Tissue Biomarkers in Gastric Cancer Treatment: Present and Future
Abstract
:1. Introduction
- (a)
- Diffuse type (poor cohesion between neoplastic cells, variable component of cells showing signet ring cell morphology);
- (b)
- Intestinal type (composed of tubular or glandular structures similar to intestinal adenocarcinoma);
- (c)
- Mixed;
- (d)
- Undifferentiated.
- (a)
- Adenocarcinoma NOS (tubular, parietal, papillary, micropapillary, mucoepidermoid, mucinous, signet ring, poorly cohesive, medullary, hepatoid, Paneth cell);
- (b)
- Squamous cell carcinoma;
- (c)
- Adenosquamous carcinoma;
- (d)
- Undifferentiated carcinoma (large cell, pleomorphic, sarcomatoid);
- (e)
- Gastroblastoma;
- (f)
- Neuroendocrine tumor.
2. Tissue Biomarkers in Standard Treatments
2.1. Programmed Cell Death Ligand 1
2.2. Human Epidermal Growth Factor Receptor 2 (HER2)
2.3. Microsatellite Instability (MSI)
2.4. Neurotrophic Tyrosine Receptor Kinases (NTRKs)
3. Future Perspectives
3.1. Epstein–Barr Virus
3.2. Claudin 18.2
3.3. Tumor Mutational Burden (TMB)
3.4. BRAF Mutations
3.5. RET
3.6. MET
3.7. FGFR2
4. Role of Liquid Biopsy
5. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Ferlay, J.; Ervik, M.; Lam, F.; Laversanne, M.; Colombet, M.; Mery, L.; Piñeros, M.; Znaor, A.; Soerjomataram, I.; Bray, F. Global Cancer Observatory: Cancer Today (Version 1.1); International Agency for Research on Cancer: Lyon, France, 2024; Available online: https://gco.iarc.who.int/today (accessed on 3 March 2024).
- Colquhoun, A.; Arnold, M.; Ferlay, J.; Goodman, K.J.; Forman, D.; Soerjomataram, I. Global patterns of cardia and non-cardia gastric cancer incidence in 2012. Gut 2015, 64, 1881–1888. [Google Scholar] [CrossRef] [PubMed]
- Allum, W.; Lordick, F.; Alsina, M.; Andritsch, E.; Ba-Ssalamah, A.; Beishon, M.; Braga, M.; Caballero, C.; Carneiro, F.; Cassinello, F.; et al. ECCO essential requirements for quality cancer care: Oesophageal and gastric cancer. Crit. Rev. Oncol. Hematol. 2018, 122, 179–193. [Google Scholar] [CrossRef] [PubMed]
- Stomach Cancer Survival Rates and Statistics—NCI. Available online: https://www.cancer.gov/types/stomach/survival (accessed on 5 March 2024).
- Heidelberger, C.; Chaudhuri, N.K.; Danneberg, P.; Mooren, D.; Griesbach, L.; Duschinsky, R.; Schnitzer, R.J.; Pleven, E.; Scheiner, J. Fluorinated Pyrimidines, A New Class of Tumour-Inhibitory Compounds. Nature 1957, 179, 663–666. [Google Scholar] [CrossRef] [PubMed]
- Cunningham, D.; Allum, W.H.; Stenning, S.P.; Thompson, J.N.; Van de Velde, C.J.H.; Nicolson, M.; Scarffe, J.H.; Lofts, F.J.; Falk, S.J.; Iveson, T.J.; et al. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N. Engl. J. Med. 2006, 355, 11–20. [Google Scholar] [CrossRef] [PubMed]
- Ychou, M.; Boige, V.; Pignon, J.-P.; Conroy, T.; Bouché, O.; Lebreton, G.; Ducourtieux, M.; Bedenne, L.; Fabre, J.-M.; Saint-Aubert, B.; et al. Perioperative chemotherapy compared with surgery alone for resectable gastroesophageal adenocarcinoma: An FNCLCC and FFCD multicenter phase III trial. J. Clin. Oncol. 2011, 29, 1715–1721. [Google Scholar] [CrossRef]
- Reynolds, J.V.; Preston, S.R.; O’Neill, B.; Lowery, M.A.; Baeksgaard, L.; Crosby, T.; Cunningham, M.; Cuffe, S.; Griffiths, G.O.; Parker, I.; et al. Trimodality therapy versus perioperative chemotherapy in the management of locally advanced adenocarcinoma of the oesophagus and oesophagogastric junction (Neo-AEGIS): An open-label, randomised, phase 3 trial. Lancet Gastroenterol. Hepatol. 2023, 8, 1015–1027. [Google Scholar] [CrossRef]
- Cunningham, D.; Starling, N.; Rao, S.; Iveson, T.; Nicolson, M.; Coxon, F.; Middleton, G.; Daniel, F.; Oates, J.; Norman, A.R. Capecitabine and Oxaliplatin for Advanced Esophagogastric Cancer. N. Engl. J. Med. 2008, 358, 36–46. [Google Scholar] [CrossRef]
- Al-Batran, S.-E.; Hartmann, J.T.; Probst, S.; Schmalenberg, H.; Hollerbach, S.; Hofheinz, R.; Rethwisch, V.; Seipelt, G.; Homann, N.; Wilhelm, G.; et al. Phase III Trial in Metastatic Gastroesophageal Adenocarcinoma with Fluorouracil, Leucovorin Plus Either Oxaliplatin or Cisplatin: A Study of the Arbeitsgemeinschaft Internistische Onkologie. J. Clin. Oncol. 2008, 26, 1435–1442. [Google Scholar] [CrossRef]
- Dank, M.; Zaluski, J.; Barone, C.; Valvere, V.; Yalcin, S.; Peschel, C.; Wenczl, M.; Goker, E.; Cisar, L.; Wang, K.; et al. Randomized phase III study comparing irinotecan combined with 5-fluorouracil and folinic acid to cisplatin combined with 5-fluorouracil in chemotherapy naive patients with advanced adenocarcinoma of the stomach or esophagogastric junction. Ann. Oncol. 2008, 19, 1450–1457. [Google Scholar] [CrossRef]
- Guimbaud, R.; Louvet, C.; Ries, P.; Ychou, M.; Maillard, E.; André, T.; Gornet, J.-M.; Aparicio, T.; Nguyen, S.; Azzedine, A.; et al. Prospective, Randomized, Multicenter, Phase III Study of Fluorouracil, Leucovorin, and Irinotecan Versus Epirubicin, Cisplatin, and Capecitabine in Advanced Gastric Adenocarcinoma: A French Intergroup (Fédération Francophone de Cancérologie Digestive, Féd. J. Clin. Oncol. 2014, 32, 3520–3526. [Google Scholar] [CrossRef]
- Janjigian, Y.Y.; Shitara, K.; Moehler, M.; Garrido, M.; Salman, P.; Shen, L.; Wyrwicz, L.; Yamaguchi, K.; Skoczylas, T.; Campos Bragagnoli, A.; 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. [Google Scholar] [CrossRef]
- Shitara, K.; Ajani, J.A.; Moehler, M.; Garrido, M.; Gallardo, C.; Shen, L.; Yamaguchi, K.; Wyrwicz, L.; Skoczylas, T.; Bragagnoli, A.C.; et al. Nivolumab plus chemotherapy or ipilimumab in gastro-oesophageal cancer. Nature 2022, 603, 942–948. [Google Scholar] [CrossRef] [PubMed]
- Smyth, E.C.; Nilsson, M.; Grabsch, H.I.; van Grieken, N.C.; Lordick, F. Gastric cancer. Lancet 2020, 396, 635–648. [Google Scholar] [CrossRef] [PubMed]
- Fuchs, C.S.; Tomasek, J.; Yong, C.J.; Dumitru, F.; Passalacqua, R.; Goswami, C.; Safran, H.; dos Santos, L.V.; Aprile, G.; Ferry, D.R.; et al. Ramucirumab monotherapy for previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (REGARD): An international, randomised, multicentre, placebo-controlled, phase 3 trial. Lancet 2014, 383, 31–39. [Google Scholar] [CrossRef] [PubMed]
- Wilke, H.; Muro, K.; Van Cutsem, E.; Oh, S.-C.; Bodoky, G.; Shimada, Y.; Hironaka, S.; Sugimoto, N.; Lipatov, O.; Kim, T.-Y.; 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, 1224–1235. [Google Scholar] [CrossRef]
- Laurén, P. The Two Histological Main Types of Gastric Carcinoma: Diffuse and So-Called Intestinal-Type Carcinoma. Acta Pathol. Microbiol. Scand. 1965, 64, 31–49. [Google Scholar] [CrossRef] [PubMed]
- Fléjou, J.-F. Classification OMS 2010 des tumeurs digestives: La quatrième édition. Ann. Pathol. 2011, 31, S27–S31. [Google Scholar] [CrossRef]
- Mariette, C.; Carneiro, F.; Grabsch, H.I.; van der Post, R.S.; Allum, W.; de Manzoni, G.; European Chapter of International Gastric Cancer Association. Consensus on the pathological definition and classification of poorly cohesive gastric carcinoma. Gastric Cancer 2019, 22, 1–9. [Google Scholar] [CrossRef] [PubMed]
- The Cancer Genome Atlas Research Network; Bass, A.J.; Thorsson, V.; Shmulevich, I.; Reynolds, S.M.; Miller, M.; Bernard, B.; Hinoue, T.; Laird, P.W.; Curtis, C.; et al. Comprehensive molecular characterization of gastric adenocarcinoma. Nature 2014, 513, 202–209. [Google Scholar] [CrossRef]
- Sohn, B.H.; Hwang, J.E.; Jang, H.J.; Lee, H.S.; Oh, S.C.; Shim, J.J.; Lee, K.-W.; Kim, E.H.; Yim, S.Y.; Lee, S.H.; et al. Clinical Significance of Four Molecular Subtypes of Gastric Cancer Identified by The Cancer Genome Atlas Project. Clin. Cancer Res. 2017, 23, 4441–4449. [Google Scholar] [CrossRef]
- Nirschl, C.J.; Drake, C.G. Molecular Pathways: Coexpression of Immune Checkpoint Molecules: Signaling Pathways and Implications for Cancer Immunotherapy. Clin. Cancer Res. 2013, 19, 4917–4924. [Google Scholar] [CrossRef] [PubMed]
- Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [PubMed]
- Kulangara, K.; Zhang, N.; Corigliano, E.; Guerrero, L.; Waldroup, S.; Jaiswal, D.; Jansson, M.; Shah, S.; Hanks, D.; Wang, J.; et al. Clinical utility of the combined positive score for programmed death ligand-1 expression and the approval of pembrolizumab for treatment of gastric cancer. Arch. Pathol. Lab. Med. 2019, 143, 330–337. [Google Scholar] [CrossRef] [PubMed]
- Lordick, F.; Carneiro, F.; Cascinu, S.; Fleitas, T.; Haustermans, K.; Piessen, G.; Vogel, A.; Smyth, E. Gastric cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann. Oncol. 2022, 33, 1005–1020. [Google Scholar] [CrossRef]
- Shah, M.A.; Kennedy, E.B.; Alarcon-Rozas, A.E.; Alcindor, T.; Bartley, A.N.; Malowany, A.B.; Bhadkamkar, N.A.; Deighton, D.C.; Janjigian, Y.; Karippot, A.; et al. Immunotherapy and Targeted Therapy for Advanced Gastroesophageal Cancer: ASCO Guideline. J. Clin. Oncol. 2023, 41, 1470–1491. [Google Scholar] [CrossRef]
- US Food and Drug Administration. List of Cleared or Approved Companion Diagnostic Devices (In Vitro and Imaging Tools). 2020. Available online: https://www.fda.gov/medicalDevices/productsandMedicalProcedures/InVitroDiagnostics/ucm301431.htm (accessed on 3 March 2024).
- Fuchs, C.S.; Doi, T.; Jang, R.W.; Muro, K.; Satoh, T.; Machado, M.; Sun, W.; Jalal, S.I.; Shah, M.A.; Metges, J.-P.; et al. Safety and efficacy of pembrolizumab monotherapy in patients with previously treated advanced gastric and gastroesophageal junction cancer: Phase 2 clinical KEYNOTE-059 trial. JAMA Oncol. 2018, 4, e180013. [Google Scholar] [CrossRef] [PubMed]
- Shitara, K.; Özgüroğlu, M.; Bang, Y.J.; Di Bartolomeo, M.; Mandalà, M.; Ryu, M.H.; Fornaro, L.; Olesiński, T.; Caglevic, C.; Muro, K.; et al. Pembrolizumab versus paclitaxel for previously treated, advanced gastric or gastro-oesophageal junction cancer (KEYNOTE-061): A randomised, open-label, controlled, phase 3 trial. Lancet 2018, 392, 123–133. [Google Scholar] [CrossRef]
- Ahn, S.; Kim, K.M. PD-L1 expression in gastric cancer: Interchangeability of 22C3 and 28-8 pharmDx assays for responses to immunotherapy. Mod. Pathol. 2021, 34, 1719–1727. [Google Scholar] [CrossRef]
- Yeong, J.; Lum, H.Y.J.; Teo, C.B.; Tan, B.K.J.; Chan, Y.H.; Tay, R.Y.K.; Choo, J.R.-E.; Jeyasekharan, A.D.; Miow, Q.H.; Loo, L.-H.; et al. Choice of PD-L1 immunohistochemistry assay influences clinical eligibility for gastric cancer immunotherapy. Gastric Cancer 2022, 25, 741–750. [Google Scholar] [CrossRef]
- Sundar, R.; Liu, D.H.W.; Hutchins, G.G.A.; Slaney, H.L.; Silva, A.N.S.; Oosting, J.; Hayden, J.D.; Hewitt, L.C.; Ng, C.C.; Mangalvedhekar, A.; et al. Spatial profiling of gastric cancer patient-matched primary and locoregional metastases reveals principles of tumour dissemination. Gut 2021, 70, 1823–1832. [Google Scholar] [CrossRef]
- Zhou, K.I.; Peterson, B.; Serritella, A.; Thomas, J.; Reizine, N.; Moya, S.; Tan, C.; Wang, Y.; Catenacci, D.V. Spatial and Temporal Heterogeneity of PD-L1 Expression and Tumor Mutational Burden in Gastroesophageal Adenocarcinoma at Baseline Diagnosis and after Chemotherapy. Clin. Cancer Res. 2020, 26, 6453–6463. [Google Scholar] [CrossRef] [PubMed]
- Yang, J.H.; Kim, H.; Roh, S.Y.; Lee, M.A.; Park, J.M.; Park, C.H.; Lee, H.H.; Jung, E.S.; Lee, S.H.; Lee, Y.J.; et al. Discordancy and changes in the pattern of programmed death ligand 1 expression before and after platinum-based chemotherapy in metastatic gastric cancer. Gastric Cancer 2019, 22, 147–154. [Google Scholar] [CrossRef] [PubMed]
- Wainberg, Z.A.; Fuchs, C.S.; Tabernero, J.; Shitara, K.; Muro, K.; Van Cutsem, E.; Bang, Y.-J.; Chung, H.C.; Yamaguchi, K.; Varga, E.; et al. Efficacy of pembrolizumab monotherapy for advanced gastric/gastroesophageal junction cancer with programmed death ligand 1 combined positive score ≥10. Clin. Cancer Res. 2021, 27, 1923–1931. [Google Scholar] [CrossRef]
- Ye, M.; Huang, D.; Zhang, Q.; Weng, W.; Tan, C.; Qin, G.; Jiang, W.; Sheng, W.; Wang, L. Heterogeneous programmed death-ligand 1 expression in gastric cancer: Comparison of tissue microarrays and whole sections. Cancer Cell Int. 2020, 20, 186. [Google Scholar] [CrossRef]
- Slamon, D.J.; Godolphin, W.; Jones, L.A.; Holt, J.A.; Wong, S.G.; Keith, D.E.; Levin, W.J.; Stuart, S.G.; Udove, J.; Ullrich, A.; et al. Studies of the HER-2/ neu Proto-Oncogene in Human Breast and Ovarian Cancer. Science 1989, 244, 707–712. [Google Scholar] [CrossRef]
- Kaptain, S.; Tan, L.K.; Chen, B. Her-2/ neu and Breast Cancer. Diagn. Mol. Pathol. 2001, 10, 139–152. [Google Scholar] [CrossRef] [PubMed]
- Tewari, M.; Kumar, A.; Mishra, R.; Kumar, M.; Shukla, H.S. HER2 Expression in Gastric and Gastroesophageal Cancer: Report from a Tertiary Care Hospital in North India. Indian J. Surg. 2015, 77, 447–451. [Google Scholar] [CrossRef]
- Polkowski, W.; van Sandick, J.W.; Offerhaus, G.J.A.; ten Kate, F.J.W.; Mulder, J.; Obertop, H.; van Lanschot, J.J.B. Prognostic Value of Laurén Classification and c-erbB-2 Oncogene Overexpression in Adenocarcinoma of the Esophagus and Gastroesophageal Junction. Ann. Surg. Oncol. 1999, 6, 290–297. [Google Scholar] [CrossRef] [PubMed]
- García, I.; Vizoso, F.; Martín, A.; Sanz, L.; Abdel-Lah, O.; Raigoso, P.; García-Muñiz, J.L. Clinical Significance of the Epidermal Growth Factor Receptor and HER2 Receptor in Resectable Gastric Cancer. Ann. Surg. Oncol. 2003, 10, 234–241. [Google Scholar] [CrossRef]
- Jonjić, N.; Kovac, K.; Krasević, M.; Valković, T.; Ernjak, N.; Sasso, F.; Melato, M. Epidermal growth factor-receptor expression correlates with tumor cell proliferation and prognosis in gastric cancer. Anticancer Res. 1997, 17, 3883–3888. [Google Scholar]
- Son, H.S.; Shin, Y.M.; Park, K.K.; Seo, K.W.; Yoon, K.Y.; Jang, H.K.; Lee, S.-H.; Yang, S.I.; Kim, J.H. Correlation between HER2 Overexpression and Clinicopathological Characteristics in Gastric Cancer Patients Who Have Undergone Curative Resection. J. Gastric Cancer 2014, 14, 180–186. [Google Scholar] [CrossRef] [PubMed]
- Takehana, T.; Kunitomo, K.; Kono, K.; Kitahara, F.; Iizuka, H.; Matsumoto, Y.; Fujino, M.A.; Ooi, A. Status of c- erb B-2 in gastric adenocarcinoma: A comparative study of immunohistochemistry, fluorescence in situ hybridization and enzyme-linked immuno-sorbent assay. Int. J. Cancer 2002, 98, 833–837. [Google Scholar] [CrossRef] [PubMed]
- Hofmann, M.; Stoss, O.; Shi, D.; Büttner, R.; Van De Vijver, M.; Kim, W.; Ochiai, A.; Rüschoff, J.; Henkel, T. Assessment of a HER2 scoring system for gastric cancer: Results from a validation study. Histopathology 2008, 52, 797–805. [Google Scholar] [CrossRef]
- Bang, Y.-J.; Van Cutsem, E.; Feyereislova, A.; Chung, H.C.; Shen, L.; Sawaki, A.; Lordick, F.; Ohtsu, A.; Omuro, Y.; Satoh, T.; 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, 687–697. [Google Scholar] [CrossRef]
- Rha, S.Y.; Lee, C.-K.; Kim, H.S.; Kang, B.; Jung, M.; Bae, W.K.; Koo, D.-H.; Shin, S.-J.; Jeung, H.-C.; Zang, D.Y.; et al. Targeting HER2 in combination with anti-PD-1 and chemotherapy confers a significant tumor shrinkage of gastric cancer: A multi-institutional phase Ib/II trial of first-line triplet regimen (pembrolizumab, trastuzumab, chemotherapy) for HER2-positive advanced gastric cancer (AGC). J. Clin. Oncol. 2020, 38, 3081. [Google Scholar] [CrossRef]
- Janjigian, Y.Y.; Maron, S.B.; Chatila, W.K.; Millang, B.; Chavan, S.S.; Alterman, C.; Chou, J.F.; Segal, M.F.; Simmons, M.Z.; Momtaz, P.; et al. First-line pembrolizumab and trastuzumab in HER2-positive oesophageal, gastric, or gastro-oesophageal junction cancer: An open-label, single-arm, phase 2 trial. Lancet Oncol. 2020, 21, 821–831. [Google Scholar] [CrossRef]
- Janjigian, Y.Y.; Kawazoe, A.; Bai, Y.; Xu, J.; Lonardi, S.; Metges, J.P.; Yanez, P.; Wyrwicz, L.S.; Shen, L.; Ostapenko, Y.; et al. Pembrolizumab plus trastuzumab and chemotherapy for HER2-positive gastric or gastro-oesophageal junction adenocarcinoma: Interim analyses from the phase 3 KEYNOTE-811 randomised placebo-controlled trial. Lancet 2023, 402, 2197–2208. [Google Scholar] [CrossRef]
- Elimova, E.; Ajani, J.A.; Iii, H.A.B.; Denlinger, C.S.; Iqbal, S.; Kang, Y.-K.; Kim, Y.H.H.; Lee, K.-W.; Lin, B.; Mehta, R.; et al. Zanidatamab + chemotherapy as first-line treatment for HER2-expressing metastatic gastroesophageal adenocarcinoma (mGEA). J. Clin. Oncol. 2023, 41, 347. [Google Scholar] [CrossRef]
- Catenacci, D.V.T.; Kang, Y.-K.; Park, H.; Uronis, H.E.; Lee, K.-W.; Ng, M.C.H.; Enzinger, P.C.; Park, S.H.; Gold, P.J.; Lacy, J.; et al. Margetuximab plus pembrolizumab in patients with previously treated, HER2-positive gastro-oesophageal adenocarcinoma (CP-MGAH22–05): A single-arm, phase 1b–2 trial. Lancet Oncol. 2020, 21, 1066–1076. [Google Scholar] [CrossRef]
- Catenacci, D.; Kang, Y.-K.; Yoon, H.; Shim, B.; Kim, S.; Oh, D.-Y.; Spira, A.; Ulahannan, S.; Avery, E.; Boland, P.; et al. Margetuximab with retifanlimab as first-line therapy in HER2+/PD-L1+ unresectable or metastatic gastroesophageal adenocarcinoma: MAHOGANY cohort A. ESMO Open 2022, 7, 100563. [Google Scholar] [CrossRef]
- Satoh, T.; Xu, R.-H.; Chung, H.C.; Sun, G.-P.; Doi, T.; Xu, J.-M.; Tsuji, A.; Omuro, Y.; Li, J.; Wang, J.-W.; et al. Lapatinib Plus Paclitaxel Versus Paclitaxel Alone in the Second-Line Treatment of HER2-Amplified Advanced Gastric Cancer in Asian Populations: TyTAN—A Randomized, Phase III Study. J. Clin. Oncol. 2014, 32, 2039–2049. [Google Scholar] [CrossRef] [PubMed]
- Tabernero, J.; Hoff, P.M.; Shen, L.; Ohtsu, A.; A Shah, M.; Cheng, K.; Song, C.; Wu, H.; Eng-Wong, J.; Kim, K.; et al. Pertuzumab plus trastuzumab and chemotherapy for HER2-positive metastatic gastric or gastro-oesophageal junction cancer (JACOB): Final analysis of a double-blind, randomised, placebo-controlled phase 3 study. Lancet Oncol. 2018, 19, 1372–1384. [Google Scholar] [CrossRef] [PubMed]
- Kang, Y.-K.; Shah, M.A.; Ohtsu, A.; Van Cutsem, E.; Ajani, J.A.; van der Horst, T.; Harle-Yge, M.-L.; Piao, Y.; Althaus, B.; Thuss-Patience, P.C. A randomized, open-label, multicenter, adaptive phase 2/3 study of trastuzumab emtansine (T-DM1) versus a taxane (TAX) in patients (pts) with previously treated HER2-positive locally advanced or metastatic gastric/gastroesophageal junction adenocarcinoma (LA/MGC/GEJC). J. Clin. Oncol. 2016, 34, 5. [Google Scholar] [CrossRef]
- Stahl, P.; Seeschaaf, C.; Lebok, P.; Kutup, A.; Bockhorn, M.; Izbicki, J.R.; Bokemeyer, C.; Simon, R.; Sauter, G.; Marx, A.H. Heterogeneity of amplification of HER2, EGFR, CCND1 and MYC in gastric cancer. BMC Gastroenterol. 2015, 15, 7. [Google Scholar] [CrossRef] [PubMed]
- Seo, S.; Ryu, M.-H.; Park, Y.S.; Ahn, J.Y.; Park, Y.; Park, S.R.; Ryoo, B.-Y.; Lee, G.H.; Jung, H.-Y.; Kang, Y.-K. Loss of HER2 positivity after anti-HER2 chemotherapy in HER2-positive gastric cancer patients: Results of the GASTric cancer HER2 reassessment study 3 (GASTHER3). Gastric Cancer 2019, 22, 527–535. [Google Scholar] [CrossRef]
- Zhang, J.; Qiu, W.; Zhang, W.; Chen, Y.; Shen, H.; Zhu, H.; Liang, X.; Shen, Z. Tracking of trastuzumab resistance in patients with HER2-positive metastatic gastric cancer by CTC liquid biopsy. Am. J. Cancer Res. 2023, 13, 5684–5697. [Google Scholar]
- Klein-Scory, S.; Ladigan-Badura, S.; Mika, T.; Verdoodt, B.; Tannapfel, A.; Pohl, M.; Schroers, R.; Baraniskin, A. Liquid biopsy based HER2 amplification status in gastric cancer patients indicates clinical response. Heliyon 2023, 9, e21339. [Google Scholar] [CrossRef]
- Makiyama, A.; Sukawa, Y.; Kashiwada, T.; Kawada, J.; Hosokawa, A.; Horie, Y.; Tsuji, A.; Moriwaki, T.; Tanioka, H.; Shinozaki, K.; et al. Randomized, Phase II Study of Trastuzumab Beyond Progression in Patients with HER2-Positive Advanced Gastric or Gastroesophageal Junction Cancer: WJOG7112G (T-ACT Study). J. Clin. Oncol. 2020, 38, 1919–1927. [Google Scholar] [CrossRef]
- Takegawa, N.; Nonagase, Y.; Yonesaka, K.; Sakai, K.; Maenishi, O.; Ogitani, Y.; Tamura, T.; Nishio, K.; Nakagawa, K.; Tsurutani, J. DS-8201a, a new HER2-targeting antibody–drug conjugate incorporating a novel DNA topoisomerase I inhibitor, overcomes HER2-positive gastric cancer T-DM1 resistance. Int. J. Cancer 2017, 141, 1682–1689. [Google Scholar] [CrossRef]
- Van Cutsem, E.; di Bartolomeo, M.; Smyth, E.; Chau, I.; Park, H.; Siena, S.; Lonardi, S.; A Wainberg, Z.; Ajani, J.; Chao, J.; et al. Trastuzumab deruxtecan in patients in the USA and Europe with HER2-positive advanced gastric or gastroesophageal junction cancer with disease progression on or after a trastuzumab-containing regimen (DESTINY-Gastric02): Primary and updated analyses from a single-arm, phase 2 study. Lancet Oncol. 2023, 24, 744–756. [Google Scholar] [CrossRef]
- Shitara, K.; Bang, Y.-J.; Iwasa, S.; Sugimoto, N.; Ryu, M.-H.; Sakai, D.; Chung, H.-C.; Kawakami, H.; Yabusaki, H.; Lee, J.; et al. Trastuzumab Deruxtecan in Previously Treated HER2-Positive Gastric Cancer. N. Engl. J. Med. 2020, 382, 2419–2430. [Google Scholar] [CrossRef] [PubMed]
- Shitara, K.; Seraj, J.; Franke, F.; Kawaguchi, Y.; Shen, L.; Kamio, T.; Meinhardt, G.; Tabernero, J. 1436TiP Trastuzumab deruxtecan (T-DXd) in patients (Pts) with HER2-positive gastric cancer (GC) or gastroesophageal junction (GEJ) adenocarcinoma who have progressed on or after a trastuzumab-containing regimen (DESTINY-gastric04, DG-04): A randomized phase III study. Ann. Oncol. 2021, 32, S1073. [Google Scholar] [CrossRef]
- Wang, Y.; Gong, J.; Wang, A.; Wei, J.; Peng, Z.; Wang, X.; Zhou, J.; Qi, C.; Liu, D.; Li, J.; et al. Disitamab vedotin (RC48) plus toripalimab for HER2-expressing advanced gastric or gastroesophageal junction and other solid tumours: A multicentre, open label, dose escalation and expansion phase 1 trial. eClinicalMedicine 2024, 68, 102415. [Google Scholar] [CrossRef]
- Holliday, R. A mechanism for gene conversion in fungi. Genet. Res. 2007, 89, 285–307. [Google Scholar] [CrossRef] [PubMed]
- Tamura, K.; Kaneda, M.; Futagawa, M.; Takeshita, M.; Kim, S.; Nakama, M.; Kawashita, N.; Tatsumi-Miyajima, J. Genetic and genomic basis of the mismatch repair system involved in Lynch syndrome. Int. J. Clin. Oncol. 2019, 24, 999–1011. [Google Scholar] [CrossRef] [PubMed]
- Schöniger, S.; Rüschoff, J. Mismatch Repair Deficiency and Microsatellite Instability. Encyclopedia 2022, 2, 1559–1576. [Google Scholar] [CrossRef]
- Bateman, A.C. DNA mismatch repair protein immunohistochemistry—An illustrated guide. Histopathology 2021, 79, 128–138. [Google Scholar] [CrossRef]
- Bartley, A.N.; Mills, A.M.; Konnick, E.; Overman, M.; Ventura, C.B.; Souter, L.; Colasacco, C.; Stadler, Z.K.; Kerr, S.; Howitt, B.E.; et al. Mismatch Repair and Microsatellite Instability Testing for Immune Checkpoint Inhibitor Therapy: Guideline From the College of American Pathologists in Collaboration With the Association for Molecular Pathology and Fight Colorectal Cancer. Arch. Pathol. Lab. Med. 2022, 146, 1194–1210. [Google Scholar] [CrossRef]
- Yu, H.Y.; Li, C.P.; Huang, Y.H.; Hsu, S.J.; Wang, Y.P.; Hsieh, Y.C.; Fang, W.L.; Huang, K.H.; Li, A.F.Y.; Lee, R.C.; et al. Microsatellite Instability, Epstein–Barr Virus, and Programmed Cell Death Ligand 1 as Predictive Markers for Immunotherapy in Gastric Cancer. Cancers 2022, 14, 218. [Google Scholar] [CrossRef]
- Tabernero, J.; Van Cutsem, E.; Bang, Y.-J.; Fuchs, C.S.; Wyrwicz, L.; Lee, K.W.; Kudaba, I.; Garrido, M.; Chung, H.C.; Salguero, H.R.C.; et al. Pembrolizumab with or without chemotherapy versus chemotherapy for advanced gastric or gastroesophageal junction (G/GEJ) adenocarcinoma: The phase III KEYNOTE-062 study. J. Clin. Oncol. 2019, 37, LBA4007. [Google Scholar] [CrossRef]
- Yoshino, T.; Pentheroudakis, G.; Mishima, S.; Overman, M.; Yeh, K.-H.; Baba, E.; Naito, Y.; Calvo, F.; Saxena, A.; Chen, L.-T.; et al. JSCO—ESMO—ASCO—JSMO—TOS: International expert consensus recommendations for tumour-agnostic treatments in patients with solid tumours with microsatellite instability or NTRK fusions. Ann. Oncol. 2020, 31, 861–872. [Google Scholar] [CrossRef] [PubMed]
- Pietrantonio, F.; Randon, G.; Di Bartolomeo, M.; Luciani, A.; Chao, J.; Smyth, E.; Petrelli, F. 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. [Google Scholar] [CrossRef]
- Marabelle, A.; Le, D.T.; Ascierto, P.A.; Di Giacomo, A.M.; De Jesus-Acosta, A.; Delord, J.-P.; Geva, R.; Gottfried, M.; Penel, N.; Hansen, A.R.; et al. Efficacy of Pembrolizumab in Patients with Noncolorectal High Microsatellite Instability/Mismatch Repair–Deficient Cancer: Results From the Phase II KEYNOTE-158 Study. J. Clin. Oncol. 2020, 38, 1–10. [Google Scholar] [CrossRef]
- Wang, Y.-L.; Gong, Y.; Lv, Z.; Li, L.; Yuan, Y. Expression of PD1/PDL1 in gastric cancer at different microsatellite status and its correlation with infiltrating immune cells in the tumor microenvironment. J. Cancer 2021, 12, 1698–1707. [Google Scholar] [CrossRef]
- De La Fouchardiere, C.; Zaanan, A.; Cohen, R.; Le Sourd, S.; Tougeron, D.; Soularue, E.; Dubreuil, O.; Willet, N.; Samalin, E.; Piessen, G.; et al. Immunotherapy for localized dMMR/MSI tumors: First interim analysis of the IMHOTEP trial. J. Clin. Oncol. 2023, 41, 2591. [Google Scholar] [CrossRef]
- André, T.; Tougeron, D.; Piessen, G.; de la Fouchardière, C.; Louvet, C.; Adenis, A.; Jary, M.; Tournigand, C.; Aparicio, T.; Desrame, J.; et al. Neoadjuvant Nivolumab Plus Ipilimumab and Adjuvant Nivolumab in Localized Deficient Mismatch Repair/Microsatellite Instability–High Gastric or Esophagogastric Junction Adenocarcinoma: The GERCOR NEONIPIGA Phase II Study. J. Clin. Oncol. 2023, 41, 255–265. [Google Scholar] [CrossRef]
- Pietrantonio, F.; Raimondi, A.; Lonardi, S.; Murgioni, S.; Cardellino, G.G.; Tamberi, S.; Strippoli, A.; Palermo, F.; Prisciandaro, M.; Randon, G.; et al. INFINITY: A multicentre, single-arm, multi-cohort, phase II trial of tremelimumab and durvalumab as neoadjuvant treatment of patients with microsatellite instability-high (MSI) resectable gastric or gastroesophageal junction adenocarcinoma (GAC/GEJAC). J. Clin. Oncol. 2023, 41, 358. [Google Scholar] [CrossRef]
- Manea, C.A.; Badiu, D.C.; Ploscaru, I.C.; Zgura, A.; Bacinschi, X.; Smarandache, C.G.; Serban, D.; Popescu, C.G.; Grigorean, V.T.; Botnarciuc, V. A review of NTRK fusions in cancer. Ann. Med. Surg. 2022, 79, 103893. [Google Scholar] [CrossRef]
- Cocco, E.; Scaltriti, M.; Drilon, A. NTRK fusion-positive cancers and TRK inhibitor therapy. Nat. Rev. Clin. Oncol. 2018, 15, 731–747. [Google Scholar] [CrossRef]
- Huygens, S.; Vellekoop, H.; Versteegh, M.; Santi, I.; Szilberhorn, L.; Zelei, T.; Nagy, B.; Tsiachristas, A.; Koleva-Kolarova, R.; Wordsworth, S.; et al. Cost-Effectiveness Analysis of Treating Patients With NTRK-Positive Cancer with the Histology-Independent Therapy Entrectinib. Value Health 2023, 26, 193–203. [Google Scholar] [CrossRef]
- Xu, Y.; Shi, X.; Wang, W.; Zhang, L.; Cheung, S.; Rudolph, M.; Brega, N.; Dong, X.; Qian, L.; Wang, L.; et al. Prevalence and clinico-genomic characteristics of patients with TRK fusion cancer in China. NPJ Precis. Oncol. 2023, 7, 75. [Google Scholar] [CrossRef] [PubMed]
- Drilon, A.; Laetsch, T.W.; Kummar, S.; Dubois, S.G.; Lassen, U.N.; Demetri, G.D.; Nathenson, M.; Doebele, R.C.; Farago, A.F.; Pappo, A.S.; et al. Efficacy of Larotrectinib in TRK Fusion–Positive Cancers in Adults and Children. N. Engl. J. Med. 2018, 378, 731–739. [Google Scholar] [CrossRef] [PubMed]
- Solomon, B.; Drilon, A.; Lin, J.; Bazhenova, L.; Goto, K.; De Langen, J.; Kim, D.-W.; Wolf, J.; Springfeld, C.; Popat, S.; et al. 1372P Repotrectinib in patients (pts) with NTRK fusion-positive (NTRK+) advanced solid tumors, including NSCLC: Update from the phase I/II TRIDENT-1 trial. Ann. Oncol. 2023, 34, S787–S788. [Google Scholar] [CrossRef]
- U.S. Food & Drug. FDA Grants Accelerated Approval to Repotrectinib for Adult and Pediatric Patients with NTRK Gene Fusion-Positive Solid Tumors. 13 June 2024. Available online: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-repotrectinib-adult-and-pediatric-patients-ntrk-gene-fusion-positive (accessed on 4 July 2024).
- Drilon, A.; Siena, S.; Ou, S.-H.I.; Patel, M.; Ahn, M.J.; Lee, J.; Bauer, T.M.; Farago, A.F.; Wheler, J.J.; Liu, S.V.; et al. Safety and Antitumor Activity of the Multitargeted Pan-TRK, ROS1, and ALK Inhibitor Entrectinib: Combined Results from Two Phase I Trials (ALKA-372-001 and STARTRK-1). Cancer Discov. 2017, 7, 400–409. [Google Scholar] [CrossRef]
- Doebele, R.C.; Drilon, A.; Paz-Ares, L.; Siena, S.; Shaw, A.T.; Farago, A.F.; Blakely, C.M.; Seto, T.; Cho, B.C.; Tosi, D.; et al. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: Integrated analysis of three phase 1–2 trials. Lancet Oncol. 2020, 21, 271–282. [Google Scholar] [CrossRef]
- Patel, M.; Siena, S.; Demetri, G.; Doebele, R.; Chae, Y.; Conkling, P.; Garrido-Laguna, I.; Longo, F.; Rolfo, C.; Sigal, D.; et al. O-3 Efficacy and safety of entrectinib in NTRK fusion-positive gastrointestinal cancers: Updated integrated analysis of three clinical trials (STARTRK-2, STARTRK-1 and ALKA-372-001). Ann. Oncol. 2020, 31, 232–233. [Google Scholar] [CrossRef]
- Sun, K.; Jia, K.; Lv, H.; Wang, S.-Q.; Wu, Y.; Lei, H.; Chen, X. EBV-Positive Gastric Cancer: Current Knowledge and Future Perspectives. Front. Oncol. 2020, 10, 583463. [Google Scholar] [CrossRef] [PubMed]
- Murphy, G.; Pfeiffer, R.; Camargo, M.C.; Rabkin, C.S. Meta-analysis shows that prevalence of Epstein–Barr virus-positive gastric cancer differs based on sex and anatomic location. Gastroenterology 2009, 137, 824–833. [Google Scholar] [CrossRef]
- Derks, S.; Liao, X.; Chiaravalli, A.M.; Xu, X.; Camargo, M.C.; Solcia, E.; Sessa, F.; Fleitas, T.; Freeman, G.J.; Rodig, S.J.; et al. Abundant PD-L1 expression in Epstein-Barr Virus-infected gastric cancers. Oncotarget 2016, 7, 32925–32932. [Google Scholar] [CrossRef]
- Bai, Y.; Xie, T.; Wang, Z.; Tong, S.; Zhao, X.; Zhao, F.; Cai, J.; Wei, X.; Peng, Z.; Shen, L. Efficacy and predictive biomarkers of immunotherapy in Epstein-Barr virus-associated gastric cancer. J. Immunother. Cancer 2022, 10, e004080. [Google Scholar] [CrossRef]
- Kim, S.Y.; Park, C.; Kim, H.-J.; Park, J.; Hwang, J.; Kim, J.-I.; Choi, M.G.; Kim, K.-M.; Kang, M.-S. Deregulation of immune response genes in patients with Epstein-Barr virus-associated gastric cancer and outcomes. Gastroenterology 2015, 148, 137–147.e9. [Google Scholar] [CrossRef] [PubMed]
- Kang, Y.-K.; Chen, L.-T.; Ryu, M.-H.; Oh, D.-Y.; Oh, S.C.; Chung, H.C.; Lee, K.-W.; Omori, T.; Shitara, K.; Sakuramoto, S.; 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, 234–247. [Google Scholar] [CrossRef] [PubMed]
- 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. Nat. Med. 2018, 24, 1449–1458. [Google Scholar] [CrossRef] [PubMed]
- Tabariès, S.; Siegel, P.M. The role of claudins in cancer metastasis. Oncogene 2016, 36, 1176–1190. [Google Scholar] [CrossRef] [PubMed]
- Niimi, T.; Nagashima, K.; Ward, J.M.; Minoo, P.; Zimonjic, D.B.; Popescu, N.C.; Kimura, S. claudin-18, a Novel Downstream Target Gene for the T/EBP/NKX2.1 Homeodomain Transcription Factor, Encodes Lung- and Stomach-Specific Isoforms through Alternative Splicing. Mol. Cell. Biol. 2001, 21, 7380–7390. [Google Scholar] [CrossRef]
- Sahin, U.; Koslowski, M.; Dhaene, K.; Usener, D.; Brandenburg, G.; Seitz, G.; Huber, C.; Türeci, O. Claudin-18 Splice Variant 2 Is a Pan-Cancer Target Suitable for Therapeutic Antibody Development. Clin. Cancer Res. 2008, 14, 7624–7634. [Google Scholar] [CrossRef]
- Kubota, Y.; Kawazoe, A.; Mishima, S.; Nakamura, Y.; Kotani, D.; Kuboki, Y.; Bando, H.; Kojima, T.; Doi, T.; Yoshino, T.; et al. Comprehensive clinical and molecular characterization of claudin 18.2 expression in advanced gastric or gastroesophageal junction cancer. ESMO Open 2023, 8, 100762. [Google Scholar] [CrossRef]
- Moran, D.; Maurus, D.; Rohde, C.; Arozullah, A. Prevalence of CLDN18.2, HER2 and PD-L1 in gastric cancer samples. Ann. Oncol. 2018, 29, viii32. [Google Scholar] [CrossRef]
- Pellino, A.; Brignola, S.; Riello, E.; Niero, M.; Murgioni, S.; Guido, M.; Nappo, F.; Businello, G.; Sbaraglia, M.; Bergamo, F.; et al. Association of CLDN18 Protein Expression with Clinicopathological Features and Prognosis in Advanced Gastric and Gastroesophageal Junction Adenocarcinomas. J. Pers. Med. 2021, 11, 1095. [Google Scholar] [CrossRef]
- Singh, P.; Toom, S.; Huang, Y. Anti-claudin 18.2 antibody as new targeted therapy for advanced gastric cancer. J. Hematol. Oncol. 2017, 10, 105. [Google Scholar] [CrossRef]
- Shitara, K.; Lordick, F.; Bang, Y.-J.; Enzinger, P.; Ilson, D.; A Shah, M.; Van Cutsem, E.; Xu, R.-H.; Aprile, G.; Xu, J.; 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. [Google Scholar] [CrossRef] [PubMed]
- Shah, M.A.; Shitara, K.; Ajani, J.A.; Bang, Y.-J.; Enzinger, P.; Ilson, D.; Lordick, F.; Van Cutsem, E.; Plazas, J.G.; Huang, J.; et al. Zolbetuximab plus CAPOX in CLDN18.2-positive gastric or gastroesophageal junction adenocarcinoma: The randomized, phase 3 GLOW trial. Nat. Med. 2023, 29, 2133–2141. [Google Scholar] [CrossRef] [PubMed]
- Study Details|A Study to Assess the Antitumor Activity, Safety, Pharmacokinetics and Biomarkers of Zolbetuximab (IMAB362) in Participants with Claudin (CLDN) 18.2 Positive, Metastatic or Advanced Unresectable Gastric and Gastroesophageal Junction (GEJ) Adenocarcinoma and Locoregional Gastric or GEJ Adenocarcinoma|ClinicalTrials.gov. Available online: https://www.clinicaltrials.gov/study/NCT03505320 (accessed on 11 May 2024).
- Klempner, S.J.; Fabrizio, D.; Bane, S.; Reinhart, M.; Peoples, T.; Ali, S.M.; Sokol, E.S.; Frampton, G.; Schrock, A.B.; Anhorn, R.; et al. Tumor Mutational Burden as a Predictive Biomarker for Response to Immune Checkpoint Inhibitors: A Review of Current Evidence. Oncologist 2020, 25, e147–e159. [Google Scholar] [CrossRef]
- Jang, J.Y.; Jeon, Y.; Jeong, S.Y.; Lim, S.H.; Kang, W.K.; Lee, J.; Kim, S.T. The Optimal Tumor Mutational Burden Cutoff Value as a Novel Marker for Predicting the Efficacy of Programmed Cell Death-1 Checkpoint Inhibitors in Advanced Gastric Cancer. J. Gastric Cancer 2023, 23, 476–486. [Google Scholar] [CrossRef]
- Chalmers, Z.R.; Connelly, C.F.; Fabrizio, D.; Gay, L.; Ali, S.M.; Ennis, R.; Schrock, A.; Campbell, B.; Shlien, A.; Chmielecki, J.; et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017, 9, 34. [Google Scholar] [CrossRef]
- Shitara, K.; Özgüroğlu, M.; Bang, Y.-J.; Di Bartolomeo, M.; Mandalà, M.; Ryu, M.-H.; Caglevic, C.; Chung, H.; Muro, K.; Van Cutsem, E.; et al. Molecular determinants of clinical outcomes with pembrolizumab versus paclitaxel in a randomized, open-label, phase III trial in patients with gastroesophageal adenocarcinoma. Ann. Oncol. 2021, 32, 1127–1136. [Google Scholar] [CrossRef]
- Lee, K.W.; Van Cutsem, E.; Bang, Y.J.; Fuchs, C.S.; Kudaba, I.; Garrido, M.; Chung, H.C.; Lee, J.; Castro, H.R.; Chao, J.; et al. Association of Tumor Mutational Burden with Efficacy of Pembrolizumab ± Chemotherapy as First-Line Therapy for Gastric Cancer in the Phase III KEYNOTE-062 Study. Clin. Cancer Res. 2022, 28, 3489–3498. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Liu, H.; Liu, W.; Zong, P.; Huang, K.; Li, Z.; Li, H.; Xiong, T.; Tian, G.; Li, C.; et al. Predicting gastric cancer tumor mutational burden from histopathological images using multimodal deep learning. Briefings Funct. Genom. 2023, 23, 228–238. [Google Scholar] [CrossRef]
- Davies, H.; Bignell, G.R.; Cox, C.; Stephens, P.; Edkins, S.; Clegg, S.; Teague, J.; Woffendin, H.; Garnett, M.J.; Bottomley, W.; et al. Mutations of the BRAF gene in human cancer. Nature 2002, 417, 949–954. [Google Scholar] [CrossRef]
- Rajagopalan, H.; Bardelli, A.; Lengauer, C.; Kinzler, K.W.; Vogelstein, B.; Velculescu, V.E. RAF/RAS oncogenes and mismatch-repair status. Nature 2002, 418, 934. [Google Scholar] [CrossRef]
- Choi, Y.Y.; Noh, S.H.; Cheong, J.-H. Molecular Dimensions of Gastric Cancer: Translational and Clinical Perspectives. J. Pathol. Transl. Med. 2016, 50, 1–9. [Google Scholar] [CrossRef] [PubMed]
- Lee, S.H.; Soung, Y.H.; Kim, H.S.; Park, W.S.; Kim, S.Y.; Lee, J.H.; Park, J.Y.; Cho, Y.G.; Kim, C.J.; Nam, S.W.; et al. BRAF and KRAS mutations in stomach cancer. Oncogene 2003, 22, 6942–6945. [Google Scholar] [CrossRef]
- Ali, S.M.; Sanford, E.M.; Klempner, S.J.; Rubinson, D.A.; Wang, K.; Palma, N.A.; Chmielecki, J.; Yelensky, R.; Palmer, G.A.; Morosini, D.; et al. Prospective Comprehensive Genomic Profiling of Advanced Gastric Carcinoma Cases Reveals Frequent Clinically Relevant Genomic Alterations and New Routes for Targeted Therapies. Oncologist 2015, 20, 499–507. [Google Scholar] [CrossRef]
- Takahashi, M.; Buma, Y.; Iwamoto, T.; Inaguma, Y.; Ikeda, H.; Hiai, H. Cloning and expression of the ret proto-oncogene encoding a tyrosine kinase with two potential transmembrane domains. Oncogene 1988, 3, 571–578. [Google Scholar] [PubMed]
- Mulligan, L.M. GDNF and the RET Receptor in Cancer: New Insights and Therapeutic Potential. Front. Physiol. 2019, 9, 1873. [Google Scholar] [CrossRef]
- Zhang, F.; Tang, J.M.; Wang, L.; Wu, P.P.; Zhang, M. Immunohistochemical detection of RET proto-oncogene product in tumoral and nontumoral mucosae of gastric cancer. Anal. Quant. Cytopathol. Histpathol. 2014, 36, 128–136. [Google Scholar] [PubMed]
- Belli, C.; Penault-Llorca, F.; Ladanyi, M.; Normanno, N.; Scoazec, J.-Y.; Lacroix, L.; Reis-Filho, J.; Subbiah, V.; Gainor, J.; Endris, V.; et al. ESMO recommendations on the standard methods to detect RET fusions and mutations in daily practice and clinical research. Ann. Oncol. 2021, 32, 337–350. [Google Scholar] [CrossRef]
- Desilets, A.; Repetto, M.; Yang, S.-R.; Sherman, E.J.; Drilon, A. RET-Altered Cancers—A Tumor-Agnostic Review of Biology, Diagnosis and Targeted Therapy Activity. Cancers 2023, 15, 4146. [Google Scholar] [CrossRef]
- Romei, C.; Cosci, B.; Renzini, G.; Bottici, V.; Molinaro, E.; Agate, L.; Passannanti, P.; Viola, D.; Biagini, A.; Basolo, F.; et al. RET genetic screening of sporadic medullary thyroid cancer (MTC) allows the preclinical diagnosis of unsuspected gene carriers and the identification of a relevant percentage of hidden familial MTC (FMTC). Clin. Endocrinol. 2011, 74, 241–247. [Google Scholar] [CrossRef]
- Yakes, F.M.; Chen, J.; Tan, J.; Yamaguchi, K.; Shi, Y.; Yu, P.; Qian, F.; Chu, F.; Bentzien, F.; Cancilla, B.; et al. Cabozantinib (XL184), a Novel MET and VEGFR2 Inhibitor, Simultaneously Suppresses Metastasis, Angiogenesis, and Tumor Growth. Mol. Cancer Ther. 2011, 10, 2298–2308. [Google Scholar] [CrossRef]
- Carlomagno, F.; Vitagliano, D.; Guida, T.; Ciardiello, F.; Tortora, G.; Vecchio, G.; Ryan, A.; Fontanini, G.; Fusco, A.; Santoro, M. ZD6474, an orally available inhibitor of KDR tyrosine kinase activity, efficiently blocks oncogenic RET kinases. Cancer Res. 2002, 62, 7284–7290. [Google Scholar] [PubMed]
- Wedge, S.R.; Ogilvie, D.J.; Dukes, M.; Kendrew, J.; Chester, R.; Jackson, J.A.; Boffey, S.J.; Valentine, P.J.; Curwen, J.O.; Musgrove, H.L.; et al. ZD6474 inhibits vascular endothelial growth factor signaling, angiogenesis, and tumor growth following oral administration. Cancer Res. 2002, 62, 4645–4655. [Google Scholar] [PubMed]
- Subbiah, V.; Gainor, J.F.; Rahal, R.; Brubaker, J.D.; Kim, J.L.; Maynard, M.; Hu, W.; Cao, Q.; Sheets, M.P.; Wilson, D.; et al. Precision Targeted Therapy with BLU-667 for RET-Driven Cancers. Cancer Discov. 2018, 8, 836–849. [Google Scholar] [CrossRef] [PubMed]
- Subbiah, V.; Velcheti, V.; Tuch, B.; Ebata, K.; Busaidy, N.; Cabanillas, M.; Wirth, L.; Stock, S.; Smith, S.; Lauriault, V.; et al. Selective RET kinase inhibition for patients with RET-altered cancers. Ann. Oncol. 2018, 29, 1869–1876. [Google Scholar] [CrossRef]
- Gainor, J.F.; Lee, D.H.; Curigliano, G.; Doebele, R.C.; Kim, D.-W.; Baik, C.S.; Tan, D.S.-W.; Lopes, G.; Gadgeel, S.M.; Cassier, P.A.; et al. Clinical activity and tolerability of BLU-667, a highly potent and selective RET inhibitor, in patients (pts) with advanced RET-fusion+ non-small cell lung cancer (NSCLC). J. Clin. Oncol. 2019, 37, 9008. [Google Scholar] [CrossRef]
- Drilon, A.E.; Subbiah, V.; Oxnard, G.R.; Bauer, T.M.; Velcheti, V.; Lakhani, N.J.; Besse, B.; Park, K.; Patel, J.D.; Cabanillas, M.E.; et al. A phase 1 study of LOXO-292, a potent and highly selective RET inhibitor, in patients with RET-altered cancers. J. Clin. Oncol. 2018, 36, 102. [Google Scholar] [CrossRef]
- Andreev-Drakhlin, A.; Roszik, J.; Subbiah, V. The landscape of RET alterations from 56,970 adult patients with cancer: Clinical implications. J. Clin. Oncol. 2019, 37, 3106. [Google Scholar] [CrossRef]
- Röcken, C. Predictive biomarkers in gastric cancer. J. Cancer Res. Clin. Oncol. 2022, 149, 467–481. [Google Scholar] [CrossRef]
- Metzger, M.; Behrens, H.; Böger, C.; Haag, J.; Krüger, S.; Röcken, C. MET in gastric cancer—Discarding a 10% cutoff rule. Histopathology 2015, 68, 241–253. [Google Scholar] [CrossRef]
- Catenacci, D.V.T.; Tebbutt, N.C.; Davidenko, I.; Murad, A.M.; Al-Batran, S.-E.; Ilson, D.H.; Tjulandin, S.; Gotovkin, E.; Karaszewska, B.; Bondarenko, I.; et al. Rilotumumab plus epirubicin, cisplatin, and capecitabine as first-line therapy in advanced MET-positive gastric or gastro-oesophageal junction cancer (RILOMET-1): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2017, 18, 1467–1482. [Google Scholar] [CrossRef]
- Iveson, T.; Donehower, R.C.; Davidenko, I.; Tjulandin, S.; Deptala, A.; Harrison, M.; Nirni, S.; Lakshmaiah, K.; Thomas, A.; Jiang, Y.; et al. Rilotumumab in combination with epirubicin, cisplatin, and capecitabine as first-line treatment for gastric or oesophagogastric junction adenocarcinoma: An open-label, dose de-escalation phase 1b study and a double-blind, randomised phase 2 study. Lancet Oncol. 2014, 15, 1007–1018. [Google Scholar] [CrossRef]
- Malka, D.; François, E.; Penault-Llorca, F.; Castan, F.; Bouché, O.; Bennouna, J.; Ghiringhelli, F.; de la Fouchardière, C.; Borg, C.; Samalin, E.; et al. FOLFOX alone or combined with rilotumumab or panitumumab as first-line treatment for patients with advanced gastroesophageal adenocarcinoma (PRODIGE 17-ACCORD 20-MEGA): A randomised, open-label, three-arm phase II trial. Eur. J. Cancer 2019, 115, 97–106. [Google Scholar] [CrossRef]
- Shah, M.A.; Bang, Y.-J.; Lordick, F.; Alsina, M.; Chen, M.; Hack, S.P.; Bruey, J.M.; Smith, D.; McCaffery, I.; Shames, D.S.; et al. Effect of Fluorouracil, Leucovorin, and Oxaliplatin with or Without Onartuzumab in HER2-Negative, MET-Positive Gastroesophageal Adenocarcinoma. JAMA Oncol. 2017, 3, 620–627. [Google Scholar] [CrossRef]
- Sakai, D.; Chung, H.C.; Oh, D.-Y.; Park, S.H.; Kadowaki, S.; Kim, Y.H.; Tsuji, A.; Komatsu, Y.; Kang, Y.-K.; Uenaka, K.; et al. A non-randomized, open-label, single-arm, Phase 2 study of emibetuzumab in Asian patients with MET diagnostic positive, advanced gastric cancer. Cancer Chemother. Pharmacol. 2017, 80, 1197–1207. [Google Scholar] [CrossRef]
- Kaji, M.; Yonemura, Y.; Harada, S.; Liu, X.; Terada, I.; Yamamoto, H. Participation of c-met in the progression of human gastric cancers: Anti-c-met oligonucleotides inhibit proliferation or invasiveness of gastric cancer cells. Cancer Gene Ther. 1996, 3, 393–404. [Google Scholar]
- Inoue, T.; Chung, Y.; Yashiro, M.; Nishimura, S.; Hasuma, T.; Otani, S.; Sowa, M. Transforming Growth Factor-β and Hepatocyte Growth Factor Produced by Gastric Fibroblasts Stimulate the Invasiveness of Scirrhous Gastric Cancer Cells. Jpn. J. Cancer Res. 1997, 88, 152–159. [Google Scholar] [CrossRef]
- Toiyama, Y.; Yasuda, H.; Saigusa, S.; Matushita, K.; Fujikawa, H.; Tanaka, K.; Mohri, Y.; Inoue, Y.; Goel, A.; Kusunoki, M. Co-expression of hepatocyte growth factor and c-Met predicts peritoneal dissemination established by autocrine hepatocyte growth factor/c-Met signaling in gastric cancer. Int. J. Cancer 2011, 130, 2912–2921. [Google Scholar] [CrossRef]
- Peng, Z.; Zhu, Y.; Wang, Q.; Gao, J.; Li, Y.; Li, Y.; Ge, S.; Shen, L. Prognostic Significance of MET Amplification and Expression in Gastric Cancer: A Systematic Review with Meta-Analysis. PLoS ONE 2014, 9, e84502. [Google Scholar] [CrossRef]
- Lee, J.; Kim, S.T.; Kim, K.; Lee, H.; Kozarewa, I.; Mortimer, P.G.; Odegaard, J.I.; Harrington, E.A.; Lee, J.; Lee, T.; et al. Tumor Genomic Profiling Guides Patients with Metastatic Gastric Cancer to Targeted Treatment: The VIKTORY Umbrella Trial. Cancer Discov. 2019, 9, 1388–1405. [Google Scholar] [CrossRef]
- Babina, I.S.; Turner, N.C. Advances and challenges in targeting FGFR signalling in cancer. Nat. Rev. Cancer 2017, 17, 318–332. [Google Scholar] [CrossRef]
- Schrumpf, T.; Behrens, H.-M.; Haag, J.; Krüger, S.; Röcken, C. FGFR2 overexpression and compromised survival in diffuse-type gastric cancer in a large central European cohort. PLoS ONE 2022, 17, e0264011. [Google Scholar] [CrossRef]
- Katoh, M. Fibroblast growth factor receptors as treatment targets in clinical oncology. Nat. Rev. Clin. Oncol. 2018, 16, 105–122. [Google Scholar] [CrossRef]
- Ahn, S.; Lee, J.; Hong, M.; Kim, S.T.; Park, S.H.; Choi, M.G.; Lee, J.-H.; Sohn, T.S.; Bae, J.M.; Kim, S.; et al. FGFR2 in gastric cancer: Protein overexpression predicts gene amplification and high H-index predicts poor survival. Mod. Pathol. 2016, 29, 1095–1103. [Google Scholar] [CrossRef]
- Hur, J.Y.; Chao, J.; Kim, K.; Kim, S.T.; Kim, K.-M.; Klempner, S.J.; Lee, J. High-level FGFR2 amplification is associated with poor prognosis and Lower response to chemotherapy in gastric cancers. Pathol. Res. Pract. 2020, 216, 152878. [Google Scholar] [CrossRef]
- Wainberg, Z.A.; Enzinger, P.C.; Kang, Y.-K.; Yamaguchi, K.; Qin, S.; Lee, K.-W.; Oh, S.C.; Li, J.; Turk, H.M.; Teixeira, A.C.; et al. Randomized double-blind placebo-controlled phase 2 study of bemarituzumab combined with modified FOLFOX6 (mFOLFOX6) in first-line (1L) treatment of advanced gastric/gastroesophageal junction adenocarcinoma (FIGHT). J. Clin. Oncol. 2021, 39, 160. [Google Scholar] [CrossRef]
- A Wainberg, Z.; 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. [Google Scholar] [CrossRef]
- Smyth, E.C.; Chao, J.; Muro, K.; Yen, P.; Yanes, R.E.; Zahlten-Kumeli, A.; Rha, S.Y. Trial in progress: Phase 3 study of bemarituzumab + mFOLFOX6 versus placebo + mFOLFOX6 in previously untreated advanced gastric or gastroesophageal junction (GEJ) cancer with FGFR2b overexpression (FORTITUDE-101). J. Clin. Oncol. 2022, 40, TPS4164. [Google Scholar] [CrossRef]
- Poulet, G.; Massias, J.; Taly, V. Liquid Biopsy: General Concepts. Acta Cytol. 2019, 63, 449–455. [Google Scholar] [CrossRef]
- Dasari, A.; Morris, V.K.; Allegra, C.J.; Atreya, C.; Benson, A.B., 3rd; Boland, P.; Chung, K.; Copur, M.S.; Corcoran, R.B.; Deming, D.A.; et al. ctDNA applications and integration in colorectal cancer: An NCI Colon and Rectal–Anal Task Forces whitepaper. Nat. Rev. Clin. Oncol. 2020, 17, 757–770. [Google Scholar] [CrossRef]
- Ignatiadis, M.; Sledge, G.W.; Jeffrey, S.S. Liquid biopsy enters the clinic—Implementation issues and future challenges. Nat. Rev. Clin. Oncol. 2021, 18, 297–312. [Google Scholar] [CrossRef]
- Rodríguez, J.; Avila, J.; Rolfo, C.; Ruíz-Patiño, A.; Russo, A.; Ricaurte, L.; Ordóñez-Reyes, C.; Arrieta, O.; Zatarain-Barrón, Z.L.; Recondo, G.; et al. When Tissue is an Issue the Liquid Biopsy is Nonissue: A Review. Oncol. Ther. 2021, 9, 89–110. [Google Scholar] [CrossRef]
- Spellman, P.T.; Gray, J.W. Detecting cancer by monitoring circulating tumor DNA. Nat. Med. 2014, 20, 474–475. [Google Scholar] [CrossRef]
- Kim, K.; Shin, D.G.; Park, M.K.; Baik, S.H.; Kim, T.H.; Kim, S.; Lee, S. Circulating cell-free DNA as a promising biomarker in patients with gastric cancer: Diagnostic validity and significant reduction of cfDNA after surgical resection. Ann. Surg. Treat. Res. 2014, 86, 136–142. [Google Scholar] [CrossRef]
- Schwarzenbach, H.; Hoon, D.S.B.; Pantel, K. Cell-free nucleic acids as biomarkers in cancer patients. Nat. Rev. Cancer 2011, 11, 426–437. [Google Scholar] [CrossRef]
- Nakamura, Y.; Fujisawa, T.; Kadowaki, S.; Takahashi, N.; Goto, M.; Yoshida, K.; Kawakami, T.; Esaki, T.; Oki, E.; Nishida, N.; et al. Characteristics of genomic alterations in circulating tumor DNA (ctDNA) in patients (Pts) with advanced gastrointestinal (GI) cancers in nationwide large-scale ctDNA screening:SCRUM-Japan Monstar-Screen. J. Clin. Oncol. 2021, 39, 106. [Google Scholar] [CrossRef]
- Jogo, T.; Nakamura, Y.; Shitara, K.; Bando, H.; Yasui, H.; Esaki, T.; Terazawa, T.; Satoh, T.; Shinozaki, E.; Nishina, T.; et al. Circulating Tumor DNA Analysis Detects FGFR2 Amplification and Concurrent Genomic Alterations Associated with FGFR Inhibitor Efficacy in Advanced Gastric Cancer. Clin. Cancer Res. 2021, 27, 5619–5627. [Google Scholar] [CrossRef]
- Li, J.; Jiang, W.; Wei, J.; Zhang, J.; Cai, L.; Luo, M.; Wang, Z.; Sun, W.; Wang, S.; Wang, C.; et al. Patient specific circulating tumor DNA fingerprints to monitor treatment response across multiple tumors. J. Transl. Med. 2020, 18, 293. [Google Scholar] [CrossRef]
- Wang, Y.; Zhao, C.; Chang, L.; Jia, R.; Liu, R.; Zhang, Y.; Gao, X.; Li, J.; Chen, R.; Xia, X.; et al. Circulating tumor DNA analyses predict progressive disease and indicate trastuzumab-resistant mechanism in advanced gastric cancer. EBioMedicine 2019, 43, 261–269. [Google Scholar] [CrossRef]
- Bratman, S.V.; Yang, S.Y.C.; Iafolla, M.A.J.; Liu, Z.; Hansen, A.R.; Bedard, P.L.; Lheureux, S.; Spreafico, A.; Razak, A.A.; Shchegrova, S.; et al. Personalized circulating tumor DNA analysis as a predictive biomarker in solid tumor patients treated with pembrolizumab. Nat. Cancer 2020, 1, 873–881. [Google Scholar] [CrossRef]
- Matsusaka, S.; Chìn, K.; Ogura, M.; Suenaga, M.; Shinozaki, E.; Mishima, Y.; Terui, Y.; Mizunuma, N.; Hatake, K. Circulating tumor cells as a surrogate marker for determining response to chemotherapy in patients with advanced gastric cancer. Cancer Sci. 2010, 101, 1067–1071. [Google Scholar] [CrossRef]
- Pantel, K.; Alix-Panabières, C. Circulating tumour cells and cell-free DNA in gastrointestinal cancer. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 73–74. [Google Scholar] [CrossRef]
- Zhang, Q.; Shan, F.; Li, Z.; Gao, J.; Li, Y.; Shen, L.; Ji, J.; Lu, M. A prospective study on the changes and clinical significance of pre-operative and post-operative circulating tumor cells in resectable gastric cancer. J. Transl. Med. 2018, 16, 171. [Google Scholar] [CrossRef]
- Ishiguro, Y.; Sakihama, H.; Yoshida, T.; Ichikawa, N.; Homma, S.; Fukai, M.; Kawamura, H.; Takahashi, N.; Taketomi, A. Prognostic Significance of Circulating Tumor Cells with Mesenchymal Phenotypes in Patients with Gastric Cancer: A Prospective Study. Ann. Surg. Oncol. 2021, 28, 1178–1186. [Google Scholar] [CrossRef]
- Mishima, Y.; Matsusaka, S.; Chin, K.; Mikuniya, M.; Minowa, S.; Takayama, T.; Shibata, H.; Kuniyoshi, R.; Ogura, M.; Terui, Y.; et al. Detection of HER2 Amplification in Circulating Tumor Cells of HER2-Negative Gastric Cancer Patients. Target. Oncol. 2017, 12, 341–351. [Google Scholar] [CrossRef]
- Wang, H.; Li, B.; Liu, Z.; Gong, J.; Shao, L.; Ren, J.; Niu, Y.; Bo, S.; Li, Z.; Lai, Y.; et al. HER2 copy number of circulating tumour DNA functions as a biomarker to predict and monitor trastuzumab efficacy in advanced gastric cancer. Eur. J. Cancer 2018, 88, 92–100. [Google Scholar] [CrossRef]
- Xie, Y.; Dang, W.; Zhang, S.; Yue, W.; Yang, L.; Zhai, X.; Yan, Q.; Lu, J. The role of exosomal noncoding RNAs in cancer. Mol. Cancer 2019, 18, 37. [Google Scholar] [CrossRef]
- Varkey, J.; Nicolaides, T. Tumor-Educated Platelets: A Review of Current and Potential Applications in Solid Tumors. Cureus 2021, 13, e19189. [Google Scholar] [CrossRef]
Drug | Trial and Phase | Determination of PDL1 Level | Design of the Study and Results | Ref. |
---|---|---|---|---|
Pembrolizumab | KEYNOTE-059, phase II | CPS ≥ 1 with Dako 22C3 | Monotherapy in heavily pretreated GC—FDA approved | [29] |
Pembrolizumab | KEYNOTE-061, phase III | CPS ≥ 1 with Dako 22C3 | Pembrolizumab vs. paclitaxel as II line—no advantages | [30] |
Nivolumab | CheckMate-649, phase III | PD-L1 CPS ≥ 5 with Dako 28-8 | Nivolumab + CT vs. CT alone as first line—FDA and EMA approved | [14] |
Drug | Trial and Phase | Design of the Study | Level of Development | Ref. |
---|---|---|---|---|
Trastuzumab | ToGA trial, phase III | Trastuzumab + CT vs. CT alone as first line | SoC | [47] |
Trastuzumab | KEYNOTE-811, phase III | Trastuzumab + pembrolizumab + CT vs. standard of care as first line in CPS ≥ 1 | FDA and EMA approved | [50] |
Zanidatamab (ZW25) | HERIZON-GEA-01, phase III | Zanidatamab plus standard CT with or without the PD-1 inhibitor as first line | Ongoing trial | [51] |
Margetuximab | MAHOGANY, phase II/III | Margetuximab with antiPD1 vs. standard of care | Ongoing | [53] |
Lapatinib | TyTAN—A, phase III | Lapatinib + paclitaxel vs. paclitaxel as second line | Failed | [54] |
Pertuzumab | JACOB, phase III | Pertuzumab + standard of care vs. standard of care as first line | Failed | [55] |
Trastuzumab emtansine (T-DM1) | GATSBY, phase II/III | T-DM1 vs. paclitaxel as second line | Failed | [56] |
Trastuzumab deruxtecan (T-DXd) | DESTINYGastric04, phase III | T-DXd vs. SoC as second line | Ongoing | [65] |
Biomarker | Assessment | Drug | Setting | Clinical Trial |
---|---|---|---|---|
PD-L1 | IHC | Nivolumab (plus standard CT) Pembrolizumab (plus trastuzumab and CT) | First line in PD-L1 CPS ≥ 5 First line in HER2-positive GC with PD-L1 CPS ≥ 1 | CheckMate-649 KEYNOTE-811 |
HER2 | IHC/FISH | Trastuzumab (plus standard CT) Trastuzumab deruxtecan | First line Third or later line | ToGA DESTINY-Gastric 0 |
MSI | IHC, PCR or NGS | Pembrolizumab | First line | KEYNOTE-158 |
NTRK | RNA-seq, IHC | Larotrectinib Entrectinib | Advanced disease | LOXO-TRK-14001, NAVIGATE STARTRK-1 and STARTRK-2 |
Claudin 18.2 | IHC | Zolbetuximab (plus standard CT) Zolbetuximab plus Capecitabine and Oxaliplatin | First line First line | SPOTLIGHT GLOW |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Airò, G.; Agnetti, V.; Pratticò, F.; Peroni, M.; Bui, S.; Mura, G.; Urbanowicz-Nijaki, M.; Lai, E.; Puzzoni, M.; Contu, F.; et al. Tissue Biomarkers in Gastric Cancer Treatment: Present and Future. Int. J. Transl. Med. 2024, 4, 640-660. https://doi.org/10.3390/ijtm4040045
Airò G, Agnetti V, Pratticò F, Peroni M, Bui S, Mura G, Urbanowicz-Nijaki M, Lai E, Puzzoni M, Contu F, et al. Tissue Biomarkers in Gastric Cancer Treatment: Present and Future. International Journal of Translational Medicine. 2024; 4(4):640-660. https://doi.org/10.3390/ijtm4040045
Chicago/Turabian StyleAirò, Giulia, Virginia Agnetti, Fabiana Pratticò, Marianna Peroni, Simona Bui, Giovanni Mura, Maria Urbanowicz-Nijaki, Eleonora Lai, Marco Puzzoni, Fabiana Contu, and et al. 2024. "Tissue Biomarkers in Gastric Cancer Treatment: Present and Future" International Journal of Translational Medicine 4, no. 4: 640-660. https://doi.org/10.3390/ijtm4040045
APA StyleAirò, G., Agnetti, V., Pratticò, F., Peroni, M., Bui, S., Mura, G., Urbanowicz-Nijaki, M., Lai, E., Puzzoni, M., Contu, F., Denaro, N., Scartozzi, M., Solinas, C., & Tommasi, C. (2024). Tissue Biomarkers in Gastric Cancer Treatment: Present and Future. International Journal of Translational Medicine, 4(4), 640-660. https://doi.org/10.3390/ijtm4040045