Putting the brakes on CTLA-4 inhibition in lung cancer?
Editorial

Putting the brakes on CTLA-4 inhibition in lung cancer?

Meghan J. Mooradian, Justin F. Gainor

Department of Medicine, Massachusetts General Hospital, Boston, MA, USA

Correspondence to: Justin F. Gainor, MD. 10 North Grove Street, LRH-238, Boston, MA 02114, USA. Email: jgainor@partners.org.

Provenance: This is an invited Editorial commissioned by Section Editor Viola Zhu, MD, PhD (Assistant Clinical Professor of Medicine, Chao Family Comprehensive Cancer Center, Division of Hematology/Oncology, University of California, Irvine School of Medicine, Orange, CA, USA).

Comment on: Govindan R, Szczesna A, Ahn MJ, et al. Phase III Trial of Ipilimumab Combined With Paclitaxel and Carboplatin in Advanced Squamous Non-Small-Cell Lung Cancer. J Clin Oncol 2017;35:3449-57.


Submitted Dec 03, 2017. Accepted for publication Jan 09, 2018.

doi: 10.21037/tlcr.2018.01.05


Immune checkpoint inhibitors have fundamentally changed the treatment landscape of non-small cell lung cancer (NSCLC). Indeed, within the last 3 years, single-agent programmed death receptor-1 (PD-1)/programmed death ligand-1 (PD-L1) inhibitors have become standard therapies in both the first- and second-line settings for patients with advanced disease (1-4). Despite these advancements, however, only a minority of patients experience durable responses to PD-1 pathway inhibition. As a result, recent efforts have focused on identifying alternative immune checkpoints and combination strategies to potentiate anti-cancer immune responses.

In the manuscript accompanying this commentary, Govindan and colleagues report findings from a phase 3 study evaluating the activity of ipilimumab, a cytotoxic T-cell lymphocyte antigen-4 (CTLA-4) inhibitor, in combination with paclitaxel and carboplatin in advanced squamous NSCLC (5). Eligible patients had recurrent or treatment-naïve stage IV squamous NSCLC. Subjects were randomly assigned in a 1:1 ratio to receive paclitaxel and carboplatin plus blinded ipilimumab 10 mg/kg or placebo every 3 weeks on a phased induction schedule that was comprised of six chemotherapy cycles, with ipilimumab or placebo administered between cycles 3 to 6. Following induction, ipilimumab or placebo maintenance was administered every 12 weeks. The primary endpoint was overall survival (OS). Of note, due to a higher than anticipated discontinuation rate of subjects prior to initiation of blinded study therapy, the primary endpoint was amended to OS in the modified intention-to-treat (mITT) population (all assigned subjects who received at least 1 dose of blinded study therapy). Importantly, in the mITT population (n=749), there was no difference in OS between study arms. Furthermore, equivocal results were seen across most pre-defined sub-groups. Secondary efficacy endpoints, including progression-free survival (PFS) and overall response rate (ORR), also did not differ between arms. Notably, there were higher rates of significant toxicity in the ipilimumab combination cohort compared with the placebo arm (grade ≥3 events, 53% vs. 36%, respectively), with more grade 5 events (n=7 vs. 1, respectively) and more patients discontinuing treatment because of toxicity (28% vs. 7%, respectively).

Given the above findings, the study by Govindan and colleagues raises several important questions. Most notably, why did CTLA-4 inhibition fail to improve outcomes when coupled with chemotherapy in this study? The background rationale for this combination is that cytotoxic chemotherapy may induce immunogenic cancer cell death, and checkpoint inhibition may consolidate this response by inducing long-lasting, immune-mediated tumor control (6,7). Early signals of activity from such an approach came from two randomized phase II studies of ipilimumab and chemotherapy (carboplatin/paclitaxel) in patients with treatment-naïve, advanced small cell and NSCLC, both of which demonstrated improvements in immune-related PFS with phased ipilimumab (i.e., beginning with cycle 3), but not concurrent ipilimumab (i.e., beginning with cycle 1) (8,9). Despite these promising results, however, recent confirmatory phase III studies of phased ipilimumab in combination with chemotherapy in both patient populations have been negative (5,10).

