Reduced intestinal-to-diffuse conversion and immunosuppressive responses underlie superiority of neoadjuvant immunochemotherapy in gastric adenocarcinoma

Lei Wang , Linghong Wan , Xu Chen , Peng Gao , Yongying Hou , Linyu Wu , Wenkang Liu , Shuoran Tian , Mengyi Han , Shiyin Peng , Yuting Tan , Yuwei Pan , Yuanfeng Ren , Jinyang Li , Haihui Wen , Qin Liu , Mengsi Zhang , Tao Wang , Zhong-Yi Qin , Junyu Xiang , Dongfeng Chen , Xianfeng Li , Shu-Nan Wang , Chuan Chen , Mengxia Li , Fan Li , Zhenning Wang , Bin Wang

MedComm ›› 2024, Vol. 5 ›› Issue (11) : e762

PDF
MedComm ›› 2024, Vol. 5 ›› Issue (11) : e762 DOI: 10.1002/mco2.762
ORIGINAL ARTICLE

Reduced intestinal-to-diffuse conversion and immunosuppressive responses underlie superiority of neoadjuvant immunochemotherapy in gastric adenocarcinoma

Author information +
History +
PDF

Abstract

Neoadjuvant immunochemotherapy (NAIC) achieves superior clinical benefits over neoadjuvant chemotherapy (NAC) in multiple types of human cancers, including gastric adenocarcinoma (GAC). However, it is poorly understood how the malignant epithelial cells and tumor immune microenvironment (TIME) might respond distinctly to NAIC and NAC that underlies therapeutic efficacy. Here treatment-naive and paired tumor tissues from multiple centers were subjected to pathological, immunological, and transcriptomic analysis. NAIC demonstrated significantly increased rate of pathological complete response compared to NAC (pCR: 25% vs. 4%, p < 0.05). Interestingly, pretreatment intestinal subtype of Lauren’s classification was predictive of pathologic regression following NAIC, but not NAC. A substantial portion of cancers underwent intestinal-to-diffuse transition, which occurred less following NAIC and correlated with treatment failure. Moreover, NAIC prevented reprogramming to an immunosuppressive TIME with less active fibroblasts and exhausted CD8+ T cells, and increased numbers of mature tertiary lymphoid structures. Mechanistically, activation of the tumor necrosis factor alpha (TNFα)/nuclear factor-kappa B (NF-κB) signaling pathway was associated with response to NAIC. Together, NAIC is superior to NAC for locally advanced GAC, likely due to reduced intestinal-to-diffuse conversion and reprogramming to an immuno-active TIME. Modulation of the histological conversion and immunosuppressive TIME could be translatable approaches to improve neoadjuvant therapeutic efficacy.

Keywords

gastric adenocarcinoma / immunochemotherapy / Lauren’s classification / neoadjuvant therapy / tumor immune microenvironment

Cite this article

Download citation ▾
Lei Wang, Linghong Wan, Xu Chen, Peng Gao, Yongying Hou, Linyu Wu, Wenkang Liu, Shuoran Tian, Mengyi Han, Shiyin Peng, Yuting Tan, Yuwei Pan, Yuanfeng Ren, Jinyang Li, Haihui Wen, Qin Liu, Mengsi Zhang, Tao Wang, Zhong-Yi Qin, Junyu Xiang, Dongfeng Chen, Xianfeng Li, Shu-Nan Wang, Chuan Chen, Mengxia Li, Fan Li, Zhenning Wang, Bin Wang. Reduced intestinal-to-diffuse conversion and immunosuppressive responses underlie superiority of neoadjuvant immunochemotherapy in gastric adenocarcinoma. MedComm, 2024, 5(11): e762 DOI:10.1002/mco2.762

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J Clin. 2021; 71(1): 7-33.

[2]

Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021; 71(3): 209-249.

[3]

Sano T, Coit DG, Kim HH, et al. Proposal of a new stage grouping of gastric cancer for TNM classification: International Gastric Cancer Association staging project. Gastric Cancer. 2017; 20(2): 217-225.

