Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1100 2023-05-22 05:08:55 |
2 format correct Meta information modification 1100 2023-05-22 07:17:34 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Li, J.J.; Rogers, J.E.; Yamashita, K.; Waters, R.E.; Blum Murphy, M.; Ajani, J.A. Emerging Therapeutics of Gastroesophageal Cancers. Encyclopedia. Available online: https://encyclopedia.pub/entry/44623 (accessed on 26 April 2024).
Li JJ, Rogers JE, Yamashita K, Waters RE, Blum Murphy M, Ajani JA. Emerging Therapeutics of Gastroesophageal Cancers. Encyclopedia. Available at: https://encyclopedia.pub/entry/44623. Accessed April 26, 2024.
Li, Jenny J., Jane E. Rogers, Kohei Yamashita, Rebecca E. Waters, Mariela Blum Murphy, Jaffer A. Ajani. "Emerging Therapeutics of Gastroesophageal Cancers" Encyclopedia, https://encyclopedia.pub/entry/44623 (accessed April 26, 2024).
Li, J.J., Rogers, J.E., Yamashita, K., Waters, R.E., Blum Murphy, M., & Ajani, J.A. (2023, May 22). Emerging Therapeutics of Gastroesophageal Cancers. In Encyclopedia. https://encyclopedia.pub/entry/44623
Li, Jenny J., et al. "Emerging Therapeutics of Gastroesophageal Cancers." Encyclopedia. Web. 22 May, 2023.
Emerging Therapeutics of Gastroesophageal Cancers
Edit

Gastroesophageal cancers are a group of aggressive malignancies that are inherently heterogeneous with poor prognosis. Esophageal squamous cell carcinoma, esophageal adenocarcinoma, gastroesophageal junction adenocarcinoma, and gastric adenocarcinoma all have distinct underlying molecular biology, which can impact available targets and treatment response. Novel therapeutic targets are under development and future treatments will be personalized based on molecular profiling.

esophageal cancer gastric cancer immunotherapy

1. Introduction

Gastroesophageal cancers, including cancers of the esophagus, gastroesophageal junction (GEJ), and stomach, represent some of the most common cancers worldwide, with an estimated 1.7 million new cases per year. When combined, they are the third leading cause of cancer-related deaths globally [1]. Esophageal cancers are comprised of two major histologic subtypes: squamous cell carcinoma (ESCC) and adenocarcinoma (EAC). While ESCC is the most common subtype worldwide, EAC is more common in the Western countries [2]. ESCC is mainly found in the upper to mid esophagus and is associated with tobacco and alcohol use, whereas EAC is mostly found in the distal esophagus and is associated with obesity, gastroesophageal reflux, and Barrett’s esophagus [3][4]. In addition to differences in risk factors and locations, these two subtypes also have distinct molecular biology and responses to treatment, which should lead to different treatment strategies [5]. In contrast, GEJ and gastric cancers are almost all adenocarcinomas (ACs). Gastric adenocarcinomas (GACs) can be further classified into intestinal and diffuse histologic subtypes based on Lauren classification [6]. The Cancer Genome Atlas Research Network has also defined four molecular subtypes of GAC (Epstein-Barr Virus [EBV]-positive, microsatellite instability [MSI], genomically stable, and chromosomal instability), which may have implications for future therapeutic development [7]. Many exciting potential new molecular targets are being explored in ongoing clinical trials, leading to a future of personalized therapies.

