Skip to main content
Top
Published in: Gastric Cancer 3/2023

22-02-2023 | Gastric Cancer | Original Article

ARID1A deficiency is targetable by AKT inhibitors in HER2-negative gastric cancer

Authors: Takahiro Sato, Motonobu Saito, Shotaro Nakajima, Katsuharu Saito, Masanori Katagata, Satoshi Fukai, Hirokazu Okayama, Wataru Sakamoto, Zenichiro Saze, Tomoyuki Momma, Kosaku Mimura, Koji Kono

Published in: Gastric Cancer | Issue 3/2023

Login to get access

Abstract

Background

The PI3K/AKT signaling pathway is frequently activated in gastric cancer (GC); however, AKT inhibitors are not effective in unselected GC patients in clinical trials. Mutations in AT-rich interactive domain 1A (ARID1A), which are found in approximately 30% of GC patients, activate PI3K/AKT signaling, suggesting that targeting the ARID1A deficiency-activated PI3K/AKT pathway is a therapeutic candidate for ARID1A-deficient GC.

Methods

The effect of AKT inhibitors was evaluated using cell viability and colony formation assays in ARID1A-deficient and ARID1A knockdown ARID1A-WT GC cells as well as in HER2-positive and HER2-negative GC. The Cancer Genome Atlas cBioPortal and Gene Expression Omnibus microarray databases were accessed to determine the extent of dependence of GC cell growth on the PI3K/AKT signaling pathway.

Results

AKT inhibitors decreased the viability of ARID1A-deficient cells and the inhibitory effect was greater in ARID1A-deficient/HER2-negative GC cells. Bioinformatics data suggested that PI3K/AKT signaling plays a greater role in proliferation and survival in ARID1A-deficient/HER2-negative GC cells than in ARID1A-deficient/HER2-positive cells, supporting the higher therapeutic efficacy of AKT inhibitors.

