Artificial intelligence for personalized therapy
Editorial

Artificial intelligence for personalized therapy

Several randomized controlled trials have demonstrated the efficacy of anti-programmed death-1 (PD-1) antibodies (1-4), anti-HER2 therapy (5,6), and anti-claudin 18.2 antibody (7,8) for gastric cancer in recent years. Although their effectiveness is limited, pharmacotherapy for gastric cancer has gradually progressed (9). The efficacy of anti-PD-1 antibodies in microsatellite instability-high patients is particularly significant, and such a predictive biomarker is extremely valuable (10). While the administration of anti-PD-1 antibodies has led to a slight increase in long-term survival, it is also known that there are more cases of long-term survival with chemotherapy alone in programmed death ligand 1 (PD-L1)-positive patients than in PD-L1-negative patients (11).

In the 2000s, as global clinical trials expanded, researchers discovered that the efficacy of the same drug varied due to differences in the medical environment and patient status across regions (12-14). To provide effective drugs for patients in each region, it is essential to establish a system for collecting data on efficacy and safety from hospitals during clinical practice after approval. Additionally, continuous reviews should ensure that only truly effective drugs are administered to patients who need them. By appropriately collecting such data and utilizing artificial intelligence (AI), machine learning, it becomes possible to predict the survival of patients who have been treated for similar conditions in the past. For example, sex differences were observed in the effects of oxaliplatin plus S-1 (15,16), and it was confirmed that excision repair cross-complementing group 1 (ERCC1), gene alterations and ERCC1 gene expression are useful in predicting the effects of cisplatin plus S-1 and triplet therapy with docetaxel and cisplatin plus S-1 (17-20) (Figure 1). The chemotherapy-sensitive group without any ERCC1 genome alterations with high expression of ERCC1 showed better survival with the doublet therapy (20).

Figure 1 Predictive and prognostic value of ERCC1 in patients with advanced gastric cancer. SNP, single nucleotide polymorphism.

Regional variations in the effectiveness of perioperative chemotherapy exist due to differences in surgical methods, medical environment, and patient status. For instance, in China, Korea, Europe, and the United States, perioperative chemotherapy is a standard treatment (21-23). In contrast, postoperative adjuvant chemotherapy is the standard treatment in Japan because the usefulness of preoperative chemotherapy has not been demonstrated (24,25). Study results also differ based on the macroscopic and histological type of patients involved. While aiming to optimize treatment using AI and leveraging existing cancer registry databases, genome analysis for cytotoxic drugs to predict the efficacy, innovative approaches to optimal targets for peritoneal dissemination and type 4 gastric cancer will improve the overall prognosis for gastric cancer patients, as many researchers have taken up this challenge.


Acknowledgments

Funding: None.


Footnote

Provenance and Peer Review: This article was commissioned by the editorial office, Chinese Clinical Oncology for the series “Progress and Future Direction to Treat Advanced Gastric Cancer”. The article did not undergo external peer review.

Conflicts of Interest: The author has completed the ICMJE uniform disclosure form (available at https://cco.amegroups.com/article/view/10.21037/cco-24-66/coif). The series “Progress and Future Direction to Treat Advanced Gastric Cancer” was commissioned by the editorial office without any funding or sponsorship. Y.Y. served as the unpaid Guest Editor of the series. Y.Y. also reported payments from Taiho, Ono and Chugai. The author has no other conflicts of interest to declare.

