51
|
Christodoulidis G, Koumarelas KE, Kouliou MN. Revolutionizing gastric cancer treatment: The potential of immunotherapy. World J Gastroenterol 2024; 30:286-289. [PMID: 38313231 PMCID: PMC10835540 DOI: 10.3748/wjg.v30.i4.286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 12/19/2023] [Accepted: 01/15/2024] [Indexed: 01/26/2024] Open
Abstract
Gastric cancer, a prevalent malignancy worldwide, ranks sixth in terms of frequency and third in fatality, causing over a million new cases and 769000 annual deaths. Predominant in Eastern Europe and Eastern Asia, risk factors include family medical history, dietary habits, tobacco use, Helicobacter pylori, and Epstein-Barr virus infections. Unfortunately, gastric cancer is often diagnosed at an advanced stage, leading to a grim prognosis, with a 5-year overall survival rate below 5%. Surgical intervention, particularly with D2 Lymphadenectomy, is the mainstay for early-stage cases but offers limited success. For advanced cases, the National Comprehensive Cancer Network recommends chemotherapy, radiation, and targeted therapy. Emerging immunotherapy presents promise, especially for unresectable or metastatic cases, with strategies like immune checkpoint inhi-bitors, tumor vaccines, adoptive immunotherapy, and nonspecific immunomodulators. In this Editorial, with regards to the article "Advances and key focus areas in gastric cancer immunotherapy: A comprehensive scientometric and clinical trial review", we address the advances in the field of immunotherapy in gastric cancer and its future prospects.
Collapse
|
52
|
Meric-Bernstam F, Makker V, Oaknin A, Oh DY, Banerjee S, González-Martín A, Jung KH, Ługowska I, Manso L, Manzano A, Melichar B, Siena S, Stroyakovskiy D, Fielding A, Ma Y, Puvvada S, Shire N, Lee JY. Efficacy and Safety of Trastuzumab Deruxtecan in Patients With HER2-Expressing Solid Tumors: Primary Results From the DESTINY-PanTumor02 Phase II Trial. J Clin Oncol 2024; 42:47-58. [PMID: 37870536 PMCID: PMC10730032 DOI: 10.1200/jco.23.02005] [Citation(s) in RCA: 105] [Impact Index Per Article: 105.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/03/2023] [Accepted: 10/12/2023] [Indexed: 10/24/2023] Open
Abstract
PURPOSE Trastuzumab deruxtecan (T-DXd) is a human epidermal growth factor 2 (HER2)-directed antibody-drug conjugate approved in HER2-expressing breast and gastric cancers and HER2-mutant non-small-cell lung cancer. Treatments are limited for other HER2-expressing solid tumors. METHODS This open-label phase II study evaluated T-DXd (5.4 mg/kg once every 3 weeks) for HER2-expressing (immunohistochemistry [IHC] 3+/2+ by local or central testing) locally advanced or metastatic disease after ≥1 systemic treatment or without alternative treatments. The primary end point was investigator-assessed confirmed objective response rate (ORR). Secondary end points included safety, duration of response, progression-free survival (PFS), and overall survival (OS). RESULTS At primary analysis, 267 patients received treatment across seven tumor cohorts: endometrial, cervical, ovarian, bladder, biliary tract, pancreatic, and other. The median follow-up was 12.75 months. In all patients, the ORR was 37.1% (n = 99; [95% CI, 31.3 to 43.2]), with responses in all cohorts; the median DOR was 11.3 months (95% CI, 9.6 to 17.8); the median PFS was 6.9 months (95% CI, 5.6 to 8.0); and the median OS was 13.4 months (95% CI, 11.9 to 15.5). In patients with central HER2 IHC 3+ expression (n = 75), the ORR was 61.3% (95% CI, 49.4 to 72.4), the median DOR was 22.1 months (95% CI, 9.6 to not reached), the median PFS was 11.9 months (95% CI, 8.2 to 13.0), and the median OS was 21.1 months (95% CI, 15.3 to 29.6). Grade ≥3 drug-related adverse events were observed in 40.8% of patients; 10.5% experienced adjudicated drug-related interstitial lung disease (ILD), with three deaths. CONCLUSION Our study demonstrates durable clinical benefit, meaningful survival outcomes, and safety consistent with the known profile (including ILD) in pretreated patients with HER2-expressing tumors receiving T-DXd. Greatest benefit was observed for the IHC 3+ population. These data support the potential role of T-DXd as a tumor-agnostic therapy for patients with HER2-expressing solid tumors.
Collapse
Affiliation(s)
- Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Vicky Makker
- Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Ana Oaknin
- Gynaecologic Cancer Programme, Vall d’Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d’Hebron, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Do-Youn Oh
- Seoul National University Hospital; Cancer Research Institute, Seoul National University College of Medicine; Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, South Korea
| | - Susana Banerjee
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, United Kingdom
| | - Antonio González-Martín
- Medical Oncology Department and Programme in Solid Tumours-CIMA, Cancer Center Clínica Universidad de Navarra, Madrid, Spain
| | - Kyung Hae Jung
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Iwona Ługowska
- Early Phase Clinical Trials Unit and Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Skłodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Luis Manso
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Aránzazu Manzano
- Experimental Therapeutics in Cancer (UTEC), Department of Medical Oncology, Hospital Clínico San Carlos, Madrid, Spain
| | - Bohuslav Melichar
- Department of Oncology, Palacký University Medical School and University Hospital, Olomouc, Czech Republic
| | - Salvatore Siena
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda and the Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Piazza dell’Ospedale Maggiore, Milan, Italy
| | | | | | - Yan Ma
- Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | | | | | - Jung-Yun Lee
- Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
53
|
Santos ES, Oliver TK, Lacchetti C, Geisel R, Wilfong LS, Fader AN, Eng C. Drug Shortages in Oncology: ASCO Clinical Guidance for Alternative Treatments. JCO Oncol Pract 2024; 20:19-32. [PMID: 37963306 DOI: 10.1200/op.23.00545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 09/26/2023] [Indexed: 11/16/2023] Open
Abstract
PURPOSE To increase awareness, outline strategies, and offer clinical guidance on navigating the complexities of treatment planning amid antineoplastic drug shortages. METHODS A multidisciplinary panel of oncologists, ethicists, and patient advocates was assembled to provide rapid clinical guidance to help providers navigate appropriate patient care in cases where rationing or alternative therapies must be considered. The groups of content experts developed general principles for resource allocation during shortages and clinical guidance on alternative therapies for specific disease sites. The recommendations are supported by evidence when available. RESULTS A total of 44 volunteers with content expertise formed the Advisory Group that developed general guidance on the prioritization of antineoplastic agents in limited supply. Disease site-specific clinical guidance was then produced by subgroups on the basis of members' specialties and expertise. The majority of alternative treatment options were developed in consideration of cisplatin and carboplatin shortages. All guidance is posted on ASCO's website. RECOMMENDATIONS The prioritization of antineoplastic agents in limited supply should be based on specific goals of the therapy where evidence-based medicine has shown survival outcome and life-extending benefit in both early and advanced stages. Recommendations for specific disease sites are presented. While management options vary according to the disease site, alternatives are presented. For settings in which there are no alternatives with comparable efficacy and safety, it is recommended that patients are referred to an area where the necessary drug is available or can be obtained.Additional information is available at asco.org/drug-shortages.
Collapse
Affiliation(s)
| | | | | | - Rachel Geisel
- American Society of Clinical Oncology, Alexandria, VA
| | | | | | - Cathy Eng
- Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
54
|
Marcinak CT, Schwartz PB, Basree MM, Hurst N, Bassetti M, Kratz JD, Uboha NV. Treatment of Oligometastatic GI Cancers. Am Soc Clin Oncol Educ Book 2024; 44:e430152. [PMID: 38190577 DOI: 10.1200/edbk_430152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
Oligometastatic state is believed to potentially represent a transitional stage between early, locoregional state disease and widely metastatic disease. Historically, locoregional approaches, particularly in advanced colorectal cancers, have demonstrated efficacy in select patients with limited burden of metastatic disease. Recent strides in systemic therapies, including biomarker-based treatments and immunotherapy, alongside innovations in surgical techniques and novel locoregional approaches such as stereotactic radiotherapy and ablation, have ushered in a new era of therapeutic possibilities across all oligometastatic GI cancers. Despite these advancements, there remains a significant gap in high-quality prospective evidence guiding patient selection and treatment decisions across various disease types. Ongoing clinical trials are anticipated to provide crucial insights into oligometastatic states, fostering the refinement of disease-specific oligometastatic state definitions and treatment algorithms. This article reviews existing data on the management of oligometastatic GI cancer, summarizes current state of knowledge for each disease state, and provides updates on ongoing studies in this space.
