Next Article in Journal
Differential Infiltration of T-Cell Populations in Tumor and Liver Tissues Predicts Recurrence-Free Survival in Surgically Resected Hepatocellular Carcinoma
Previous Article in Journal
Reciprocal Modulation of Tumour and Immune Cell Motility: Uncovering Dynamic Interplays and Therapeutic Approaches
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

The Attractiveness of B7-H3 as a Target for Lung Cancer Treatment

by
Kostas A. Papavassiliou
1,
Amalia A. Sofianidi
2 and
Athanasios G. Papavassiliou
2,*
1
First University Department of Respiratory Medicine, ‘Sotiria’ Chest Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
2
Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
*
Author to whom correspondence should be addressed.
Cancers 2025, 17(9), 1546; https://doi.org/10.3390/cancers17091546
Submission received: 28 April 2025 / Revised: 30 April 2025 / Accepted: 30 April 2025 / Published: 1 May 2025
(This article belongs to the Section Cancer Pathophysiology)
In 2015, the U.S. Food and Drug Administration (FDA) approved nivolumab, a programmed cell death protein-1 (PD-1) immune checkpoint inhibitor (ICI), for squamous non-small-cell lung cancer (NSCLC) treatment, making it the first immunotherapy officially approved for lung cancer [1]. During the past 10 years, immunotherapy has revolutionized the therapeutic landscape of lung cancer. However, the overall response rate of current immunotherapy options in NSCLC is only 20–30% [2,3], while acquired resistance to current ICIs is ubiquitous, occurring in more than 60% of initial responders [4]. Novel immune checkpoint molecules are urgently needed in order to strengthen our armamentarium against this deadly disease.
B7 homolog 3 protein (B7-H3), also known as cluster of differentiation 276 (CD276), is a transmembrane protein, a member of the B7 family of immune checkpoint proteins, and closely related to programmed death receptor ligand 1 (PD-L1, also termed B7-H1) [5]. B7-H3 expression in healthy tissues is limited to immune cells [5]; nevertheless, B7-H3 is aberrantly expressed on tumor cells, tumor vasculature, and other tumor microenvironment (TME) components [5]. Notably, B7-H3 expression was detected in cancer-associated fibroblasts (CAFs) in renal cell carcinoma tissue [6].
Research has recently established the expression of B7-H3 across various human malignancies, including small-cell lung cancer (SCLC) and NSCLC [7]. In NSCLC, the expression of the B7-H3 protein was found in approximately 80% of the studied cases [8], while in SCLC, B7-H3 was detected in nearly 65% of the samples in one study [9] and in 75% of the cases in another study [10]. What is universally accepted is that the expression of the B7-H3 protein in lung cancer tissue is associated with higher metastatic potential [11] and poor clinical outcomes for the patients [7,8,10].
Notably, an immunohistochemical evaluation of B7-H3 expression on lung cancer biopsies derived from patients under diverse antitumor regimens showed a stable expression of B7-H3 in patients with squamous-cell carcinoma (SCC) following treatment [12]. This stable expression of B7-H3 in this particular histological subtype of lung cancer patients makes them perfect candidates for therapies targeting B7-H3. Due to the lower incidence of actionable molecular targets for SCC and the late detection stage, the average five-year survival rate for these patients is low [13]. Additionally, the aforementioned immunohistochemical analysis demonstrated a higher B7-H3 expression in wild-type epidermal growth factor receptor (EGFR) compared to EGFR-mutated adenocarcinoma patients [12]. Therefore, B7-H3 could become a valuable target in these subtypes of lung cancer patients. Interestingly, B7-H3 expression was detected in solid tumors with negligible or negative PD-L1 expression, like pulmonary invasive mucinous adenocarcinoma [14], which highlights that there is room for novel immunotherapeutic options where current ICIs fail.
To be able to take full advantage of B7-H3 protein as a target for lung cancer treatment, we must first elucidate its biological and molecular profile. Although its receptor has not yet been fully unraveled, the multifaceted roles of B7-H3 in tumorigenesis are well established [5]. T-lymphocytes express a receptor unknown to date that binds B7-H3 in antigen-presenting cells (APCs) or tumor cells. This connection supports an immunosuppressive TME, which is molded through the production of associated immunosuppressive cytokines and the polarization of M1 tumor-associated macrophages (TAMs) to an M2 phenotype [5]. On the other hand, B7-H3 possesses pro-tumorigenic capabilities such as tumor migration, invasion, metastasis, treatment resistance, and metabolic reprogramming. B7-H3 is engaged in the potentiation of signal transduction cascades such as phosphatidylinositol-3 kinases (PI3Ks), extracellular signal-regulated kinases (ERKs), and signal transducer and activator of transcription 3 (STAT3) pathways in cancer cells, which may result in augmented tumor cell proliferation and growth [5]. Research efforts are currently trying to decipher these signaling pathways in lung cancer, demonstrating that in NSCLC, B7-H3 triggers epigenetic modifications via the PI3K/AKT pathway, which in turn promotes metastasis through the process of epithelial-to-mesenchymal transition (EMT) [15].
Several B7-H3-based cancer immunotherapy strategies are currently being evaluated in clinical trials enrolling lung cancer patients. Antibody–drug conjugates (ADCs), monoclonal antibodies (mAbs), bispecific antibodies, and chimeric antigen receptor (CAR)-T cells are listed amongst strategic approaches targeting the B7-H3 protein. What is worth mentioning is that the dual checkpoint targeting of B7-H3 and other immune checkpoint molecules is under development [16]. The expression of B7-H3 tends to be mutually exclusive to PD-L1 and cytotoxic T-lymphocyte-associated protein 4 (CTLA4, also known as CD152) expressions [17]. Dual targeting approaches could result in the co-efficient collaboration of the immune system to accelerate tumor shrinkage in solid tumors [16]. Results from a phase I/II clinical trial are encouraging, with an objective response rate (ORR) of 35.7%, progression-free survival (PFS) of 4.83 months, and overall survival (OS) of 12.32 months in NSCLC patients who have not received ICIs in previous therapeutic modalities [16]. However, attention should be drawn, as the dual checkpoint targeting of B7-family proteins is associated with a higher incidence of adverse events, especially immune-related ones [16].
ADCs are shaping the future of cancer treatment. HS-20093, an ADC composed of a human immunoglobulin G1 mAb directed against B7-H3, connected to an as-yet-undefined cytotoxic agent, is currently assessed as monotherapy in a phase I study (NCT05276609) in patients with different solid malignancies, including lung tumors. This study, which recruited 29 patients with NSCLC and 11 patients with SCLC, revealed a favorable toxicity profile accompanied by encouraging anticancer potency, mainly in the SCLC group, where partial responses were attained in 7 out of 9 patients (response rate: 77.8%) [18]. In SCLC, Ifinatamab deruxtecan (I-DXd), a B7-H3-directed ADC comprising an anti-B7-H3 mAb linked to a topoisomerase I inhibitor payload, is under investigation in the phase III IDeate-Lung02 (NCT06203210) clinical trial [19]. Other innovative approaches aiming to exploit B7-H3 protein expression in NSCLC are B7-H3-targeting gold nanocage pH-sensitive conjugates that can precisely attack tumor cells [20], and CAR-engineered natural killer (NK) cells targeting B7-H3 that present enhanced cytotoxicity alongside fewer adverse events [21].
In conclusion, B7-H3 appears to be a promising target that displays elevated and more vigorous expression in lung cancer tissues in comparison to presently available checkpoint protein species. Developing strategies for the synchronous targeting of different proteins of the B7 family could help overcome resistance to current ICIs. Identifying the receptors and intracellular binding domains of the B7-H3 protein in both immune and cancer cells could accelerate this process. Artificial intelligence (AI) is expected to be a valuable ally in this effort, facilitated by AI-driven protein interaction prediction tools [22]. Nevertheless, even though therapeutic modalities targeting B7-H3 in lung cancer are eventually developed, there is an urgent need to identify which subset of patients will most likely profit from them. Shared guidelines assessing B7-H3 expression and quantitative immunostaining scoring systems in different solid tumors are of utmost importance [12]. Delving deeper into the prognostic and predictive role of B7-H3 in lung cancer will provide a patient-tailored and biomarker-guided treatment strategy.

