Skip to main content
Top
Published in: BMC Cancer 1/2024

Open Access 01-12-2024 | NSCLC | Research

Down-regulated HHLA2 enhances neoadjuvant immunotherapy efficacy in patients with non-small cell lung cancer (NSCLC) with chronic obstructive pulmonary disease (COPD)

Authors: Ao Zeng, Yanze Yin, Zhilong Xu, Abudumijiti Abuduwayiti, Fujun Yang, Mohammed Saud Shaik, Chao Wang, Keyi Chen, Chao Wang, Xinyun Fang, Jie Dai

Published in: BMC Cancer | Issue 1/2024

Login to get access

Abstract

Background

Emerging data suggested a favorable outcome in advanced non-small cell lung cancer (NSCLC) with chronic obstructive pulmonary disease (COPD) patients treated by immunotherapy. The objective of this study was to investigate the effectiveness of neoadjuvant immunotherapy among NSCLC with COPD versus NSCLC without COPD and explore the potential mechanistic links.

Patients and methods

Patients with NSCLC receiving neoadjuvant immunotherapy and surgery at Shanghai Pulmonary Hospital between November 2020 and January 2023 were reviewed. The assessment of neoadjuvant immunotherapy’s effectiveness was conducted based on the major pathologic response (MPR). The gene expression profile was investigated by RNA sequencing data. Immune cell proportions were examined using flow cytometry. The association between gene expression, immune cells, and pathologic response was validated by immunohistochemistry and single-cell data.

Results

A total of 230 NSCLC patients who received neoadjuvant immunotherapy were analyzed, including 60 (26.1%) with COPD. Multivariate logistic regression demonstrated that COPD was a predictor for MPR after neoadjuvant immunotherapy [odds ratio (OR), 2.490; 95% confidence interval (CI), 1.295–4.912; P = 0.007]. NSCLC with COPD showed a down-regulation of HERV–H LTR-associating protein 2 (HHLA2), which was an immune checkpoint molecule, and the HHLA2low group demonstrated the enrichment of CD8+CD103+ tissue-resident memory T cells (TRM) compared to the HHLA2high group (11.9% vs. 4.2%, P = 0.013). Single-cell analysis revealed TRM enrichment in the MPR group. Similarly, NSCLC with COPD exhibited a higher proportion of CD8+CD103+TRM compared to NSCLC without COPD (11.9% vs. 4.6%, P = 0.040).

Conclusions

The study identified NSCLC with COPD as a favorable lung cancer type for neoadjuvant immunotherapy, offering a new perspective on the multimodality treatment of this patient population. Down-regulated HHLA2 in NSCLC with COPD might improve the MPR rate to neoadjuvant immunotherapy owing to the enrichment of CD8+CD103+TRM.