Conclusions

The effect of AKT inhibitors on cell proliferation and survival is affected by HER2 status, providing a rationale for exploring targeted therapy using AKT inhibitors in ARID1A-deficient/HER2-negative GC.
Appendix
Available only for authorised users
Literature
1.
go back to reference Jim MA, Pinheiro PS, Carreira H, Espey DK, Wiggins CL, Weir HK. Stomach cancer survival in the United States by race and stage (2001–2009): findings from the CONCORD-2 study. Cancer. 2017;123(Suppl 24):4994–5013.PubMedCrossRef Jim MA, Pinheiro PS, Carreira H, Espey DK, Wiggins CL, Weir HK. Stomach cancer survival in the United States by race and stage (2001–2009): findings from the CONCORD-2 study. Cancer. 2017;123(Suppl 24):4994–5013.PubMedCrossRef
2.
go back to reference Taniyama Y, Katanoda K, Charvat H, Hori M, Ohno Y, Sasazuki S, et al. Estimation of lifetime cumulative incidence and mortality risk of gastric cancer. Jpn J Clin Oncol. 2017;47:1097–102.PubMedPubMedCentralCrossRef Taniyama Y, Katanoda K, Charvat H, Hori M, Ohno Y, Sasazuki S, et al. Estimation of lifetime cumulative incidence and mortality risk of gastric cancer. Jpn J Clin Oncol. 2017;47:1097–102.PubMedPubMedCentralCrossRef
3.
go back to reference Kono K, Nakajima S, Mimura K. Current status of immune checkpoint inhibitors for gastric cancer. Gastric Cancer. 2020;23:565–78.PubMedCrossRef Kono K, Nakajima S, Mimura K. Current status of immune checkpoint inhibitors for gastric cancer. Gastric Cancer. 2020;23:565–78.PubMedCrossRef
4.
go back to reference Janjigian YY, Shitara K, Moehler M, Garrido M, Salman P, Shen L, 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.PubMedPubMedCentralCrossRef Janjigian YY, Shitara K, Moehler M, Garrido M, Salman P, Shen L, 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.PubMedPubMedCentralCrossRef
5.
go back to reference Cancer Genome Atlas Research N. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513:202–9.CrossRef Cancer Genome Atlas Research N. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513:202–9.CrossRef
6.
go back to reference Cristescu R, Lee J, Nebozhyn M, Kim KM, Ting JC, Wong SS, et al. Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat Med. 2015;21:449–56.PubMedCrossRef Cristescu R, Lee J, Nebozhyn M, Kim KM, Ting JC, Wong SS, et al. Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat Med. 2015;21:449–56.PubMedCrossRef
8.
go back to reference Dijksterhuis WPM, Verhoeven RHA, Meijer SL, Slingerland M, Haj Mohammad N, de Vos-Geelen J, et al. Increased assessment of HER2 in metastatic gastroesophageal cancer patients: a nationwide population-based cohort study. Gastric Cancer. 2020;23:579–90.PubMedPubMedCentralCrossRef Dijksterhuis WPM, Verhoeven RHA, Meijer SL, Slingerland M, Haj Mohammad N, de Vos-Geelen J, et al. Increased assessment of HER2 in metastatic gastroesophageal cancer patients: a nationwide population-based cohort study. Gastric Cancer. 2020;23:579–90.PubMedPubMedCentralCrossRef
9.
go back to reference Van Cutsem E, Bang YJ, Feng-Yi F, Xu JM, Lee KW, Jiao SC, et al. HER2 screening data from ToGA: targeting HER2 in gastric and gastroesophageal junction cancer. Gastric Cancer. 2015;18:476–84.PubMedCrossRef Van Cutsem E, Bang YJ, Feng-Yi F, Xu JM, Lee KW, Jiao SC, et al. HER2 screening data from ToGA: targeting HER2 in gastric and gastroesophageal junction cancer. Gastric Cancer. 2015;18:476–84.PubMedCrossRef
11.
go back to reference Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki 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.PubMedCrossRef Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki 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.PubMedCrossRef
12.
go back to reference Shinozaki-Ushiku A, Ishikawa S, Komura D, Seto Y, Aburatani H, Ushiku T. The first case of gastric carcinoma with NTRK rearrangement: identification of a novel ATP1B-NTRK1 fusion. Gastric Cancer. 2020;23:944–7.PubMedCrossRef Shinozaki-Ushiku A, Ishikawa S, Komura D, Seto Y, Aburatani H, Ushiku T. The first case of gastric carcinoma with NTRK rearrangement: identification of a novel ATP1B-NTRK1 fusion. Gastric Cancer. 2020;23:944–7.PubMedCrossRef
14.
