020.586449

36. Desilets A, Blanc-Durand F, Lau S, et al. Durvalumab therapy following chemoradiation compared with a historical cohort treated with chemoradiation alone in patients with stage III non-small cell lung cancer: a real-world multicentre study. Eur J Cancer. 2021;142:83–91. doi:10.1016/j.ejca.2020.10.008

37. Faehling M, Schumann C, Christopoulos P, et al. Durvalumab after definitive chemoradiotherapy in locally advanced unresectable non-small cell lung cancer (NSCLC): real-world data on survival and safety from the German expanded-access program (EAP). Lung Cancer. 2020;150:114–122. doi:10.1016/j.lungcan.2020.10.006

38. Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45(2):228–247. doi:10.1016/j.ejca.2008.10.026

39. Taugner J, Käsmann L, Eze C, et al. Real-world prospective analysis of treatment patterns in durvalumab maintenance after chemoradiotherapy in unresectable, locally advanced NSCLC patients. Invest New Drugs. 2021;39(4):1189–1196. doi:10.1007/s10637-021-01091-9

40. Forde PM, Chaft JE, Smith KN, et al. Neoadjuvant PD-1 blockade in resectable lung cancer. N Engl J Med. 2018;378(21):1976–1986. doi:10.1056/NEJMoa1716078

41. Provencio M, Nadal E, Insa A, et al. Neoadjuvant chemotherapy and nivolumab in resectable non-small-cell lung cancer (NADIM): an open-label, multicentre, single-arm, phase 2 trial. Lancet Oncol. 2020;21(11):1413–1422. doi:10.1016/S1470-2045(20)30453-8

42. Cascone T, William WN, Weissferdt A, et al. Neoadjuvant nivolumab or nivolumab plus ipilimumab in operable non-small cell lung cancer: the phase 2 randomized NEOSTAR trial. Nat Med. 2021;27(3):504–514. doi:10.1038/s41591-020-01224-2

43. Jabbour SK, Lee KH, Frost N, et al. Pembrolizumab plus concurrent chemoradiation therapy in patients with unresectable, locally advanced, stage III non-small cell lung cancer: the phase 2 KEYNOTE-799 nonrandomized trial. JAMA Oncol. 2021;7(9):1351. doi:10.1001/jamaoncol.2021.2301

44. Durm GA, Althouse SK, Sadiq AA, et al. Phase II trial of concurrent chemoradiation with consolidation pembrolizumab in patients with unresectable stage III non-small cell lung cancer: Hoosier Cancer Research Network LUN 14-179. J Clin Oncol. 2018;36:8500. doi:10.1200/jco.2018.36.15_suppl.8500

45. Ross HJ, Kozono DE, Urbanic JJ, et al. AFT-16: phase II trial of atezolizumab before and after definitive chemoradiation (CRT) for unresectable stage III non-small cell lung cancer (NSCLC). J Clin Oncol. 2020;38:9045. doi:10.1200/jco.2020.38.15_suppl.9045

46. Rothschild SI, Zippelius A, Eboulet EI, et al. SAKK 16/14: durvalumab in addition to neoadjuvant chemotherapy in patients with stage IIIA(N2) non-small-cell lung cancer-a multicenter single-arm phase II trial. J Clin Oncol. 2021;39:2872–2880.

47. Aredo JV, Mambetsariev I, Hellyer JA, et al. Durvalumab for stage III EGFR-mutated NSCLC after definitive chemoradiotherapy. J Thorac Oncol. 2021;16(6):1030–1041. doi:10.1016/j.jtho.2021.01.1628

48. Hellyer JA, Aredo JV, Das M, et al. Role of consolidation durvalumab in patients with EGFR- and HER2-mutant unresectable stage III NSCLC. J Thorac Oncol. 2021;16(5):868–872. doi:10.1016/j.jtho.2020.12.020

49. Spiers L, Coupe N, Payne M. Toxicities associated with checkpoint inhibitors—an overview. Rheumatology. 2019;58(Suppl7):vii7. doi:10.1093/rheumatology/kez418

50. Puzanov I, Diab A, Abdallah K, et al.; Society for Immunotherapy of Cancer Toxicity Management Working Group. Managing toxicities associated with immune checkpoint inhibitors: consensus recommendations from the Society for Immunotherapy of Cancer (SITC) Toxicity Management Working Group. J Immunother Cancer. 2017;5. doi:10.1186/s40425-017-0300-z

51. Cousin S, Seneschal J, Italiano A. Toxicity profiles of immunotherapy. Pharmacol Ther. 2018;181:91–100. doi:10.1016/j.pharmthera.2017.07.005

52. Belum VR, Benhuri B, Postow MA, et al. Characterisation and management of dermatologic adverse events to agents targeting the PD-1 receptor. Eur J Cancer. 2016;60:12–25. doi:10.1016/j.ejca.2016.02.010

53. Baraibar I, Melero I, Ponz-Sarvise M, Castanon E. Safety and tolerability of immune checkpoint inhibitors (PD-1 and PD-L1) in Cancer. Drug Saf. 2019;42(2):281–294. doi:10.1007/s40264-018-0774-8

54. Villadolid J, Amin A. Immune checkpoint inhibitors in clinical practice: update on management of immune-related toxicities. Transl Lung Cancer Res. 2015;4(5):560–575.