[4]

Cunningham D, Allum WH, Stenning SP, et al. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med. 2006; 355(1): 11-20.

[5]

Al-Batran SE, Homann N, Pauligk C, et al. Perioperative chemotherapy with fluorouracil plus leucovorin, oxaliplatin, and docetaxel versus fluorouracil or capecitabine plus cisplatin and epirubicin for locally advanced, resectable gastric or gastro-oesophageal junction adenocarcinoma (FLOT4): a randomised, phase 2/3 trial. Lancet. 2019; 393(10184): 1948-1957.

[6]

Zhang X, Liang H, Li Z, et al. Perioperative or postoperative adjuvant oxaliplatin with S-1 versus adjuvant oxaliplatin with capecitabine in patients with locally advanced gastric or gastro-oesophageal junction adenocarcinoma undergoing D2 gastrectomy (RESOLVE): an open-label, superiority and non-inferiority, phase 3 randomised controlled trial. Lancet Oncol. 2021; 22(8): 1081-1092.

[7]

Wang FH, Zhang XT, Tang L, et al. The Chinese Society of Clinical Oncology (CSCO): clinical guidelines for the diagnosis and treatment of gastric cancer, 2023. Cancer Commun. 2024; 44(1): 127-172.

[8]

NCCN. The NCCN Gastric Cancer Clinical Practice Guidelines in Oncology (Version 1.2024) [EB/OL]. Fort Washington: NCCN. March 7, 2024. http://www.nccn.org/professionals/physician_gls/pdf/gastric.pdf

[9]

Janjigian YY, Shitara K, Moehler M, et al. First-line nivolumab plus chemotherapy versus chemotherapy alone for advanced gastric, gastro-oesophageal junction, and oesophageal adenocarcinoma (CheckMate 649): a randomised, open-label, phase 3 trial. Lancet. 2021; 398(10294): 27-40.

[10]

Wang FH, Zhang XT, Li YF, et al. The Chinese Society of Clinical Oncology (CSCO): clinical guidelines for the diagnosis and treatment of gastric cancer, 2021. Cancer Commun (Lond). 2021; 41(8): 747-795.

[11]

Ajani JA, D’Amico TA, Bentrem DJ, et al. Gastric cancer, version 2.2022, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2022; 20(2): 167-192.

[12]

Janjigian YY, Kawazoe A, Yañez P, et al. The KEYNOTE-811 trial of dual PD-1 and HER2 blockade in HER2-positive gastric cancer. Nature. 2021; 600(7890): 727-730.

[13]

Lordick F, Carneiro F, Cascinu S, et al. Gastric cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol. 2022; 33(10): 1005-1020.

[14]

Verschoor YL, van de Haar J, van den Berg JG, et al. Neoadjuvant atezolizumab plus chemotherapy in gastric and gastroesophageal junction adenocarcinoma: the phase 2 PANDA trial. Nat Med. 2024; 30(2): 519-530.

[15]

Jiang H, Yu X, Li N, et al. Efficacy and safety of neoadjuvant sintilimab, oxaliplatin and capecitabine in patients with locally advanced, resectable gastric or gastroesophageal junction adenocarcinoma: early results of a phase 2 study. J Immunother Cancer. 2022; 10(3): e003635.

[16]

Guo H, Ding P, Sun C, et al. Efficacy and safety of sintilimab plus XELOX as a neoadjuvant regimen in patients with locally advanced gastric cancer: a single-arm, open-label, phase II trial. Front Oncol. 2022; 12: 927781.

[17]

Lin JL, Lin JX, Lin JP, et al. Safety and efficacy of camrelizumab in combination with nab-paclitaxel plus S-1 for the treatment of gastric cancer with serosal invasion. Front Immunol. 2021; 12: 783243.

[18]

Kim R, An M, Lee H, et al. Early tumor-immune microenvironmental remodeling and response to first-line fluoropyrimidine and platinum chemotherapy in advanced gastric cancer. Cancer Discov. 2022; 12(4): 984-1001.