Collapse
Affiliation(s)
- Clayton T Marcinak
- Department of Surgery, University of Wisconsin School of Medicine and Public Health University of Wisconsin-Madison, Madison, WI
| | - Patrick B Schwartz
- Department of Surgery, University of Wisconsin School of Medicine and Public Health University of Wisconsin-Madison, Madison, WI
| | - Mustafa M Basree
- Department of Human Oncology, University of Wisconsin Hospital and Clinics, Madison, WI
| | - Newton Hurst
- Department of Human Oncology, University of Wisconsin Hospital and Clinics, Madison, WI
| | - Michael Bassetti
- Department of Human Oncology, University of Wisconsin Hospital and Clinics, Madison, WI
| | - Jeremy D Kratz
- University of Wisconsin-Madison, Madison, WI
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI
- Center for Human Genomics and Precision Medicine, University of Wisconsin, Madison, WI
- William S. Middleton Memorial Veterans Hospital, Madison, WI
| | - Nataliya V Uboha
- University of Wisconsin-Madison, Madison, WI
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI
| |
Collapse
|
55
|
Wang HH, Steffens EN, Kats-Ugurlu G, van Etten B, Burgerhof JGM, Hospers GAP, Plukker JTM. Potential Predictive Immune and Metabolic Biomarkers of Tumor Microenvironment Regarding Pathological and Clinical Response in Esophageal Cancer After Neoadjuvant Chemoradiotherapy: A Systematic Review. Ann Surg Oncol 2024; 31:433-451. [PMID: 37777688 PMCID: PMC10695872 DOI: 10.1245/s10434-023-14352-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 09/13/2023] [Indexed: 10/02/2023]
Abstract
INTRODUCTION The tumor microenvironment (TME) plays a crucial role in therapy response and modulation of immunologic surveillance. Adjuvant immunotherapy has recently been introduced in post-surgery treatment of locally advanced esophageal cancer (EC) with residual pathological disease after neoadjuvant chemoradiotherapy (nCRT). F-18 fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG-PET/CT) remains a valuable imaging tool to assess therapy response and to visualize metabolic TME; however, there is still a paucity in understanding the interaction between the TME and nCRT response. This systematic review investigated the potential of TME biomarkers and 18F-FDG-PET/CT features to predict pathological and clinical response (CR) after nCRT in EC. METHODS A literature search of the Medline and Embase electronic databases identified 4190 studies. Studies regarding immune and metabolic TME biomarkers and 18F-FDG-PET/CT features were included for predicting pathological response (PR) and/or CR after nCRT. Separate analyses were performed for 18F-FDG-PET/CT markers and these TME biomarkers. RESULTS The final analysis included 21 studies-10 about immune and metabolic markers alone and 11 with additional 18F-FDG-PET/CT features. High CD8 infiltration before and after nCRT, and CD3 and CD4 infiltration after nCRT, generally correlated with better PR. A high expression of tumoral or stromal programmed death-ligand 1 (PD-L1) after nCRT was generally associated with poor PR. Moreover, total lesion glycolysis (TLG) and metabolic tumor volume (MTV) of the primary tumor were potentially predictive for clinical and PR. CONCLUSION CD8, CD4, CD3, and PD-L1 are promising immune markers in predicting PR, whereas TLG and MTV are potential 18F-FDG-PET/CT features to predict clinical and PR after nCRT in EC.
Collapse
Affiliation(s)
- H H Wang
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - E N Steffens
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - G Kats-Ugurlu
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - B van Etten
- Department of Surgical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - J G M Burgerhof
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - G A P Hospers
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - J T M Plukker
- Department of Surgical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
56
|
Kang C. Trastuzumab Deruxtecan: A Review in Gastric or Gastro-Oesophageal Junction Adenocarcinoma. Target Oncol 2023; 18:981-989. [PMID: 37787931 DOI: 10.1007/s11523-023-00998-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2023] [Indexed: 10/04/2023]
Abstract
Trastuzumab deruxtecan (Enhertu®) is a human epidermal growth factor receptor type 2 (HER2)-directed antibody-drug conjugate that is approved in several countries globally for adults with advanced HER2-positive gastric or gastro-oesophageal junction (GOJ) adenocarcinoma who have received a prior trastuzumab-based regime. In the phase II DESTINY-Gastric01 trial, intravenous trastuzumab deruxtecan was significantly more effective than standard chemotherapy (physician's choice of intravenous irinotecan or paclitaxel) in achieving objective response and improving overall survival in Japanese or South Korean adults with advanced HER2-positive gastric or GOJ adenocarcinoma who had received two or more previous therapies. In the phase II DESTINY-Gastric02 trial, trastuzumab deruxtecan was able to induce durable response in adults from the USA or Europe with unresectable or metastatic HER2-positive gastric or GOJ adenocarcinoma. Trastuzumab deruxtecan was generally tolerable in these patients; the most common adverse events included nausea, neutropenia, fatigue and decreased appetite. Trastuzumab deruxtecan carries regulatory warnings (including boxed warnings in the USA) for interstitial lung disease/pneumonitis and embryo-foetal toxicity. Current evidence indicates that trastuzumab deruxtecan is an effective treatment option, and is generally tolerable, in previously treated adults with advanced HER2-positive gastric or GOJ adenocarcinoma.
Collapse
Affiliation(s)
- Connie Kang
- Springer Nature, Mairangi Bay, Private Bag 65901, Auckland, 0754, New Zealand.
| |
Collapse
|
57
|
Baretton GB, Lordick F, Gaiser T, Hofheinz R, Horst D, Lorenzen S, Moehler M, Röcken C, Schirmacher P, Stahl M, Thuss-Patience P, Tiemann K. Standardized and quality-assured predictive PD-L1 testing in the upper gastrointestinal tract. J Cancer Res Clin Oncol 2023; 149:16231-16238. [PMID: 37874352 PMCID: PMC10620316 DOI: 10.1007/s00432-023-05180-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 10/25/2023]
Abstract
As a result of the high approval dynamics and the growing number of immuno-oncological concepts, the complexity of treatment decisions and control in the area of cancers of the esophagus, gastroesophageal junction and stomach is constantly increasing. Since the treatment indication for PD-1 inhibitors that are currently approved in the European Union is often linked to the expression of PD-L1 (programmed cell death-ligand 1), the evaluation of tissue-based predictive markers by the pathologist is of crucial importance for treatment stratification. Even though the immunohistochemical analysis of the PD-L1 expression status is one of the best studied, therapy-relevant biomarkers for an immuno-oncological treatment, due to the high heterogeneity of carcinomas of the upper gastrointestinal tract, there are challenges in daily clinical diagnostic work with regard to implementation, standardization and interpretation of testing. An interdisciplinary group of experts from Germany has taken a position on relevant questions from daily pathological and clinical practice, which concern the starting material, quality-assured testing and the interpretation of pathological findings, and has developed recommendations for structured reporting.
Collapse
Affiliation(s)
- Gustavo B Baretton
- Institute for Pathology, University Hospital Carl Gustav Carus, TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
| | - Florian Lordick
- Department of Medicine II (Oncology, Gastroenterology, Hepatology and Pulmonology) and University Cancer Center Leipzig, University of Leipzig Medical Center, Leipzig, Germany.