55. Naidoo J, Schindler K, Querfeld C, et al. Autoimmune bullous skin disorders with immune checkpoint inhibitors targeting PD-1 and PD-L1. Cancer Immunol Res. 2016;4(5):383–389. doi:10.1158/2326-6066.CIR-15-0123

56. Naidoo J, Page DB, Li BT, et al. Toxicities of the anti-PD-1 and anti-PD-L1 immune checkpoint antibodies. Ann Oncol. 2015;26(12):2375–2391. doi:10.1093/annonc/mdv383

57. Suresh K, Voong KR, Shankar B, et al. Pneumonitis in non-small cell lung cancer patients receiving immune checkpoint immunotherapy: incidence and risk factors. J Thorac Oncol. 2018;13(12):1930–1939. doi:10.1016/j.jtho.2018.08.2035

58. McGovern K, Ghaly M, Esposito M, Barnaby K, Seetharamu N. Radiation recall pneumonitis in the setting of immunotherapy and radiation: a focused review. Future Sci OA. 2019;5(5):FSO378. doi:10.2144/fsoa-2018-0123

59. Naidoo J, Wang X, Woo KM, et al. Pneumonitis in patients treated with anti-programmed death-1/programmed death ligand 1 therapy. J Clin Oncol. 2017;35(7):709–717. doi:10.1200/JCO.2016.68.2005

60. Chen X, Sheikh K, Nakajima E, et al. Radiation versus immune checkpoint inhibitor associated pneumonitis: distinct radiologic morphologies. Oncologist. 2021;26(10). doi:10.1002/onco.13900

61. Naidoo J, Reuss JE, Suresh K, et al. Immune-related (IR)-pneumonitis during the COVID-19 pandemic: multidisciplinary recommendations for diagnosis and management. J Immunother Cancer. 2020;8(1):e000984. doi:10.1136/jitc-2020-000984

62. Naidoo J, Nishino M, Patel SP, et al. Immune-related pneumonitis after chemoradiotherapy and subsequent immune checkpoint blockade in unresectable stage III non–small-cell lung cancer. Clin Lung Cancer. 2020;21:e435–44. doi:10.1016/j.cllc.2020.02.025

63. O’Kane GM, Labbé C, Doherty MK, Young K, Albaba H, Leighl NB. Monitoring and management of immune‐related adverse events associated with programmed cell death protein‐1 axis inhibitors in lung cancer. Oncologist. 2017;22(1):70. doi:10.1634/theoncologist.2016-0164

64. Franzen D, Schad K, Kowalski B, et al. Ipilimumab and early signs of pulmonary toxicity in patients with metastastic melanoma: a prospective observational study. Cancer Immunol Immunother. 2018;67(1):127–134. doi:10.1007/s00262-017-2071-2

65. Cui P, Liu Z, Wang G, et al. Risk factors for pneumonitis in patients treated with anti-programmed death-1 therapy: a case-control study. Cancer Med. 2018;7(8):4115–4120. doi:10.1002/cam4.1579

66. Cho JY, Kim J, Lee JS, et al. Characteristics, incidence, and risk factors of immune checkpoint inhibitor-related pneumonitis in patients with non-small cell lung cancer. Lung Cancer. 2018;125:150–156. doi:10.1016/j.lungcan.2018.09.015

67. Okada N, Matsuoka R, Sakurada T, et al. Risk factors of immune checkpoint inhibitor-related interstitial lung disease in patients with lung cancer: a single-institution retrospective study. Sci Rep. 2020;10(1):13773. doi:10.1038/s41598-020-70743-2

68. Criss SD, Mooradian MJ, Sheehan DF, et al. Cost-effectiveness and budgetary consequence analysis of durvalumab consolidation therapy vs no consolidation therapy after chemoradiotherapy in stage III non-small cell lung cancer in the context of the US health care system. JAMA Oncol. 2019;5(3):358–365. doi:10.1001/jamaoncol.2018.5449