[19]

Li S, Yu W, Xie F, et al. Neoadjuvant therapy with immune checkpoint blockade, antiangiogenesis, and chemotherapy for locally advanced gastric cancer. Nat Commun. 2023; 14(1): 8.

[20]

Bagaev A, Kotlov N, Nomie K, et al. Conserved pan-cancer microenvironment subtypes predict response to immunotherapy. Cancer Cell. 2021; 39(6): 845-865.

[21]

Chen DS, Mellman I. Elements of cancer immunity and the cancer–immune set point. Nature. 2017; 541(7637): 321-330.

[22]

Bejarano L, Jordāo MJC, Joyce JA. Therapeutic targeting of the tumor microenvironment. Cancer Discov. 2021; 11(4): 933-959.

[23]

Jiang M, Yang Z, Dai J, et al. Intratumor microbiome: selective colonization in the tumor microenvironment and a vital regulator of tumor biology. MedComm. 2023; 4(5): e376.

[24]

Chu T, Wang Z, Pe’er D, Danko CG. Cell type and gene expression deconvolution with BayesPrism enables Bayesian integrative analysis across bulk and single-cell RNA sequencing in oncology. Nat Cancer. 2022; 3(4): 505-517.

[25]

Fridman WH, Meylan M, Petitprez F, Sun CM, Italiano A, Sautès-Fridman C. B cells and tertiary lymphoid structures as determinants of tumour immune contexture and clinical outcome. Nat Rev Clin Oncol. 2022; 19(7): 441-457.

[26]

Cabrita R, Lauss M, Sanna A, et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature. 2020; 577(7791): 561-565.

[27]

Jiang Q, Liu W, Zeng X, et al. Safety and efficacy of tislelizumab plus chemotherapy versus chemotherapy alone as neoadjuvant treatment for patients with locally advanced gastric cancer: real-world experience with a consecutive patient cohort. Front Immunol. 2023; 14: 1122121.

[28]

Zhang X, Zhang C, Hou H, et al. Neoadjuvant PD-1 blockade plus chemotherapy versus chemotherapy alone in locally advanced stage II–III gastric cancer: a single-centre retrospective study. Transl Oncol. 2023; 31: 101657.

[29]

Su J, Guo W, Chen Z, et al. Safety and short-term outcomes of laparoscopic surgery for advanced gastric cancer after neoadjuvant immunotherapy: a retrospective cohort study. Front Immunol. 2022; 13: 1078196.

[30]

Yuan SQ, Nie RC, Jin Y, et al. Perioperative toripalimab and chemotherapy in locally advanced gastric or gastro-esophageal junction cancer: a randomized phase 2 trial. Nat Med. 2024; 30(2): 552-559.

[31]

Yuan S, Nie RC, Jin Y, et al. Perioperative PD-1 antibody toripalimab plus SOX or XELOX chemotherapy versus SOX or XELOX alone for locally advanced gastric or gastro-oesophageal junction cancer: results from a prospective, randomized, open-label, phase II trial. J Clin Oncol. 2023; 41(_suppl16): 4001.

[32]

Lin X, Zong C, Zhang Z, Fang W, Xu P. Progresses in biomarkers for cancer immunotherapy. MedComm. 2023; 4(5): e387.

[33]

Le Magnen C, Shen MM, Abate-Shen C. Lineage plasticity in cancer progression and treatment. Annu Rev Cancer Biol. 2018; 2: 271-289.

[34]

Quintanal-Villalonga Á, Chan JM, Yu HA, et al. Lineage plasticity in cancer: a shared pathway of therapeutic resistance. Nat Rev Clin Oncol. 2020; 17(6): 360-371.

[35]

Liu K, Tian F, Chen X, et al. Stabilization of TGF-β receptor 1 by a receptor-associated adaptor dictates feedback activation of the TGF-β signaling pathway to maintain liver cancer stemness and drug resistance. Adv Sci (Weinh). 2024:e2402327.

