Skip to main content

Advertisement

Log in

Serum immune modulators associated with immune-related toxicities and efficacy of atezolizumab in patients with non-small cell lung cancer

  • Research
  • Published:
Journal of Cancer Research and Clinical Oncology Aims and scope Submit manuscript

Abstract

Purpose

Identifying patients at high risk of immune-related adverse events (irAEs) that impede the achievement of durable efficacy of programmed cell death 1 (PD-1)/programmed death ligand 1 (PD-L1) blockade therapy is important in improving their management. Identification of a novel predictive factor of therapeutic benefit is also important in improving patient selection for treatment with PD-1/PD-L1 inhibitors. Further determinants driving response and linking with irAEs are urgently required.

Methods

To address these unmet needs in the field, we explored whether 27 soluble checkpoint proteins and immunomodulatory proteins in serum at the therapy baseline and after week 3 were associated with irAE onset and therapeutic efficacy using MILLIPLEX Human Immuno-Oncology Checkpoint Protein Panel assays in a prospective, multicenter cohort of 81 patients with non-small cell lung cancer (NSCLC) receiving atezolizumab monotherapy.

Results

By competing-risks regression analysis, we identified that high levels of B cell-activating factor (BAFF) at baseline were a significant and strong risk factor of irAEs (hazard ratio, 5.61; 95% confidence interval, 2.43–12.96; P < 0.0001). We also identified that increased inducible T cell co-stimulator (ICOS) during the first therapeutic cycle was an independent factor associated with prolonged progression-free survival and overall survival.

Conclusion

These findings are in keeping with the reported mechanistic basis of these molecules and may provide potential guidance for clinical decision-making to improve patient care. Further validation studies are warranted.

Trial registration UMIN000035616 (January 28, 2019)

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

Data availability

The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.

Abbreviations

BAFF:

B cell-activating factor

BTLA:

B and T lymphocyte attenuator

CD40L:

CD40 ligand

CI:

Confidence interval

CTLA-4:

Cytotoxic T lymphocyte-associated protein 4

DNAM-1:

DNAX accessory molecule 1

FGL1:

Fibrinogen-like protein 1

GITR:

Glucocorticoid-induced TNFR-related protein

GITRL:

GITR ligand

HR:

Hazard ratio

HVEM:

Herpesvirus entry mediator

ICI:

Immune-checkpoint inhibitor

ICOS:

Inducible T cell co-stimulator

ICOSL:

ICOS ligand

irAE:

Immune-related adverse event

LAG-3:

Lymphocyte-activation gene 3

LIPI:

Lung immune prognostic index

NSCLC:

Non-small cell lung cancer

OS:

Overall survival

PD-1:

Programmed cell death 1

PD-L1:

Programmed death ligand 1

PD-L2:

Programmed death ligand 2

PFS:

Progression-free survival

PVR:

Poliovirus receptor cell adhesion molecule

TIM-3:

T cell immunoglobulin and mucin-domain containing-3

TLR-2:

Toll-like receptor 2

4-1BBL:

4-1BB ligand

References

  • Abu-Sbeih H, Faleck DM, Ricciuti B et al (2020) Immune checkpoint inhibitor therapy in patients with preexisting inflammatory bowel disease. J Clin Oncol 38:576–583

    CAS  PubMed  Google Scholar 

  • Akbari O, Freeman GJ, Meyer EH et al (2002) Antigen-specific regulatory T cells develop via the ICOS-ICOS-ligand pathway and inhibit allergen-induced airway hyperreactivity. Nat Med 8:1024–1032

    CAS  PubMed  Google Scholar 

  • Amatore F, Gorvel L, Olive D (2020) Role of inducible co-stimulator (ICOS) in cancer immunotherapy. Expert Opin Biol Ther 20:141–150

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  • Bar N, Costa F, Das R et al (2020) Differential effects of PD-L1 versus PD-1 blockade on myeloid inflammation in human cancer. JCI Insight 5:e129353

    PubMed  PubMed Central  Google Scholar 

  • Bomze D, Hasan Ali O, Bate A, Flatz L (2019) Association between immune-related adverse events during anti-PD-1 therapy and tumor mutational burden. JAMA Oncol 5:1633–1635

    PubMed  PubMed Central  Google Scholar 

  • Cabrita R, Lauss M, Sanna A et al (2020) Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature 577:561–565