Trial registration

Approval for the collection and utilization of clinical samples was granted by the Ethics Committee of Shanghai Pulmonary Hospital (Approval number: K23-228).
Appendix
Available only for authorised users
Literature
1.
go back to reference Yi YS, Ban WH, Sohng KY. Effect of COPD on symptoms, quality of life and prognosis in patients with advanced non-small cell lung cancer. BMC Cancer. 2018;18(1):1053.CrossRefPubMedPubMedCentral Yi YS, Ban WH, Sohng KY. Effect of COPD on symptoms, quality of life and prognosis in patients with advanced non-small cell lung cancer. BMC Cancer. 2018;18(1):1053.CrossRefPubMedPubMedCentral
2.
go back to reference Zhai R, Yu X, Shafer A, Wain JC, Christiani DC. The impact of coexisting COPD on survival of patients with early-stage non-small cell lung cancer undergoing surgical resection. Chest. 2014;145(2):346–53.CrossRefPubMed Zhai R, Yu X, Shafer A, Wain JC, Christiani DC. The impact of coexisting COPD on survival of patients with early-stage non-small cell lung cancer undergoing surgical resection. Chest. 2014;145(2):346–53.CrossRefPubMed
3.
go back to reference Haslam A, Prasad V. Estimation of the percentage of US patients with Cancer who are eligible for and respond to checkpoint inhibitor immunotherapy drugs. JAMA Netw Open. 2019;2(5):e192535.CrossRefPubMedPubMedCentral Haslam A, Prasad V. Estimation of the percentage of US patients with Cancer who are eligible for and respond to checkpoint inhibitor immunotherapy drugs. JAMA Netw Open. 2019;2(5):e192535.CrossRefPubMedPubMedCentral
4.
go back to reference Sholl LM, Hirsch FR, Hwang D, Botling J, Lopez-Rios F, Bubendorf L, et al. The promises and challenges of Tumor Mutation Burden as an Immunotherapy Biomarker: a perspective from the International Association for the Study of Lung Cancer Pathology Committee. J Thorac Oncol. 2020;15(9):1409–24.CrossRefPubMedPubMedCentral Sholl LM, Hirsch FR, Hwang D, Botling J, Lopez-Rios F, Bubendorf L, et al. The promises and challenges of Tumor Mutation Burden as an Immunotherapy Biomarker: a perspective from the International Association for the Study of Lung Cancer Pathology Committee. J Thorac Oncol. 2020;15(9):1409–24.CrossRefPubMedPubMedCentral
5.
go back to reference Zhou J, Chao Y, Yao D, Ding N, Li J, Gao L, et al. Impact of chronic obstructive pulmonary disease on immune checkpoint inhibitor efficacy in advanced lung cancer and the potential prognostic factors. Transl Lung Cancer Res. 2021;10(5):2148–62.CrossRefPubMedPubMedCentral Zhou J, Chao Y, Yao D, Ding N, Li J, Gao L, et al. Impact of chronic obstructive pulmonary disease on immune checkpoint inhibitor efficacy in advanced lung cancer and the potential prognostic factors. Transl Lung Cancer Res. 2021;10(5):2148–62.CrossRefPubMedPubMedCentral
6.
go back to reference Noda Y, Shiroyama T, Masuhiro K, Amiya S, Enomoto T, Adachi Y, et al. Quantitative evaluation of emphysema for predicting immunotherapy response in patients with advanced non-small-cell lung cancer. Sci Rep. 2022;12(1):8881.CrossRefPubMedPubMedCentral Noda Y, Shiroyama T, Masuhiro K, Amiya S, Enomoto T, Adachi Y, et al. Quantitative evaluation of emphysema for predicting immunotherapy response in patients with advanced non-small-cell lung cancer. Sci Rep. 2022;12(1):8881.CrossRefPubMedPubMedCentral
7.
go back to reference Takayama Y, Nakamura T, Fukushiro Y, Mishima S, Masuda K, Shoda H. Coexistence of Emphysema with non-small-cell Lung Cancer predicts the therapeutic efficacy of Immune Checkpoint inhibitors. Vivo. 2021;35(1):467–74.CrossRef Takayama Y, Nakamura T, Fukushiro Y, Mishima S, Masuda K, Shoda H. Coexistence of Emphysema with non-small-cell Lung Cancer predicts the therapeutic efficacy of Immune Checkpoint inhibitors. Vivo. 2021;35(1):467–74.CrossRef