[36]

Xiang J, Zhang N, Du A, et al. A ubiquitin-dependent switch on MEF2D senses pro-metastatic niche signals to facilitate intrahepatic metastasis of liver cancer. Adv Sci (Weinh). 2023; 10(35): e2305550.

[37]

Al-Batran SE, Lorenzen S, Thuss-Patience PC, et al. Surgical and pathological outcome, and pathological regression, in patients receiving perioperative atezolizumab in combination with FLOT chemotherapy versus FLOT alone for resectable esophagogastric adenocarcinoma: interim results from DANTE, a randomized, multicenter, phase IIb trial of the FLOT-AIO German Gastric Cancer Group and Swiss SAKK. J Clin Oncol. 2022; 40(_suppl16): 4003.

[38]

Quintanal-Villalonga A, Taniguchi H, Zhan YA, et al. Multiomic analysis of lung tumors defines pathways activated in neuroendocrine transformation. Cancer Discov. 2021; 11(12): 3028-3047.

[39]

Wang M, Chen X, Tan P, et al. Acquired semi-squamatization during chemotherapy suggests differentiation as a therapeutic strategy for bladder cancer. Cancer Cell. 2022; 40(9): 1044-1059.

[40]

Zou Q, Liu M, Liu K, Zhang Y, North BJ, Wang B. E3 ubiquitin ligases in cancer stem cells: key regulators of cancer hallmarks and novel therapeutic opportunities. Cell Oncol (Dordr). 2023; 46(3): 545-570.

[41]

Lauren P. The two histological main types of gastric carcinoma: diffuse and so-called intestinal-type carcinoma. An attempt at a histo-clinical classification. Acta Pathol Microbiol Scand. 1965; 64: 31-49.

[42]

Liu M, Liu Q, Zou Q, et al. The composition and roles of gastric stem cells in epithelial homeostasis, regeneration, and tumorigenesis. Cell Oncol. 2023; 46(4):867-883.

[43]

Smyth EC, Nilsson M, Grabsch HI, van Grieken NC, Lordick F. Gastric cancer. Lancet. 2020; 396(10251): 635-648.

[44]

Wu X, Wu MY, Jiang M, et al. TNF-α sensitizes chemotherapy and radiotherapy against breast cancer cells. Cancer Cell Int. 2017; 17: 13.

[45]

Dobin A, Davis CA, Schlesinger F, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013; 29(1): 15-21.

[46]

Li H, Handsaker B, Wysoker A, et al. The sequence alignment/map format and SAMtools. Bioinformatics. 2009; 25(16): 2078-2079.

[47]

Harrow J, Frankish A, Gonzalez JM, et al. GENCODE: the reference human genome annotation for the ENCODE project. Genome Res. 2012; 22(9): 1760-1774.

[48]

Liao Y, Smyth GK, Shi W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics. 2014; 30(7): 923-930.

[49]

Johnson WE, Li C, Rabinovic A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics. 2007; 8(1): 118-127.

[50]

Kwon M, An M, Klempner SJ, et al. Determinants of response and intrinsic resistance to PD-1 blockade in microsatellite instability-high gastric cancer. Cancer Discov. 2021; 11(9): 2168-2185.

[51]

Wolf FA, Angerer P, Theis FJ. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 2018; 19(1): 15.

[52]

Traag VA, Waltman L, van Eck NJ. From Louvain to Leiden: guaranteeing well-connected communities. Sci Rep. 2019; 9(1): 5233.

[53]

Kang B, Camps J, Fan B, et al. Parallel single-cell and bulk transcriptome analyses reveal key features of the gastric tumor microenvironment. Genome Biol. 2022; 23(1): 265.

RIGHTS & PERMISSIONS

2024 The Author(s). MedComm published by Sichuan International Medical Exchange & Promotion Association (SCIMEA) and John Wiley & Sons Australia, Ltd.

AI Summary AI Mindmap
PDF

164

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/