Ethical Statement: The author is accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. 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;398:27-40. [Crossref] [PubMed]
  2. Sun JM, Shen L, Shah MA, et al. Pembrolizumab plus chemotherapy versus chemotherapy alone for first-line treatment of advanced oesophageal cancer (KEYNOTE-590): a randomised, placebo-controlled, phase 3 study. Lancet 2021;398:759-71. [Crossref] [PubMed]
  3. Kang YK, Chen LT, Ryu MH, et al. Nivolumab plus chemotherapy versus placebo plus chemotherapy in patients with HER2-negative, untreated, unresectable advanced or recurrent gastric or gastro-oesophageal junction cancer (ATTRACTION-4): a randomised, multicentre, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2022;23:234-47. [Crossref] [PubMed]
  4. Lu Z, Wang J, Shu Y, et al. Sintilimab versus placebo in combination with chemotherapy as first line treatment for locally advanced or metastatic oesophageal squamous cell carcinoma (ORIENT-15): multicentre, randomised, double blind, phase 3 trial. BMJ 2022;377:e068714. [Crossref] [PubMed]
  5. Bang YJ, Van Cutsem E, Feyereislova A, et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet 2010;376:687-97. [Crossref] [PubMed]
  6. Shitara K, Bang YJ, Iwasa S, et al. Trastuzumab Deruxtecan in Previously Treated HER2-Positive Gastric Cancer. N Engl J Med 2020;382:2419-30. [Crossref] [PubMed]
  7. Inamoto R, Takahashi N, Yamada Y. Claudin18.2 in Advanced Gastric Cancer. Cancers (Basel) 2023;15:5742. [Crossref] [PubMed]
  8. Shitara K, Lordick F, Bang YJ, et al. Zolbetuximab plus mFOLFOX6 in patients with CLDN18.2-positive, HER2-negative, untreated, locally advanced unresectable or metastatic gastric or gastro-oesophageal junction adenocarcinoma (SPOTLIGHT): a multicentre, randomised, double-blind, phase 3 trial. Lancet 2023;401:1655-1668. Lancet 2023;401:1655-68. [Crossref] [PubMed]
  9. Yamada Y. Present status and perspective of chemotherapy for patients with unresectable advanced or metastatic gastric cancer in Japan. Glob Health Med 2020;2:156-63. [Crossref] [PubMed]
  10. Pietrantonio F, Randon G, Di Bartolomeo M, et al. Predictive role of microsatellite instability for PD-1 blockade in patients with advanced gastric cancer: a meta-analysis of randomized clinical trials. ESMO Open 2021;6:100036. [Crossref] [PubMed]
  11. Hirano H, Yamada Y, Nagashima K, et al. Impact of PD-L1 expression on survival in patients with unresectable/recurrent gastric cancer receiving first-line chemotherapy without immune checkpoint inhibitors. J Clin Oncol 2024;42:suppl.391.
  12. Ohtsu A, Shah MA, Van Cutsem E, et al. Bevacizumab in combination with chemotherapy as first-line therapy in advanced gastric cancer: a randomized, double-blind, placebo-controlled phase III study. J Clin Oncol 2011;29:3968-76. [Crossref] [PubMed]
  13. Shen L, Li J, Xu J, et al. Bevacizumab plus capecitabine and cisplatin in Chinese patients with inoperable locally advanced or metastatic gastric or gastroesophageal junction cancer: randomized, double-blind, phase III study (AVATAR study). Gastric Cancer 2015;18:168-76. [Crossref] [PubMed]
  14. Wilke H, Muro K, Van Cutsem E, et al. Ramucirumab plus paclitaxel versus placebo plus paclitaxel in patients with previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (RAINBOW): a double-blind, randomised phase 3 trial. Lancet Oncol 2014;15:1224-35. [Crossref] [PubMed]
  15. Yamada Y, Higuchi K, Nishikawa K, et al. Phase III study comparing oxaliplatin plus S-1 with cisplatin plus S-1 in chemotherapy-naïve patients with advanced gastric cancer. Ann Oncol 2015;26:141-8. [Crossref] [PubMed]
  16. Yamada Y, Koizumi W, Nishikawa K, et al. Sex differences in the safety of S-1 plus oxaliplatin and S-1 plus cisplatin for patients with metastatic gastric cancer. Cancer Sci 2019;110:2875-83. [Crossref] [PubMed]
  17. Matsubara J, Nishina T, Yamada Y, et al. Impacts of excision repair cross-complementing gene 1 (ERCC1), dihydropyrimidine dehydrogenase, and epidermal growth factor receptor on the outcomes of patients with advanced gastric cancer. Br J Cancer 2008;98:832-9. [Crossref] [PubMed]
  18. Yamada Y, Boku N, Nishina T, et al. Impact of excision repair cross-complementing gene 1 (ERCC1) on the outcomes of patients with advanced gastric cancer: correlative study in Japan Clinical Oncology Group Trial JCOG9912. Ann Oncol 2013;24:2560-5. [Crossref] [PubMed]
  19. Yamada Y, Boku N, Mizusawa J, et al. Docetaxel plus cisplatin and S-1 versus cisplatin and S-1 in patients with advanced gastric cancer (JCOG1013): an open-label, phase 3, randomised controlled trial. Lancet Gastroenterol Hepatol 2019;4:501-10. [Crossref] [PubMed]
  20. Yamada Y, Nagashima K, Azuma M, et al. Predictive and prognostic value of excision repair cross-complementing group 1 in patients with advanced gastric cancer. BJC Reports 2024;2:18. [Crossref]
  21. Al-Batran SE, Homann N, Pauligk C, et al. Effect of Neoadjuvant Chemotherapy Followed by Surgical Resection on Survival in Patients With Limited Metastatic Gastric or Gastroesophageal Junction Cancer: The AIO-FLOT3 Trial. JAMA Oncol 2017;3:1237-44. [Crossref] [PubMed]
  22. Kang YK, Yook JH, Park YK, et al. PRODIGY: A Phase III Study of Neoadjuvant Docetaxel, Oxaliplatin, and S-1 Plus Surgery and Adjuvant S-1 Versus Surgery and Adjuvant S-1 for Resectable Advanced Gastric Cancer. J Clin Oncol 2021;39:2903-13. [Crossref] [PubMed]
  23. 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;22:1081-92. Erratum in: Lancet Oncol 2021;22:e347. [Crossref] [PubMed]
  24. Sakuramoto S, Sasako M, Yamaguchi T, et al. Adjuvant chemotherapy for gastric cancer with S-1, an oral fluoropyrimidine. N Engl J Med 2007;357:1810-20. [Crossref] [PubMed]
  25. Yoshida K, Kodera Y, Kochi M, et al. Addition of Docetaxel to Oral Fluoropyrimidine Improves Efficacy in Patients With Stage III Gastric Cancer: Interim Analysis of JACCRO GC-07, a Randomized Controlled Trial. J Clin Oncol 2019;37:1296-304. [Crossref] [PubMed]
Yasuhide Yamada

Yasuhide Yamada, MD, PhD, FACP

Department of Medical Research, National Center for Global Health and Medicine, Tokyo, Japan

(Email: yayamada@hosp.ncgm.go.jp)

Keywords: Programmed death ligand 1 (PD-L1); gastric cancer; HER2; claudin 18.2; ERCC1

Submitted May 17, 2024. Accepted for publication Jun 04, 2024. Published online Jul 18, 2024.

doi: 10.21037/cco-24-66

Cite this article as: Yamada Y. Artificial intelligence for personalized therapy. Chin Clin Oncol 2024;13(4):47. doi: 10.21037/cco-24-66

Download Citation