8.
go back to reference Kerdidani D, Magkouta S, Chouvardas P, Karavana V, Glynos K, Roumelioti F, et al. Cigarette smoke-Induced Emphysema exhausts early cytotoxic CD8(+) T cell responses against nascent Lung Cancer cells. J Immunol. 2018;201(5):1558–69.CrossRefPubMed Kerdidani D, Magkouta S, Chouvardas P, Karavana V, Glynos K, Roumelioti F, et al. Cigarette smoke-Induced Emphysema exhausts early cytotoxic CD8(+) T cell responses against nascent Lung Cancer cells. J Immunol. 2018;201(5):1558–69.CrossRefPubMed
9.
go back to reference Zhao R, Chinai JM, Buhl S, Scandiuzzi L, Ray A, Jeon H, et al. HHLA2 is a member of the B7 family and inhibits human CD4 and CD8 T-cell function. Proc Natl Acad Sci U S A. 2013;110(24):9879–84.CrossRefPubMedPubMedCentral Zhao R, Chinai JM, Buhl S, Scandiuzzi L, Ray A, Jeon H, et al. HHLA2 is a member of the B7 family and inhibits human CD4 and CD8 T-cell function. Proc Natl Acad Sci U S A. 2013;110(24):9879–84.CrossRefPubMedPubMedCentral
10.
go back to reference Janakiram M, Chinai JM, Fineberg S, Fiser A, Montagna C, Medavarapu R, et al. Expression, clinical significance, and receptor identification of the newest B7 family Member HHLA2 protein. Clin Cancer Res. 2015;21(10):2359–66.CrossRefPubMed Janakiram M, Chinai JM, Fineberg S, Fiser A, Montagna C, Medavarapu R, et al. Expression, clinical significance, and receptor identification of the newest B7 family Member HHLA2 protein. Clin Cancer Res. 2015;21(10):2359–66.CrossRefPubMed
11.
go back to reference Li Y, Lv C, Yu Y, Wu B, Zhang Y, Lang Q et al. KIR3DL3-HHLA2 and TMIGD2-HHLA2 pathways: the dual role of HHLA2 in immune responses and its potential therapeutic approach for cancer immunotherapy. J Adv Res. 2022. Li Y, Lv C, Yu Y, Wu B, Zhang Y, Lang Q et al. KIR3DL3-HHLA2 and TMIGD2-HHLA2 pathways: the dual role of HHLA2 in immune responses and its potential therapeutic approach for cancer immunotherapy. J Adv Res. 2022.
12.
go back to reference Wei Y, Ren X, Galbo PM Jr., Moerdler S, Wang H, Sica RA et al. KIR3DL3-HHLA2 is a human immunosuppressive pathway and a therapeutic target. Sci Immunol. 2021;6(61). Wei Y, Ren X, Galbo PM Jr., Moerdler S, Wang H, Sica RA et al. KIR3DL3-HHLA2 is a human immunosuppressive pathway and a therapeutic target. Sci Immunol. 2021;6(61).
13.
go back to reference Rieder SA, Wang J, White N, Qadri A, Menard C, Stephens G, et al. B7-H7 (HHLA2) inhibits T-cell activation and proliferation in the presence of TCR and CD28 signaling. Cell Mol Immunol. 2021;18(6):1503–11.CrossRefPubMed Rieder SA, Wang J, White N, Qadri A, Menard C, Stephens G, et al. B7-H7 (HHLA2) inhibits T-cell activation and proliferation in the presence of TCR and CD28 signaling. Cell Mol Immunol. 2021;18(6):1503–11.CrossRefPubMed
14.
go back to reference Cheng H, Borczuk A, Janakiram M, Ren X, Lin J, Assal A, et al. Wide expression and significance of Alternative Immune Checkpoint molecules, B7x and HHLA2, in PD-L1-Negative human lung cancers. Clin Cancer Res. 2018;24(8):1954–64.CrossRefPubMedPubMedCentral Cheng H, Borczuk A, Janakiram M, Ren X, Lin J, Assal A, et al. Wide expression and significance of Alternative Immune Checkpoint molecules, B7x and HHLA2, in PD-L1-Negative human lung cancers. Clin Cancer Res. 2018;24(8):1954–64.CrossRefPubMedPubMedCentral
15.
go back to reference Okla K, Farber DL, Zou W. Tissue-resident memory T cells in tumor immunity and immunotherapy. J Exp Med. 2021;218(4). Okla K, Farber DL, Zou W. Tissue-resident memory T cells in tumor immunity and immunotherapy. J Exp Med. 2021;218(4).
16.
