Next Article in Journal
Comparison of Survival Outcomes of Single- and Five-Fraction Schedules of Stereotactic Body Radiation Therapy for Early-Stage Central or Peripheral NSCLC
Previous Article in Journal
Tumor-Stroma Ratio in Basaloid and Conventional Laryngeal Squamous Cell Carcinoma: Prognostic Significance and Concordance in Paired Biopsies and Surgical Samples
Previous Article in Special Issue
Dissecting the Immunological Profiles in NSD3-Amplified LUSC through Integrative Multi-Scale Analyses
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Immunotherapy for Solid Tumors

by
Hortense de Saint Basile
1,
Zineb Maaradji
1 and
Elizabeth Fabre
1,2,*
1
APHP, Paris Cancer Institute CARPEM, Department of Thoracic Oncology, European Hospital Georges Pompidou, 75015 Paris, France
2
INSERM Team UMR_S970, Immunotherapy and Antiangiogenic Treatment in Cancerology, Paris Descartes University, 75015 Paris, France
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(6), 1646; https://doi.org/10.3390/cancers15061646
Submission received: 27 February 2023 / Accepted: 1 March 2023 / Published: 8 March 2023
(This article belongs to the Special Issue Immunotherapy for Solid Tumors)
The development of immune checkpoint inhibitors (ICIs) constitutes a major therapeutic advance in the treatment of a number of malignancies. However, there remains a crucial need to explore the underlying mechanisms which impair the efficacy of immunotherapy.
This Special Issue includes four original articles and one review to investigate new strategies and predictors in order to rationalize the use of immunotherapy in the treatment of solid tumors.
Recently, the histone H3 lysine 36 (H3K36) methyltransferase NSD3, a neighbor gene of FGFR1, was identified as a key genetic driver of lung squamous cell carcinoma (LUSQ) [1]. By utilizing multi-scale analyses (genome, transcriptome, proteome, and TMA array), ref. [2] provides the first evidence that NSD3 gene amplification defines a non-immunogenic phenotype in LUSC and is associated with poor immunotherapy outcomes. Additionally, the authors demonstrate that high unfolded protein response (UPR) promotes the “cold” tumor immune microenvironment of NSD3-amplified LUSC. UPR signaling represents an intrinsic survival mechanism for tumor cells and has the capacity to potentially modify anti-tumor immunity [3].
By blocking immune checkpoints, ICIs disrupt immune homeostasis and can therefore potentially induce high immune reactivity and immune-related adverse events (irAEs). N. Guezour et al. [4] investigated the prognostic role of Grade 3–4 (irAE)s on overall survival in advanced NSCLC and showed that high-grade irAEs are clearly associated with better outcomes. The correlation between severe immune-related toxicity and outcomes in NSCLC brings important data for a better understanding of ICI mechanisms of actions.
Anti-programmed cell death protein-1 (PD1)/anti-programmed cell death protein-1 ligand (PDL1) have yielded disappointing results in a variety of situations, notably because of immune tolerance and low immunogenicity, as described in Ewing sarcoma (EwS) [5], an aggressive variety of bone tumor occurring in adolescents and young adults.
The development of challenging immune-based approaches, such as T-cell engagers, increases the interest in the identification of targetable tumoral surface antigenes. The article by K. Nagamura et al. [6] identifies G-protein-coupled receptor (GPCRs) GPR64 for the first time as a potential antibody-based therapeutic target for Ewing sarcoma. This research article offers perspectives for promising treatments in sarcomas through the identification of a targetable tumor antigens.
M. Kuske et al. [7] supported this Special Issue with a detailed review of the immunomodulatory effects of anti-PD1/PDL1 in immune cell types other than the T-cell compartment.
By describing the compensatory upregulation of non-targeted ICs, this article helps to understand resistance and irAEs mechanisms. Furthermore, the identification of interactions between different immune cell subsets provides the rationale for emerging ICI combinations.
Antacid agents such as proton pump inhibitors (PPIs) and histamine-2-receptor antagonists (H2RAs) are commonly prescribed owing to their potential capacity to modify the activity of anticancer therapies a variety of several mechanisms, including gut microbiome changes [8].
Rizzo et al. [9] performed a systematic literature review and meta-analysis to investigate the outcome of NSCLC patients receiving concomitantly antiacid agents and immunotherapy. The authors present pooled results of six studies into NSCLC patients treated by ICI, and report a reduced OS and PFS in the cases of PPIs or H2RAs use. The authors conclude that special consideration should be given to antiacid agents prescription in order to preserve the anti-tumor immunity through the promotion of microbiote integrity.
In conclusion, this Special Issue reports recent, highly relevant data to discuss how to predict and enhance responses to immunotherapy and thereby define new therapeutic strategies.

Author Contributions

Conceptualization, writing and editing: H.d.S.B., Z.M. and E.F. All authors have read and agreed to the published version of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Yuan, G.; Flores, N.M.; Hausmann, S.; Lofgren, S.M.; Kharchenko, V.; Angulo-Ibanez, M.; Sengupta, D.; Lu, X.; Czaban, I.; Azhibek, D.; et al. Elevated NSD3 histone methylation activity drives squamous cell lung cancer. Nature 2021, 590, 504–508. [Google Scholar] [CrossRef] [PubMed]
  2. Xu, D.; Liu, S.; Wu, X.; Marti, T.M.; Dorn, P.; Schmid, R.A.; Peng, R.-W.; Shu, Y. Dissecting the immunological profiles in NSD3-amplified LUSC through integrative multi-scale analyses. Cancers 2022, 14, 4997. [Google Scholar] [CrossRef] [PubMed]
  3. Walter, P.; Ron, D. The unfolded protein response: From stress pathway to homeostatic regulation. Science 2011, 334, 1081–1086. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Guezour, N.; Soussi, G.; Brosseau, S.; Abbar, B.; Naltet, C.; Vauchier, C.; Poté, N.; Hachon, L.; Namour, C.; Khalil, A.; et al. Grade 3–4 immune-related adverse events induced by immune checkpoint inhibitors in non-small-cell lung cancer (NSCLC) patients are correlated with better outcome: A real-life observational study. Cancers 2022, 14, 3878. [Google Scholar] [CrossRef] [PubMed]
  5. Evdokimova, V.; Gassmann, H.; Radvanyi, L.; Burdach, S.E.G. Current state of immunotherapy and mechanisms of immune evasion in ewing sarcoma and osteosarcoma. Cancers 2022, 15, 272. [Google Scholar] [CrossRef] [PubMed]
  6. Nakamura, K.; Asanuma, K.; Okamoto, T.; Yoshida, K.; Matsuyama, Y.; Kita, K.; Hagi, T.; Nakamura, T.; Sudo, A. GPR64, Screened from ewing sarcoma cells, is a potential target for antibody-based therapy for various sarcomas. Cancers 2022, 14, 814. [Google Scholar] [CrossRef] [PubMed]
  7. Kuske, M.; Haist, M.; Jung, T.; Grabbe, S.; Bros, M. Immunomodulatory properties of immune checkpoint inhibitors—More than boosting t-cell responses? Cancers 2022, 14, 1710. [Google Scholar] [CrossRef] [PubMed]
  8. Weersma, R.K.; Zhernakova, A.; Fu, J. Interaction between drugs and the gut microbiome. Gut 2020, 69, 1510–1519. [Google Scholar] [CrossRef] [PubMed]
  9. Rizzo, A.; Cusmai, A.; Giovannelli, F.; Acquafredda, S.; Rinaldi, L.; Misino, A.; Montagna, E.S.; Ungaro, V.; Lorusso, M.; Palmiotti, G. Impact of proton pump inhibitors and histamine-2-receptor antagonists on non-small cell lung cancer immunotherapy: A systematic review and meta-analysis. Cancers 2022, 14, 1404. [Google Scholar] [CrossRef] [PubMed]
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

de Saint Basile, H.; Maaradji, Z.; Fabre, E. Immunotherapy for Solid Tumors. Cancers 2023, 15, 1646. https://doi.org/10.3390/cancers15061646

AMA Style

de Saint Basile H, Maaradji Z, Fabre E. Immunotherapy for Solid Tumors. Cancers. 2023; 15(6):1646. https://doi.org/10.3390/cancers15061646

Chicago/Turabian Style

de Saint Basile, Hortense, Zineb Maaradji, and Elizabeth Fabre. 2023. "Immunotherapy for Solid Tumors" Cancers 15, no. 6: 1646. https://doi.org/10.3390/cancers15061646

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