    CAS  PubMed  Google Scholar 

  • Cascone T, William WN Jr, Weissferdt A et al (2021) Neoadjuvant nivolumab or nivolumab plus ipilimumab in operable non-small cell lung cancer: the phase 2 randomized NEOSTAR trial. Nat Med 27:504–514

    CAS  PubMed  PubMed Central  Google Scholar 

  • Chen J, Wang J, Xu H (2021) Comparison of atezolizumab, durvalumab, pembrolizumab, and nivolumab as first-line treatment in patients with extensive-stage small cell lung cancer: a systematic review and network meta-analysis. Medicine 100:e25180

    CAS  PubMed  PubMed Central  Google Scholar 

  • Das R, Bar N, Ferreira M et al (2018) Early B cell changes predict autoimmunity following combination immune checkpoint blockade. J Clin Invest 128:715–720

    PubMed  PubMed Central  Google Scholar 

  • Dubin K, Callahan MK, Ren B et al (2016) Intestinal microbiome analyses identify melanoma patients at risk for checkpoint-blockade-induced colitis. Nat Commun 7:10391

    CAS  PubMed  PubMed Central  Google Scholar 

  • Felip E, Altorki N, Zhou C et al (2021) Adjuvant atezolizumab after adjuvant chemotherapy in resected stage IB-IIIA non-small-cell lung cancer (IMpower010): a randomised, multicentre, open-label, phase 3 trial. Lancet 398:1344–1357

    CAS  PubMed  Google Scholar 

  • Giannicola R, D’Arrigo G, Botta C et al (2019) Early blood rise in auto-antibodies to nuclear and smooth muscle antigens is predictive of prolonged survival and autoimmunity in metastatic-non-small cell lung cancer patients treated with PD-1 immune-check point blockade by nivolumab. Mol Clin Oncol 11:81–90

    CAS  PubMed  PubMed Central  Google Scholar 

  • Gong B, Kiyotani K, Sakata S et al (2019) Secreted PD-L1 variants mediate resistance to PD-L1 blockade therapy in non-small cell lung cancer. J Exp Med 216:982–1000

    CAS  PubMed  PubMed Central  Google Scholar 

  • Gorelik L, Gilbride K, Dobles M et al (2003) Normal B cell homeostasis requires B cell activation factor production by radiation-resistant cells. J Exp Med 198:937–945

    CAS  PubMed  PubMed Central  Google Scholar 

  • Gorgulho J, Roderburg C, Heymann F et al (2021) Serum levels of soluble B and T lymphocyte attenuator predict overall survival in patients undergoing immune checkpoint inhibitor therapy for solid malignancies. Int J Cancer 149:1189–1198

    CAS  PubMed  Google Scholar 

  • Groom J, Kalled SL, Cutler AH et al (2002) Association of BAFF/BLyS overexpression and altered B cell differentiation with Sjogren’s syndrome. J Clin Invest 109:59–68

    CAS  PubMed  PubMed Central  Google Scholar 

  • Haslam A, Prasad V (2019) Estimation of the percentage of US patients with cancer who are eligible for and respond to checkpoint inhibitor immunotherapy drugs. JAMA Netw Open 2:e192535

    PubMed  PubMed Central  Google Scholar 

  • Havel JJ, Chowell D, Chan TA (2019) The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy. Nat Rev Cancer 19:133–150

    CAS  PubMed  PubMed Central  Google Scholar 

  • Helmink BA, Reddy SM, Gao J et al (2020) B cells and tertiary lymphoid structures promote immunotherapy response. Nature 577:549–555

    CAS  PubMed  PubMed Central  Google Scholar 

  • Hutloff A, Dittrich AM, Beier KC et al (1999) ICOS is an inducible T-cell co-stimulator structurally and functionally related to CD28. Nature 397:263–266

    CAS  PubMed  Google Scholar 

  • Kanda Y (2013) Investigation of the freely available easy-to-use software “EZR” for medical statistics. Bone Marrow Transpl 48:452–458

    CAS  Google Scholar 

  • Kreuzaler M, Rauch M, Salzer U et al (2012) Soluble BAFF levels inversely correlate with peripheral B cell numbers and the expression of BAFF receptors. J Immunol 188:497–503