2. Claudin 18.2

Claudin 18 isoform 2 (CLDN 18.2) is a tight junction molecule that is orthotopically expressed in gastric cancers and ectopically activated in multiple other cancer types, including pancreatic, esophageal, ovarian, and lung [8]. Zolbetuximab, a monoclonal antibody against CLDN 18.2, has previously shown activity when added to chemotherapy in the phase 2 FAST trial, where the addition of zolbetuximab to chemotherapy increased survival of patients with advanced gastric/GEJ AC [9]. During the 2023 ASCO Gastrointestinal Cancer Symposium, results from the phase 3 SPOTLIGHT trial were presented. In HER2-negative, CLDN 18.2-positive, advanced gastric or GEJ AC patients, first line treatment with zolbetuximab + FOLFOX (5-fluorouracil + oxaliplatin) (n = 283) significantly improved progression free survival (PFS; 10.6 months vs. 8.7 months, hazard ratio 0.75, p = 0.0066) and overall survival (OS; 18.2 months vs. 15.5 months, hazard ratio 0.75, p = 0.0053) compared to placebo + FOLFOX (n = 282) [10]. GLOW, another phase 3 trial of zolbetuximab + chemotherapy (capecitabine + oxaliplatin) in CLDN 18.2-positive HER2-negative advanced gastric/GEJ AC patients, recently announced that it met its the primary endpoint of PFS and secondary end point of OS [11]. These positive trials could establish CLDN 18.2 as a new predictive biomarker and zolbetuximab + chemotherapy as a new standard of care treatment for CLDN18.2-positive advanced gastric/GEJ cancer patients. It is important to note that in the SPOTLIGHT trial, only 13.2% patients who were CLDN 18.2-positive also had PD-L1 CPS ≥ 5, and all enrolled patients were HER2-negative. The CLDN 18.2-positive population represents a unique subset of patients who were previously mostly considered triple negative and would not have qualified for any targeted therapy options.
There are now multiple ongoing trials of novel monoclonal antibodies, antibody-drug conjugates, bispecific antibodies, and cellular therapies targeting CLDN 18.2 (NCT04856150, NCT04400383, NCT04632108, NCT04805307, NCT04495296, NCT04404595). Zolbetuximab is also being studied in combination with immunotherapy (IO) agents (NCT03505320).

3. FGFR2

Fibroblast growth factor receptors (FGFRs) have been found to be therapeutic targets in multiple cancer types, and FGFR2 specifically is a promising target for gastroesophageal cancers [12][13]. Bemarituzumab is a monoclonal antibody that targets FGFR2b [14]. The FIGHT trial, a phase 2 randomized placebo-controlled study, evaluated bemarituzumab + FOLFOX (n = 77) vs. placebo + FOLFOX (n = 78) in patients with FGFR2b overexpressed/amplified advanced gastric or GEJ AC [15]. While the trial did not meet its primary endpoint of PFS, the bemarituzumab group trended toward longer PFS (9.5 months vs. 7.4 months, hazard ratio 0.68, p = 0.073), longer OS (not reached vs. 12.9 months, hazard ratio 0.58, p = 0.027), and higher objective response rate (ORR; 47% vs. 33%) compared to the placebo group. The FIGHT trial also showed that around 30% patients with HER2-negative advanced gastric cancer had FGFR2b overexpression or FGFR2 amplification, which supports the development of FGFR2 as a therapeutic target. The phase 3 FORTITUDE-101 trial is currently underway to further evaluate bemarituzumab + FOLFOX as first line treatment of FGFR2b-selected advanced gastric/GEJ AC patients (NCT05052801).

4. DKK1

The canonical Wnt/β-catenin signaling pathway plays an important role in cancer cell proliferation, survival, and migration [16][17][18]. Dickkopf-1 protein (DKK1) is an antagonist of the Wnt signaling pathway, and overexpression of DKK1 has been associated with tumor growth, angiogenesis, and a more immunosuppressive tumor microenvironment [19][20][21]. DKN-01 is a monoclonal antibody against DKK1, and it is being studied in combination with tislelizumab (anti-PD-1) and chemotherapy (FOLFOX or CAPOX) in the phase 2 DisTinGuish trial (NCT04363801). Data from this combination as first line therapy for HER2-negative advanced gastric/GEJ cancer patients (n = 25) showed promising activity [22][23]. The ORR for all patients is 68% and the ORR for the DKK1 high group is 90%.