69. Mehra R, Yong C, Seal B, van Keep M, Raad A, Zhang Y. Cost-effectiveness of durvalumab after chemoradiotherapy in unresectable stage III NSCLC: a US healthcare perspective. J Natl Compr Canc Netw. 2021;19(2):153–162. doi:10.6004/jnccn.2020.7621

70. Armeni P, Borsoi L, Fornaro G, Jommi C, Grossi F, Costa F. Cost-effectiveness and net monetary benefit of durvalumab consolidation therapy versus no consolidation therapy after chemoradiotherapy in stage III non-small cell lung cancer in the Italian national health service. Clin Ther. 2020;42(5):830–847. doi:10.1016/j.clinthera.2020.03.012

71. Sakaguchi T, Ito K, Furuhashi K, et al. Patients with unresectable stage III non-small cell lung cancer eligible to receive consolidation therapy with durvalumab in clinical practice based on PACIFIC study criteria. Respir Investig. 2019;57(5):466–471. doi:10.1016/j.resinv.2019.03.011

72. Patel P, Alrifai D, McDonald F, Forster M. Beyond chemoradiotherapy: improving treatment outcomes for patients with stage III unresectable non-small-cell lung cancer through immuno-oncology and durvalumab (Imfinzi®▼, AstraZeneca UK Limited). Br J Cancer. 2020;123(Suppl 1):18–27. doi:10.1038/s41416-020-01071-5

73. Hosoya K, Fujimoto D, Kawachi H, et al. Ineligibility for the PACIFIC trial in unresectable stage III non-small cell lung cancer patients. Cancer Chemother Pharmacol. 2019;84(2):275–280. doi:10.1007/s00280-019-03885-4

74. Gettinger S, Horn L, Jackman D, et al. Five-year follow-up of nivolumab in previously treated advanced non-small-cell lung cancer: results from the CA209-003 study. J Clin Oncol. 2018;36(17):1675–1684. doi:10.1200/JCO.2017.77.0412

75. Waterhouse DM, Garon EB, Chandler J, et al. Continuous versus 1-year fixed-duration nivolumab in previously treated advanced non-small-cell lung cancer: checkMate 153. J Clin Oncol. 2020;38(33):3863–3873. doi:10.1200/JCO.20.00131

76. Borghaei H, Gettinger S, Vokes EE, et al. Five-year outcomes from the randomized, phase III trials checkMate 017 and 057: nivolumab versus docetaxel in previously treated non-small-cell lung cancer. J Clin Oncol. 2021;39(7):723–733. doi:10.1200/JCO.20.01605

77. Garon EB, Hellmann MD, Rizvi NA, et al. Five-year overall survival for patients with advanced non‒small-cell lung cancer treated with pembrolizumab: results from the phase I KEYNOTE-001 study. J Clin Oncol. 2019;37(28):2518–2527. doi:10.1200/JCO.19.00934

78. Herbst R, Garon E, Kim D, et al. FP13.01 5-year survival update from KEYNOTE-010: pembrolizumab versus docetaxel in previously treated, PD-L1–positive advanced NSCLC. J Thorac Oncol. 2021;16(3):S223–S224. doi:10.1016/j.jtho.2021.01.140

79. von Pawel J, Bordoni R, Satouchi M, et al. Long-term survival in patients with advanced non-small-cell lung cancer treated with atezolizumab versus docetaxel: results from the randomised phase III OAK study. Eur J Cancer. 2019;107:124–132. doi:10.1016/j.ejca.2018.11.020

80. Vijayvergia N, Shah PC, Denlinger CS. Survivorship in non–small cell lung cancer: challenges faced and steps forward. J Natl Compr Canc Netw. 2015;13:1151–1161. doi:10.6004/jnccn.2015.0140

81. Hall KH, Liu Y, Jiang C, Harvey RD. New and worsening long-term immune-related adverse events with PD-1/PD-L1 pathway agents in patients with cancer. Pharmacotherapy. 2020;40(2):133–141. doi:10.1002/phar.2354

82. Naidoo J, Zhang J, Lipson EJ, et al. A multidisciplinary toxicity team for cancer immunotherapy-related adverse events. J Natl Compr Canc Netw. 2019;17(6):712–720. doi:10.6004/jnccn.2018.7268

83. Perloff T, Dawkins M, Hurd M, Wang J, Hallmeyer S. Planning for survivorship after checkpoint inhibitors: a multidisicplinary education initiative. J Clin Oncol. 2019;37:98. doi:10.1200/jco.2019.37.8_suppl.98

84. O’Reilly A, Hughes P, Mann J, et al. An immunotherapy survivor population: health-related quality of life and toxicity in patients with metastatic melanoma treated with immune checkpoint inhibitors. Support Care Cancer. 2020;28(2):561–570. doi:10.1007/s00520-019-04818-w

Creative Commons License © 2021 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.