go back to reference Banchereau R, Chitre AS, Scherl A, Wu TD, Patil NS, de Almeida P et al. Intratumoral CD103 + CD8 + T cells predict response to PD-L1 blockade. J Immunother Cancer. 2021;9(4). Banchereau R, Chitre AS, Scherl A, Wu TD, Patil NS, de Almeida P et al. Intratumoral CD103 + CD8 + T cells predict response to PD-L1 blockade. J Immunother Cancer. 2021;9(4).
17.
go back to reference Corgnac S, Malenica I, Mezquita L, Auclin E, Voilin E, Kacher J, et al. CD103(+)CD8(+) T(RM) cells accumulate in tumors of Anti-PD-1-Responder Lung Cancer patients and are tumor-reactive lymphocytes enriched with Tc17. Cell Rep Med. 2020;1(7):100127.CrossRefPubMedPubMedCentral Corgnac S, Malenica I, Mezquita L, Auclin E, Voilin E, Kacher J, et al. CD103(+)CD8(+) T(RM) cells accumulate in tumors of Anti-PD-1-Responder Lung Cancer patients and are tumor-reactive lymphocytes enriched with Tc17. Cell Rep Med. 2020;1(7):100127.CrossRefPubMedPubMedCentral
18.
go back to reference Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, et al. Robust enumeration of cell subsets from tissue expression profiles. Nat Methods. 2015;12(5):453–7.CrossRefPubMedPubMedCentral Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, et al. Robust enumeration of cell subsets from tissue expression profiles. Nat Methods. 2015;12(5):453–7.CrossRefPubMedPubMedCentral
19.
go back to reference Caushi JX, Zhang J, Ji Z, Vaghasia A, Zhang B, Hsiue EH, et al. Transcriptional programs of neoantigen-specific TIL in anti-PD-1-treated lung cancers. Nature. 2021;596(7870):126–32.CrossRefPubMedPubMedCentral Caushi JX, Zhang J, Ji Z, Vaghasia A, Zhang B, Hsiue EH, et al. Transcriptional programs of neoantigen-specific TIL in anti-PD-1-treated lung cancers. Nature. 2021;596(7870):126–32.CrossRefPubMedPubMedCentral
20.
go back to reference Chu Y, Dai E, Li Y, Han G, Pei G, Ingram DR, et al. Pan-cancer T cell atlas links a cellular stress response state to immunotherapy resistance. Nat Med. 2023;29(6):1550–62.CrossRefPubMed Chu Y, Dai E, Li Y, Han G, Pei G, Ingram DR, et al. Pan-cancer T cell atlas links a cellular stress response state to immunotherapy resistance. Nat Med. 2023;29(6):1550–62.CrossRefPubMed
21.
go back to reference Ma H, Zhang Q, Zhao Y, Zhang Y, Zhang J, Chen G, et al. Molecular and clinicopathological characteristics of Lung Cancer Concomitant Chronic Obstructive Pulmonary Disease (COPD). Int J Chron Obstruct Pulmon Dis. 2022;17:1601–12.CrossRefPubMedPubMedCentral Ma H, Zhang Q, Zhao Y, Zhang Y, Zhang J, Chen G, et al. Molecular and clinicopathological characteristics of Lung Cancer Concomitant Chronic Obstructive Pulmonary Disease (COPD). Int J Chron Obstruct Pulmon Dis. 2022;17:1601–12.CrossRefPubMedPubMedCentral
22.
go back to reference Zhang Q, Feng X, Hu W, Li C, Sun D, Peng Z, et al. Chronic obstructive pulmonary disease alters the genetic landscape and tumor immune microenvironment in lung cancer patients. Front Oncol. 2023;13:1169874.CrossRefPubMedPubMedCentral Zhang Q, Feng X, Hu W, Li C, Sun D, Peng Z, et al. Chronic obstructive pulmonary disease alters the genetic landscape and tumor immune microenvironment in lung cancer patients. Front Oncol. 2023;13:1169874.CrossRefPubMedPubMedCentral
23.
go back to reference Shin SH, Park HY, Im Y, Jung HA, Sun JM, Ahn JS, et al. Improved treatment outcome of pembrolizumab in patients with nonsmall cell lung cancer and chronic obstructive pulmonary disease. Int J Cancer. 2019;145(9):2433–9.CrossRefPubMed Shin SH, Park HY, Im Y, Jung HA, Sun JM, Ahn JS, et al. Improved treatment outcome of pembrolizumab in patients with nonsmall cell lung cancer and chronic obstructive pulmonary disease. Int J Cancer. 2019;145(9):2433–9.CrossRefPubMed
24.