5. Tyrosine Kinase Inhibitors

The combination of multi-kinase inhibitor (MKI) and IO has been successful in the treatment of other tumor types, including endometrial cancer and renal cell carcinoma. This combination is also being actively explored in gastroesophageal cancers. Prior studies have shown that oncogenic kinases have immunomodulatory activity and can lead to an immunosuppressive tumor microenvironment [24]. Inhibition of oncogenic kinases with MKIs have been shown to increase cytotoxic T-cell infiltration and decrease tumor associated macrophages, which leads to a more immune-permissive tumor microenvironment [25]. The impact of MKIs on the tumor microenvironment can synergize with IO use to improve antitumor efficacy [26]. The EPOC1706 trial was a single-arm phase 2 trial that evaluated lenvatinib + pembrolizumab in patients with advanced GAC (n = 29) and ORR was 69% [27]. There are now phase 3 trials to evaluate pembrolizumab + lenvatinib + chemotherapy in advanced ESCC and gastroesophageal AC (LEAP-014, LEAP-015). Similarly, trials to evaluate other combinations of MKIs and IOs are also underway (atezolizumab + cabozantinib, tislelizumab + sitravatinib; NCT05007613, NCT05461794).

6. Others

The EBV-positive molecular subgroup of GAC patients may present a unique opportunity for treatment with IO therapy. A prospective phase 2 trial of pembrolizumab in patients with advanced gastric or GEJ AC who progressed after first line chemotherapy found that EBV-positive patients (n = 6) had an ORR of 100% [28]. Another prospective observational study noted that EBV-positivity is a biomarker for immunotherapy in metastatic gastric cancer [29]. Due to the small sample sizes, larger prospective trials will be needed to fully evaluate the role of IO in EBV-positive patients. Nonetheless, targeting EBV-positive patients with IO therapy may be a novel option for the management of these patients.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249.
  2. Morgan, E.; Soerjomataram, I.; Rumgay, H.; Coleman, H.G.; Thrift, A.P.; Vignat, J.; Laversanne, M.; Ferlay, J.; Arnold, M. The Global Landscape of Esophageal Squamous Cell Carcinoma and Esophageal Adenocarcinoma Incidence and Mortality in 2020 and Projections to 2040: New Estimates From GLOBOCAN 2020. Gastroenterology 2022, 163, 649–658.e2.
  3. Freedman, N.D.; Abnet, C.C.; Leitzmann, M.F.; Mouw, T.; Subar, A.F.; Hollenbeck, A.R.; Schatzkin, A. A Prospective Study of Tobacco, Alcohol, and the Risk of Esophageal and Gastric Cancer Subtypes. Am. J. Epidemiol. 2007, 165, 1424–1433.
  4. Coleman, H.G.; Xie, S.H.; Lagergren, J. The Epidemiology of Esophageal Adenocarcinoma. Gastroenterology 2018, 154, 390–405.
  5. Kim, J.; Bowlby, R.; Mungall, A.J.; Robertson, A.G.; Odze, R.D.; Cherniack, A.D.; Shih, J.; Pedamallu, C.S.; Cibulskis, C.; Dunford, A.; et al. Integrated Genomic Characterization of Oesophageal Carcinoma. Nature 2017, 541, 169–174.
  6. 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.