go back to reference Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357:409–13.PubMedPubMedCentralCrossRef Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357:409–13.PubMedPubMedCentralCrossRef
16.
go back to reference Yamada L, Saito M, Thar Min AK, Saito K, Ashizawa M, Kase K, et al. Selective sensitivity of EZH2 inhibitors based on synthetic lethality in ARID1A-deficient gastric cancer. Gastric Cancer. 2021;24:60–71.PubMedCrossRef Yamada L, Saito M, Thar Min AK, Saito K, Ashizawa M, Kase K, et al. Selective sensitivity of EZH2 inhibitors based on synthetic lethality in ARID1A-deficient gastric cancer. Gastric Cancer. 2021;24:60–71.PubMedCrossRef
17.
18.
go back to reference Wang K, Kan J, Yuen ST, Shi ST, Chu KM, Law S, et al. Exome sequencing identifies frequent mutation of ARID1A in molecular subtypes of gastric cancer. Nat Genet. 2011;43:1219–23.PubMedCrossRef Wang K, Kan J, Yuen ST, Shi ST, Chu KM, Law S, et al. Exome sequencing identifies frequent mutation of ARID1A in molecular subtypes of gastric cancer. Nat Genet. 2011;43:1219–23.PubMedCrossRef
19.
20.
go back to reference Sasaki T, Kuniyasu H. Significance of AKT in gastric cancer (review). Int J Oncol. 2014;45:2187–92.PubMedCrossRef Sasaki T, Kuniyasu H. Significance of AKT in gastric cancer (review). Int J Oncol. 2014;45:2187–92.PubMedCrossRef
23.
go back to reference Kase K, Saito M, Nakajima S, Takayanagi D, Saito K, Yamada L, et al. ARID1A deficiency in EBV-positive gastric cancer is partially regulated by EBV-encoded miRNAs, but not by DNA promotor hypermethylation. Carcinogenesis. 2021;42:21–30.PubMedCrossRef Kase K, Saito M, Nakajima S, Takayanagi D, Saito K, Yamada L, et al. ARID1A deficiency in EBV-positive gastric cancer is partially regulated by EBV-encoded miRNAs, but not by DNA promotor hypermethylation. Carcinogenesis. 2021;42:21–30.PubMedCrossRef
24.
go back to reference Nakajima S, Mimura K, Matsumoto T, Thar Min AK, Ito M, Nakano H, et al. The effects of T-DXd on the expression of HLA class I and chemokines CXCL9/10/11 in HER2-overexpressing gastric cancer cells. Sci Rep. 2021;11:16891.PubMedPubMedCentralCrossRef Nakajima S, Mimura K, Matsumoto T, Thar Min AK, Ito M, Nakano H, et al. The effects of T-DXd on the expression of HLA class I and chemokines CXCL9/10/11 in HER2-overexpressing gastric cancer cells. Sci Rep. 2021;11:16891.PubMedPubMedCentralCrossRef
25.
go back to reference Nakano H, Saito M, Nakajima S, Saito K, Nakayama Y, Kase K, et al. PD-L1 overexpression in EBV-positive gastric cancer is caused by unique genomic or epigenomic mechanisms. Sci Rep. 2021;11:1982.PubMedPubMedCentralCrossRef Nakano H, Saito M, Nakajima S, Saito K, Nakayama Y, Kase K, et al. PD-L1 overexpression in EBV-positive gastric cancer is caused by unique genomic or epigenomic mechanisms. Sci Rep. 2021;11:1982.PubMedPubMedCentralCrossRef
26.
go back to reference Lei Z, Tan IB, Das K, Deng N, Zouridis H, Pattison S, et al. Identification of molecular subtypes of gastric cancer with different responses to PI3-kinase inhibitors and 5-fluorouracil. Gastroenterology. 2013;145:554–65.PubMedCrossRef Lei Z, Tan IB, Das K, Deng N, Zouridis H, Pattison S, et al. Identification of molecular subtypes of gastric cancer with different responses to PI3-kinase inhibitors and 5-fluorouracil. Gastroenterology. 2013;145:554–65.PubMedCrossRef
27.
go back to reference Qian Z, Zhu G, Tang L, Wang M, Zhang L, Fu J, et al. Whole genome gene copy number profiling of gastric cancer identifies PAK1 and KRAS gene amplification as therapy targets. Genes Chromosom Cancer. 2014;53:883–94.PubMedCrossRef Qian Z, Zhu G, Tang L, Wang M, Zhang L, Fu J, et al. Whole genome gene copy number profiling of gastric cancer identifies PAK1 and KRAS gene amplification as therapy targets. Genes Chromosom Cancer. 2014;53:883–94.PubMedCrossRef
28.
go back to reference George J, Saito M, Tsuta K, Iwakawa R, Shiraishi K, Scheel AH, et al. Genomic amplification of CD274 (PD-L1) in small-cell lung cancer. Clin Cancer Res. 2017;23:1220–6.PubMedCrossRef George J, Saito M, Tsuta K, Iwakawa R, Shiraishi K, Scheel AH, et al. Genomic amplification of CD274 (PD-L1) in small-cell lung cancer. Clin Cancer Res. 2017;23:1220–6.PubMedCrossRef
29.