One potential explanation for these negative findings is that compared to other disease settings (e.g., advanced melanoma), the dose of ipilimumab in these studies (10 mg/kg) was too high and contributed to excess treatment-related toxicity. Indeed, Govindan et al. postulate that the increased toxicity and treatment discontinuation rates in the ipilimumab arm, along with corresponding reductions in exposure to chemotherapy, may have contributed to the lack of efficacy.

An alternative explanation is that the study was performed without a biomarker selection strategy. Based upon the experience with other forms of immunotherapy, it’s possible that only a small subset of patients with lung cancer derive benefit from the addition of CTLA-4 inhibition to chemotherapy, but such benefit may have been obscured in an unselected patient population. For example, in melanoma, only ~10–15% of patients experience objective responses to single-agent ipilimumab (11,12). In NSCLC, the activity is likely to be even lower. Indeed, in early studies, CTLA-4 inhibitors had minimal anti-tumor activity as single agents in NSCLC (13). While chemotherapy has been hypothesized to augment this activity, it is unlikely that such benefit would extend beyond a subgroup of patients. Therefore, biomarker selection would ideally be incorporated into such trials, but unfortunately, predictive biomarkers for CTLA-4 inhibition have proven elusive to date.

Beyond toxicity considerations and the lack of an effective predictive biomarker, an additional hypothesis for the limited activity of ipilimumab in NSCLC revolves around the mechanism of action of CTLA-4 inhibition. Ipilimumab is believed to stimulate early-stage T-cell activation in the lymphoid compartment; however, this may be insufficient to generate an effective antitumor response within the tumor microenvironment. To induce the latter, stimulation of effector T-cell function within the tumor microenvironment may also be necessary (14).

When taking the above factors together, we are left with a fundamental question: is there still a place for CTLA-4 inhibition in NSCLC? While the above data by Govindan and colleagues certainly dampens enthusiasm for use of CTLA-4 inhibitors with chemotherapy, CTLA-4 inhibition may still have a role in certain settings—particularly when used in combination with other immune checkpoint inhibitors. Indeed, multiple studies evaluating combinations of CTLA-4 and PD-1 pathway inhibitors are now ongoing with the rationale that blockade of CTLA-4 may enhance early immune activation in lymphoid tissue while PD-1 inhibition may enhance tumor cell kill in the periphery (15). Thus, blockade of both CTLA-4 and PD-1/PD-L1 may produce non-overlapping effects (16). Indeed, in vivo studies demonstrate synergy with dual checkpoint blockade compared with either single-agent alone (17).

In the clinic, combinations of PD-1/PD-L1 and CTLA-4 inhibitors were initially explored in advanced melanoma, and initial data from Checkmate-067 was suggestive that combination therapy was destined to become the new standard of care. In this phase III trial, nivolumab 3 mg/kg alone or nivolumab 1 mg/kg plus ipilimumab 3 mg/kg was compared to single agent ipilimumab 3 mg/kg in patients with metastatic melanoma. Despite robust PFS and ORR data (18), enthusiasm for the front-line combination has been tempered in part due to concerns regarding the tolerability of combined checkpoint blockade. For example, treatment-related grade 3 or 4 toxicity exceeded 50% with combination nivolumab and ipilimumab, and 36% of patients discontinued therapy (18). Furthermore, though the study was not powered for direct comparison of combination therapy vs. single-agent PD-1 inhibition, the recently published 3-year OS rates (combination therapy 58%, nivolumab alone 52%) forces clinicians to weigh the risks and benefits of dual checkpoint blockade and carefully select patients for the combinatorial approach (19).