  7. Bass, A.J.; Thorsson, V.; Shmulevich, I.; Reynolds, S.M.; Miller, M.; Bernard, B.; Hinoue, T.; Laird, P.W.; Curtis, C.; Shen, H.; et al. Comprehensive Molecular Characterization of Gastric Adenocarcinoma. Nature 2014, 513, 202–209.
  8. Sahin, U.; Koslowski, M.; Dhaene, K.; Usener, D.; Brandenburg, G.; Seitz, G.; Huber, C.; Turecil, O. Claudin-18 Splice Variant 2 Is a Pan-Cancer Target Suitable for Therapeutic Antibody Development. Clin. Cancer Res. 2008, 14, 7624–7634.
  9. Sahin, U.; Türeci, Ö.; Manikhas, G.; Lordick, F.; Rusyn, A.; Vynnychenko, I.; Dudov, A.; Bazin, I.; Bondarenko, I.; Melichar, B.; et al. FAST: A Randomised Phase II Study of Zolbetuximab (IMAB362) plus EOX versus EOX Alone for First-Line Treatment of Advanced CLDN18.2-Positive Gastric and Gastro-Oesophageal Adenocarcinoma. Ann. Oncol. 2021, 32, 609–619.
  10. Shitara, K.; Lordick, F.; Bang, Y.-J.; Enzinger, P.C.; Ilson, D.H.; Shah, M.A.; Van Cutsem, E.; Xu, R.; Aprile, G.; Xu, J.; et al. Zolbetuximab + MFOLFOX6 as First-Line (1L) Treatment for Patients (Pts) Withclaudin-18.2+ (CLDN18.2+) / HER2− Locally Advanced (LA) Unresectable or Metastatic Gastric or Gastroesophageal Junction (MG/GEJ) Adenocarcinoma: Primary Results from Phase 3 SPOTLIGHT Study. J. Clin. Oncol. 2023, 41, LBA292.
  11. Contests, A.W.; Kill, F.T.; Live, W.U.; Schedule, T.V.; Investigate, T.V.; Bureau, G.D. Astellas Announces Zolbetuximab Meets Primary Endpoint in Phase 3 GLOW Trial as First-Line Treatment in Claudin 18.2 Positive, HER2-Negative Locally Advanced Unresectable or Metastatic Gastric and Gastroesophageal Junction (GEJ) Cancers. Available online: https://www.astellas.com/en/news/26891 (accessed on 26 February 2023).
  12. Katoh, M. Fibroblast Growth Factor Receptors as Treatment Targets in Clinical Oncology. Nat. Rev. Clin. Oncol. 2018, 16, 105–122.
  13. Zhang, J.; Tang, P.M.K.; Zhou, Y.; Cheng, A.S.L.; Yu, J.; Kang, W.; To, K.F. Targeting the Oncogenic FGF-FGFR Axis in Gastric Carcinogenesis. Cells 2019, 8, 637.
  14. Xiang, H.; Chan, A.G.; Ahene, A.; Bellovin, D.I.; Deng, R.; Hsu, A.W.; Jeffry, U.; Palencia, S.; Powers, J.; Zanghi, J.; et al. Preclinical Characterization of Bemarituzumab, an Anti-FGFR2b Antibody for the Treatment of Cancer. Food Add. Contam. Part A 2021, 13, 1982–1992.
  15. Wainberg, Z.A.; Enzinger, P.C.; Kang, Y.K.; Qin, S.; Yamaguchi, K.; Kim, I.H.; Saeed, A.; Oh, S.C.; Li, J.; Turk, H.M.; et al. Bemarituzumab in Patients with FGFR2b-Selected Gastric or Gastro-Oesophageal Junction Adenocarcinoma (FIGHT): A Randomised, Double-Blind, Placebo-Controlled, Phase 2 Study. Lancet Oncol. 2022, 23, 1430–1440.
  16. Logan, C.Y.; Nusse, R. The Wnt Signaling Pathway In Development And Disease. Annu. Rev. Cell Dev. Biol. 2004, 20, 781–810.
  17. MacDonald, B.T.; Tamai, K.; He, X. Wnt/β-Catenin Signaling: Components, Mechanisms, and Diseases. Dev. Cell 2009, 17, 9–26.
  18. Barker, N.; Clevers, H. Mining the Wnt Pathway for Cancer Therapeutics. Nature Rev. Drug. Discov. 2006, 5, 997–1014.
  19. Krause, U.; Ryan, D.M.; Clough, B.H.; Gregory, C.A. An Unexpected Role for a Wnt-Inhibitor: Dickkopf-1 Triggers a Novel Cancer Survival Mechanism through Modulation of Aldehyde-Dehydrogenase-1 Activity. Cell Death Dis. 2014, 5, e1093.
  20. Smadja, D.M.; D’Audigier, C.; Weiswald, L.B.; Badoual, C.; Dangles-Marie, V.; Mauge, L.; Evrard, S.; Laurendeau, I.; Lallemand, F.; Germain, S.; et al. The Wnt Antagonist Dickkopf-1 Increases Endothelial Progenitor Cell Angiogenic Potential. Arterioscler. Thromb. Vasc. Biol. 2010, 30, 2544–2552.
  21. Malladi, S.; MacAlinao, D.G.; Jin, X.; He, L.; Basnet, H.; Zou, Y.; De Stanchina, E.; Massagué, J. Metastatic Latency and Immune Evasion through Autocrine Inhibition of WNT. Cell 2016, 165, 45–60.
  22. Klempner, S.J.; Chao, J.; Uronis, H.E.; Sirard, C.A.; Kagey, M.; Baum, J.; Song, J.; Wang, J.; Kim, I.-H.; Lee, K.W.; et al. DKN-01 and Tislelizumab ± Chemotherapy as a First-Line (1L) and Second-Line (2L) Investigational Therapy in Advanced Gastroesophageal Adenocarcinoma (GEA): DisTinGuish Trial. J. Clin. Oncol. 2022, 40, 292.
  23. Klempner, S.; Chao, J.; Uronis, H.; Sirard, C.; Kagey, M.; Baum, J.; Song, J.; Wang, J.; Sonbol, M.; Wainberg, Z.; et al. DKN-01 and Tislelizumab + Chemotherapy as First-Line (1L) Investigational Therapy in Advanced Gastroesophageal Adenocarcinoma (GEA): DisTinGuish Trial. Ann. Oncol. 2022, 33, S555–S580.
  24. Nishida, N.; Rodríguez, M. Role of Oncogenic Pathways on the Cancer Immunosuppressive Microenvironment and Its Clinical Implications in Hepatocellular Carcinoma. Cancers 2021, 13, 3666.
  25. Kato, Y.; Tabata, K.; Kimura, T.; Yachie-Kinoshita, A.; Ozawa, Y.; Yamada, K.; Ito, J.; Tachino, S.; Hori, Y.; Matsuki, M.; et al. Lenvatinib plus Anti-PD-1 Antibody Combination Treatment Activates CD8+ T Cells through Reduction of Tumor-Associated Macrophage and Activation of the Interferon Pathway. PLoS ONE 2019, 14, e0212513.
  26. Kwilas, A.R.; Donahue, R.N.; Tsang, K.Y.; Hodge, J.W. Immune Consequences of Tyrosine Kinase Inhibitors That Synergize with Cancer Immunotherapy. Cancer Cell Microenviron. 2015, 2, e677.
  27. Kawazoe, A.; Fukuoka, S.; Nakamura, Y.; Kuboki, Y.; Wakabayashi, M.; Nomura, S.; Mikamoto, Y.; Shima, H.; Fujishiro, N.; Higuchi, T.; et al. Lenvatinib plus Pembrolizumab in Patients with Advanced Gastric Cancer in the First-Line or Second-Line Setting (EPOC1706): An Open-Label, Single-Arm, Phase 2 Trial. Lancet Oncol. 2020, 21, 1057–1065.
  28. Kim, S.T.; Cristescu, R.; Bass, A.J.; Kim, K.M.; Odegaard, J.I.; Kim, K.; Liu, X.Q.; Sher, X.; Jung, H.; Lee, M.; et al. Comprehensive Molecular Characterization of Clinical Responses to PD-1 Inhibition in Metastatic Gastric Cancer. Nature Med. 2018, 24, 1449–1458.
  29. Xie, T.; Liu, Y.; Zhang, Z.; Zhang, X.; Gong, J.; Qi, C.; Li, J.; Shen, L.; Peng, Z. Positive Status of Epstein-Barr Virus as a Biomarker for Gastric Cancer Immunotherapy: A Prospective Observational Study. J. Immunother. 2020, 43, 139–144.
More
Information
Subjects: Oncology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , ,
View Times: 215
Entry Collection: Gastrointestinal Disease
Revisions: 2 times (View History)
Update Date: 22 May 2023
1000/1000