go back to reference Song M, Bode AM, Dong Z, Lee MH. AKT as a therapeutic target for cancer. Cancer Res. 2019;79:1019–31.PubMedCrossRef Song M, Bode AM, Dong Z, Lee MH. AKT as a therapeutic target for cancer. Cancer Res. 2019;79:1019–31.PubMedCrossRef
30.
go back to reference Dong X, Song S, Li Y, Fan Y, Wang L, Wang R, et al. Loss of ARID1A activates mTOR signaling and SOX9 in gastric adenocarcinoma-rationale for targeting ARID1A deficiency. Gut. 2022;71:467–78.PubMedCrossRef Dong X, Song S, Li Y, Fan Y, Wang L, Wang R, et al. Loss of ARID1A activates mTOR signaling and SOX9 in gastric adenocarcinoma-rationale for targeting ARID1A deficiency. Gut. 2022;71:467–78.PubMedCrossRef
31.
go back to reference Lee D, Yu EJ, Ham IH, Hur H, Kim YS. AKT inhibition is an effective treatment strategy in ARID1A-deficient gastric cancer cells. Onco Targets Ther. 2017;10:4153–9.PubMedPubMedCentralCrossRef Lee D, Yu EJ, Ham IH, Hur H, Kim YS. AKT inhibition is an effective treatment strategy in ARID1A-deficient gastric cancer cells. Onco Targets Ther. 2017;10:4153–9.PubMedPubMedCentralCrossRef
32.
go back to reference Bang YJ, Kang YK, Ng M, Chung HC, Wainberg ZA, Gendreau S, et al. A phase II, randomised study of mFOLFOX6 with or without the Akt inhibitor ipatasertib in patients with locally advanced or metastatic gastric or gastroesophageal junction cancer. Eur J Cancer. 2019;108:17–24.PubMedCrossRef Bang YJ, Kang YK, Ng M, Chung HC, Wainberg ZA, Gendreau S, et al. A phase II, randomised study of mFOLFOX6 with or without the Akt inhibitor ipatasertib in patients with locally advanced or metastatic gastric or gastroesophageal junction cancer. Eur J Cancer. 2019;108:17–24.PubMedCrossRef
33.
go back to reference Sweeney C, Bracarda S, Sternberg CN, Chi KN, Olmos D, Sandhu S, et al. Ipatasertib plus abiraterone and prednisolone in metastatic castration-resistant prostate cancer (IPATential150): a multicentre, randomised, double-blind, phase 3 trial. Lancet. 2021;398:131–42.PubMedCrossRef Sweeney C, Bracarda S, Sternberg CN, Chi KN, Olmos D, Sandhu S, et al. Ipatasertib plus abiraterone and prednisolone in metastatic castration-resistant prostate cancer (IPATential150): a multicentre, randomised, double-blind, phase 3 trial. Lancet. 2021;398:131–42.PubMedCrossRef
34.
go back to reference Isakoff SJ, Tabernero J, Molife LR, Soria JC, Cervantes A, Vogelzang NJ, et al. Antitumor activity of ipatasertib combined with chemotherapy: results from a phase Ib study in solid tumors. Ann Oncol. 2020;31:626–33.PubMedCrossRef Isakoff SJ, Tabernero J, Molife LR, Soria JC, Cervantes A, Vogelzang NJ, et al. Antitumor activity of ipatasertib combined with chemotherapy: results from a phase Ib study in solid tumors. Ann Oncol. 2020;31:626–33.PubMedCrossRef
35.
go back to reference Oliveira M, Saura C, Nuciforo P, Calvo I, Andersen J, Passos-Coelho JL, et al. FAIRLANE, a double-blind placebo-controlled randomized phase II trial of neoadjuvant ipatasertib plus paclitaxel for early triple-negative breast cancer. Ann Oncol. 2019;30:1289–97.PubMedCrossRef Oliveira M, Saura C, Nuciforo P, Calvo I, Andersen J, Passos-Coelho JL, et al. FAIRLANE, a double-blind placebo-controlled randomized phase II trial of neoadjuvant ipatasertib plus paclitaxel for early triple-negative breast cancer. Ann Oncol. 2019;30:1289–97.PubMedCrossRef
36.
go back to reference Lee J, Kim ST, Kim K, Lee H, Kozarewa I, Mortimer PGS, et al. Tumor genomic profiling guides patients with metastatic gastric cancer to targeted treatment: the VIKTORY umbrella trial. Cancer Discov. 2019;9:1388–405.PubMedCrossRef Lee J, Kim ST, Kim K, Lee H, Kozarewa I, Mortimer PGS, et al. Tumor genomic profiling guides patients with metastatic gastric cancer to targeted treatment: the VIKTORY umbrella trial. Cancer Discov. 2019;9:1388–405.PubMedCrossRef
37.
go back to reference Li J, Davies BR, Han S, Zhou M, Bai Y, Zhang J, et al. The AKT inhibitor AZD5363 is selectively active in PI3KCA mutant gastric cancer, and sensitizes a patient-derived gastric cancer xenograft model with PTEN loss to Taxotere. J Transl Med. 2013;11:241.PubMedPubMedCentralCrossRef Li J, Davies BR, Han S, Zhou M, Bai Y, Zhang J, et al. The AKT inhibitor AZD5363 is selectively active in PI3KCA mutant gastric cancer, and sensitizes a patient-derived gastric cancer xenograft model with PTEN loss to Taxotere. J Transl Med. 2013;11:241.PubMedPubMedCentralCrossRef