    CAS  PubMed  Google Scholar 

  • Lavie F, Miceli-Richard C, Quillard J et al (2004) Expression of BAFF (BLyS) in T cells infiltrating labial salivary glands from patients with Sjogren’s syndrome. J Pathol 202:496–502

    CAS  PubMed  Google Scholar 

  • Lozano AX, Chaudhuri AA, Nene A et al (2022) T cell characteristics associated with toxicity to immune checkpoint blockade in patients with melanoma. Nat Med 28:353–362

    CAS  PubMed  PubMed Central  Google Scholar 

  • Mackay F, Browning JL (2002) BAFF: a fundamental survival factor for B cells. Nat Rev Immunol 2:465–475

    CAS  PubMed  Google Scholar 

  • Mackay F, Woodcock SA, Lawton P et al (1999) Mice transgenic for BAFF develop lymphocytic disorders along with autoimmune manifestations. J Exp Med 190:1697–1710

    CAS  PubMed  PubMed Central  Google Scholar 

  • Mezquita L, Auclin E, Ferrara R et al (2018) Association of the lung immune prognostic index with immune checkpoint inhibitor outcomes in patients with advanced non-small cell lung cancer. JAMA Oncol 4:351–357

    PubMed  PubMed Central  Google Scholar 

  • Moore PA, Belvedere O, Orr A et al (1999) BLyS: member of the tumor necrosis factor family and B lymphocyte stimulator. Science 285:260–263

    CAS  PubMed  Google Scholar 

  • Mukama T, Fortner RT, Katzke V et al (2022) Prospective evaluation of 92 serum protein biomarkers for early detection of ovarian cancer. Br J Cancer 126:1301–1309

    CAS  PubMed  PubMed Central  Google Scholar 

  • Nardelli B, Belvedere O, Roschke V et al (2001) Synthesis and release of B-lymphocyte stimulator from myeloid cells. Blood 97:198–204

    CAS  PubMed  Google Scholar 

  • Petitprez F, de Reynies A, Keung EZ et al (2020) B cells are associated with survival and immunotherapy response in sarcoma. Nature 577:556–560

    CAS  PubMed  Google Scholar 

  • Rittmeyer A, Barlesi F, Waterkamp D et al (2017) Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. Lancet 389:255–265

    PubMed  Google Scholar 

  • Sarantopoulos S, Stevenson KE, Kim HT et al (2007) High levels of B-cell activating factor in patients with active chronic graft-versus-host disease. Clin Cancer Res 13:6107–6114

    CAS  PubMed  PubMed Central  Google Scholar 

  • Schneider P, MacKay F, Steiner V et al (1999) BAFF, a novel ligand of the tumor necrosis factor family, stimulates B cell growth. J Exp Med 189:1747–1756

    CAS  PubMed  PubMed Central  Google Scholar 

  • Solinas C, Gu-Trantien C, Willard-Gallo K (2020) The rationale behind targeting the ICOS-ICOS ligand costimulatory pathway in cancer immunotherapy. ESMO Open 5:e000544

    PubMed  PubMed Central  Google Scholar 

  • Sullivan RJ, Weber JS (2021) Immune-related toxicities of checkpoint inhibitors: mechanisms and mitigation strategies. Nat Rev Drug Discov. https://doi.org/10.1038/s41573-021-00259-5

    Article  PubMed  Google Scholar 

  • Suzuki Y, Karayama M, Uto T et al (2020) Assessment of immune-related interstitial lung disease in patients with NSCLC treated with immune checkpoint inhibitors: a multicenter prospective study. J Thorac Oncol 15:1317–1327

    CAS  PubMed  Google Scholar 

  • Xiao Z, Mayer AT, Nobashi TW, Gambhir SS (2020) ICOS Is an indicator of T-cell-mediated response to cancer immunotherapy. Cancer Res 80:3023–3032

    CAS  PubMed  Google Scholar 

  • Xu S, Shukuya T, Tamura J et al (2022) Heterogeneous outcomes of immune checkpoint inhibitor rechallenge in patients with NSCLC: a systematic review and meta-analysis. JTO Clin Res Rep 3:100309

    PubMed  PubMed Central  Google Scholar 

  • Yanaba K, Asano Y, Noda S et al (2013) Increased production of soluble inducible costimulator in patients with diffuse cutaneous systemic sclerosis. Arch Dermatol Res 305:17–23

    CAS  PubMed  Google Scholar 

  • Yarchoan M, Ho WJ, Mohan A et al (2020) Effects of B cell-activating factor on tumor immunity. JCI Insight 5:e136417

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors would like to thank the patients, their families, and all the investigators who participated in this study. We also thank H. Nikki March, Ph.D., from Edanz (https://jp.edanz.com/ac) for editing a draft of this manuscript.