In NSCLC, PD-(L)1 plus CTLA-4 combinations have also shown initial promise (20-22). For example, in Checkmate-012, the combination of nivolumab and ipilimumab produced objective responses in 38–47% of treatment-naïve patients, including responses among PD-L1 negative patients (22). Given the experience of such combinations in melanoma, several different doses and schedules were evaluated in Checkmate-012 in an effort to improve tolerability. Ultimately, a dose-schedule of nivolumab 3 mg/kg every 2 weeks plus ipilimumab 1 mg/kg every 6 weeks was selected for further evaluation, including in an ongoing randomized phase III study (Checkmate-227; NCT02477826).

Despite the initial promising results of combined CTLA-4 plus PD-1 pathway inhibition above, enthusiasm for this strategy has been somewhat stifled more recently with a preliminary report from the phase III MYSTIC trial (23). In this randomized, open-label, global study, patients with treatment-naïve, advanced NSCLC were randomized 1:1:1 to receive the PD-L1 inhibitor durvalumab, durvalumab plus tremelimumab (a CTLA-4 antagonist), or platinum-based chemotherapy. Notably, this study failed to meet its primary endpoint of improved PFS in the durvalumab/tremelimumab arm compared to chemotherapy among patients with high PD-L1 expression (defined as PD-L1 expression on 25% or more of their cancer cells). Data pertaining to ORR, duration of response, as well as OS of durvalumab +/− tremelimumab is pending.

CTLA-4-based combination strategies are also under investigation in small-cell lung cancer (SCLC). Interestingly, despite the high mutational burden of SCLC, single agent PD-1/PD-L1 therapy has produced ORRs of only ~10%. However, the addition of ipilimumab 3 mg/kg to nivolumab 1 mg/kg was associated with a doubling of the ORR compared to nivolumab monotherapy (24). Moreover, OS rates at 1 and 2 years were also significantly higher with the combination compared to nivolumab monotherapy (25). Importantly, and in contrast to NSCLC, PD-L1 expression did not predict for response in SCLC; however, emerging data suggests that a high tumor mutational burden (TMB) may serve as a predictive biomarker for combined checkpoint blockade in this setting (26). Further prospective validation and optimization of TMB cut-off values will be required before it is incorporated in clinical practice.

While the therapeutic landscape for patients with advanced lung cancer has evolved dramatically over the last 5 years, a majority of patients still fail to respond to immune checkpoint blockade. It is hoped that combination approaches may enhance efficacy and durability of immune-based therapies while keeping toxicity at a minimum. The important work by Govindan and colleagues (5) demonstrates that combination cytotoxic chemotherapy and CTLA-4 antagonism is not the best path forward. With this knowledge, investigators can now focus on alternative strategies, including the investigation of novel immunotherapies and new combinations.

Based upon its single-agent activity and tolerability, PD-1/PD-L1 inhibition is the logical backbone of any front-line combination strategy moving forward. It remains to be seen however whether cytotoxic chemotherapy or a second immune checkpoint inhibitor (e.g., CTLA-4 inhibitor) is the most effective and tolerable partner for PD-1 blockade. Recently, data from two randomized studies, KEYNOTE 021G and IMpower 150, demonstrated improved outcomes with PD-1/PD-L1 inhibition plus chemotherapy compared to chemotherapy alone, and one regimen (carboplatin/pemetrexed/pembrolizumab) recently gained regulatory approval in the United States for the first-line treatment of nonsquamous NSCLC (27,28). As a result, data from several ongoing phase III studies evaluating PD-1 inhibition added to platinum-based chemotherapy [e.g., KEYNOTE-189 (NCT02578680), KEYNOTE-407 (NCT02775435), Checkmate 227 (NCT02477826)] are highly anticipated. In addition, a host of clinical trials are now underway exploring various co-stimulatory, co-inhibitory, and immune-metabolic agents. While studies of these combinations mature, it appears CTLA-4 inhibition will remain in the conversion, but for how long remains unclear.


Acknowledgements

None.


Footnote

Conflicts of Interest: JF Gainor has served as a compensated consultant or received honoraria from Bristol-Myers Squibb, Genentech/Roche, Merck, Incyte, Array Biopharma, Novartis, Pfizer, Ariad/Takeda, Loxo and Theravance. MJ Mooradian has no conflicts of interest to declare.


References

  1. Reck M, Rodriguez-Abreu D, Robinson AG, et al. Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. N Engl J Med 2016;375:1823-33. [Crossref] [PubMed]
  2. Brahmer J, Reckamp KL, Baas P, et al. Nivolumab versus Docetaxel in Advanced Squamous-Cell Non-Small-Cell Lung Cancer. N Engl J Med 2015;373:123-35. [Crossref] [PubMed]
  3. Borghaei H, Brahmer J, Horn L, et al. P2.35: Nivolumab vs Docetaxel in Advanced NSCLC: CheckMate 017/057 2-Y Update and Exploratory Cytokine Profile Analysis: Track: Immunotherapy. J Thorac Oncol 2016;11:S237-8. [Crossref] [PubMed]
  4. Fehrenbacher L, Spira A, Ballinger M, et al. Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): a multicentre, open-label, phase 2 randomised controlled trial. Lancet 2016;387:1837-46. [Crossref] [PubMed]
  5. Govindan R, Szczesna A, Ahn MJ, et al. Phase III Trial of Ipilimumab Combined With Paclitaxel and Carboplatin in Advanced Squamous Non-Small-Cell Lung Cancer. J Clin Oncol 2017;35:3449-57. [Crossref] [PubMed]
  6. Emens LA, Middleton G. The interplay of immunotherapy and chemotherapy: harnessing potential synergies. Cancer Immunol Res 2015;3:436-43. [Crossref] [PubMed]
  7. Hato SV, Khong A, de Vries IJ, et al. Molecular pathways: the immunogenic effects of platinum-based chemotherapeutics. Clin Cancer Res 2014;20:2831-7. [Crossref] [PubMed]
  8. Lynch TJ, Bondarenko I, Luft A, et al. Ipilimumab in combination with paclitaxel and carboplatin as first-line treatment in stage IIIB/IV non-small-cell lung cancer: results from a randomized, double-blind, multicenter phase II study. J Clin Oncol 2012;30:2046-54. [Crossref] [PubMed]
  9. Reck M, Bondarenko I, Luft A, et al. Ipilimumab in combination with paclitaxel and carboplatin as first-line therapy in extensive-disease-small-cell lung cancer: results from a randomized, double-blind, multicenter phase 2 trial. Ann Oncol 2013;24:75-83. [Crossref] [PubMed]
  10. Reck M, Luft A, Szczesna A, et al. Phase III Randomized Trial of Ipilimumab Plus Etoposide and Platinum Versus Placebo Plus Etoposide and Platinum in Extensive-Stage Small-Cell Lung Cancer. J Clin Oncol 2016;34:3740-8. [Crossref] [PubMed]
  11. Hodi FS, O'Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 2010;363:711-23. [Crossref] [PubMed]
  12. Robert C, Thomas L, Bondarenko I, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med 2011;364:2517-26. [Crossref] [PubMed]
  13. Zatloukal P, Heo DS, Park K, et al. Randomized phase II clinical trial comparing tremelimumab (CP-675,206) with best supportive care (BSC) following first-line platinum-based therapy in patients (pts) with advanced non-small cell lung cancer (NSCLC). J Clin Oncol 2009;27:8071.
  14. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012;12:252-64. [Crossref] [PubMed]
  15. Fife BT, Bluestone JA. Control of peripheral T-cell tolerance and autoimmunity via the CTLA-4 and PD-1 pathways. Immunol Rev 2008;224:166-82. [Crossref] [PubMed]
  16. Buchbinder EI, Desai A. CTLA-4 and PD-1 Pathways: Similarities, Differences, and Implications of Their Inhibition. Am J Clin Oncol 2016;39:98-106. [Crossref] [PubMed]
  17. Das R, Verma R, Sznol M, et al. Combination therapy with anti-CTLA-4 and anti-PD-1 leads to distinct immunologic changes in vivo. J Immunol 2015;194:950-9. [Crossref] [PubMed]
  18. Larkin J, Chiarion-Sileni V, Gonzalez R, et al. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N Engl J Med 2015;373:23-34. [Crossref] [PubMed]
  19. Wolchok JD, Chiarion-Sileni V, Gonzalez R, et al. Overall Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N Engl J Med 2017;377:1345-56. [Crossref] [PubMed]
  20. Patnaik A, Kang SP, Rasco D, et al. Phase I Study of Pembrolizumab (MK-3475; Anti-PD-1 Monoclonal Antibody) in Patients with Advanced Solid Tumors. Clin Cancer Res 2015;21:4286-93. [Crossref] [PubMed]
  21. Antonia S, Goldberg SB, Balmanoukian A, et al. Safety and antitumour activity of durvalumab plus tremelimumab in non-small cell lung cancer: a multicentre, phase 1b study. Lancet Oncol 2016;17:299-308. [Crossref] [PubMed]
  22. Hellmann MD, Rizvi NA, Goldman JW, et al. Nivolumab plus ipilimumab as first-line treatment for advanced non-small-cell lung cancer (CheckMate 012): results of an open-label, phase 1, multicohort study. Lancet Oncol 2017;18:31-41. [Crossref] [PubMed]
  23. AstraZeneca reports initial results from the ongoing MYSTIC trial in Stage IV lung cancer. Available online: https://www.astrazeneca.com/media-centre/press-releases/2017/astrazeneca-reports-initial-results-from-the-ongoing-mystic-trial-in-stage-iv-lung-cancer-27072017.html
  24. Antonia SJ, Lopez-Martin JA, Bendell J, et al. Nivolumab alone and nivolumab plus ipilimumab in recurrent small-cell lung cancer (CheckMate 032): a multicentre, open-label, phase 1/2 trial. Lancet Oncol 2016;17:883-95. [Crossref] [PubMed]
  25. Hellmann MD, Ott PA, Zugazagoitia J, et al. Nivolumab (nivo) ± ipilimumab (ipi) in advanced small-cell lung cancer (SCLC): First report of a randomized expansion cohort from CheckMate 032. J Clin Oncol 2017;35:8503.
  26. Rizvi N, Antonia S, Callahan MK, et al. Impact of tumor mutation burden on the efficacy of nivolumab or nivolumab plus ipilimumab in small cell lung cancer: An exploratory analysis of CheckMate 032. 2017 World Conference on Lung Cancer, 2017. abstract OA 07.03a.
  27. Langer CJ, Gadgeel SM, Borghaei H, et al. Carboplatin and pemetrexed with or without pembrolizumab for advanced, non-squamous non-small-cell lung cancer: a randomised, phase 2 cohort of the open-label KEYNOTE-021 study. Lancet Oncol 2016;17:1497-508. [Crossref] [PubMed]
  28. Reck M, Socinski MA, Cappuzzo F, et al. Primary PFS and safety analyses of a randomised Phase III study of carboplatin + paclitaxel +/− bevacizumab, with or without atezolizumab in 1L non-squamous metastatic NSCLC (IMpower150). ESMO Immuno-oncology Conference, 2017.
Cite this article as: Mooradian MJ, Gainor JF. Putting the brakes on CTLA-4 inhibition in lung cancer? Transl Lung Cancer Res 2018;7(Suppl 1):S35-S38. doi: 10.21037/tlcr.2018.01.05

Download Citation