38.
go back to reference Wilson MR, Reske JJ, Holladay J, Wilber GE, Rhodes M, Koeman J, et al. ARID1A and PI3-kinase pathway mutations in the endometrium drive epithelial transdifferentiation and collective invasion. Nat Commun. 2019;10:3554.PubMedPubMedCentralCrossRef Wilson MR, Reske JJ, Holladay J, Wilber GE, Rhodes M, Koeman J, et al. ARID1A and PI3-kinase pathway mutations in the endometrium drive epithelial transdifferentiation and collective invasion. Nat Commun. 2019;10:3554.PubMedPubMedCentralCrossRef
39.
go back to reference Shitara K, Yatabe Y, Matsuo K, Sugano M, Kondo C, Takahari D, et al. Prognosis of patients with advanced gastric cancer by HER2 status and trastuzumab treatment. Gastric Cancer. 2013;16:261–7.PubMedCrossRef Shitara K, Yatabe Y, Matsuo K, Sugano M, Kondo C, Takahari D, et al. Prognosis of patients with advanced gastric cancer by HER2 status and trastuzumab treatment. Gastric Cancer. 2013;16:261–7.PubMedCrossRef
40.
go back to reference Lordick F, Carneiro F, Cascinu S, Fleitas T, Haustermans K, Piessen G, et al. Gastric cancer: ESMO clinical practice guideline for diagnosis, treatment and follow-up. Ann Oncol. 2022;33:1005–20.PubMedCrossRef Lordick F, Carneiro F, Cascinu S, Fleitas T, Haustermans K, Piessen G, et al. Gastric cancer: ESMO clinical practice guideline for diagnosis, treatment and follow-up. Ann Oncol. 2022;33:1005–20.PubMedCrossRef
41.
go back to reference Shitara K, Baba E, Fujitani K, Oki E, Fujii S, Yamaguchi K. Discovery and development of trastuzumab deruxtecan and safety management for patients with HER2-positive gastric cancer. Gastric Cancer. 2021;24:780–9.PubMedPubMedCentralCrossRef Shitara K, Baba E, Fujitani K, Oki E, Fujii S, Yamaguchi K. Discovery and development of trastuzumab deruxtecan and safety management for patients with HER2-positive gastric cancer. Gastric Cancer. 2021;24:780–9.PubMedPubMedCentralCrossRef
42.
go back to reference Aoki M, Iwasa S, Boku N. Trastuzumab deruxtecan for the treatment of HER2-positive advanced gastric cancer: a clinical perspective. Gastric Cancer. 2021;24:567–76.PubMedCrossRef Aoki M, Iwasa S, Boku N. Trastuzumab deruxtecan for the treatment of HER2-positive advanced gastric cancer: a clinical perspective. Gastric Cancer. 2021;24:567–76.PubMedCrossRef
43.
go back to reference Mosele F, Remon J, Mateo J, Westphalen CB, Barlesi F, Lolkema MP, et al. Recommendations for the use of next-generation sequencing (NGS) for patients with metastatic cancers: a report from the ESMO Precision Medicine Working Group. Ann Oncol. 2020;31:1491–505.PubMedCrossRef Mosele F, Remon J, Mateo J, Westphalen CB, Barlesi F, Lolkema MP, et al. Recommendations for the use of next-generation sequencing (NGS) for patients with metastatic cancers: a report from the ESMO Precision Medicine Working Group. Ann Oncol. 2020;31:1491–505.PubMedCrossRef
44.
go back to reference Diaz-Serrano A, Angulo B, Dominguez C, Pazo-Cid R, Salud A, Jimenez-Fonseca P, et al. Genomic profiling of HER2-positive gastric cancer: PI3K/Akt/mTOR pathway as predictor of outcomes in her2-positive advanced gastric cancer treated with trastuzumab. Oncologist. 2018;23:1092–102.PubMedPubMedCentralCrossRef Diaz-Serrano A, Angulo B, Dominguez C, Pazo-Cid R, Salud A, Jimenez-Fonseca P, et al. Genomic profiling of HER2-positive gastric cancer: PI3K/Akt/mTOR pathway as predictor of outcomes in her2-positive advanced gastric cancer treated with trastuzumab. Oncologist. 2018;23:1092–102.PubMedPubMedCentralCrossRef
Metadata
Title
ARID1A deficiency is targetable by AKT inhibitors in HER2-negative gastric cancer
Authors
Takahiro Sato
Motonobu Saito
Shotaro Nakajima
Katsuharu Saito
Masanori Katagata
Satoshi Fukai
Hirokazu Okayama
Wataru Sakamoto
Zenichiro Saze
Tomoyuki Momma
Kosaku Mimura
Koji Kono
Publication date
22-02-2023
Publisher
Springer Nature Singapore
Published in
Gastric Cancer / Issue 3/2023
Print ISSN: 1436-3291
Electronic ISSN: 1436-3305
DOI
https://doi.org/10.1007/s10120-023-01373-6

Other articles of this Issue 3/2023

Gastric Cancer 3/2023 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine