Skip to main content
  • Letter to the Editor
  • Open access
  • Published:

The current status and future of ADC therapy for small cell Lung Cancer: a promising approach

To the editor


Antibody-drug conjugates (ADCs) are a promising targeted therapy for cancer treatment, combining the specificity of monoclonal antibodies with the cytotoxic potency of small-molecule drugs. ADCs in small cell lung cancer (SCLC) were initially used to target DLL3, a Notch inhibitory ligand [1]. The first clinical trial showed promising objective response rates (ORR). Although, subsequent ADCs targeting DLL3 did not replicate the same clinical benefits, sparked a surge of interest in the development of ADCs [2]. In order to provide a concise summary of the current research status in this field, we have analyzed data from preclinical studies and clinical trials conducted since 2020.

Findings of clinical trials since 2020

In our analysis of ongoing clinical trials, as presented in Table 1, we identified four trials that reported relevant findings for SCLC. Notably, the most encouraging outcomes were observed with HS-20,093 and ABBV-011. As of March 10, 2023, a total of 53 participants, including SCLC patients and individuals with other solid tumors, were enrolled in these trials. Among the subset of 9 patients, a remarkable response rate of 77.8% was observed, with 7 patients showing partial responses (PRs). These findings suggest that HS-20,093 demonstrated promising anti-tumor activity in SCLC patients [3]. Seizure-related homolog protein 6 (SEZ6) is another promising target that has demonstrated efficacy. It is a surface-expressed target with broad expression in SCLC cells and minimal expression in normal tissues, as confirmed by RNA-seq and immunohistochemistry (IHC) analysis. Subsequently, a first-in-human study of ABBV-011 in patients with SCLC was conducted [4]. In this trial, the confirmed objective response rate was 25%, with 10 patients showing PR. The median duration of response was 4.2 months. The median progression-free survival was 3.5 months.

Table 1 Summary of ADC trials targeting SCLC since 2020

In addition, a few trials have reported results related to SCLC, primarily with single-case PR reports. For example, in the case of BL-B01D1, an EGFRxHER3 bispecific ADC utilizing a DNA topoisomerase I inhibitor as the payload, out of 7 SCLC patients, 1 patient showed a complete partial response (cPR) [5]. In terms of combination therapy, a phase I trial was conducted utilizing a combination of Sacituzumab Govitecan and Berzosertib, an ATR inhibitor, in SCLC patients. Tumor regressions were observed in two patients, including one patient with SCLC that had transformed from EGFR-mutant non-small cell lung cancer [6].

Preclinical findings of SCLC targeted ADC

Pre-clinical studies suggested some potential yet unexplored targets, such as c-Kit, Glypican 2, JAM3, etc. Overexpression of c-Kit is observed in 70% of SCLC patients. In this particular investigation, researchers assessed the efficacy of an ADC specifically designed to target c-Kit with DM1 as payload. The results demonstrated that 4C9-DM1 effectively induced apoptosis in SCLC cells [7]. In a recent study, DNA-damaging dimers were used to specifically target Glypican 2 [8]. The dimers induced DNA damage, apoptosis, and bystander cell killing, resulting in regression of SCLC tumors. Higher expression levels of junctional adhesion molecule 3, known as JAM3, has been also validated in SCLC cell lines. JAM3 was detected at both mRNA and protein levels in all three SCLC cell lines. Although an ADC targeting JAM3 has not been developed yet, these findings suggest that JAM3 could be a promising therapeutic target for SCLC [9].

Summary

The promising results of HS-20,093 and ABBV-011 demonstrate the feasibility of ADC as a therapeutic strategy. Overall, ADCs targeting SCLC are mostly in the early stages of development, including phase I or phase I/II trials. TROP2 is the most commonly targeted molecule in SCLC (7 out of 19), followed by B7-H3 (3 out of 19). Other targets are scattered. The diversity of targets is good, with a total of 10 different targets, including one advanced bi-specific ADCs in the field. In terms of payload, microtubule inhibitors and TOP1 inhibitors are the two main categories, but TOP1 inhibitors have shown efficacy in SCLC. Further research and development of ADCs could provide new treatment options for SCLC patients and improve clinical outcomes. In the development strategy for ADCs in SCLC, several aspects can be considered:

  1. 1.

    Target selection: It is important to avoid blindly following trends and instead consider solid preclinical data as a backup for subsequent clinical. Additionally, there are several ADCs in preclinical stages or targeting potent targets that show promising clinical effects, and their potential should be explored.

  2. 2.

    Payload selection: Although, microtubule inhibitors and TOP1 inhibitors are the main choices, considering the physiological characteristics of SCLC, the development of targeted drugs addressing resistance mechanisms could be an important way to achieve differentiation.

  3. 3.

    Beyond expression: Recent studies in related tumor types (non-small cell lung cancer, NSCLC) have reported the impact HER2 amplification and mutation on receptor internalization [10]. It is worth investigating similar mechanisms exist for current targets in SCLC as well. Expression may no longer be the criterion for ADC application.

In conclusion, ADCs offer a promising and rapidly evolving therapeutic strategy for the treatment of SCLC. To maximize their potential, a comprehensive approach is needed, including careful target selection, optimization of payloads to overcome drug resistance, and exploration of mechanisms to enhance ADC efficiency. Continued research and development efforts in these areas are expected to further improve the efficacy and safety of ADCs, ultimately benefiting patients with SCLC in clinical practice.

Availability of data and materials

Not Applicable.

Abbreviations

ADCs:

Antibody-drug conjugates

SCLC:

Small cell lung cancer

ORR:

Objective response rates

PRs:

Partial responses

SEZ6:

Seizure-related homolog protein 6

IHC:

Immunohistochemistry

cPR:

Complete partial response

NSCLC:

Non-small cell lung cancer

References

  1. Pérez-Herrero E, Fernández-Medarde A. Advanced targeted therapies in cancer: drug nanocarriers, the future of chemotherapy. Eur J Pharm Biopharm. 2015;93:52–79.

    Article  PubMed  Google Scholar 

  2. Rudin CM, Pietanza MC, Bauer TM, Ready N, Morgensztern D, Glisson BS, et al. Rovalpituzumab tesirine, a DLL3-targeted antibody-drug conjugate, in recurrent small-cell lung cancer: a first-in-human, first-in-class, open-label, phase 1 study. Lancet Oncol. 2017;18(1):42–51.

    Article  CAS  PubMed  Google Scholar 

  3. Wang J, Xing L, Sun Y, Guo W, Wang H, Chen J, 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(16suppl):3017.

    Article  Google Scholar 

  4. Wiedemeyer WR, Gavrilyuk J, Schammel A, Zhao X, Sarvaiya H, Pysz M, et al. ABBV-011, a Novel, Calicheamicin-Based antibody-drug Conjugate, targets SEZ6 to Eradicate Small Cell Lung Cancer Tumors. Mol Cancer Ther. 2022;21(6):986–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Ma Y, Zhao Y, Fang W, Zhao H, Huang Y, Yang Y, et al. BL-B01D1, a first-in-class EGFRxHER3 bispecific antibody-drug conjugate (ADC), in patients with locally advanced or metastatic solid tumor: results from a first-in-human phase 1 study. J Clin Oncol. 2023;41(16suppl):3001.

    Google Scholar 

  6. Abel ML, Takahashi N, Peer C, Redon CE, Nichols S, Vilimas R, et al. Targeting replication stress and chemotherapy resistance with a combination of sacituzumab govitecan and berzosertib: a phase I clinical trial. Clin Cancer Res. 2023;29(18):3603–11.

    Article  CAS  PubMed  Google Scholar 

  7. Kim KH, Kim JO, Park JY, Seo MD, Park SG. Antibody-drug conjugate targeting c-kit for the treatment of small cell lung cancer. Int J Mol Sci. 2022;23(4):2264.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Raman S, Buongervino SN, Lane MV, Zhelev DV, Zhu Z, Cui H, et al. A GPC2 antibody-drug conjugate is efficacious against neuroblastoma and small-cell lung cancer via binding a conformational epitope. Cell Rep Med. 2021;2(7): 100344.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Yamaguchi M, Hirai S, Idogawa M, Sumi T, Uchida H, Fujitani N, et al. Junctional adhesion molecule 3 is a potential therapeutic target for small cell lung carcinoma. Exp Cell Res. 2023;426(2): 113570.

    Article  CAS  PubMed  Google Scholar 

  10. Li BT, Michelini F, Misale S, Cocco E, Baldino L, Cai Y, et al. HER2-Mediated internalization of cytotoxic agents in ERBB2 amplified or mutant lung cancers. Cancer Discov. 2020;10(5):674–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not Applicable.

Funding

This work was supported by the grant Beijing Municipal Health Commission (Beijing Demonstration Research Ward BCRW20200303); National Natural Science Foundation of China (Mechanism investigation of CAR-γδT cells therapy in the treatment of malignant pleural mesothelioma, 82272951); National Natural Science Foundation of China (Mechanism exploration of hybrid membrane vaccine for immunotherapy of malignant schwannomas, 82272953); Chinese Academy of Medical Sciences (2022-I2M-C&T-B-070).

Author information

Authors and Affiliations

Authors

Contributions

JZ, PM, and QT wrote the first draft, prepared the figures and edited the final version. SW and NL contributed to the revision. All of the authors have seen and approved the final version.

Corresponding authors

Correspondence to Shuhang Wang or Ning Li.

Ethics declarations

Ethics approval and consent to participate

Not Applicable.

Consent for publication

Not Applicable.

Competing interests

Not Applicable.

Additional information

Publisher’s Note

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

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhou, J., Ma, P., Tang, Q. et al. The current status and future of ADC therapy for small cell Lung Cancer: a promising approach. J Transl Med 21, 808 (2023). https://doi.org/10.1186/s12967-023-04471-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12967-023-04471-2