go back to reference Xu L, Li F, Jiang M, Li Z, Xu D, Jing J, et al. Immunosuppression by Inflammation-Stimulated Amplification of Myeloid-Derived Suppressor Cells and changes in expression of Immune Checkpoint HHLA2 in Chronic Obstructive Pulmonary Disease. Int J Chron Obstruct Pulmon Dis. 2023;18:139–53.CrossRefPubMedPubMedCentral Xu L, Li F, Jiang M, Li Z, Xu D, Jing J, et al. Immunosuppression by Inflammation-Stimulated Amplification of Myeloid-Derived Suppressor Cells and changes in expression of Immune Checkpoint HHLA2 in Chronic Obstructive Pulmonary Disease. Int J Chron Obstruct Pulmon Dis. 2023;18:139–53.CrossRefPubMedPubMedCentral
25.
go back to reference Su Q, Du J, Xiong X, Xie X, Wang L. B7-H7: a potential target for cancer immunotherapy. Int Immunopharmacol. 2023;121:110403.CrossRefPubMed Su Q, Du J, Xiong X, Xie X, Wang L. B7-H7: a potential target for cancer immunotherapy. Int Immunopharmacol. 2023;121:110403.CrossRefPubMed
26.
go back to reference Zhou QH, Li KW, Chen X, He HX, Peng SM, Peng SR et al. HHLA2 and PD-L1 co-expression predicts poor prognosis in patients with clear cell renal cell carcinoma. J Immunother Cancer. 2020;8(1). Zhou QH, Li KW, Chen X, He HX, Peng SM, Peng SR et al. HHLA2 and PD-L1 co-expression predicts poor prognosis in patients with clear cell renal cell carcinoma. J Immunother Cancer. 2020;8(1).
27.
go back to reference Xia C, Huang W, Chen YL, Fu HB, Tang M, Zhang TL, et al. Coexpression of HHLA2 and PD-L1 on Tumor cells independently predicts the survival of spinal Chordoma patients. Front Immunol. 2021;12:797407.CrossRefPubMed Xia C, Huang W, Chen YL, Fu HB, Tang M, Zhang TL, et al. Coexpression of HHLA2 and PD-L1 on Tumor cells independently predicts the survival of spinal Chordoma patients. Front Immunol. 2021;12:797407.CrossRefPubMed
28.
go back to reference Gaud G, Lesourne R, Love PE. Regulatory mechanisms in T cell receptor signalling. Nat Rev Immunol. 2018;18(8):485–97.CrossRefPubMed Gaud G, Lesourne R, Love PE. Regulatory mechanisms in T cell receptor signalling. Nat Rev Immunol. 2018;18(8):485–97.CrossRefPubMed
29.
go back to reference Palmer WH, Leaton LA, Campos Codo A, Crute B, Roest J, Zhu S, et al. Polymorphic KIR3DL3 expression modulates tissue-resident and innate-like T cells. Sci Immunol. 2023;8(84):eade5343.CrossRefPubMed Palmer WH, Leaton LA, Campos Codo A, Crute B, Roest J, Zhu S, et al. Polymorphic KIR3DL3 expression modulates tissue-resident and innate-like T cells. Sci Immunol. 2023;8(84):eade5343.CrossRefPubMed
30.
go back to reference Luoma AM, Suo S, Wang Y, Gunasti L, Porter CBM, Nabilsi N, et al. Tissue-resident memory and circulating T cells are early responders to pre-surgical cancer immunotherapy. Cell. 2022;185(16):2918–e3529.CrossRefPubMedPubMedCentral Luoma AM, Suo S, Wang Y, Gunasti L, Porter CBM, Nabilsi N, et al. Tissue-resident memory and circulating T cells are early responders to pre-surgical cancer immunotherapy. Cell. 2022;185(16):2918–e3529.CrossRefPubMedPubMedCentral
31.
go back to reference Tissue-Resident Memory T. Cells underlie Neoadjuvant Immunotherapy Response. Cancer Discov. 2022;12(9):OF2.CrossRef Tissue-Resident Memory T. Cells underlie Neoadjuvant Immunotherapy Response. Cancer Discov. 2022;12(9):OF2.CrossRef
32.
go back to reference Virassamy B, Caramia F, Savas P, Sant S, Wang J, Christo SN, et al. Intratumoral CD8(+) T cells with a tissue-resident memory phenotype mediate local immunity and immune checkpoint responses in breast cancer. Cancer Cell. 2023;41(3):585–601. e8.CrossRefPubMed Virassamy B, Caramia F, Savas P, Sant S, Wang J, Christo SN, et al. Intratumoral CD8(+) T cells with a tissue-resident memory phenotype mediate local immunity and immune checkpoint responses in breast cancer. Cancer Cell. 2023;41(3):585–601. e8.CrossRefPubMed
33.