Author Contributions

Conceptualization, K.A.P., A.A.S. and A.G.P.; writing—original draft preparation, K.A.P. and A.A.S.; literature search and preparation of all references, A.A.S.; supervision, A.G.P.; writing—review and editing, K.A.P. and A.G.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Brahmer, J.; Reckamp, K.L.; Baas, P.; Crinò, L.; Eberhardt, W.E.E.; Poddubskaya, E.; Antonia, S.; Pluzanski, A.; Vokes, E.E.; Holgado, E.; et al. Nivolumab versus Docetaxel in Advanced Squamous-Cell Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2015, 373, 123–135. [Google Scholar] [CrossRef] [PubMed]
  2. Gogishvili, M.; Melkadze, T.; Makharadze, T.; Giorgadze, D.; Dvorkin, M.; Penkov, K.; Laktionov, K.; Nemsadze, G.; Nechaeva, M.; Rozhkova, I.; et al. Cemiplimab plus Chemotherapy versus Chemotherapy Alone in Non-Small Cell Lung Cancer: A Randomized, Controlled, Double-Blind Phase 3 Trial. Nat. Med. 2022, 28, 2374–2380. [Google Scholar] [CrossRef] [PubMed]
  3. Hellmann, M.D.; Paz-Ares, L.; Bernabe Caro, R.; Zurawski, B.; Kim, S.-W.; Carcereny Costa, E.; Park, K.; Alexandru, A.; Lupinacci, L.; De La Mora Jimenez, E.; et al. Nivolumab plus Ipilimumab in Advanced Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2019, 381, 2020–2031. [Google Scholar] [CrossRef] [PubMed]
  4. Memon, D.; Schoenfeld, A.J.; Ye, D.; Fromm, G.; Rizvi, H.; Zhang, X.; Keddar, M.R.; Mathew, D.; Yoo, K.J.; Qiu, J.; et al. Clinical and Molecular Features of Acquired Resistance to Immunotherapy in Non-Small Cell Lung Cancer. Cancer Cell 2024, 42, 209–224.e9. [Google Scholar] [CrossRef]
  5. Getu, A.A.; Tigabu, A.; Zhou, M.; Lu, J.; Fodstad, Ø.; Tan, M. New Frontiers in Immune Checkpoint B7-H3 (CD276) Research and Drug Development. Mol. Cancer 2023, 22, 43. [Google Scholar] [CrossRef]
  6. Zhang, S.; Zhou, C.; Zhang, D.; Huang, Z.; Zhang, G. The Anti-Apoptotic Effect on Cancer-Associated Fibroblasts of B7-H3 Molecule Enhancing the Cell Invasion and Metastasis in Renal Cancer. OncoTargets Ther. 2019, 12, 4119–4127. [Google Scholar] [CrossRef]
  7. Miller, C.D.; Lozada, J.R.; Zorko, N.A.; Elliott, A.; Makovec, A.; Radovich, M.; Heath, E.I.; Agarwal, N.; Mckay, R.R.; Garje, R.; et al. Pan-Cancer Interrogation of B7-H3 (CD276) as an Actionable Therapeutic Target Across Human Malignancies. Cancer Res. Commun. 2024, 4, 1369–1379. [Google Scholar] [CrossRef]
  8. Altan, M.; Pelekanou, V.; Schalper, K.A.; Toki, M.; Gaule, P.; Syrigos, K.; Herbst, R.S.; Rimm, D.L. B7-H3 Expression in NSCLC and Its Association with B7-H4, PD-L1 and Tumor-Infiltrating Lymphocytes. Clin. Cancer Res. 2017, 23, 5202–5209. [Google Scholar] [CrossRef]
  9. Carvajal-Hausdorf, D.; Altan, M.; Velcheti, V.; Gettinger, S.N.; Herbst, R.S.; Rimm, D.L.; Schalper, K.A. Expression and Clinical Significance of PD-L1, B7-H3, B7-H4 and TILs in Human Small Cell Lung Cancer (SCLC). J. Immunother. Cancer 2019, 7, 65. [Google Scholar] [CrossRef]
  10. Qiu, M.; Xia, Q.; Chen, Y.; Fang, X.; Li, Q.; Zhu, L.; Jiang, X.; Xiong, Z.; Yang, S. The Expression of Three Negative Co-Stimulatory B7 Family Molecules in Small Cell Lung Cancer and Their Effect on Prognosis. Front. Oncol. 2021, 11, 600238. [Google Scholar] [CrossRef]
  11. Zhang, D.; Huang, H.; Gao, X.; Yu, G.; Zhang, X.; Jin, H.; Xu, R.; Wang, Z.; Zhang, G. High Expression of B7-H3 on Monocyte/Macrophages in Tumor Microenvironment Promotes Lung Cancer Progression by Inhibiting Apoptosis. Transl. Oncol. 2024, 41, 101874. [Google Scholar] [CrossRef] [PubMed]
  12. Omori, S.; Muramatsu, K.; Kawata, T.; Miyawaki, E.; Miyawaki, T.; Mamesaya, N.; Kawamura, T.; Kobayashi, H.; Nakashima, K.; Wakuda, K.; et al. Immunohistochemical Analysis of B7-H3 Expression in Patients with Lung Cancer Following Various Anti-Cancer Treatments. Investig. New Drugs 2023, 41, 356–364. [Google Scholar] [CrossRef]
  13. Sabbula, B.R.; Gasalberti, D.P.; Mukkamalla, S.K.R.; Anjum, F. Squamous Cell Lung Cancer. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2025; Available online: https://www.ncbi.nlm.nih.gov/books/nbk564510/ (accessed on 24 April 2025).
  14. Nakagomi, T.; Goto, T.; Hirotsu, Y.; Shikata, D.; Yokoyama, Y.; Higuchi, R.; Otake, S.; Amemiya, K.; Oyama, T.; Mochizuki, H.; et al. Genomic Characteristics of Invasive Mucinous Adenocarcinomas of the Lung and Potential Therapeutic Targets of B7-H3. Cancers 2018, 10, 478. [Google Scholar] [CrossRef]
  15. Liao, H.; Ding, M.; Zhou, N.; Yang, Y.; Chen, L. B7-H3 Promotes the Epithelial-mesenchymal Transition of NSCLC by Targeting SIRT1 through the PI3K/AKT Pathway. Mol. Med. Rep. 2022, 25, 79. [Google Scholar] [CrossRef]
  16. Aggarwal, C.; Prawira, A.; Antonia, S.; Rahma, O.; Tolcher, A.; Cohen, R.B.; Lou, Y.; Hauke, R.; Vogelzang, N.P.; Zandberg, D.; et al. Dual Checkpoint Targeting of B7-H3 and PD-1 with Enoblituzumab and Pembrolizumab in Advanced Solid Tumors: Interim Results from a Multicenter Phase I/II Trial. J. Immunother. Cancer 2022, 10, e004424. [Google Scholar] [CrossRef]
  17. Fabrizio, F.P.; Muscarella, L.A.; Rossi, A. B7-H3/CD276 and Small-Cell Lung Cancer: What’s New? Transl. Oncol. 2024, 39, 101801. [Google Scholar] [CrossRef] [PubMed]
  18. Wang, J.; Duan, J.; Xing, L.; Sun, Y.; Guo, W.; Wang, H.; Chen, J.; Han, L.; Liu, B.; Wang, Q.; et al. ARTEMIS-001: Phase 1 Study of HS-20093, a B7-H3–Targeting Antibody-Drug Conjugate, in Patients with Advanced Solid Tumor. J. Clin. Oncol. 2023, 41, 3017. [Google Scholar] [CrossRef]
  19. Owonikoko, T.K.; Byers, L.A.; Cheng, Y.; Hayashi, H.; Paz-Ares, L.G.; Perol, M.; Turner, J.; Qian, M.; Garcia, C.R.; Godard, J.; et al. IDeate-Lung02: A Phase 3, Randomized, Open-Label Study of Ifinatamab Deruxtecan (I-DXd) vs Treatment of Physician’s Choice (TPC) in Relapsed Small Cell Lung Cancer (SCLC). J. Clin. Oncol. 2024, 42, TPS8126. [Google Scholar] [CrossRef]
  20. Chen, B.; Zheng, K.; Fang, S.; Huang, K.; Chu, C.; Zhuang, J.; Lin, J.; Li, S.; Yao, H.; Liu, A.; et al. B7H3 Targeting Gold Nanocage pH-Sensitive Conjugates for Precise and Synergistic Chemo-Photothermal Therapy against NSCLC. J. Nanobiotechnol. 2023, 21, 378. [Google Scholar] [CrossRef]
  21. Yang, S.; Cao, B.; Zhou, G.; Zhu, L.; Wang, L.; Zhang, L.; Kwok, H.F.; Zhang, Z.; Zhao, Q. Targeting B7-H3 Immune Checkpoint With Chimeric Antigen Receptor-Engineered Natural Killer Cells Exhibits Potent Cytotoxicity Against Non-Small Cell Lung Cancer. Front. Pharmacol. 2020, 11, 1089. [Google Scholar] [CrossRef]
  22. Guo, Y.; Wang, X.; Zhang, C.; Chen, W.; Fu, Y.; Yu, Y.; Chen, Y.; Shao, T.; Zhang, J.; Ding, G. Tumor Immunotherapy Targeting B7-H3: From Mechanisms to Clinical Applications. ImmunoTargets Ther. 2025, 14, 291–320. [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

Papavassiliou, K.A.; Sofianidi, A.A.; Papavassiliou, A.G. The Attractiveness of B7-H3 as a Target for Lung Cancer Treatment. Cancers 2025, 17, 1546. https://doi.org/10.3390/cancers17091546

AMA Style

Papavassiliou KA, Sofianidi AA, Papavassiliou AG. The Attractiveness of B7-H3 as a Target for Lung Cancer Treatment. Cancers. 2025; 17(9):1546. https://doi.org/10.3390/cancers17091546

Chicago/Turabian Style

Papavassiliou, Kostas A., Amalia A. Sofianidi, and Athanasios G. Papavassiliou. 2025. "The Attractiveness of B7-H3 as a Target for Lung Cancer Treatment" Cancers 17, no. 9: 1546. https://doi.org/10.3390/cancers17091546

APA Style

Papavassiliou, K. A., Sofianidi, A. A., & Papavassiliou, A. G. (2025). The Attractiveness of B7-H3 as a Target for Lung Cancer Treatment. Cancers, 17(9), 1546. https://doi.org/10.3390/cancers17091546

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