Funding

This work was supported by Chugai Pharmaceutical Co., Ltd. (Tokyo, Japan). The funder had no role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

Y.I.: Conceptualization, acquisition of data, data curation, formal analysis, investigation, methodology, project administration, resources, supervision, validation, visualization, writing—original draft, writing—review and editing. N.I.: Conceptualization, acquisition of data, data curation, funding acquisition, investigation, methodology, project administration, resources, supervision, validation, visualization, writing—original draft, writing—review and editing. M.K, K.A., S.M., M.I., T.U., M.F., D.H., T.M., H.M., N.I., M.T., Y.K., H.Y., H.H., Y.S., K.F., N.E., T.F., T.S: Acquisition of data, data curation, supervision, validation, writing—review and editing.

Corresponding author

Correspondence to Yusuke Inoue.

Ethics declarations

Competing interests

The authors have no relevant financial or non-financial interests to disclose.

Consent to participate

All patients provided written informed consent to participate in the study.

Ethics approval

This study was approved by the institutional review board at each site (Hamamatsu University School of Medicine, #18-164) and was conducted in accordance with the International Council for Harmonisation Good Clinical Practice guidelines and the Declaration of Helsinki. This study followed the Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) reporting guidelines. This study was registered at the UMIN Clinical Trials Registry as UMIN000035616.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Below is the link to the electronic supplementary material.

432_2022_4193_MOESM1_ESM.tif

Supplementary file1 (TIF 961 KB) Supplementary Fig. S1. Subset analyses of BAFF according to EGFR genotype and prior treatment with immune checkpoint inhibitors (ICIs). a Serum BAFF values at therapeutic baseline in patients with EGFR wild-type (WT) tumors without prior ICI treatment (EGFR-WT, N = 56), EGFR-WT tumors with prior ICI treatment (ICI-rechallenged; N = 10), and EGFR-mutant tumors (N = 15). The Kruskal–Wallis test followed by adjustment using the method of Holm was applied. b Cumulative incidence curves for the onset of any-grade immune-related adverse events (irAEs) according to EGFR mutation status and history of prior ICI treatment. c and d Cumulative incidence curves for the onset of (c) any-grade irAEs and (d) pneumonitis according to serum BAFF levels in patients with EGFR-WT tumors without prior ICI treatment. e and f Cumulative incidence curves for the onset of (e) any-grade irAEs and (f) pneumonitis according to serum BAFF levels in patients with EGFR-mutant tumors.

432_2022_4193_MOESM2_ESM.tif

Supplementary file2 (TIF 937 KB) Supplementary Fig. S2. Prognostic impact of serum ICOS variation 3 weeks after atezolizumab initiation on progression-free survival (PFS) and overall survival (OS) in adenocarcinoma patients according to EGFR genotype. a. Kaplan–Meier curves of PFS according to serum ICOS variation in patients with EGFR-WT adenocarcinoma. b Kaplan–Meier curves of OS according to serum ICOS variation in patients with EGFR-WT adenocarcinoma. c Kaplan–Meier curves of PFS according to serum ICOS variation in patients with EGFR-mutant adenocarcinoma. d Kaplan–Meier curves of OS according to serum ICOS variation in patients with EGFR-mutant adenocarcinoma.

Supplementary file3 (DOCX 16 KB)

Supplementary file4 (DOCX 18 KB)

Supplementary file5 (DOCX 19 KB)

Supplementary file6 (DOCX 19 KB)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Inoue, Y., Inui, N., Karayama, M. et al. Serum immune modulators associated with immune-related toxicities and efficacy of atezolizumab in patients with non-small cell lung cancer. J Cancer Res Clin Oncol 149, 2963–2974 (2023). https://doi.org/10.1007/s00432-022-04193-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00432-022-04193-w

Keywords

Navigation