| | - T Gaiser
- Institute of Applied Pathology, 67346, Speyer, Germany
| | - R Hofheinz
- University Medicine Mannheim, Mannheim, Germany
| | - D Horst
- Institute of Pathology of the Charité-University Medicine Berlin, Berlin, Germany
| | - S Lorenzen
- Department of Medicine III, Klinikum Rechts der Isar, Munich, Germany
| | - M Moehler
- Department of Medicine I, University Medicine Mainz, Mainz, Germany
| | - C Röcken
- Institute of Pathology, Christian-Albrechts University, Kiel, Germany
| | - P Schirmacher
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - M Stahl
- Department of Oncology and Palliative Care, Kliniken Essen Mitte, Evangelische Huyssens-Stiftung, Essen-Huttrop, Essen, Germany
| | - P Thuss-Patience
- Charité Center of Tumor Medicine CC14, Charité Campus Virchow-University Medicine Berlin, Berlin, Germany
| | - K Tiemann
- Institute of Hematopathology, Hamburg, Germany
| |
Collapse
|
58
|
Yoshihara K, Kobayashi Y, Endo S, Fukae M, Hennig S, Kastrissios H, Kamiyama E, Garimella T, Abutarif M. Trastuzumab Deruxtecan Dosing in Human Epidermal Growth Factor Receptor 2-Positive Gastric Cancer: Population Pharmacokinetic Modeling and Exposure-Response Analysis. J Clin Pharmacol 2023; 63:1232-1243. [PMID: 37393579 DOI: 10.1002/jcph.2295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 06/21/2023] [Indexed: 07/04/2023]
Abstract
This study evaluated the benefit/risk of trastuzumab deruxtecan (T-DXd) 6.4 mg/kg in patients with human epidermal growth factor receptor 2 (HER2)-positive gastric cancer using pharmacometrics. A population pharmacokinetic (PopPK) model was developed using data from patients with gastric cancer, breast cancer, or other tumors in T-DXd clinical trials, primarily conducted in Asia. Post hoc model-estimated pharmacokinetic metrics were used in exposure-efficacy (objective response rates, ORRs) and exposure-safety analyses. The PopPK analysis included 808 patients (217 with gastric cancer, 512 with breast cancer, and 79 with other cancers). In gastric cancer, the T-DXd 6.4 mg/kg steady-state exposure metrics were lower compared with 6.4 mg/kg in breast cancer, but were similar to 5.4 mg/kg in breast cancer. Tumor type was selected as a significant covariate on T-DXd clearance. In exposure-efficacy analysis among 160 patients with gastric cancer, the T-DXd steady-state minimum concentration was associated with a confirmed ORR in univariate logistic regression analysis (P = .023). The model-predicted confirmed ORRs in gastric cancer were 36.0% (90%CI 29.3% to 43.7%) with 5.4 mg/kg and 40.0% (90%CI 33.1% to 47.6%) with 6.4 mg/kg. Among 808 patients in the exposure-safety analyses, the model-predicted estimates for the rates of any-grade interstitial lung disease (ILD) over a period of 180 days were 10.2% (90%CI 8.7% to 12.8%) with 6.4 mg/kg in gastric cancer and 9.7% (90%CI 8.2% to 11.8%) with 5.4 mg/kg in breast cancer. In gastric cancer, the efficacy of T-DXd was higher at 6.4 mg/kg than at 5.4 mg/kg. Exposure and ILD rates were comparable between 6.4 mg/kg in gastric cancer and 5.4 mg/kg in breast cancer. This study identified T-DXd 6.4 mg/kg as the recommended dose in HER2-positive gastric cancer.
Collapse
Affiliation(s)
- Kazutaka Yoshihara
- Quantitative Clinical Pharmacology, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | | | - Seiko Endo
- Quantitative Clinical Pharmacology, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Masato Fukae
- Quantitative Clinical Pharmacology, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | | | - Helen Kastrissios
- Certara Inc., Melbourne, Australia
- QuanTx Consulting, Mountain View, CA, USA
| | - Emi Kamiyama
- Quantitative Clinical Pharmacology, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Tushar Garimella
- Quantitative Clinical Pharmacology, Daiichi Sankyo Inc., Basking Ridge, NJ, USA
| | - Malaz Abutarif
- Quantitative Clinical Pharmacology, Daiichi Sankyo Inc., Basking Ridge, NJ, USA
| |
Collapse
|
59
|
Li YN, Xie B, Zhang Y, He MH, Xing Y, Mu DM, Wang H, Guo R. Advances and key focus areas in gastric cancer immunotherapy: A comprehensive scientometric and clinical trial review (1999-2023). World J Gastroenterol 2023; 29:5593-5617. [PMID: 37970478 PMCID: PMC10642438 DOI: 10.3748/wjg.v29.i40.5593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/25/2023] [Accepted: 10/17/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) is the sixth most common cancer and third leading cause of cancer-related deaths worldwide. Current treatments mainly rely on surgery- and chemotherapy-based systemic; however, the prognosis remains poor for advanced disease. Recent studies have suggested that immunotherapy has significant potential in cancer therapy; thus, GC immunotherapy may improve quality of life and survival for patients with this disease. AIM To provide a comprehensive overview of the knowledge structure and research hotspots of GC immunotherapy. METHODS We conducted a bibliometric analysis of publications on immunotherapy related to GC in the Web of Science Core Collection database. We analyzed 2013 pub-lications from 1999 to February 1, 2023, using the VOSviewer and CiteSpace software. We assessed publication and citation distributions using the WoS platform and explored research countries, institutions, journals, authors, references, and keywords (co-occurrence, timeline view, and burst analysis). In addition, we examined 228 trials on immunotherapy, 137 on adoptive cell therapy, 274 on immune checkpoint inhibitors (ICIs), and 23 on vaccines from ClinicalTrials.gov and the International Clinical Trials Registry Platform. The Impact Index Per Article for the top ten high-cited papers collected from Reference Citation Analysis (RCA) are presented. RESULTS Our bibliometric analysis revealed that the study of immunotherapy in GC has developed rapidly in recent years. China accounted for almost half the publications, followed by the United States. The number of publications in recent years has been growing continuously, and most institutions and authors with the most publications are from China. The main keywords or clusters identified were "tumor microenvironment", "adoptive immunotherapy", "dendritic therapy", and "microsatellite instability". CONCLUSION Our analysis of 2013 publications indicated that immunotherapy for GC has led to several new developments in recent years. Considerable progress has been made in vaccinations, immune checkpoint therapy, and adoptive cellular therapy. In particular, ICIs and chimeric antigen receptor T-cells are novel options for the treatment of GC. We suggest that the combination of ICIs, chemotherapy, targeted therapy, and other immunotherapies should be the primary research direction in the future.
Collapse
Affiliation(s)
- Yao-Nan Li
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Bin Xie
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ying Zhang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Ming-Hua He
- College of Computer Science and Technology, Jilin University, Changchun 130012, Jilin Province, China
| | - Yang Xing
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Dong-Mei Mu
- Division of Clinical Research, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Hong Wang
- Cancer Center, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| | - Rui Guo
- Clinical Laboratory, The First Hospital of Jilin University, Changchun 130012, Jilin Province, China
| |
Collapse
|
60
|
Désilets A, Elkhoury R, Gebai A, Tehfe M. Current and Emerging Role of Monoclonal Antibody-Based First-Line Treatment in Advanced Gastro-Esophageal and Gastric Cancer. Curr Oncol 2023; 30:9304-9316. [PMID: 37887572 PMCID: PMC10605724 DOI: 10.3390/curroncol30100672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/12/2023] [Accepted: 10/19/2023] [Indexed: 10/28/2023] Open
Abstract
Gastric cancer is the fifth most common malignancy worldwide and one of the main causes of cancer-related death. While surgical treatment is the only curative option for early disease, many have inoperable or advanced disease at diagnosis. Treatment in this case would be a combination of chemotherapy and immunotherapy. Gastro-esophageal (GEJ) and gastric cancer (GC) genetic profiling with current molecular diagnostic techniques has significantly changed the therapeutic landscape in advanced cancers. The identification of key players in GEJ and GC survival and proliferation, such as human epidermal growth factor 2 (HER2), vascular endothelial growth factor (VEGF), and programmed cell death protein 1 (PD-1)/programmed cell death ligand-1 (PD-L1), has allowed for the individualization of advanced cancer treatment and significant improvement in overall survival and progression-free survival of patients. This review comprehensively examines the current and emerging role of monoclonal antibody-based first-line treatments in advanced GEJ and GC. We explore the impact of monoclonal antibodies targeting HER2, VEGF, PD-1/PD-L1, and Claudin 18.2 (CLDN18.2) on the first-line treatment landscape by talking about key clinical trials. This review emphasizes the importance of biomarker testing for optimal treatment selection and provides practical recommendations based on ASCO guidelines.
Collapse
Affiliation(s)
- Audrey Désilets
- Department of Medicine, Université de Montreal, Montreal, QC H3T 1J4, Canada
| | - Reem Elkhoury
- Department of Medicine, Université de Montreal, Montreal, QC H3T 1J4, Canada
- Hematology-Oncology, Oncology Center-Centre Hospitalier de l’Universite de Montreal, Montreal, QC H2X 0C1, Canada
| | - Ahmad Gebai
- Department of Medicine, Université de Montreal, Montreal, QC H3T 1J4, Canada
| | - Mustapha Tehfe
- Department of Medicine, Université de Montreal, Montreal, QC H3T 1J4, Canada
- Hematology-Oncology, Oncology Center-Centre Hospitalier de l’Universite de Montreal, Montreal, QC H2X 0C1, Canada
| |
Collapse
|
61
|
Klempner SJ, Lee KW, Shitara K, Metges JP, Lonardi S, Ilson DH, Fazio N, Kim TY, Bai LY, Moran D, Yang J, Arozullah A, Park JW, Raizer JJ, Bang YJ, Shah MA. ILUSTRO: Phase II Multicohort Trial of Zolbetuximab in Patients with Advanced or Metastatic Claudin 18.2-Positive Gastric or Gastroesophageal Junction Adenocarcinoma. Clin Cancer Res 2023; 29:3882-3891. [PMID: 37490286 PMCID: PMC10543966 DOI: 10.1158/1078-0432.ccr-23-0204] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/20/2023] [Accepted: 07/20/2023] [Indexed: 07/26/2023]
Abstract
PURPOSE Zolbetuximab, an IgG1 monoclonal antibody, binds to claudin 18.2 (CLDN18.2) and mediates tumor cell death through antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity. We sought to examine zolbetuximab combinations in CLDN18.2-positive HER2-negative gastric/gastroesophageal junction (G/GEJ) adenocarcinoma. PATIENTS AND METHODS This phase II study assessed efficacy and safety of zolbetuximab, alone or with modified FOLFOX6 (mFOLFOX6) or pembrolizumab, in CLDN18.2-positive advanced/metastatic G/GEJ adenocarcinoma. Patients received zolbetuximab as monotherapy in third/later-line (Cohort 1A, n = 30), with mFOLFOX6 in first-line (Cohort 2, n = 21), or with pembrolizumab in third/later-line (Cohort 3A, n = 3) treatment. The primary endpoint for Cohort 1A was objective response rate (ORR). Key secondary endpoints were ORR (Cohorts 2 and 3A), overall survival (OS; Cohort 1A), and progression-free survival (PFS) and safety (all cohorts). RESULTS ORR was 0% in Cohorts 1A and 3A, and 71.4% [95% confidence interval (CI), 47.82-88.72] in Cohort 2. Median PFS was 1.54 months (95% CI, 1.31-2.56) in Cohort 1A, 2.96 months (95% CI, 1.48-4.44) in Cohort 3A, and 17.8 months (95% CI, 8.05-25.69) in Cohort 2. Median OS in Cohort 1A was 5.62 months (95% CI, 2.27-11.53). Gastrointestinal adverse events occurred across cohorts [nausea, 63%-90% (grade ≥ 3, 4.8%-6.7%) and vomiting, 33%-67% (grade ≥ 3, 6.7%-9.5%)]. CONCLUSIONS Zolbetuximab plus mFOLFOX6 demonstrated promising efficacy in previously untreated patients with CLDN18.2-positive G/GEJ adenocarcinoma. These data support the first-line development of zolbetuximab in patients whose tumors are CLDN18.2-positive. Across cohorts, zolbetuximab treatment was tolerable with no new safety signals.
Collapse
Affiliation(s)
- Samuel J. Klempner
- Department of Medicine, Division of Hematology-Oncology, Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Keun-Wook Lee
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, South Korea
| | - Kohei Shitara
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | | | - Sara Lonardi
- Medical Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - David H. Ilson
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nicola Fazio
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO, IRCCS, Milan, Italy
| | - Tae Yong Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Li-Yuan Bai
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, and College of Medicine, China Medical University, Taichung, Taiwan
| | | | | | | | | | - Jeffrey J. Raizer
- Clinical Sciences, Oncology, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts
| | - Yung-Jue Bang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Manish A. Shah
- Division of Hematology and Medical Oncology, Weill Cornell Medical College, New York, New York
| |
Collapse
|
62
|
Wei Z, Zhao X, Chen J, Sun Q, Wang Z, Wang Y, Ye Z, Yuan Y, Sun L, Jing J. Deep Learning-Based Stratification of Gastric Cancer Patients from Hematoxylin and Eosin-Stained Whole Slide Images by Predicting Molecular Features for Immunotherapy Response. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1517-1527. [PMID: 37356573 DOI: 10.1016/j.ajpath.2023.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 06/01/2023] [Accepted: 06/08/2023] [Indexed: 06/27/2023]
Abstract
Determining the molecular characteristics of cancer patients is crucial for optimal immunotherapy decisions. The aim of this study was to screen immunotherapy beneficiaries by predicting key molecular features from hematoxylin and eosin-stained images based on deep learning models. An independent data set from Asian gastric cancer patients was included for external validation. In addition, a segmentation model (Horizontal-Vertical Network) was used to quantify the cellular composition of tumor stroma. The model performance was evaluated by measuring the area under the curve (AUC). The tumor extraction model achieved an AUC of 0.9386 and 0.9062 in the internal and external test sets, respectively. The stratification model could predict the immunotherapy-sensitive subtypes (AUC range, 0.8685 to 0.9461), the genetic mutations (AUC range, 0.8283 to 0.9225), and the pathway activity (AUC range, 0.7568 to 0.8612) fairly accurately. In external validation, the prediction performance of Epstein-Barr virus and programmed cell death ligand 1 expression status achieved AUCs of 0.7906 and 0.6384, respectively. The segmentation model identified a relatively high proportion of inflammatory cells and connective cells in some immunotherapy-sensitive subtypes. The deep learning-based models potentially may serve as a valuable tool to screen for the beneficiaries of immunotherapy in gastric cancer patients.
Collapse
Affiliation(s)
- Zheng Wei
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, China; Key Laboratory of Cancer Etiology and Prevention, Liaoning Education Department, The First Hospital of China Medical University, Shenyang, China; Key Laboratory of GI Cancer Etiology and Prevention, Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - Xu Zhao
- Mathematical Computer Teaching and Research Office, Liaoning Vocational College of Medicine, Shenyang, China
| | - Jing Chen
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, China; Key Laboratory of Cancer Etiology and Prevention, Liaoning Education Department, The First Hospital of China Medical University, Shenyang, China; Key Laboratory of GI Cancer Etiology and Prevention, Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - Qiuyan Sun
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, China; Key Laboratory of Cancer Etiology and Prevention, Liaoning Education Department, The First Hospital of China Medical University, Shenyang, China; Key Laboratory of GI Cancer Etiology and Prevention, Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - Zeyang Wang
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, China; Key Laboratory of Cancer Etiology and Prevention, Liaoning Education Department, The First Hospital of China Medical University, Shenyang, China; Key Laboratory of GI Cancer Etiology and Prevention, Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - Yanli Wang
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, China; Key Laboratory of Cancer Etiology and Prevention, Liaoning Education Department, The First Hospital of China Medical University, Shenyang, China; Key Laboratory of GI Cancer Etiology and Prevention, Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - Zhiyi Ye
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, China; Key Laboratory of Cancer Etiology and Prevention, Liaoning Education Department, The First Hospital of China Medical University, Shenyang, China; Key Laboratory of GI Cancer Etiology and Prevention, Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - Yuan Yuan
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, China; Key Laboratory of Cancer Etiology and Prevention, Liaoning Education Department, The First Hospital of China Medical University, Shenyang, China; Key Laboratory of GI Cancer Etiology and Prevention, Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - Liping Sun
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, China; Key Laboratory of Cancer Etiology and Prevention, Liaoning Education Department, The First Hospital of China Medical University, Shenyang, China; Key Laboratory of GI Cancer Etiology and Prevention, Liaoning Province, The First Hospital of China Medical University, Shenyang, China.
| | - Jingjing Jing
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, China; Key Laboratory of Cancer Etiology and Prevention, Liaoning Education Department, The First Hospital of China Medical University, Shenyang, China; Key Laboratory of GI Cancer Etiology and Prevention, Liaoning Province, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
63
|
Cammarota AL, Falco A, Basile A, Molino C, Chetta M, D’Angelo G, Marzullo L, De Marco M, Turco MC, Rosati A. Pancreatic Cancer-Secreted Proteins: Targeting Their Functions in Tumor Microenvironment. Cancers (Basel) 2023; 15:4825. [PMID: 37835519 PMCID: PMC10571538 DOI: 10.3390/cancers15194825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is a ravaging disease with a poor prognosis, requiring a more detailed understanding of its biology to foster the development of effective therapies. The unsatisfactory results of treatments targeting cell proliferation and its related mechanisms suggest a shift in focus towards the inflammatory tumor microenvironment (TME). Here, we discuss the role of cancer-secreted proteins in the complex TME tumor-stroma crosstalk, shedding lights on druggable molecular targets for the development of innovative, safer and more efficient therapeutic strategies.
Collapse
Affiliation(s)
- Anna Lisa Cammarota
- Department of Medicine, Surgery and Dentistry “Schola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (A.L.C.); (A.F.); (A.B.); (L.M.); (M.C.T.)
| | - Antonia Falco
- Department of Medicine, Surgery and Dentistry “Schola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (A.L.C.); (A.F.); (A.B.); (L.M.); (M.C.T.)
| | - Anna Basile
- Department of Medicine, Surgery and Dentistry “Schola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (A.L.C.); (A.F.); (A.B.); (L.M.); (M.C.T.)
| | - Carlo Molino
- General Surgery Unit, A.O.R.N. Cardarelli, 80131 Naples, Italy;
| | - Massimiliano Chetta
- Medical and Laboratory Genetics Unit, A.O.R.N., Cardarelli, 80131 Naples, Italy;
| | - Gianni D’Angelo
- Department of Computer Science, University of Salerno, 84084 Fisciano, Italy;
| | - Liberato Marzullo
- Department of Medicine, Surgery and Dentistry “Schola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (A.L.C.); (A.F.); (A.B.); (L.M.); (M.C.T.)
- FIBROSYS s.r.l., University of Salerno, 84081 Baronissi, Italy
| | - Margot De Marco
- Department of Medicine, Surgery and Dentistry “Schola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (A.L.C.); (A.F.); (A.B.); (L.M.); (M.C.T.)
- FIBROSYS s.r.l., University of Salerno, 84081 Baronissi, Italy
| | - Maria Caterina Turco
- Department of Medicine, Surgery and Dentistry “Schola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (A.L.C.); (A.F.); (A.B.); (L.M.); (M.C.T.)
- FIBROSYS s.r.l., University of Salerno, 84081 Baronissi, Italy
| | - Alessandra Rosati
- Department of Medicine, Surgery and Dentistry “Schola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (A.L.C.); (A.F.); (A.B.); (L.M.); (M.C.T.)
- FIBROSYS s.r.l., University of Salerno, 84081 Baronissi, Italy
| |
Collapse
|
64
|
Nakamura H, Shionoya A, Arihara Y, Hayasaka N, Kubo T, Usami M, Sugita S, Uhara H, Takada K. Pemphigus vulgaris as an immune-related adverse event in recurrent metastatic esophageal squamous cell carcinoma treated with ipilimumab plus nivolumab: a case report and literature review. Front Immunol 2023; 14:1259071. [PMID: 37753079 PMCID: PMC10518453 DOI: 10.3389/fimmu.2023.1259071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 08/28/2023] [Indexed: 09/28/2023] Open
Abstract
Ipilimumab plus nivolumab therapy is approved for patients with unresectable advanced esophageal squamous cell carcinoma (ESCC). Although a combination of immune checkpoint inhibitors (ICIs), compared to conventional chemotherapy, can improve overall survival in patients with advanced ESCC, this increases the incidence of immune-related adverse events (irAEs). Here, we describe an ESCC case that developed pemphigus vulgaris (PV), an extremely rare cutaneous irAE, during ipilimumab plus nivolumab treatment. The patient achieved a partial response to treatment. The PV was successfully managed after the cessation of ipilimumab and the use of a topical steroid. We should thus re-treat ESCC with nivolumab monotherapy. In the era of ICIs as standard cancer therapeutics, diagnostic criteria for blistering diseases need to be established to properly manage patients with cutaneous irAEs.
Collapse
Affiliation(s)
- Hajime Nakamura
- Department of Medical Oncology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Aika Shionoya
- Department of Dermatology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yohei Arihara
- Department of Medical Oncology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Naotaka Hayasaka
- Department of Medical Oncology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tomohiro Kubo
- Department of Medical Oncology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Makoto Usami
- Department of Medical Oncology, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Medical Oncology, Steel Memorial Muroran Hospital, Muroran, Japan
| | - Shintaro Sugita
- Department of Surgical Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hisashi Uhara
- Department of Dermatology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kohichi Takada
- Department of Medical Oncology, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
65
|
Huang Y, You M, Wu Q, Chen R. Cost-effectiveness analysis of zolbetuximab plus mFOLFOX6 as the first-line treatment for CLDN18.2-positive, HER2-negative advanced gastric or Gastroesophageal Adenocarcinoma. Front Pharmacol 2023; 14:1238009. [PMID: 37719841 PMCID: PMC10500349 DOI: 10.3389/fphar.2023.1238009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 07/27/2023] [Indexed: 09/19/2023] Open
Abstract
Background: The SPOTLIGHT trial demonstrated that zolbetuximab plus mFOLFOX6 (ZOL-FO) as a first-line regimen compared with placebo plus mFOLFOX6 (PLB-FO) conferred clinical benefits to patients with CLDN18.2-positive, HER2-negative advanced gastric or gastroesophageal junction (G/GEJ) adenocarcinoma. However, due to the high cost of zolbetuximab, whether ZOL-FO is cost-effective compared with PLB-FO is unclear. This study aimed to evaluate the cost-effectiveness of ZOL-FO as a first-line treatment option for CLDN18.2-positive, HER2-negative advanced G/GEJ adenocarcinoma from the perspective of the Chinese healthcare system. Methods: Markov models with three different health states were developed to assess the cost-effectiveness of ZOL-FO as a first-line treatment option for CLDN18.2-positive, HER2-negative advanced G/GEJ adenocarcinoma. Clinical efficacy data were obtained from the SPOTLIGHT trial; the drug's cost was calculated at national bid prices, and other costs and utility values were obtained from the published literature. Outcomes included total costs, quality-adjusted life years (QALYs), and incremental cost-effectiveness ratios (ICERs). The model's robustness was verified using one-way sensitivity and probabilistic sensitivity analyses. Results: The ZOL-FO group gained 1.64 QALYs at $87,746.35, while the PLB-FO group gained 1.23 QALYs at $11,947.81. The ICER for ZOL-FO versus PLB-FO was $185,353.28 per QALY gained. The parameters exerting an important impact on the model results were the price of zolbetuximab, body surface area, and progression-free survival utility. At a willingness-to-pay threshold of $38,201/QALY, ZOL-FO had a 0% probability of cost-effectiveness compared with PLB-FO. Conclusion: From the perspective of the Chinese healthcare system, ZOL-FO is unlikely to be cost-effective as the first-line treatment option for CLDN18.2-positive, HER2-negative advanced G/GEJ adenocarcinoma.
Collapse
Affiliation(s)
- Yufan Huang
- Department of Pharmacy, Mindong Hospital Affiliated to Fujian Medical University, Ningde, Fujian, China
| | - Maojin You
- Department of Pharmacy, Mindong Hospital Affiliated to Fujian Medical University, Ningde, Fujian, China
| | - Qundan Wu
- Department of Pharmacy, Quanzhou Skin Disease Prevention and Treatment Hospital, Quanzhou, Fujian, China
| | - Ruijia Chen
- Department of Pharmacy, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
66
|
Shah MA, Shitara K, Ajani JA, Bang YJ, Enzinger P, Ilson D, Lordick F, Van Cutsem E, Gallego Plazas J, Huang J, Shen L, Oh SC, Sunpaweravong P, Soo Hoo HF, Turk HM, Oh M, Park JW, Moran D, Bhattacharya P, Arozullah A, Xu RH. Zolbetuximab plus CAPOX in CLDN18.2-positive gastric or gastroesophageal junction adenocarcinoma: the randomized, phase 3 GLOW trial. Nat Med 2023; 29:2133-2141. [PMID: 37524953 PMCID: PMC10427418 DOI: 10.1038/s41591-023-02465-7] [Citation(s) in RCA: 115] [Impact Index Per Article: 115.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 06/15/2023] [Indexed: 08/02/2023]
Abstract
There is an urgent need for first-line treatment options for patients with human epidermal growth factor receptor 2 (HER2)-negative, locally advanced unresectable or metastatic gastric or gastroesophageal junction (mG/GEJ) adenocarcinoma. Claudin-18 isoform 2 (CLDN18.2) is expressed in normal gastric cells and maintained in malignant G/GEJ adenocarcinoma cells. GLOW (closed enrollment), a global, double-blind, phase 3 study, examined zolbetuximab, a monoclonal antibody that targets CLDN18.2, plus capecitabine and oxaliplatin (CAPOX) as first-line treatment for CLDN18.2-positive, HER2-negative, locally advanced unresectable or mG/GEJ adenocarcinoma. Patients (n = 507) were randomized 1:1 (block sizes of two) to zolbetuximab plus CAPOX or placebo plus CAPOX. GLOW met the primary endpoint of progression-free survival (median, 8.21 months versus 6.80 months with zolbetuximab versus placebo; hazard ratio (HR) = 0.687; 95% confidence interval (CI), 0.544-0.866; P = 0.0007) and key secondary endpoint of overall survival (median, 14.39 months versus 12.16 months; HR = 0.771; 95% CI, 0.615-0.965; P = 0.0118). Grade ≥3 treatment-emergent adverse events were similar with zolbetuximab (72.8%) and placebo (69.9%). Zolbetuximab plus CAPOX represents a potential new first-line therapy for patients with CLDN18.2-positive, HER2-negative, locally advanced unresectable or mG/GEJ adenocarcinoma. ClinicalTrials.gov identifier: NCT03653507 .
Collapse
Affiliation(s)
- Manish A Shah
- Weill Cornell Medical College, New York City, NY, USA
| | - Kohei Shitara
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa City, Japan
| | - Jaffer A Ajani
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yung-Jue Bang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Peter Enzinger
- Center for Esophageal and Gastric Cancer, Dana-Farber Cancer Institute, Boston, MA, USA
| | - David Ilson
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Florian Lordick
- Department of Medicine and University Cancer Center Leipzig, University of Leipzig Medical Center, Leipzig, Germany
| | - Eric Van Cutsem
- Digestive Oncology, University Hospitals Gasthuisberg, Leuven and KULeuven, Leuven, Belgium
| | - Javier Gallego Plazas
- Department of Medical Oncology, Hospital General Universitario de Elche, Elche, Spain
| | - Jing Huang
- Department of Medical Oncology, National Cancer Center / National Clinical Research Center for Cancer / Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Sang Cheul Oh
- Department of Internal Medicine, Korea University Guro Hospital, Seoul, Republic of Korea
| | - Patrapim Sunpaweravong
- Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Hwoei Fen Soo Hoo
- Department of Oncology and Radiotherapy, Penang Hospital, Penang, Malaysia
| | - Haci Mehmet Turk
- Department of Medical Oncology, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
| | - Mok Oh
- Astellas Pharma Global Development, Inc., Northbrook, IL, USA
| | - Jung Wook Park
- Astellas Pharma Global Development, Inc., Northbrook, IL, USA
| | - Diarmuid Moran
- Astellas Pharma Global Development, Inc., Northbrook, IL, USA
| | | | - Ahsan Arozullah
- Astellas Pharma Global Development, Inc., Northbrook, IL, USA
| | - Rui-Hua Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
67
|
Shitara K, Lordick F, Bang YJ, Enzinger P, Ilson D, Shah MA, Van Cutsem E, Xu RH, Aprile G, Xu J, Chao J, Pazo-Cid R, Kang YK, Yang J, Moran D, Bhattacharya P, Arozullah A, Park JW, Oh M, Ajani JA. 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. [PMID: 37068504 DOI: 10.1016/s0140-6736(23)00620-7] [Citation(s) in RCA: 192] [Impact Index Per Article: 192.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/17/2023] [Accepted: 03/19/2023] [Indexed: 04/19/2023]
Abstract
BACKGROUND Zolbetuximab, a monoclonal antibody targeting claudin-18 isoform 2 (CLDN18.2), has shown efficacy in patients with CLDN18.2-positive, human epidermal growth factor receptor 2 (HER2)-negative, locally advanced unresectable or metastatic gastric or gastro-oesophageal junction adenocarcinoma. We report the results of the SPOTLIGHT trial, which investigated the efficacy and safety of first-line zolbetuximab plus mFOLFOX6 (modified folinic acid [or levofolinate], fluorouracil, and oxaliplatin regimen) versus placebo plus mFOLFOX6 in patients with CLDN18.2-positive, HER2-negative, locally advanced unresectable or metastatic gastric or gastro-oesophageal junction adenocarcinoma. METHODS SPOTLIGHT is a global, randomised, placebo-controlled, double-blind, phase 3 trial that enrolled patients from 215 centres in 20 countries. Eligible patients were aged 18 years or older with CLDN18.2-positive (defined as ≥75% of tumour cells showing moderate-to-strong membranous CLDN18 staining), HER2-negative (based on local or central evaluation), previously untreated, locally advanced unresectable or metastatic gastric or gastro-oesophageal junction adenocarcinoma, with radiologically evaluable disease (measurable or non-measurable) according to Response Evaluation Criteria in Solid Tumors version 1.1; an Eastern Cooperative Oncology Group performance status score of 0 or 1; and adequate organ function. Patients were randomly assigned (1:1) via interactive response technology and stratified according to region, number of organs with metastases, and previous gastrectomy. Patients received zolbetuximab (800 mg/m2 loading dose followed by 600 mg/m2 every 3 weeks) plus mFOLFOX6 (every 2 weeks) or placebo plus mFOLFOX6. The primary endpoint was progression-free survival assessed by independent review committee in all randomly assigned patients. Safety was assessed in all treated patients. The study is registered with ClinicalTrials.gov, NCT03504397, and is closed to new participants. FINDINGS Between June 21, 2018, and April 1, 2022, 565 patients were randomly assigned to receive either zolbetuximab plus mFOLFOX6 (283 patients; the zolbetuximab group) or placebo plus mFOLFOX6 (282 patients; the placebo group). At least one dose of treatment was administered to 279 (99%) of 283 patients in the zolbetuximab group and 278 (99%) of 282 patients in the placebo group. In the zolbetuximab group, 176 (62%) patients were male and 107 (38%) were female. In the placebo group, 175 (62%) patients were male and 107 (38%) were female. The median follow-up duration for progression-free survival was 12·94 months in the zolbetuximab group versus 12·65 months in the placebo group. Zolbetuximab treatment showed a significant reduction in the risk of disease progression or death compared with placebo (hazard ratio [HR] 0·75, 95% CI 0·60-0·94; p=0·0066). The median progression-free survival was 10·61 months (95% CI 8·90-12·48) in the zolbetuximab group versus 8·67 months (8·21-10·28) in the placebo group. Zolbetuximab treatment also showed a significant reduction in the risk of death versus placebo (HR 0·75, 95% CI 0·60-0·94; p=0·0053). Treatment-emergent grade 3 or worse adverse events occurred in 242 (87%) of 279 patients in the zolbetuximab group versus 216 (78%) of 278 patients in the placebo group. The most common grade 3 or worse adverse events were nausea, vomiting, and decreased appetite. Treatment-related deaths occurred in five (2%) patients in the zolbetuximab group versus four (1%) patients in the placebo group. No new safety signals were identified. INTERPRETATION Targeting CLDN18.2 with zolbetuximab significantly prolonged progression-free survival and overall survival when combined with mFOLFOX6 versus placebo plus mFOLFOX6 in patients with CLDN18.2-positive, HER2-negative, locally advanced unresectable or metastatic gastric or gastro-oesophageal junction adenocarcinoma. Zolbetuximab plus mFOLFOX6 might represent a new first-line treatment in these patients. FUNDING Astellas Pharma, Inc.
Collapse
Affiliation(s)
- Kohei Shitara
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa City, Japan
| | - Florian Lordick
- Department of Medicine and University Cancer Center Leipzig, University of Leipzig Medical Center, Leipzig, Germany
| | - Yung-Jue Bang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Peter Enzinger
- Center for Esophageal and Gastric Cancer, Dana-Farber Cancer Institute, Boston, MA, USA
| | - David Ilson
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Manish A Shah
- Weill Cornell Medical College, New York City, NY, USA
| | - Eric Van Cutsem
- Department of Digestive Oncology, University Hospitals Gasthuisberg, Leuven, and KULeuven, Leuven, Belgium
| | - Rui-Hua Xu
- Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Giuseppe Aprile
- Department of Oncology, Azienda ULSS 8 Berica, Veneto, Italy
| | - Jianming Xu
- Department of Gastrointestinal Oncology, The Fifth Medical Center of the PLA General Hospital, Beijing, China
| | - Joseph Chao
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | | | - Yoon-Koo Kang
- Department of Oncology, Asan Medical Center, University of Ulsan, Seoul, South Korea
| | - Jianning Yang
- Astellas Pharma Global Development, Inc, Northbrook, IL, USA
| | - Diarmuid Moran
- Astellas Pharma Global Development, Inc, Northbrook, IL, USA
| | | | - Ahsan Arozullah
- Astellas Pharma Global Development, Inc, Northbrook, IL, USA
| | - Jung Wook Park
- Astellas Pharma Global Development, Inc, Northbrook, IL, USA
| | - Mok Oh
- Astellas Pharma Global Development, Inc, Northbrook, IL, USA
| | - Jaffer A Ajani
- The University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
68
|
Gong X, Hou D, Zhou S, Tan J, Zhong G, Yang B, Xie L, Han F, Zhong L. FMO family may serve as novel marker and potential therapeutic target for the peritoneal metastasis in gastric cancer. Front Oncol 2023; 13:1144775. [PMID: 37274237 PMCID: PMC10234505 DOI: 10.3389/fonc.2023.1144775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 04/18/2023] [Indexed: 06/06/2023] Open
Abstract
Objective To explore the relationship between flavin-containing monooxygenases (FMOs) and peritoneal metastasis (PM) in gastric cancer (GC). Materials and methods TIMER 2.0 was used to perform pan-cancer analysis and assess the correlation between the expression of FMOs and cancers. A dataset from The Cancer Genome Atlas (TCGA) was used to analyze the correlation between FMOs and clinicopathological features of GC. PM is well established as the most common mode of metastasis in GC. To further analyze the correlation between FMOs and PM of GC, a dataset was obtained from the National Center for Biotechnology Information Gene Expression Omnibus (GEO) database. The results were validated by immunohistochemistry. The relationship between FMOs and PM of GC was explored, and a novel PM risk signature was constructed by least absolute shrinkage and selection operator (LASSO) regression analysis. The regression model's validity was tested by multisampling. A nomogram was established based on the model for predicting PM in GC patients. The mechanism of FMOs in GC patients presenting with PM was assessed by conducting Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses in TCGA and GEO datasets. Finally, the potential relationship between FMOs and immunotherapy was analyzed. Results The pan-cancer analysis in TCGA and GEO datasets showed that FMO1 was upregulated, while FMO2 and FMO4 were downregulated in GC. Moreover, FMO1 and FMO2 correlated positively with the T and N stage of GC in the TCGA dataset. FMO1 and FMO2 expression was a risk factor for GC (hazard ratio: 1.112 and 1.185). The overexpression of FMO1 was significantly correlated with worse disease-free-survival (DFS) and overall survival (OS). However, no relationship was found between FMO2 expression in GC and DFS and OS. PM was highly prevalent among GC patients and typically associated with a worse prognosis. FMO1 was highly expressed in GC with PM. FMO1 and FMO2 were positively correlated with PM in GC. We identified a 12-gene panel for predicting the PM risk signature by LASSO (Area Under Curve (AUC) = 0.948, 95%CI: 0.896-1.000). A 10-gene panel for PM prediction was identified (AUC = 0.932, 95%CI: 0.874-0.990), comprising FMO1 and FMO2. To establish a model for clinical application, a 7-gene panel was established (AUC = 0.927, 95% CI: 0.877-0.977) and successfully validated by multisampling. (AUC = 0.892, 95% CI: 0.878-0.906). GO and KEGG analyses suggest that FMO1 and FMO2 regulate the extracellular matrix and cell adhesion. FMO1 and FMO2 were positively correlated with the immune score of GC, and their expression was associated with the infiltration of immune cells. Conclusion PM in GC is strongly correlated with FMOs. Overall, FMO1 and FMO2 have huge prospects for application as novel diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Xumeng Gong
- Department of Surgical Oncology, Yuebei People’s Hospital, Shaoguan, Guangdong, China
- Department of Head-Neck and Breast Surgery, Yuebei People’s Hospital of Shantou University, Shaoguan, Guangdong, China
| | - Dong Hou
- Department of Gastrointestinal Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shengning Zhou
- Department of Gastrointestinal Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jianan Tan
- Department of Gastrointestinal Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Guangyu Zhong
- Department of Gastrointestinal Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Bing Yang
- Department of Gastrointestinal Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Lang Xie
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Fanghai Han
- Department of Gastrointestinal Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Lin Zhong
- Department of Gastrointestinal Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
69
|
Li JJ, Rogers JE, Yamashita K, Waters RE, Blum Murphy M, Ajani JA. Therapeutic Advances in the Treatment of Gastroesophageal Cancers. Biomolecules 2023; 13:biom13050796. [PMID: 37238666 DOI: 10.3390/biom13050796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023] Open
Abstract
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. Multimodality therapy is needed in the localized setting and treatment decisions require multidisciplinary discussions. Systemic therapies for treatment of advanced/metastatic disease should be biomarker-driven, when appropriate. Current FDA approved treatments include HER2-targeted therapy, immunotherapy, and chemotherapy. However, novel therapeutic targets are under development and future treatments will be personalized based on molecular profiling. Herein, we review the current treatment approaches and discuss promising advances in targeted therapies for gastroesophageal cancers.
Collapse
Affiliation(s)
- Jenny J Li
- Department of Gastrointestinal Medical Oncology, University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Jane E Rogers
- Department of Pharmacy Clinical Program, University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Kohei Yamashita
- Department of Gastrointestinal Medical Oncology, University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Rebecca E Waters
- Department of Pathology, Division of Pathology/Lab Medicine, University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Mariela Blum Murphy
- Department of Gastrointestinal Medical Oncology, University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| |
Collapse
|
70
|
Shah MA, Altorki N, Patel P, Harrison S, Bass A, Abrams JA. Improving outcomes in patients with oesophageal cancer. Nat Rev Clin Oncol 2023; 20:390-407. [PMID: 37085570 DOI: 10.1038/s41571-023-00757-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2023] [Indexed: 04/23/2023]
Abstract
The care of patients with oesophageal cancer or of individuals who have an elevated risk of oesophageal cancer has changed dramatically. The epidemiology of squamous cell and adenocarcinoma of the oesophagus has diverged over the past several decades, with a marked increase in incidence only for oesophageal adenocarcinoma. Only in the past decade, however, have molecular features that distinguish these two forms of the disease been identified. This advance has the potential to improve screening for oesophageal cancers through the development of novel minimally invasive diagnostic technologies predicated on cancer-specific genomic or epigenetic alterations. Surgical techniques have also evolved towards less invasive approaches associated with less morbidity, without compromising oncological outcomes. With improvements in multidisciplinary care, advances in radiotherapy and new tools to detect minimal residual disease, certain patients may no longer even require surgical tumour resection. However, perhaps the most anticipated advance in the treatment of patients with oesophageal cancer is the advent of immune-checkpoint inhibitors, which harness and enhance the host immune response against cancer. In this Review, we discuss all these advances in the management of oesophageal cancer, representing only the beginning of a transformation in our quest to improve patient outcomes.
Collapse
Affiliation(s)
- Manish A Shah
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| | - Nasser Altorki
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Pretish Patel
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Sebron Harrison
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Adam Bass
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Julian A Abrams
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
71
|
Maeng CH. Clinical Implication of Liver Metastasis in the Treatment of Gastric Cancer. J Gastric Cancer 2023; 23:251-252. [PMID: 37129149 PMCID: PMC10154135 DOI: 10.5230/jgc.2023.23.e21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/03/2023] Open
Affiliation(s)
- Chi Hoon Maeng
- Division of Medical Oncology-Hematology, Department of Internal Medicine, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, Korea.
| |
Collapse
|
72
|
Liu S, Dou L, Li S. Cost-effectiveness analysis of PD-1 inhibitors combined with chemotherapy as first-line therapy for advanced esophageal squamous-cell carcinoma in China. Front Pharmacol 2023; 14:1055727. [PMID: 36937861 PMCID: PMC10017726 DOI: 10.3389/fphar.2023.1055727] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
Objective: This study was aimed to investigate the cost-effectiveness of all available programmed death 1 (PD-1) inhibitors combined with chemotherapy in the first-line treatment of advanced esophageal squamous-cell carcinoma (ESCC) from the Chinese healthcare system perspective. Methods: A partitioned survival model with a 3-week cycle and a 10-year time horizon was constructed based on a network meta-analysis. The survival data and utility values were derived from clinical trials, and the direct medical costs were collected from public drug bidding database and published literature. Total costs, quality-adjusted life-years (QALYs) and incremental cost-effectiveness ratios (ICERs) were calculated. Scenario, one-way and probabilistic sensitivity analyses were performed to assess the uncertainty around model parameters. Results: Compared with mono-chemotherapy, toripalimab, sintilimab and camrelizumab plus chemotherapy were cost-effective treatment regimens, while serplulimab, pembrolizumab and nivolumab plus chemotherapy were not cost-effective options. Toripalimab plus chemotherapy provided the highest QALYs of 0.95 with the lower cost of $8,110.53 compared to other competing alternatives. The robustness of the base-case results was confirmed by scenario and one-way sensitivity analysis. At a willingness-to-pay threshold of three times per capita gross domestic product ($38,351.20) in 2021, the probability of toripalimab plus chemotherapy being the optimal option was 74.25% compared with other six competing alternatives. Conclusion: Toripalimab plus chemotherapy represented the most cost-effective option as the first-line therapy for advanced ESCC patients in China.
Collapse
Affiliation(s)
- Shixian Liu
- Centre for Health Management and Policy Research, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- NHC Key Laboratory of Health Economics and Policy Research (Shandong University), Jinan, China
- Center for Health Preference Research, Shandong University, Jinan, China
| | - Lei Dou
- Centre for Health Management and Policy Research, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- NHC Key Laboratory of Health Economics and Policy Research (Shandong University), Jinan, China
- Center for Health Preference Research, Shandong University, Jinan, China
| | - Shunping Li
- Centre for Health Management and Policy Research, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- NHC Key Laboratory of Health Economics and Policy Research (Shandong University), Jinan, China
- Center for Health Preference Research, Shandong University, Jinan, China
| |
Collapse
|
73
|
Lavacchi D, Fancelli S, Buttitta E, Vannini G, Guidolin A, Winchler C, Caliman E, Vannini A, Giommoni E, Brugia M, Cianchi F, Pillozzi S, Roviello G, Antonuzzo L. Perioperative Tailored Treatments for Gastric Cancer: Times Are Changing. Int J Mol Sci 2023; 24:4877. [PMID: 36902306 PMCID: PMC10003389 DOI: 10.3390/ijms24054877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/24/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
Resectable gastric or gastroesophageal (G/GEJ) cancer is a heterogeneous disease with no defined molecularly based treatment strategy. Unfortunately, nearly half of patients experience disease recurrence despite standard treatments (neoadjuvant and/or adjuvant chemotherapy/chemoradiotherapy and surgery). In this review, we summarize the evidence of potential tailored approaches in perioperative treatment of G/GEJ cancer, with a special focus on patients with human epidermal growth factor receptor-2(HER2)-positive and microsatellite instability-high (MSI-H) tumors. In patients with resectable MSI-H G/GEJ adenocarcinoma, the ongoing INFINITY trial introduces the concept of non-operative management for patients with complete clinical-pathological-molecular response, and this could be a novel and potential practice changing strategy. Other pathways involving vascular endothelial growth factor receptor (VEGFR), fibroblast growth factor receptor (FGFR), claudin18 isoform 2 (CLDN18.2), and DNA damage repair proteins are also described, with limited evidence until now. Although tailored therapy appears to be a promising strategy for resectable G/GEJ cancer, there are several methodological issues to address: inadequate sample size for pivotal trials, underestimation of subgroup effects, and choice of primary endpoint (tumor-centered vs. patient-centered endpoints). A better optimization of G/GEJ cancer treatment allows maximizing patient outcomes. In the perioperative phase, although caution is mandatory, times are changing and tailored strategies could introduce new treatment concepts. Overall, MSI-H G/GEJ cancer patients possess the characteristics to be the subgroup that could receive the most benefit from a tailored approach.
Collapse
Affiliation(s)
- Daniele Lavacchi
- Clinical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Sara Fancelli
- Clinical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Eleonora Buttitta
- Clinical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Gianmarco Vannini
- Clinical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Alessia Guidolin
- Clinical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Costanza Winchler
- Clinical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Enrico Caliman
- Clinical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Agnese Vannini
- Clinical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Elisa Giommoni
- Medical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Marco Brugia
- Medical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Fabio Cianchi
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
- Unit of Digestive Surgery, Careggi University Hospital, 50134 Florence, Italy
| | - Serena Pillozzi
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
- Medical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| | | | - Lorenzo Antonuzzo
- Clinical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
- Medical Oncology Unit, Careggi University Hospital, 50134 Florence, Italy
| |
Collapse
|
74
|
Kawaguchi D, Kawakami T, Kakuda Y, Yamazaki K. Rare Case of Advanced Gastric Cancer Complicated with Fibrinogen Storage Disease Treated with Chemotherapy plus Immune Checkpoint Inhibitor: A Case Report. Case Rep Oncol 2023; 16:1267-1273. [PMID: 37928864 PMCID: PMC10622163 DOI: 10.1159/000534145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 09/12/2023] [Indexed: 11/07/2023] Open
Abstract
The administration of chemotherapy to cancer patients with organ dysfunction raises concerns regarding its safety. The safety profile of patients with organ dysfunction due to rare diseases treated with chemotherapy plus immune checkpoint inhibitor is limited. Fibrinogen storage disease (FSD) is a rare disease that causes liver dysfunction through endoplasmic reticulum stress response due to abnormal accumulation of fibrinogen in the endoplasmic reticulum of hepatocytes. Although chemotherapy plus nivolumab is recommended as a standard first-line treatment for patients with advanced gastric cancer (AGC), its safety profile for patients with FSD is rarely available. In this study, an 80-year-old male with gastric cancer with positive lavage cytology was scheduled to receive palliative chemotherapy. This case had liver dysfunction of unknown cause, and a liver biopsy was performed. Histopathological findings revealed a diagnosis of type II/III fibrinogen inclusion based on morphology and immunohistochemistry. Liver function was recovered by administering ursodeoxycholic acid. Therefore, the combination chemotherapy of S-1, oxaliplatin, with nivolumab as palliative chemotherapy was initiated. The case responded well to chemotherapy and achieved conversion surgery without worsening of liver function. We report a case of AGC with fibrinogen inclusion complication where chemotherapy was safely administered with a good outcome. The combination therapy of cytotoxic drugs and immune checkpoint inhibitors may be safely and effectively administered to such patients.
Collapse
Affiliation(s)
- Daiki Kawaguchi
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Nagaizumi, Japan
- Division of Gastroenterology, Shizuoka City Shizuoka Hospital, Shizuoka, Japan
| | - Takeshi Kawakami
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Nagaizumi, Japan
| | - Yuko Kakuda
- Division of Pathology, Shizuoka Cancer Center, Nagaizumi, Japan
| | - Kentaro Yamazaki
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Nagaizumi, Japan
| |
Collapse
|
75
|
Qu J, He X, Luo Y, Yu P, Chen Y, Liu J, Wang X, Wang C, Liang T, Bai Y, Han Y, Man L, Leng C, Zhou C, He L, Wang X, Liu Y, Qu X. Evaluation of second-line apatinib plus irinotecan as a treatment for advanced gastric adenocarcinoma or gastroesophageal conjunction adenocarcinoma: a prospective, multicenter phase II trial. Front Oncol 2023; 13:1072943. [PMID: 37168383 PMCID: PMC10166633 DOI: 10.3389/fonc.2023.1072943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 03/27/2023] [Indexed: 05/13/2023] Open
Abstract
Objective Apatinib and irinotecan are used as systematic therapies for advanced gastric adenocarcinoma (GAC) and gastroesophageal junction adenocarcinoma (GEJA), while the evidence for their combination as second-line therapy in these patients is limited. This study aimed to evaluate the efficacy and safety of second-line apatinib plus irinotecan for the treatment of GAC and GEJA. Methods In this prospective, multicenter phase II clinical study, 28 patients with advanced GAC or GEJA who received second-line apatinib plus irinotecan were recruited. Results In total, 1 (3.6%) patient achieved complete response, 7 (25.0%) patients achieved partial response, 13 (46.4%) patients had stable disease, and 4 (14.3%) patients showed progressive disease, while clinical response was not evaluable or not assessed in 3 (10.7%) patients. The objective response rate and disease control rate were 28.6% and 75.0%, respectively. Meanwhile, the median (95% confidence interval (CI)) progression-free survival (PFS) was 4.5 (3.9-5.1) months, and the median (95% CI) overall survival (OS) was 11.3 (7.4-15.1) months. By multivariate Cox regression analysis, male sex, liver metastasis, and peritoneal metastasis were independently associated with worse PFS or OS, while treatment duration ≥5 months was independently associated with better OS. In terms of the safety profile, 89.3% of patients experienced treatment-emergent adverse events of any grade, among which 82.1% of patients had grade 1-2 adverse events and 64.3% of patients had grade 3-4 adverse events. Conclusion Apatinib plus irinotecan as second-line therapy achieves a good treatment response and satisfactory survival with tolerable safety in patients with advanced GAC or GEJA.
Collapse
Affiliation(s)
- Jinglei Qu
- Department of Medical Oncology, The First Hospital of China Medical University, China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Liaoning Province Clinical Research Center for Cancer, Shenyang, China
- Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, China
| | - Xin He
- Department of Medical Oncology, The First Hospital of China Medical University, China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Liaoning Province Clinical Research Center for Cancer, Shenyang, China
- Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, China
| | - Ying Luo
- Department of Medical Oncology, The First Hospital of China Medical University, China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Liaoning Province Clinical Research Center for Cancer, Shenyang, China
- Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, China
| | - Ping Yu
- Department of Medical Oncology, The First Hospital of China Medical University, China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Liaoning Province Clinical Research Center for Cancer, Shenyang, China
- Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, China
| | - Ying Chen
- Department of Medical Oncology, The First Hospital of China Medical University, China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Liaoning Province Clinical Research Center for Cancer, Shenyang, China
- Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, China
| | - Jing Liu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xin Wang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Chang Wang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Tingting Liang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Yuxian Bai
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yu Han
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Li Man
- Department of Medical Oncology, Anshan Cancer Hospital, Anshan, China
| | - Chuanchun Leng
- Department of Medical Oncology, The Central Hospital of Anshan, Anshan, China
| | - Caiyun Zhou
- The Fourth Oncology Departments, Huludao Central Hospital, Huludao, China
| | - Lijie He
- Department of Medical Oncology, People’s Hospital of Liaoning Province, Shenyang, China
| | - Xin Wang
- Department of Medical Oncology, General Hospital of Benxi Iron and Steel Industry Group of Liaoning Health Industry Group, Benxi, China
| | - Yunpeng Liu
- Department of Medical Oncology, The First Hospital of China Medical University, China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Liaoning Province Clinical Research Center for Cancer, Shenyang, China
- Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, China
- *Correspondence: Xiujuan Qu, ; Yunpeng Liu,
| | - Xiujuan Qu
- Department of Medical Oncology, The First Hospital of China Medical University, China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Liaoning Province Clinical Research Center for Cancer, Shenyang, China
- Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, China
- *Correspondence: Xiujuan Qu, ; Yunpeng Liu,
| |
Collapse
|