go back to reference Liang M, Wang X, Cai D, Guan W, Shen X. Tissue-resident memory T cells in gastrointestinal tumors: turning immune desert into immune oasis. Front Immunol. 2023;14:1119383.CrossRefPubMedPubMedCentral Liang M, Wang X, Cai D, Guan W, Shen X. Tissue-resident memory T cells in gastrointestinal tumors: turning immune desert into immune oasis. Front Immunol. 2023;14:1119383.CrossRefPubMedPubMedCentral
34.
go back to reference Edwards J, Wilmott JS, Madore J, Gide TN, Quek C, Tasker A, et al. CD103(+) Tumor-Resident CD8(+) T Cells Are Associated with Improved Survival in Immunotherapy-Naive Melanoma patients and Expand significantly during Anti-PD-1 treatment. Clin Cancer Res. 2018;24(13):3036–45.CrossRefPubMed Edwards J, Wilmott JS, Madore J, Gide TN, Quek C, Tasker A, et al. CD103(+) Tumor-Resident CD8(+) T Cells Are Associated with Improved Survival in Immunotherapy-Naive Melanoma patients and Expand significantly during Anti-PD-1 treatment. Clin Cancer Res. 2018;24(13):3036–45.CrossRefPubMed
35.
go back to reference Abdeljaoued S, Arfa S, Kroemer M, Ben Khelil M, Vienot A, Heyd B et al. Tissue-resident memory T cells in gastrointestinal cancer immunology and immunotherapy: ready for prime time? J Immunother Cancer. 2022;10(4). Abdeljaoued S, Arfa S, Kroemer M, Ben Khelil M, Vienot A, Heyd B et al. Tissue-resident memory T cells in gastrointestinal cancer immunology and immunotherapy: ready for prime time? J Immunother Cancer. 2022;10(4).
36.
go back to reference Biton J, Ouakrim H, Dechartres A, Alifano M, Mansuet-Lupo A, Si H, et al. Impaired tumor-infiltrating T cells in patients with chronic obstructive Pulmonary Disease Impact Lung Cancer response to PD-1 blockade. Am J Respir Crit Care Med. 2018;198(7):928–40.CrossRefPubMed Biton J, Ouakrim H, Dechartres A, Alifano M, Mansuet-Lupo A, Si H, et al. Impaired tumor-infiltrating T cells in patients with chronic obstructive Pulmonary Disease Impact Lung Cancer response to PD-1 blockade. Am J Respir Crit Care Med. 2018;198(7):928–40.CrossRefPubMed
37.
go back to reference Mark NM, Kargl J, Busch SE, Yang GHY, Metz HE, Zhang H, et al. Chronic obstructive Pulmonary Disease alters Immune Cell Composition and Immune checkpoint inhibitor efficacy in Non-small Cell Lung Cancer. Am J Respir Crit Care Med. 2018;197(3):325–36.CrossRefPubMedPubMedCentral Mark NM, Kargl J, Busch SE, Yang GHY, Metz HE, Zhang H, et al. Chronic obstructive Pulmonary Disease alters Immune Cell Composition and Immune checkpoint inhibitor efficacy in Non-small Cell Lung Cancer. Am J Respir Crit Care Med. 2018;197(3):325–36.CrossRefPubMedPubMedCentral
38.
go back to reference Corleis B, Cho JL, Gates SJ, Linder AH, Dickey A, Lisanti-Park AC, et al. Smoking and human immunodeficiency virus 1 infection promote Retention of CD8(+) T cells in the Airway Mucosa. Am J Respir Cell Mol Biol. 2021;65(5):513–20.CrossRefPubMedPubMedCentral Corleis B, Cho JL, Gates SJ, Linder AH, Dickey A, Lisanti-Park AC, et al. Smoking and human immunodeficiency virus 1 infection promote Retention of CD8(+) T cells in the Airway Mucosa. Am J Respir Cell Mol Biol. 2021;65(5):513–20.CrossRefPubMedPubMedCentral
Metadata
Title
Down-regulated HHLA2 enhances neoadjuvant immunotherapy efficacy in patients with non-small cell lung cancer (NSCLC) with chronic obstructive pulmonary disease (COPD)
Authors
Ao Zeng
Yanze Yin
Zhilong Xu
Abudumijiti Abuduwayiti
Fujun Yang
Mohammed Saud Shaik
Chao Wang
Keyi Chen
Chao Wang
Xinyun Fang
Jie Dai
Publication date
01-12-2024
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2024
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-024-12137-5

Other articles of this Issue 1/2024

BMC Cancer 1/2024 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine