1
|
He M, Ji C, Li Z, Chen S, Gao J, Shen L, Zhang C. Circulating tumor DNA predicts clinical benefits of immune checkpoint blockade in HER2-negative patients with advanced gastric cancer. Gastric Cancer 2025:10.1007/s10120-025-01621-x. [PMID: 40372586 DOI: 10.1007/s10120-025-01621-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 04/29/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are becoming more prominent in the treatment of gastric cancer (GC). However, predictive biomarkers of response to ICIs in HER2-negative patients remain incompletely understood. METHODS A total of 47 patients diagnosed with HER2-negative advanced GC who underwent ICI regimens were eligible for this study. Plasma samples with paired white blood cells prior to treatments were collected from these 47 patients. Variations of circulating tumor DNA (ctDNA) was evaluated by next-generation sequencing followed by its significance analysis. RESULTS A total of 658 somatic mutations involving 203 genes were identified in all ctDNA. Mutations in MEN1, MLH1, CEBPA, ATR, GNAQ, and FOXL2 genes were more frequent in responders (P < 0.05). Compared with wild-type patients, patients with CEBPA or IRS2 mutations had prolonged median progression-free survival (mPFS, P = 0.0056). Patients with co-occurring mutations in IRS2/CEBPA, IRS2/POLD1, TP53/PIK3CA, or POLD1/CEBPA had longer mPFS compared with others (P = 0.003; 0.006; 0.0166; 0.0315; respectively). Both alteration of CDKN2A alone and co-mutations with MSH6 were significantly associated with superior overall survival (OS, P = 0.0289; 0.0355; respectively). In addition, higher on-treatment ctDNA concentration or variant allele frequency (VAF) were associated with poorer response (P < 0.05). Additionally, the increased molecular alterations of POLE, FGFR2 and MDC1 seemed to indicate the acquired resistance to ICIs. CONCLUSIONS Variation signatures captured by ctDNA as well as the kinetics of ctDNA could predict the clinical benefits of ICB in HER2-negative GC patients, which was worth further validated in large cohort.
Collapse
Affiliation(s)
- Mei He
- Department of Oncology, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen-Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China
| | - Congcong Ji
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Zhiwei Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Shiqing Chen
- Department of Clinical and Translational Medicine, 3D Medicines Inc., Shanghai, 201114, China
| | - Jing Gao
- Department of Oncology, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen-Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China
| | - Lin Shen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Cheng Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
2
|
Meric-Bernstam F, Rha SY, Hamilton E, Kang YK, Hanna DL, Iqbal S, Lee KW, Lee J, Beeram M, Oh DY, Chaves J, Goodwin RA, Ajani JA, Yang L, Oza R, Elimova E. Zanidatamab monotherapy or combined with chemotherapy in HER2-expressing gastroesophageal adenocarcinoma: a phase 1 trial. Nat Commun 2025; 16:4293. [PMID: 40341383 PMCID: PMC12062219 DOI: 10.1038/s41467-025-59279-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 04/15/2025] [Indexed: 05/10/2025] Open
Abstract
There is a need for novel therapies for patients with previously treated HER2-positive gastroesophageal adenocarcinoma (GEA). This phase 1 (NCT02892123) dose-escalation and expansion trial evaluated zanidatamab (a dual HER2-targeted bispecific antibody) ± chemotherapy in previously treated patients with HER2-expressing, locally advanced/metastatic cancers. Here, we report the outcomes for GEA cohorts receiving zanidatamab monotherapy or with chemotherapy (paclitaxel or capecitabine). The primary endpoint was safety and tolerability. Secondary endpoints were objective response rate (ORR), disease control rate, progression-free survival, pharmacokinetics, and immunogenicity. Seventy patients were enrolled (n = 29 monotherapy; n = 41 combination therapy); most received prior HER2-targeted agents (monotherapy, 93%; combination therapy, 95%). With monotherapy, 69% of patients had any-grade treatment-related AEs (TRAEs); 17% had grade ≥ 3 TRAEs. The most common any-grade TRAEs were diarrhea (41%) and infusion-related reactions (24%). With combination therapy, 98% of patients had any-grade TRAEs; 51% had grade ≥ 3 TRAEs. The most common any-grade TRAEs were diarrhea (68%) and fatigue (44%). Confirmed ORR was 32.1% (95% confidence interval [CI] 15.9-52.4) with monotherapy and 48.6% (95% CI 31.9-65.6) with combination therapy. In heavily pre-treated patients with HER2-expressing GEA, zanidatamab ± chemotherapy had a manageable safety profile and promising antitumor activity.
Collapse
Affiliation(s)
- Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Sun Young Rha
- Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | | | - Yoon-Koo Kang
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Diana L Hanna
- USC Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Syma Iqbal
- USC Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Keun-Wook Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Jeeyun Lee
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | | - 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
| | | | - Rachel A Goodwin
- Ottawa Hospital Cancer Centre, Ottawa Hospital Research Centre, Ottawa, ON, Canada
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lin Yang
- Jazz Pharmaceuticals, Palo Alto, CA, USA
| | - Rajen Oza
- Jazz Pharmaceuticals, Palo Alto, CA, USA
| | - Elena Elimova
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| |
Collapse
|
3
|
Zeng S, Yin S, Lian S, Luo M, Feng L, Liao Y, Huang Z, Zheng Y, Xie C, Zhuo S. A Clinical-Radiomic Combined Model based on Dual-Layer Spectral CT for Predicting Pathological T4 in Gastric Cancer. Acad Radiol 2025:S1076-6332(25)00383-6. [PMID: 40328540 DOI: 10.1016/j.acra.2025.04.035] [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: 03/20/2025] [Revised: 04/11/2025] [Accepted: 04/12/2025] [Indexed: 05/08/2025]
Abstract
RATIONALE AND OBJECTIVES This study aimed to develop and validate a dual-layer spectral CT based clinical-radiomic model for pre-treatment prediction of pathological T4 (pT4) in gastric cancer (GC) patients. MATERIALS AND METHODS This retrospective study included 148 surgically confirmed GC patients who underwent dual-layer spectral CT scanning before surgery and were divided into a training (n=104) and test (n=44) cohorts. Subjective assessments were performed based on conventional 120-kV CT images by two readers. Clinical models were developed using patient demographics, serum tumor markers, and image features from CT scans. Radiomics model included features extracted from conventional 120-kV CT and dual-layer CT-derived spectral base image (SBI), such as virtual monoenergetic images (40 keV, 70 keV, 100 keV), iodine density (ID), effective atomic number (Zeff), and electron density (ED) images for both the arterial phase (AP) and portal venous phase (PVP). A clinical-radiomic combined model was developed and visualized using a nomogram. RESULTS Tumor thickness on CT and serum level of CA19-9 levels were identified as independent predictors. The clinical-radiomic combined model demonstrated superior performance compared to subjective image interpretation and other models, with an AUC of 0.906 (95% CI, 0.848-0.963) in the training cohort and 0.873 in the test cohort. The nomogram was significantly associated with pT4 status, supporting its potential utility in clinical prediction. CONCLUSION The integration of clinical characteristics with radiomic features from conventional CT and dual-layer CT-derived SBI achieved a high diagnostic accuracy for predicting pT4 in GC patients. This combined approach could assist in treatment planning and patient management in GC.
Collapse
Affiliation(s)
- Sihui Zeng
- Department of Radiology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.); State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.)
| | - Shaohan Yin
- Department of Radiology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.); State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.)
| | - Shanshan Lian
- Department of Radiology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.); State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.)
| | - Ma Luo
- Department of Radiology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.); State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.)
| | - Lili Feng
- Department of Radiology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.); State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.)
| | - Yuting Liao
- Philips Healthcare, Guangzhou 510000, PR China (Y.L., Z.H.)
| | - Zhijie Huang
- Philips Healthcare, Guangzhou 510000, PR China (Y.L., Z.H.)
| | - Yuquan Zheng
- Department of Radiology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.); State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.)
| | - Chuanmiao Xie
- Department of Radiology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.); State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.).
| | - Shuiqing Zhuo
- Department of Radiology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.); State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China (S.Z., S.Y., S.L., M.L., L.F., Y.Z., C.X.).
| |
Collapse
|
4
|
Lee J, Choi J, Kang MS, Yu SY, Kim K. Bilateral optic neuropathy in Krukenberg tumor treated with FOLFOX plus nivolumab: a case report. BMC Ophthalmol 2025; 25:262. [PMID: 40307761 PMCID: PMC12042502 DOI: 10.1186/s12886-025-04102-y] [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: 03/01/2025] [Accepted: 04/23/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND A combination of FOLFOX and nivolumab is a first-line treatment for HER2-negative advanced gastric cancer, significantly improving survival. However, this regimen is associated with potential neurotoxicity. 5-Fluorouracil and oxaliplatin in FOLFOX have been linked to optic neuropathy, whereas nivolumab, an immune checkpoint inhibitor, may cause immune-related optic neuropathy. Although FOLFOX plus nivolumab provides considerable survival benefits, careful monitoring for ocular complications is essential. We report a case of bilateral optic neuropathy in a patient who received FOLFOX plus nivolumab. Despite discontinuation of chemotherapy and treated with high-dose corticosteroid pulse therapy, the patient's symptoms did not improve. CASE PRESENTATION A 48-year-old woman with a Krukenberg tumor developed progressive bilateral visual impairment during treatment with FOLFOX plus nivolumab. Her ophthalmologic history included branch retinal vein occlusion in the right eye but no systemic diseases. She presented with bilateral central scotomas and visual field defects. On examination, best-corrected visual acuity (BCVA) was 20/40 in the right eye and 20/25 in the left eye, with bilateral optic disc swelling and hemorrhage. Fluorescein angiography confirmed optic disc leakage, and a visual evoked potential (VEP) test indicated axonal loss. Brain magnetic resonance imaging (MRI) and positron emission tomography-computed tomography (PET-CT) ruled out metastatic disease, and autoimmune markers were negative. High-dose intravenous methylprednisolone was administered, and chemotherapy was discontinued. Despite initial stabilization, vision deteriorated, ultimately progressing to light perception in both eyes. Repeated steroid pulse therapy failed to improve outcomes. CONCLUSIONS This case highlights the potential for severe bilateral optic neuropathy associated with FOLFOX plus nivolumab, leading to irreversible vision loss. These findings suggest that close ophthalmologic monitoring is warranted in patients receiving FOLFOX plus nivolumab for early recognition of bilateral visual impairment. Further research is needed to elucidate the mechanisms and identify agents responsible for the development of optic neuropathy.
Collapse
Affiliation(s)
- Junwoo Lee
- Department of Ophthalmology, Gachon University Gil Medical Center, Incheon, Korea
| | - Jaehwan Choi
- Department of Ophthalmology, Kyung Hee University Hospital, Kyung Hee University, 23, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Min Seok Kang
- Department of Ophthalmology, Kyung Hee University Hospital, Kyung Hee University, 23, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Seung-Young Yu
- Department of Ophthalmology, Kyung Hee University Hospital, Kyung Hee University, 23, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Kiyoung Kim
- Department of Ophthalmology, Kyung Hee University Hospital, Kyung Hee University, 23, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| |
Collapse
|
5
|
Feng Z, Ou L, Li H, Hao Y, Wei R, Zhang G, Yao M. Unveiling the therapeutic potential of HZQYF: exploring the inhibitory impact of a clinical herbal formula on gastric cancer through network pharmacology and transcript analysis. BMC Complement Med Ther 2025; 25:142. [PMID: 40247271 PMCID: PMC12004866 DOI: 10.1186/s12906-025-04871-5] [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: 03/28/2024] [Accepted: 03/27/2025] [Indexed: 04/19/2025] Open
Abstract
Hezi Qingyou Formula (HZQYF) is a clinical formulation known for its efficacy in treating gastrointestinal diseases. Nevertheless, its specific impact and underlying mechanism of action in gastric cancer remain to be fully elucidated. The major components of the formula were precisely identified and characterized using ultra-high-performance liquid chromatography coupled with a tandem mass spectrometer (UHPLC-MS/MS). Network pharmacology and transcript analysis were utilized to identify the targets associated with drug-disease interactions. Subsequently, Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Reactome analyses were conducted to unravel the pivotal pathways involved. Furthermore, in vitro experiments were performed to validate the anti-gastric cancer activity of HZQYF, including assessments of cell viability and clonogenic potential. These results revealed that 260 co-expressed targets were identified as shared between HZQYF and gastric cancer. These genes were significantly enriched in biological processes and pathways related to steroid metabolism, gamma-aminobutyric acid (GABA)-A receptor complex, steroid binding activity, extracellular ligand-gated ion channel activity, chemical carcinogenesis-reactive oxygen species, and GABAergic synapse. Furthermore, the principal components of the formula were characterized. Subsequent cell experiments confirmed the formula's ability to inhibit gastric cancer activity and suppress colony formation in vitro. In conclusion, these findings suggest that Hezi Qingyou Formula may exert its anti-gastric cancer activity by influencing reactive oxygen species and modulating GABAergic synapses in-silico methods. This study provides a foundation for further exploration of HZQYF as a potential therapeutic agent for gastric cancer.
Collapse
Affiliation(s)
- Zhong Feng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen, 518107, China
- International Pharmaceutical Engineering Lab of Shandong Province, Feixian, Shandong, 273400, China
| | - Ling Ou
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen, 518107, China.
| | - Hui Li
- International Pharmaceutical Engineering Lab of Shandong Province, Feixian, Shandong, 273400, China
| | - Yajie Hao
- International Pharmaceutical Engineering Lab of Shandong Province, Feixian, Shandong, 273400, China
| | - Ruixia Wei
- Lunan Pharmaceutical Group Co., Ltd, Linyi, Shandong, 276000, China
| | - Guimin Zhang
- Lunan Pharmaceutical Group Co., Ltd, Linyi, Shandong, 276000, China.
| | - Meicun Yao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen, 518107, China.
| |
Collapse
|
6
|
Lemay F, Sandhu AS, Stein BD, Goodwin R. A Canadian algorithm for upper gastrointestinal cancer management. Front Oncol 2025; 15:1548637. [PMID: 40297809 PMCID: PMC12034531 DOI: 10.3389/fonc.2025.1548637] [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: 12/19/2024] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Recent advances in immunotherapy have changed the treatment landscape for cancers of the upper gastrointestinal (GI) system. Immune checkpoint inhibitors can lead to better survival and improved quality of life for affected individuals. Adopting new treatment strategies in real-world practice can be challenging, and algorithms that are easy to implement in Canadian oncology practices would benefit clinicians and patients. In this study, we present expert opinion on best practices for upper GI cancer management, including a new algorithm that integrates the latest evidence for screening, workup, diagnosis, treatment, and survivorship. The algorithm is based on a novel approach comprising a case-based, accredited educational program with asynchronous discussion among clinicians practicing across Canada, with the input of expert medical oncologists and gastroenterologists. A needs assessment was employed to determine current areas of educational need in the field of upper GI cancers, and a patient representative provided insights into patient concerns and priorities. The best practices described here include seeking patient input throughout treatment, integrating immune checkpoint inhibitors into systemic therapy for both localized and advanced disease, and providing comprehensive supportive care throughout the treatment and survivorship journey.
Collapse
Affiliation(s)
- Frédéric Lemay
- Division of Gastroenterology, Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Amindeep S. Sandhu
- Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | | | - Rachel Goodwin
- Division of Medical Oncology, Department of Medicine, University of Ottawa, The Ottawa Hospital Regional Cancer Centre, Ottawa, ON, Canada
| |
Collapse
|
7
|
Son SM, Woo CG, Lee OJ, Lee SK, Cho M, Lee YP, Kim H, Kim HK, Yang Y, Kwon J, Lee KH, Kim DH, Yun HY, Han HS. Discordance in Claudin 18.2 Expression Between Primary and Metastatic Lesions in Patients With Gastric Cancer. J Gastric Cancer 2025; 25:303-317. [PMID: 40200874 PMCID: PMC11982508 DOI: 10.5230/jgc.2025.25.e2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/21/2024] [Accepted: 11/09/2024] [Indexed: 04/10/2025] Open
Abstract
PURPOSE Claudin 18.2 (CLDN18.2) has emerged as a promising therapeutic target for CLDN18.2-expressing gastric cancer (GC). We sought to examine the heterogeneity of CLDN18.2 expression between primary GC (PGC) and metastatic GC (MGC) using various scoring methods. MATERIALS AND METHODS We retrospectively analyzed data from 102 patients with pathologically confirmed paired primary and metastatic gastric or gastroesophageal junction adenocarcinomas. CLDN18.2 expression was evaluated through immunohistochemistry on formalin-fixed paraffin-embedded tissue samples. We assessed CLDN18.2 positivity using multiple scoring approaches, including the immunoreactivity score, H-score, and the percentage of tumor cells showing moderate-to-strong staining intensity. We analyzed the concordance rates between PGC and MGC and the association of CLDN18.2 positivity with clinicopathological features. RESULTS CLDN18.2 positivity varied from 25% to 65% depending on the scoring method, with PGC consistently showing higher expression levels than MGC. Intratumoral heterogeneity was noted in 25.5% of PGCs and 19.6% of MGCs. Intertumoral heterogeneity, manifesting as discordance in CLDN18.2 positivity between PGC and MGC, was observed in about 20% of cases, with moderate agreement across scoring methods (κ=0.47 to 0.60). In PGC, higher CLDN18.2 positivity correlated with synchronous metastasis, presence of peritoneal metastasis, poorly differentiated grade, and biopsy specimens. In MGC, positivity was associated with synchronous metastasis, presence of peritoneal metastasis, and metastatic peritoneal tissues. CONCLUSIONS CLDN18.2 expression demonstrates significant heterogeneity between PGC and MGC, with a 20% discordance rate. Comprehensive tissue sampling and reassessment of CLDN18.2 status are crucial, especially before initiating CLDN18.2-targeted therapies.
Collapse
Affiliation(s)
- Seung-Myoung Son
- Department of Pathology, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Chang Gok Woo
- Department of Pathology, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Ok-Jun Lee
- Department of Pathology, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Sun Kyung Lee
- Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, Korea
| | - Minkwan Cho
- Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, Korea
| | - Yong-Pyo Lee
- Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, Korea
| | - Hongsik Kim
- Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, Korea
| | - Hee Kyung Kim
- Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, Korea
- Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Yaewon Yang
- Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, Korea
- Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Jihyun Kwon
- Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, Korea
- Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Ki Hyeong Lee
- Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, Korea
- Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Dae Hoon Kim
- Department of Surgery, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Hyo Yung Yun
- Department of Surgery, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Hye Sook Han
- Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, Korea
- Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju, Korea.
| |
Collapse
|
8
|
Miyajima Y, Kawakami T. Treatment Selection for Patients with HER2-Negative Metastatic Gastric Cancer Expressing Claudin 18.2 and PD-L1. Cancers (Basel) 2025; 17:1120. [PMID: 40227631 PMCID: PMC11987827 DOI: 10.3390/cancers17071120] [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: 02/05/2025] [Revised: 03/13/2025] [Accepted: 03/25/2025] [Indexed: 04/15/2025] Open
Abstract
Combination therapy of chemotherapy and zolbetuximab demonstrated a significant survival benefit compared to chemotherapy alone in patients with human epidermal growth factor receptor 2 (HER2)-negative, claudin (CLDN) 18.2-positive metastatic gastric cancer (mGC). Consequently, it has been approved as a standard first-line therapy for these patients. Combination therapy of chemotherapy and immune checkpoint inhibitors (ICIs)-either nivolumab or pembrolizumab-is a standard first-line therapy for patients with HER2-negative mGCs that are positive for programmed death-ligand 1 (PD-L1) expression, as defined by a combined positive score (CPS). Although approximately 13-22% of CLDN-positive mGCs are also CPS-positive, optimal treatment for mGC patients expressing both CLDN and PD-L1 remains undetermined due to the absence of direct comparative studies between zolbetuximab and ICIs. Treatment selection under this condition has become a critical issue. In this review, we discuss the appropriate treatment selection for HER2-negative mGC patients who are double-positive for CLDN 18.2 and PD-L1 based on clinical data and differences in the mechanism of action and safety profile between zolbetuximab and ICI.
Collapse
Affiliation(s)
| | - Takeshi Kawakami
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Shizuoka 411-8777, Japan;
| |
Collapse
|
9
|
Bonfill Cosp X, Savall-Esteve O, Bracchiglione J, Requeijo C, Santero M. Mismatch between evidence and related clinical recommendations about the treatment of advanced esophageal cancer patients with anticancer drugs: A critical historical review. J Cancer Policy 2025; 44:100580. [PMID: 40147630 DOI: 10.1016/j.jcpo.2025.100580] [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: 07/04/2024] [Revised: 02/19/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
PURPOSE to analyze the most robust research and recommendations that have informed the potential superiority of treatments with anticancer drugs over any type of supportive care for advanced esophageal cancer (EC). METHODS We conducted a critical historical review. First, we identified randomized clinical trials (RCTs) from a previous scoping review conducted by our research group, ASTAC, updating the search strategy. Second, we searched for the most important and recognized international clinical practice guidelines (CPGs) in advanced EC. Finally, we performed a systematic document analysis to compare whether the recommendations proposed in the CPGs were supported by the previously identified relevant evidence. RESULTS We identified and assessed 15 RCTs and 11 CPGs from ESMO (eight), ASCO (two), and NICE (one) published over the last 40 years. There is a clear mismatch between these guidelines' recommendations and the available RCTs regarding the efficacy of anticancer drugs compared to best supportive care (BSC). CONCLUSION There is a lack of consistent evidence to support the treatment of advanced EC patients with anticancer drugs, and a notable mismatch exists between the available evidence and the recommendations made by relevant CPGs. As a result, these guidelines may be biased in favoring the use of anticancer drugs over supportive care and in consequence it is advisable to be very prudent when proposing systemic treatments to patients with advanced EC. Further rigorous and independent research is needed to better evaluate the true benefits of anticancer treatments in advanced EC and to update the CPGs accordingly.
Collapse
Affiliation(s)
- Xavier Bonfill Cosp
- Iberoamerican Cochrane Centre, Barcelona, Spain; Universitat Autònoma Barcelona (UAB), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain; Institut de Recerca Sant Pau (IR Sant Pau), Barcelona, Spain
| | - Olga Savall-Esteve
- Iberoamerican Cochrane Centre, Barcelona, Spain; Institut de Recerca Sant Pau (IR Sant Pau), Barcelona, Spain
| | - Javier Bracchiglione
- Iberoamerican Cochrane Centre, Barcelona, Spain; Universitat Autònoma Barcelona (UAB), Barcelona, Spain; Interdisciplinary Centre for Health Studies (CIESAL), Universidad de Valparaíso, Viña del Mar, Chile; CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain; Institut de Recerca Sant Pau (IR Sant Pau), Barcelona, Spain
| | - Carolina Requeijo
- Iberoamerican Cochrane Centre, Barcelona, Spain; Institut de Recerca Sant Pau (IR Sant Pau), Barcelona, Spain
| | - Marilina Santero
- Iberoamerican Cochrane Centre, Barcelona, Spain; Universitat Autònoma Barcelona (UAB), Barcelona, Spain; Institut de Recerca Sant Pau (IR Sant Pau), Barcelona, Spain.
| |
Collapse
|
10
|
Leigh J, Ahmed A, Aubin F, Berry S, Boucher M, Campeau MP, Colwell B, Connors S, Corbett J, Dadwal S, Dudani S, Elimova E, Falkson C, Galvis L, Goel R, Gotfrit J, Hyde A, Febbraro M, Laidley DT, Locke G, Mahmud A, Baccili Cury Megid T, Michael J, Nair VJ, Quigley S, Ramjeesingh R, Samimi S, Seal M, Snow S, Spadafora S, Stuckless T, Wilson B, Asmis T, Goodwin R, Vickers M. Eastern Canadian Gastrointestinal Cancer Consensus Conference 2024. Curr Oncol 2025; 32:175. [PMID: 40136379 PMCID: PMC11941643 DOI: 10.3390/curroncol32030175] [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] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/05/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025] Open
Abstract
The Eastern Canadian Gastrointestinal Cancer Consensus Conference was an annual meeting that was held in St. John's, Newfoundland and Labrador, from 26 to 28 September 2024. This included experts in medical oncology, radiation oncology, surgical oncology, nuclear medicine, and general practitioners in oncology (GPO) from across the eastern Canadian provinces who are involved in the management of patients with gastrointestinal malignancies. This consensus statement generated by the conference addresses multiple topics, including the management of localized rectal cancer, liver-limited colorectal cancer, systemic therapy for advanced biliary tract cancers, radioligand therapy for gastroenteropancreatic neuroendocrine tumors (GEP-NETs), systemic therapy for pancreatic and midgut well-differentiated NETs, and systemic therapy for HER2-positive gastroesophageal cancers.
Collapse
Affiliation(s)
| | - Arwa Ahmed
- The Ottawa Hospital Cancer Centre, Ottawa, ON K1H 8L6, Canada
| | - Francine Aubin
- Centre Hospitalier de l’Universite de Montreal, Montreal, QC H2X 3E4, Canada
| | - Scott Berry
- Trillium Health Partners, Mississauga, ON L5A 4G1, Canada
| | - Melanie Boucher
- Prince Edward Island Cancer Treatment Center, Charlottetown, PE C1A 8T5, Canada
| | | | - Bruce Colwell
- Queen Elizabeth II Health Sciences Center, Halifax, NS B3H 3A7, Canada
| | | | - Jessica Corbett
- Prince Edward Island Cancer Treatment Center, Charlottetown, PE C1A 8T5, Canada
| | | | - Shaan Dudani
- William Osler Health System, Brampton, ON L6R 3J7, Canada
| | - Elena Elimova
- Princess Margaret Cancer Center, Toronto, ON M5G 2M9, Canada
| | - Conrad Falkson
- Kingston Health Sciences Center, Kingston, ON K7L 2V7, Canada
| | - Luisa Galvis
- Horizon Health Network, Fredericton, NB E3B 4R3, Canada
| | - Rakesh Goel
- The Ottawa Hospital Cancer Centre, Ottawa, ON K1H 8L6, Canada
| | - Joanna Gotfrit
- The Ottawa Hospital Cancer Centre, Ottawa, ON K1H 8L6, Canada
| | - Angela Hyde
- Dr. H. Bliss Murphy Cancer Center, St. John’s, NL A1B 3X5, Canada
| | - Michela Febbraro
- Algoma District Cancer Program, Sault Ste. Marie, ON P6B 0A8, Canada
| | | | - Gordon Locke
- The Ottawa Hospital Cancer Centre, Ottawa, ON K1H 8L6, Canada
| | - Aamer Mahmud
- Kingston Health Sciences Center, Kingston, ON K7L 2V7, Canada
| | | | - James Michael
- Saint John Regional Hospital Oncology Center, Saint John, NB E2L 4L2, Canada
| | - Vimoj J. Nair
- The Ottawa Hospital Cancer Centre, Ottawa, ON K1H 8L6, Canada
| | - Stephen Quigley
- Health Sciences Center-Eastern Health, St. John’s, NL A1B 3V6, Canada
| | - Ravi Ramjeesingh
- Queen Elizabeth II Health Sciences Center, Halifax, NS B3H 3A7, Canada
| | - Setareh Samimi
- Hopital du Sacre-Coeur de Montreal, Montreal, QC H4J 1C5, Canada
| | - Melanie Seal
- Dr. H. Bliss Murphy Cancer Center, St. John’s, NL A1B 3X5, Canada
| | - Stephanie Snow
- Queen Elizabeth II Health Sciences Center, Halifax, NS B3H 3A7, Canada
| | - Silvana Spadafora
- Algoma District Cancer Program, Sault Ste. Marie, ON P6B 0A8, Canada
| | - Teri Stuckless
- Dr. H. Bliss Murphy Cancer Center, St. John’s, NL A1B 3X5, Canada
| | - Brooke Wilson
- Kingston Health Sciences Center, Kingston, ON K7L 2V7, Canada
| | - Timothy Asmis
- The Ottawa Hospital Cancer Centre, Ottawa, ON K1H 8L6, Canada
| | - Rachel Goodwin
- The Ottawa Hospital Cancer Centre, Ottawa, ON K1H 8L6, Canada
| | - Michael Vickers
- The Ottawa Hospital Cancer Centre, Ottawa, ON K1H 8L6, Canada
| |
Collapse
|
11
|
Li Y, Ren W, Li S, Zheng J, Gu H, Gao L, Zhi K. The clinicopathological significance and prognostic value of PD-L1 in oral squamous cell carcinoma. Oral Dis 2025; 31:815-825. [PMID: 39092614 DOI: 10.1111/odi.15096] [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: 01/06/2024] [Revised: 05/22/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024]
Abstract
OBJECTIVE To investigate the relationship between the expression of PD-L1 in OSCC and the clinicopathological features and prognosis of patients. METHODS We retrospectively analyzed the clinicopathological data and prognosis of 381 OSCC patients. Immunohistochemical staining was performed on OSCC tumor specimens, and the expression level of PD-L1 was evaluated according to the combined positive score (CPS). Kaplan-Meier analysis was used to identify the effect of PD-L1 expression and clinicopathological features on the prognosis of patients. Univariate and multivariate Cox regression analyses were conducted to determine the hazard factors affecting the prognosis of patients. RESULTS PD-L1 overexpression was significantly associated with cervical lymph node metastasis (p = 0.018), worse clinical stage (p = 0.022), worse tumor differentiation (p = 0.046), and worse depth of invasion (DOI) (p = 0.003). Poorer clinical stage and degree of tumor differentiation were significantly associated with poorer OS and DSS in patients. PD-L1 expression was not associated with prognosis in patients with OSCC. CONCLUSIONS High PD-L1 expression was significantly associated with higher tumor malignancy in OSCC patients. Poorer clinical stage and degree of tumor differentiation were associated with poor prognosis in OSCC patients. Our results may help clinicians develop more appropriate individualized treatment strategies for their patients, thus improving their outcomes.
Collapse
Affiliation(s)
- Yizhan Li
- Department of Oral and Maxillofacial Reconstruction, The Affiliated Hospital of Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
- Key Lab of Oral Clinical Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenhao Ren
- Department of Oral and Maxillofacial Reconstruction, The Affiliated Hospital of Qingdao University, Qingdao, China
- Key Lab of Oral Clinical Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shaoming Li
- Department of Oral and Maxillofacial Reconstruction, The Affiliated Hospital of Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
- Key Lab of Oral Clinical Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jingjing Zheng
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Haiyan Gu
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Ling Gao
- Department of Oral and Maxillofacial Reconstruction, The Affiliated Hospital of Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
- Key Lab of Oral Clinical Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Keqian Zhi
- Department of Oral and Maxillofacial Reconstruction, The Affiliated Hospital of Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
- Key Lab of Oral Clinical Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
12
|
Papageorgiou GI, Skouteris N, Grenzelia M, Maragkoudakis E, Eleftheriou K. Is it time to revisit the significance of PD-L1 expression in assisting our treatment decisions? Immunotherapy 2025; 17:223-227. [PMID: 40116402 PMCID: PMC12013450 DOI: 10.1080/1750743x.2025.2483152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 03/17/2025] [Indexed: 03/23/2025] Open
Affiliation(s)
| | | | - Maria Grenzelia
- Radiation Oncology Center, Iaso General Clinic, Marousi, Greece
| | | | | |
Collapse
|
13
|
Wei J, Zhang P, Hu Q, Cheng X, Shen C, Chen Z, Zhuang W, Yin Y, Zhang B, Gou H, Yang K, Bi F, Liu M. Nab-paclitaxel combined with cadonilimab (AK104) as second-line treatment for advanced gastric cancer: protocol for a phase II prospective, multicenter, single-arm clinical trial. Front Immunol 2025; 16:1519545. [PMID: 40070819 PMCID: PMC11893503 DOI: 10.3389/fimmu.2025.1519545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 01/31/2025] [Indexed: 03/14/2025] Open
Abstract
Background Gastric cancer (GC) is one of the most prevalent malignant tumors worldwide, often diagnosed at an advanced stage with a poor prognosis. Paclitaxel, nab-paclitaxel, and irinotecan, either as monotherapies or in combination with ramucirumab, are currently standard second-line treatments for GC. However, the efficacy of these therapies is limited, necessitating the development of new combination strategies to improve response rates. Immune checkpoint inhibitors (ICIs) have shown success in first-line treatment for advanced GC, leading to interest in immune rechallenge strategies for second-line treatment. Re-challenging patients with ICIs after progression on first-line treatment may restore immune responses and provide additional clinical benefit. Recently, cadonilimab (AK104), a bispecific antibody targeting PD-1 and CTLA-4, has demonstrated promising antitumor activity when combined with chemotherapy in advanced gastric and gastroesophageal junction (GEJ) adenocarcinoma. However, the efficacy and safety of nab-paclitaxel combined with AK104 for the treatment of advanced GC remain unclear. Furthermore, identifying predictive biomarkers of efficacy is essential to developing personalized treatment strategies. This study aims to explore the safety and efficacy of nab-paclitaxel combined with AK104 as a second-line treatment for patients who have progressed after first-line chemoimmunotherapy, focusing on evaluating the therapeutic effect of ICIs rechallenge in gastric cancer. Methods This is a prospective, multicenter, open-label, single-arm Phase II clinical study. Eligible patients were histologically or cytologically diagnosed with unresectable recurrent or metastatic GC, failed first-line chemotherapy in combination with immune checkpoint inhibitor, aged between 18-75 years old, expected survival ≥3 months, and with a physical status of 0 or 1 in the Eastern Cooperative Cancer Group (ECOG). Enrolled patients will receive intravenous cadonilimab (AK104) 6 mg/kg on days 1, and 15, and intravenous nab-paclitaxel 100 mg/m2 every four weeks on days 1, 8, and 15. The primary endpoints were objective response rate (ORR), and secondary endpoints were disease control rate (DCR), progression-free survival (PFS), and overall survival (OS). The exploratory objective was to identify biomarkers associated with efficacy, mechanism of action, and safety. A total of 59 participants were planned to be recruited using Simon's two-stage design. The trial was initiated in June 2024 in China. Discussion This study is the first prospective trial to evaluate the combination of nab-paclitaxel and cadonilimab as second-line treatment after first-line chemoimmunotherapy failure. By investigating immune rechallenge, it aims to reactivate anti-tumor immune responses and improve clinical outcomes in GC patients. The exploration of predictive biomarkers, such as ctDNA, TMB, MSI, PD-L1 expression, TIL profiles, and gut microbiota, will help personalize treatment and identify patients most likely to benefit from immune rechallenge. This trial could provide valuable insights into overcoming immune resistance and contribute to developing a promising second-line therapeutic strategy for advanced GC. Clinical trial registration ClinicalTrials.Gov, identifier NCT06349967.
Collapse
Affiliation(s)
- Jing Wei
- Gastric Cancer Center, Department of Medical Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Pengfei Zhang
- Gastric Cancer Center, Department of Medical Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiancheng Hu
- Gastric Cancer Center, Department of Medical Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaolong Cheng
- Department of General Surgery/Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chaoyong Shen
- Department of General Surgery/Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhixin Chen
- Department of General Surgery/Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wen Zhuang
- Department of General Surgery/Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuan Yin
- Department of General Surgery/Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bo Zhang
- Department of General Surgery/Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hongfeng Gou
- Gastric Cancer Center, Department of Medical Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kun Yang
- Department of General Surgery/Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feng Bi
- Division of Abdominal Cancer, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Ming Liu
- Gastric Cancer Center, Department of Medical Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
14
|
Cheng N, Wang B, Xu J, Xue L, Ying J. Tumor stroma ratio, tumor stroma maturity, tumor-infiltrating immune cells in relation to prognosis, and neoadjuvant therapy response in esophagogastric junction adenocarcinoma. Virchows Arch 2025; 486:257-266. [PMID: 38383941 DOI: 10.1007/s00428-024-03755-2] [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: 10/31/2023] [Revised: 01/14/2024] [Accepted: 01/27/2024] [Indexed: 02/23/2024]
Abstract
Accurate predictions on prognosis and neoadjuvant therapy response are crucial for esophagogastric junction adenocarcinoma (EGJA) patients. Therefore, we aimed to investigate the predictive abilities of several indicators, including tumor stroma ratio (TSR), tumor stroma maturity (TSM), and the density and spatial distribution of tumor-infiltrating immune cells (TIICs), such as T cells, B cells, and tumor-associated macrophages (TAMs). Resection and biopsy specimens of a total of 695 patients were included, obtained from the National Cancer Center (NCC) and The Cancer Genome Atlas (TCGA) cohorts. TSR and TSM were evaluated based on histological assessment. TIICs were quantified by QuPath following immunohistochemical (IHC) staining in resection specimens, while the Klintrup-Mäkinen (KM) grade was employed for evaluating TIIC in biopsy specimens. Patients with high stromal levels or immature stroma had relatively worse prognoses. Furthermore, high CD8+T cell count in the tumor periphery, as well as low CD68+ TAM count either in the tumor center or in the tumor periphery, was an independent favorable prognostic factor. Significantly, the combination model incorporating TSM and CD163+TAMs emerged as an independent prognostic factor in both two independent cohorts (HR 3.644, 95% CI 1.341-9.900, p = 0.011 and HR 1.891, 95% CI 1.195-2.99, p = 0.006, respectively). Additionally, high stromal levels in preoperative biopsies correlated with poor neoadjuvant therapy response (p < 0.05). In conclusion, our findings suggest that TSR, TSM, CD8+T cell, CD68+TAMs, and CD163+TAMs predict the prognosis to some extent in patients with EGJA. Notably, the combined model incorporating TSM and CD163+TAM can contribute significantly to prognostic stratification. Additionally, high stromal levels evaluated in preoperative biopsy specimens correlated with poor neoadjuvant therapy response.
Collapse
Affiliation(s)
- Na Cheng
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan, Chaoyang District, Beijing, 100021, China
| | - Bingzhi Wang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan, Chaoyang District, Beijing, 100021, China
| | - Jiaqi Xu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan, Chaoyang District, Beijing, 100021, China
| | - Liyan Xue
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan, Chaoyang District, Beijing, 100021, China.
| | - Jianming Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
15
|
Guo T, Hui Y, Zhu W, Ke F, Zhou T, Qiu W, Li X, Li L, Cheng H. Extended survival of a patient with gastrointestinal multiple malignancies managed with anti-PD-1 immunotherapy: a case report. Immunotherapy 2025; 17:95-101. [PMID: 39935272 PMCID: PMC11901419 DOI: 10.1080/1750743x.2025.2463309] [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: 08/28/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
INTRODUCTION The annual rise in gastrointestinal cancer cases is evident, yet the occurrence of multiple primary malignancies remains comparatively uncommon. In recent years, immunotherapy has swiftly emerged as the leading treatment for several solid tumors, including gastrointestinal cancers. Single treatments might be ineffective, necessitating the need for comprehensive integrative medicine. CASE DESCRIPTION This study reports a case of multiple cancers, including colorectal and gastric cancers. Diverse systemic treatments, like capecitabine, the combination of capecitabine and paclitaxel liposome, as well as capecitabine with toripalimab, were unsuccessful. Nevertheless, prolonged survival was attained through anti-PD-1 immunotherapy complemented by alternative medicine approaches. The patient has exceeded a 35-month survival post-initial diagnosis and 20-month survival since the subsequent diagnosis, markedly surpassing the prognosis often associated with advanced-stage multiple cancers. CONCLUSION In summary, this case underscores the potential effectiveness of a holistic, integrative medical approach in managing advanced multiple malignancies amid drug resistance and disease progression.
Collapse
Affiliation(s)
- Tianhao Guo
- Institute of Health and Regimen, Jiangsu Open University, Nanjing, Jiangsu, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, Jiangsu, China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yifan Hui
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, Jiangsu, China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Wenjian Zhu
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, Jiangsu, China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Fei Ke
- Department of Pathology, Jiangsu Provincial Hospital of Traditional Chinese Medicine (Affiliated Hospital of Nanjing University of Chinese Medicine), Nanjing, Jiangsu, China
| | - Tingting Zhou
- Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Wenli Qiu
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xuan Li
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Liu Li
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, Jiangsu, China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Haibo Cheng
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, Jiangsu, China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
16
|
Tsimafeyeu I, Musayeva G, Mahmudova S, Otkhozoria N, Abbasov B, Kahharov A, Guliyev F. Nivolumab Combined with Chemotherapy in FGFR2 and PD-L1 Co-Expressing Metastatic Gastric Cancer: A Prospective Phase 2 NIVOFGFR2 Study. J Gastrointest Cancer 2025; 56:40. [PMID: 39799264 DOI: 10.1007/s12029-025-01172-5] [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] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND Immunotherapy is increasingly significant in treating metastatic gastric cancer. This prospective phase 2 study investigates the efficacy and safety of combining nivolumab with chemotherapy in patients with metastatic gastric cancer co-expressing FGFR2 and PD-L1. METHODS Eligible patients were aged 18 years or older, with previously untreated HER-2 negative, PD-L1 positive, and FGFR2 positive metastatic gastric adenocarcinoma. Patients received nivolumab (360 mg every 3 weeks) in combination with chemotherapy (CAPOX: capecitabine 1000 mg/m2 twice daily on days 1-14 and oxaliplatin 130 mg/m2 on day 1, every 3 weeks). Tumor assessments were conducted using RECIST v1.1 every 8 weeks for 48 weeks, then every 12 weeks. The primary endpoint was the 1-year progression-free survival (PFS) rate. Secondary endpoints included median PFS, overall survival (OS), objective response rate (ORR), and grade ≥ 3 adverse events (AEs). RESULTS From June 2022 to October 2023, 194 patients were assessed for eligibility, with 23 patients enrolled and treated. At a median follow-up of 17.3 months, the 1-year PFS rate was 30.4%, with a median PFS of 6.0 months (95% CI, 4.3-7.7). The median OS was 15.1 months (95% CI, 13.2-16.8). The ORR was 21.7%, with one complete response and four partial responses. Grade 3 or higher TRAEs were reported in 34.8% of patients, primarily associated with chemotherapy. No treatment-related deaths occurred. CONCLUSIONS While the primary endpoint of improved 1-year PFS rate was not met, the study offers valuable insights into the potential benefits of combining nivolumab with chemotherapy in FGFR2 and PD-L1 co-expressing metastatic gastric cancer. Future research should optimize patient selection, assess combined immunotherapy and targeted anti-FGFR2 therapy, and further investigate the role of subsequent treatments to maximize therapeutic benefits.
Collapse
Affiliation(s)
- Ilya Tsimafeyeu
- Bureau for Cancer Research - BUCARE, 526 W 158Th Str, New York, NY, 10032, USA.
| | | | | | | | | | | | | |
Collapse
|
17
|
Wu Z, Wang T, Lan J, Wang J, Chen G, Tong T, Zhang H. Deep learning-based prediction of HER2 status and trastuzumab treatment efficacy of gastric adenocarcinoma based on morphological features. J Transl Med 2025; 23:13. [PMID: 39762854 PMCID: PMC11702172 DOI: 10.1186/s12967-024-06034-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 12/25/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND First-line treatment for advanced gastric adenocarcinoma (GAC) with human epidermal growth factor receptor 2 (HER2) is trastuzumab combined with chemotherapy. In clinical practice, HER2 positivity is identified through immunohistochemistry (IHC) or fluorescence in situ hybridization (FISH), whereas deep learning (DL) can predict HER2 status based on tumor histopathological features. However, it remains uncertain whether these deep learning-derived features can predict the efficacy of anti-HER2 therapy. METHODS We analyzed a cohort of 300 consecutive surgical specimens and 101 biopsy specimens, all undergoing HER2 testing, along with 41 biopsy specimens receiving trastuzumab-based therapy for HER2-positive GAC. RESULTS We developed a convolutional neural network (CNN) model using surgical specimens that achieved an area under the curve (AUC) value of 0.847 in predicting HER2 amplification, and achieved an AUC of 0.903 in predicting HER2 status specifically in patients with HER2 2 + expression. The model also predicted HER2 status in gastric biopsy specimens, achieving an AUC of 0.723. Furthermore, our classifier was trained using 41 HER2-positive gastric biopsy specimens that had undergone trastuzumab treatment, our model demonstrated an AUC of 0.833 for the (CR + PR) / (SD + PD) subgroup. CONCLUSION This work explores an algorithm that utilizes hematoxylin and eosin (H&E) staining to accurately predict HER2 status and assess the response to trastuzumab in GAC, potentially facilitating clinical decision-making.
Collapse
Affiliation(s)
- Zhida Wu
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Tao Wang
- College of Physics and Information Engineering, Fuzhou University, Xueyuan Road No.2, Fuzhou, Fujian, 350108, China
| | - Junlin Lan
- College of Physics and Information Engineering, Fuzhou University, Xueyuan Road No.2, Fuzhou, Fujian, 350108, China
| | - Jianchao Wang
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Gang Chen
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Tong Tong
- College of Physics and Information Engineering, Fuzhou University, Xueyuan Road No.2, Fuzhou, Fujian, 350108, China.
| | - Hejun Zhang
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China.
| |
Collapse
|
18
|
Despotidis M, Lyros O, Driva TS, Sarantis P, Kapetanakis EI, Mylonakis A, Mamilos A, Sakellariou S, Schizas D. DKK1 and Its Receptors in Esophageal Adenocarcinoma: A Promising Molecular Target. Diagnostics (Basel) 2025; 15:85. [PMID: 39795613 PMCID: PMC11720708 DOI: 10.3390/diagnostics15010085] [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: 11/07/2024] [Revised: 12/26/2024] [Accepted: 01/01/2025] [Indexed: 01/13/2025] Open
Abstract
Esophageal adenocarcinoma (EAC) is an aggressive gastrointestinal (GI) malignancy with increasing incidence. Despite the recent progress in targeted therapies and surgical approaches, the survival rates of esophageal adenocarcinoma patients remain poor. The Dickkopf (DKK) proteins are secretory proteins known mainly as antagonists of the Wnt/β-catenin signaling pathway, which is considered an oncogene. However, it has been shown that in several GI cancers, including esophageal cancer, DKK1 may act as an oncogene itself through Wnt-independent signaling pathways. LRP5\6 and Kremen1/2 (Krm1/2) are transmembrane receptors to which the DKK proteins are mainly known to bind. CKAP4 (cytoskeleton-associated protein 4) is a novel receptor of DKK1, and the DKK1-CKAP4 pathway seems to be crucial in the role of DKK1 as an oncogene. The aim of this review is to feature the essential role of DKK1 and its receptors in carcinogenesis with a focus on EAC in an era of urgent need for specific biomarkers along with improved targeted therapies.
Collapse
Affiliation(s)
- Markos Despotidis
- First Department of Surgery, National and Kapodistrian University of Athens, Laikon General Hospital, 11527 Athens, Greece; (A.M.); (D.S.)
| | - Orestis Lyros
- Fourth Department of Surgery, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Tatiana S. Driva
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (T.S.D.); (S.S.)
| | - Panagiotis Sarantis
- Department of Biological Chemistry, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Emmanouil I. Kapetanakis
- Department of Thoracic Surgery, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Adam Mylonakis
- First Department of Surgery, National and Kapodistrian University of Athens, Laikon General Hospital, 11527 Athens, Greece; (A.M.); (D.S.)
| | - Andreas Mamilos
- Institute of Pathology, University of Regensburg, 93053 Bavaria, Germany;
- Department of Pathology, German Oncology Center, Limassol 4108, Cyprus
| | - Stratigoula Sakellariou
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (T.S.D.); (S.S.)
| | - Dimitrios Schizas
- First Department of Surgery, National and Kapodistrian University of Athens, Laikon General Hospital, 11527 Athens, Greece; (A.M.); (D.S.)
| |
Collapse
|
19
|
Voutsadakis IA. Molecular alterations in claudin 18 suppressed and non-suppressed gastric adenocarcinomas to guide targeted therapies. Tissue Barriers 2025; 13:2348852. [PMID: 38713052 PMCID: PMC11970779 DOI: 10.1080/21688370.2024.2348852] [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: 02/03/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND Gastric adenocarcinoma represents an aggressive type of cancer and an important cause of cancer mortality. Progress in gastric cancer therapeutics has resulted from a better understanding of the molecular pathogenesis of the disease and introduction of targeted therapies, but most gastric cancer patients still rely on non-targeted chemotherapy as the mainstay of treatment for advanced disease. METHODS An analysis of publicly available series from The Cancer Genome Atlas (TCGA) gastric cancer cohort was undertaken to delineate the clinical and genomic landscape of gastric cancers with suppressed expression of claudin 18 compared with cancers with non-suppressed claudin 18. Claudin 18 suppressed cancers were defined as having an mRNA expression z-score relative to normal samples (log RNA Seq V2) of less than -1. Claudin 18 non-suppressed cancers were defined as having an mRNA expression z-score relative to normal samples (log RNA Seq V2) above 0.5. RESULTS Gastric cancers with claudin 18 mRNA suppression represented 7.7% of the gastric adenocarcinomas of TCGA cohort, while non-suppressed cancers represented 46.6% of the cases. The two groups did not differ in clinical and genomic characteristics, such as mean age, histology, grade, and stage. The mutation landscape of claudin 18 suppressed cases included high mutation rates of TP53, of genes of the WNT/β-catenin pathway and of ubiquitin ligase FBXW7. Moreover, a subset of both claudin 18 suppressed and non-suppressed cancers displayed mutations in Mismatch Repair (MMR) associated genes or a high tumor mutation burden (TMB). At the mRNA expression level, claudin 18 suppressed gastric cancers showed up-regulation of EMT core transcription factor Snail 2 and down-regulation of genes of HLA cluster. The survival of gastric cancer patients with claudin 18 mRNA suppression was not significantly different compared with patients with non-suppressed claudin 18. CONCLUSION Sub-sets of gastric cancers with claudin 18 mRNA suppression displayed characteristics of potential therapeutic interest, such as mutations in WNT and PI3K pathways and MMR defects. These may guide the development of alternative targeted therapies, in this sub-set of gastric cancers which are not candidates for claudin 18 targeting therapies.
Collapse
Affiliation(s)
- Ioannis A. Voutsadakis
- Algoma District Cancer Program, Sault Area Hospital, Sault Ste Marie, Ontario, Canada
- Division of Clinical Sciences, Section of Internal Medicine, Northern Ontario School of Medicine, Sudbury, ON, Canada
| |
Collapse
|
20
|
Tippareddy C, Martinez OM, Benza AR, Bera K, Ramaiya N, Tirumani SH. From guidelines to radiology practice: navigating the 2023 ASCO guidelines for advanced gastroesophageal cancer and beyond. Abdom Radiol (NY) 2025; 50:78-93. [PMID: 39123051 PMCID: PMC11711647 DOI: 10.1007/s00261-024-04499-y] [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: 04/07/2024] [Revised: 07/09/2024] [Accepted: 07/13/2024] [Indexed: 08/12/2024]
Abstract
The American Society of Clinical Oncology (ASCO) updated the guidelines for the treatment of advanced gastroesophageal (GE) cancer in 2023, signifying a major shift towards targeted therapeutics and precision medicine. This article serves as an imaging-based review of recent developments in the care of patients with GE cancer. We cover the epidemiology, the developing treatment paradigms, and the imaging assessment of GE malignancy. In addition, this review aims to familiarize radiologists with the unique adverse effects pertaining to therapeutics, surgeries, radiation therapies, and associated imaging corollaries. A case-based approach will be used to both explore the efficacy of modern treatments and demonstrate their adverse effects, such as chemotherapy-associated pneumonitis, radiation esophagitis, and anastomotic failure. With this comprehensive exploration of gastroesophageal cancer, radiologists will be equipped with the essential tools to inform the treatment decisions made by medical oncologists, radiation oncologists, and surgical oncologists in the new era of precision medicine.
Collapse
Affiliation(s)
- Charit Tippareddy
- Department of Radiology, University Hospitals Cleveland Medical Center, Case Western Reserve University, 1110 Euclid Ave, Cleveland, OH, 44106, USA.
- Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| | | | - Andrew R Benza
- Department of Radiology, University Hospitals Cleveland Medical Center, Case Western Reserve University, 1110 Euclid Ave, Cleveland, OH, 44106, USA
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Kaustav Bera
- Department of Radiology, University Hospitals Cleveland Medical Center, Case Western Reserve University, 1110 Euclid Ave, Cleveland, OH, 44106, USA
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Nikhil Ramaiya
- Department of Radiology, University Hospitals Cleveland Medical Center, Case Western Reserve University, 1110 Euclid Ave, Cleveland, OH, 44106, USA
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Sree Harsha Tirumani
- Department of Radiology, University Hospitals Cleveland Medical Center, Case Western Reserve University, 1110 Euclid Ave, Cleveland, OH, 44106, USA
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
21
|
Dong Z, Yang Z, Ren J, Li F, Wang G, Wang Y. Envafolimab Inhibits the Growth of Gastric Cancer Cells with Low PD-L1 Expression through the DDX20/NF-κB/TNF-α Signaling Pathway. Curr Cancer Drug Targets 2025; 25:648-664. [PMID: 39021191 DOI: 10.2174/0115680096314855240619181909] [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: 03/27/2024] [Revised: 05/01/2024] [Accepted: 05/06/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND The mechanism of action of envafolimab (also known as KN035), a programmed death ligand 1 (PD-L1) inhibitor, in gastric adenocarcinoma patients with low PD-L1 expression is not well understood. OBJECTIVES The objective of this study was to explore the underlying mechanism of envafolimab in gastric cancer with low PD-L1 expression. METHODS Cytotoxicity and proliferation were evaluated by a CCK8 assay. Transwell assays were used to detect the migration and invasion ability of gastric cancer cells. The effect of envafolimab on the apoptosis of gastric cancer cells was detected by flow cytometry. The effect of envafolimab on gastric cancer cells with low PD-L1 expression was investigated via proteomics and bioinformatics analysis. RESULTS A total of 19 patients with advanced gastric adenocarcinoma who received envafolimab monotherapy or combination therapy were reviewed. Among them, 4 patients had low PD-L1 expression, the objective response rate (ORR) was 75% (3/4), and the disease control rate (DCR) was 100% (4/4). In vitro experiments showed that envafolimab inhibited the proliferation, invasion, and migration of gastric cancer cells with low expression of PD-L1 and induced cell apoptosis. DDX20 may be the target of envafolimab in gastric cancer cells, and it is related to the NF-κB signaling pathway. Western blot results showed that the protein expressions of DDX20, NF-κB p65, and TNF-α in gastric cancer cells were decreased after adding envafolimab. Furthermore, the DDX20 gene was silenced by small interfering RNA to further study the effect of DDX20 on PDL1 low expression in gastric cancer cells. CONCLUSION This study confirmed that envafolimab could inhibit the growth of gastric cancer cells with low PD-L1 expression by down-regulating DDX20 expression and regulating the NF- κB/TNF-α signaling pathway.
Collapse
Affiliation(s)
- Zhuanxia Dong
- First Hospital of Shanxi Medical University, Shanxi Medical University,, Taiyuan, Shanxi, 030001, China
| | - Zefeng Yang
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Jing Ren
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150040, China
| | - Feng Li
- Central laboratory, Shanxi Provincial Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, 030013, China
| | - Guangyu Wang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150040, China
| | - Yusheng Wang
- Department of Oncology Digestive, First Hospital of Shanxi Medical University, Taiyuan City, 030001, China
| |
Collapse
|
22
|
Han Y, Yang W, Meng Y, Cai Z, Zhang B. Chemotherapy combined with immunotherapy in patients with gastric cancer: protocol for a systematic review. BMJ Open 2024; 14:e086986. [PMID: 39675826 PMCID: PMC11647349 DOI: 10.1136/bmjopen-2024-086986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 11/22/2024] [Indexed: 12/17/2024] Open
Abstract
INTRODUCTION The integration of chemotherapy and immunotherapy is continually making new advances in the field of gastric cancer treatment and has already become the new standard of care for the disease. However, few systematic reviews cover a wide array of immune checkpoint inhibitors (ICIs), antibody types and therapeutic modalities (perioperative or systemic) in this domain. Our aim is to incorporate the most recent clinical studies on combination therapy for the treatment of gastric cancer into a systematic review. This will comprehensively assess the benefits and drawbacks for patients with gastric cancer and will guide future research in this area. Ultimately, this will provide evidence-based support for patients with gastric cancer. METHODS AND ANALYSIS This protocol for a systematic review adheres to the Preferred Reporting Items for Systematic Review and Meta-analysis - Protocols (PRISMA-P) guidelines. We will conduct a search of the MEDLINE, Embase, and CENTRAL databases from database inception, all retrieved literature will be screened by two authors in two rounds. Inclusion criteria will be randomised clinical trials, reported in English, that compare immunotherapy-combined therapy with chemotherapy alone in the perioperative and systematic first-line treatment of gastric cancer. The primary outcomes will be progression-free survival (PFS) and overall survival (OS). Secondary outcomes will include objective response rate (ORR), event-free survival (EFS), disease-free survival (DFS), pathologic complete response (pCR), major pathologic response (MPR), and the R0 resection rate. Heterogeneity and publication bias of included literature will be investigated. Where included literatures allow, we will conduct meta-analyses and subgroup analyses to further refine the pros and cons of combined therapy, providing evidence-based foundations for subsequent research. ETHICS AND DISSEMINATION This work is a review based on existing literature and no ethical review is required. Dissemination of the results will occur via academic journal publication, conference presentations, and multiple media platforms. PROSPERO REGISTRATION NUMBER CRD42023477353.
Collapse
Affiliation(s)
- Yihui Han
- Department of General Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Gastric Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Wenming Yang
- Department of Pediatric Urology, Sichuan University West China Second University Hospital, Chengdu, Sichuan, China
| | - Yang Meng
- State Key Laboratory of Biotherapy and Cancer Center, Frontiers Science Center for Disease-Related Molecular Network, and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Zhaolun Cai
- Department of General Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Gastric Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Bo Zhang
- Department of General Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Gastric Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
23
|
Voutsadakis IA. Gastric Adenocarcinomas with CDX2 Induction Show Higher Frequency of TP53 and KMT2B Mutations and MYC Amplifications but Similar Survival Compared with Cancers with No CDX2 Induction. J Clin Med 2024; 13:7635. [PMID: 39768557 PMCID: PMC11727917 DOI: 10.3390/jcm13247635] [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: 11/11/2024] [Revised: 12/07/2024] [Accepted: 12/13/2024] [Indexed: 01/16/2025] Open
Abstract
Background: Gastric cancer is one of the most prevalent gastrointestinal cancers. Mortality is high, and improved treatments are needed. A better understanding of the pathophysiology of the disease and discovery of biomarkers for targeted therapies are paramount for therapeutic progress. CDX2, a transcription factor of hindgut specification, is induced in several gastric cancers, especially with intestinal differentiation, and could be helpful for defining sub-types with particular characteristics. Methods: Gastric cancers with induced CDX2 mRNA expression were identified from the gastric cohort of The Cancer Genome Atlas (TCGA) and were compared with cancers that had no CDX2 mRNA induction. Induced CDX2 mRNA expression was defined as mRNA expression z-score relative to all samples above 0, and non-induced CDX2 mRNA expression was defined as mRNA expression z-score relative to all samples below -1. Results: Patients with gastric cancers with CDX2 mRNA induction were older, had less frequently diffuse histology, and more often had mutations in TP53 and KMT2B and amplifications in MYC. CDX2 induction was correlated with HNF4α induction and was reversely correlated with SOX2. Gastric cancers with CDX2 mRNA induction showed lower PD-L1 expression than cancers with lower CDX2 expression but did not differ in CLDN18 mRNA expression. Progression-free and overall survival of the two groups was also not significantly different. Conclusion: Gastric cancers with CDX2 mRNA induction displayed specific characteristics that differentiate them from cancers with no CDX2 induction and could be of interest for optimizing current and future therapies.
Collapse
Affiliation(s)
- Ioannis A. Voutsadakis
- Algoma District Cancer Program, Sault Area Hospital, 750 Great Northern Road, Sault Ste Marie, ON P6B 0A8, Canada; or
- Division of Clinical Sciences, Section of Internal Medicine, Northern Ontario School of Medicine, Sudbury, ON P3E 2C6, Canada
| |
Collapse
|
24
|
Lin F, Zhu LX, Ye ZM, Peng F, Chen MC, Li XM, Zhu ZH, Zhu Y. Computed Tomography-Based Intratumor Heterogeneity Predicts Response to Immunotherapy Plus Chemotherapy in Esophageal Squamous Cell Carcinoma. Acad Radiol 2024; 31:4886-4899. [PMID: 38981774 DOI: 10.1016/j.acra.2024.06.032] [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: 04/18/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 07/11/2024]
Abstract
RATIONALE AND OBJECTIVES This study explored the intratumor heterogeneity (ITH) of esophageal squamous cell carcinoma (ESCC) using computed tomography (CT) and investigated the value of CT-based ITH in predicting the response to immune checkpoint inhibitor (ICI) plus chemotherapy in patients with ESCC. MATERIALS AND METHODS This retrospective study included 416 patients with ESCC who received ICI plus chemotherapy at two independent hospitals between January 2019 and July 2022. Multiparametric CT features were extracted from ESCC lesions and screened using hierarchical clustering and dimensionality reduction algorithms. Logistic regression and machine learning models based on selected features were developed to predict treatment response and validated in separate datasets. ITH was quantified using the score calculated by the best-performing model and visualized through feature clustering and feature contribution heatmaps. A gene set enrichment analysis (GSEA) was performed to identify the biological pathways underlying the CT-based ITH. RESULTS The extreme gradient boosting model based on CT-derived ITH had higher discriminative power, with areas under the receiver operating characteristic curve of 0.864 (95% confidence interval [CI]: 0.774-0.954) and 0.796 (95% CI: 0.698-0.893) in the internal and external validation sets. The CT-based ITH pattern differed significantly between responding and non-responding patients. The GSEA indicated that CT-based ITH was associated with immunity-, keratinization-, and epidermal cell differentiation-related pathways. CONCLUSION CT-based ITH is an effective biomarker for identifying patients with ESCC who could benefit from ICI plus chemotherapy. Immunity-, keratinization-, and epidermal cell differentiation-related pathways may influence the patient's response to ICI plus chemotherapy.
Collapse
Affiliation(s)
- Fangzeng Lin
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong Province, People's Republic of China (F.L., M.C.C., Y.Z.)
| | - Lian-Xin Zhu
- Medical College of Nanchang University, Nanchang 330000, Jiangxi Province, People's Republic of China (L.X.Z.); Queen Mary University of London, London, United Kingdom (L.X.Z.)
| | - Zi-Ming Ye
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou 510060, Guangdong Province, People's Republic of China (Z.M.Y., Z.H.Z.)
| | - Fang Peng
- Department of Radiation Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong Province, People's Republic of China (F.P.)
| | - Mei-Cheng Chen
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong Province, People's Republic of China (F.L., M.C.C., Y.Z.)
| | - Xiang-Min Li
- Department of Radiology, Hui Ya Hospital of The First Affiliated Hospital, Sun Yat-sen University, Huizhou 516080, Guangdong Province, People's Republic of China (X.M.L.)
| | - Zhi-Hua Zhu
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou 510060, Guangdong Province, People's Republic of China (Z.M.Y., Z.H.Z.)
| | - Ying Zhu
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong Province, People's Republic of China (F.L., M.C.C., Y.Z.).
| |
Collapse
|
25
|
Tian B, Bian Y, Pang Y, Gao Y, Yu C, Zhang X, Zhou S, Li Z, Xin L, Lin H, Wang L. Dysregulated inclusion of BOLA3 exon 3 promoted by HNRNPC accelerates the progression of esophageal squamous cell carcinoma. Front Med 2024; 18:1035-1053. [PMID: 39455467 DOI: 10.1007/s11684-024-1068-4] [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: 10/05/2023] [Accepted: 01/31/2024] [Indexed: 10/28/2024]
Abstract
Dysregulated RNA splicing events produce transcripts that facilitate esophageal squamous cell carcinoma (ESCC) progression, but how this splicing process is abnormally regulated remains elusive. Here, we unveiled a novel alternative splicing axis of BOLA3 transcripts and its regulator HNRNPC in ESCC. The long-form BOLA3 (BOLA3-L) containing exon 3 exhibited high expression levels in ESCC and was associated with poor prognosis. Functional assays demonstrated the protumorigenic function of BOLA3-L in ESCC cells. Additionally, HNRNPC bound to BOLA3 mRNA and promoted BOLA3 exon 3 inclusion forming BOLA3-L. High HNRNPC expression was positively correlated with the presence of BOLA3-L and associated with an unfavorable prognosis. HNRNPC knockdown effectively suppressed the malignant biological behavior of ESCC cells, which were significantly rescued by BOLA3-L overexpression. Moreover, BOLA3-L played a significant role in mitochondrial structural and functional stability. E2F7 acted as a key transcription factor that promoted the upregulation of HNRNPC and inclusion of BOLA3 exon 3. Our findings provided novel insights into how alternative splicing contributes to ESCC progression.
Collapse
Affiliation(s)
- Bo Tian
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yan Bian
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yanan Pang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Ye Gao
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Chuting Yu
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Xun Zhang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Siwei Zhou
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Zhaoshen Li
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Lei Xin
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| | - Han Lin
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| | - Luowei Wang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
26
|
Randon G, Lonardi S, Fassan M, Palermo F, Tamberi S, Giommoni E, Ceccon C, Di Donato S, Fornaro L, Brunetti O, De Vita F, Bittoni A, Chini C, Spallanzani A, Nappo F, Bethaz V, Strippoli A, Latiano T, Cardellino GG, Giuliani F, Morano F, Niger M, Raimondi A, Prisciandaro M, Pircher CC, Sciortino C, Marchesi S, Garattini SK, Airò G, Miceli R, Di Bartolomeo M, Pietrantonio F. Ramucirumab plus paclitaxel as switch maintenance versus continuation of first-line oxaliplatin-based chemotherapy in patients with advanced HER2-negative gastric or gastro-oesophageal junction cancer (ARMANI): a randomised, open-label, multicentre, phase 3 trial. Lancet Oncol 2024; 25:1539-1550. [PMID: 39557058 DOI: 10.1016/s1470-2045(24)00580-1] [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: 08/28/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 11/20/2024]
Abstract
BACKGROUND Paclitaxel plus ramucirumab is recommended as a second-line treatment regimen in patients with advanced HER2-negative gastric or gastro-oesophageal junction cancer. We aimed to assess whether switch maintenance or early second-line therapy with paclitaxel plus ramucirumab improved outcomes compared with continuation of oxaliplatin and fluoropyrimidine doublet chemotherapy as a first-line strategy. METHODS ARMANI was a multicentre, open-label, randomised, phase 3 trial done in 31 hospitals in Italy. We enrolled patients aged 18 years or older with an Eastern Cooperative Oncology Group performance status of 0 or 1 and locally advanced unresectable or metastatic HER2-negative gastric or gastro-oesophageal junction cancer, who had disease control after 3 months of FOLFOX (leucovorin, fluorouracil, and oxaliplatin) or CAPOX (capecitabine and oxaliplatin). Patients were randomly assigned (1:1) to either paclitaxel 80 mg/m2 on days 1, 8, and 15 plus ramucirumab at 8 mg/kg on days 1 and 15 every 28 days intravenously (switch maintenance group) or continuation of oxaliplatin-based doublet chemotherapy (FOLFOX or CAPOX) for an additional 12 weeks, followed by fluoropyrimidine monotherapy maintenance (control group). Randomisation was stratified by previous gastrectomy (no vs yes), peritoneal carcinomatosis (yes vs no), and primary tumour location (gastro-oesophageal junction vs gastric). Treatment group allocation was done using a web-based system with a minimisation algorithm implementing a random component. The primary endpoint was progression-free survival, analysed on an intention-to-treat basis. The safety population included patients who received at least one dose of the study treatment. This study is registered with ClinicalTrials.gov, NCT02934464, and is complete. FINDINGS Between Jan 1, 2017, and Oct 2, 2023, 280 patients were randomly assigned to receive paclitaxel plus ramucirumab (switch maintenance group; n=144) or to continue FOLFOX or CAPOX (control group; n=136). All patients were White. 180 (64%) of 280 patients were male and 100 (36%) were female. At a median follow-up of 43·7 months (IQR 24·0-57·9), 253 (90%) of 280 patients had a progression-free survival event: 131 (91%) of 144 patients in the switch maintenance group and 122 (90%) of 136 patients in the control group. Median progression-free survival was 6·6 months (95% CI 5·9-7·8) in the switch maintenance group and 3·5 months (2·8-4·2) in the control group (HR 0·61, 95% CI 0·48-0·79; p=0·0002). The assumption of proportional hazards was violated; in an analysis of 24-month restricted mean survival time, restricted mean progression-free survival was 8·8 months (95% CI 7·7-9·9) in the switch maintenance group and 6·1 months (5·0-7·2) in the control group (p=0·0010). The most frequent grade 3-4 treatment-related adverse events were neutropenia (37 [26%] patients in the switch maintenance group vs 13 [10%] patients in the control group), peripheral neuropathy (eight [6%] vs nine [7%]) and arterial hypertension (nine [6%] vs none). Serious adverse events occurred in 28 (20%) of 141 patients in the experimental group and 15 (11%) of 135 patients in the control group; these events were treatment-related in two (1%) patients in the switch maintenance group (pulmonary embolism) and two (1%) patients in the control group (mucositis and anaemia). No treatment-related deaths occurred. INTERPRETATION Paclitaxel and ramucirumab switch maintenance could be a potential treatment strategy in patients with advanced HER2-negative gastric or gastro-oesophageal junction cancer who are not eligible for immunotherapy or targeted agents. FUNDING Partly funded by Eli Lilly.
Collapse
Affiliation(s)
- Giovanni Randon
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Sara Lonardi
- Department of Oncology, Veneto Institute of Oncology Istituto Oncologico Veneto-IRCCS, Padua, Italy
| | - Matteo Fassan
- Surgical Pathology Unit, Department of Medicine, University Hospital of Padua, Padua, Italy; Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Federica Palermo
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Stefano Tamberi
- Oncology Unit, Santa Maria delle Croci Hospital, Azienda Unità Sanitaria Locale della Romagna, Ravenna, Italy
| | - Elisa Giommoni
- Medical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - Carlotta Ceccon
- Surgical Pathology Unit, Department of Medicine, University Hospital of Padua, Padua, Italy
| | - Samantha Di Donato
- Medical Oncology Department ASL Toscana Centro, Santo Stefano Hospital, Prato, Italy
| | - Lorenzo Fornaro
- Unit of Medical Oncology, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
| | - Oronzo Brunetti
- Istituto Tumori "Giovanni Paolo II" of Bari IRCCS, Bari, Italy
| | - Ferdinando De Vita
- Division of Medical Oncology, Department of Precision Medicine, University of Campania "L Vanvitelli", Naples, Italy
| | - Alessandro Bittoni
- Oncology Unit, Istituto Romagnolo Per Lo Studio Dei Tumori "Dino Amadori" IRCCS, Meldola, Italy
| | - Claudio Chini
- Department of Medical Oncology, ASST Sette Laghi, Ospedale di Circolo e Fondazione Macchi, Varese, Italy
| | - Andrea Spallanzani
- Department of Oncology and Haematology, Division of Oncology, University Hospital of Modena, Modena, Italy
| | - Floriana Nappo
- Department of Oncology, Veneto Institute of Oncology Istituto Oncologico Veneto-IRCCS, Padua, Italy
| | - Valerie Bethaz
- Department of Oncology, "San Luigi Gonzaga" University Hospital, University of Turin, Orbassano, Italy
| | - Antonia Strippoli
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Tiziana Latiano
- Department of Oncology, Fondazione IRCCS "Casa Sollievo della Sofferenza" Hospital, San Giovanni Rotondo, Foggia, Italy
| | | | - Francesco Giuliani
- Istituto Tumori "Giovanni Paolo II" of Bari IRCCS, Bari, Italy; Medical Oncology Unit, San Paolo Hospital, Bari, Italy
| | - Federica Morano
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Monica Niger
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alessandra Raimondi
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Michele Prisciandaro
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Carolina Sciortino
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvia Marchesi
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvio Ken Garattini
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Department of Oncology, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | - Giulia Airò
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Rosalba Miceli
- Unit of Biostatistics for Clinical Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Maria Di Bartolomeo
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| |
Collapse
|
27
|
Kahana N, Boaz E, Horesh N, Emile SH, Dourado J, Aeschbacher P, Rogers P, Gefen R, Lo Menzo E, Rosenthal RJ. Evaluation of the robustness of randomized controlled trials for the treatment modalities of esophageal cancer using the fragility index - a systematic review. Surg Endosc 2024; 38:7037-7044. [PMID: 39443379 DOI: 10.1007/s00464-024-11343-3] [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: 03/16/2024] [Accepted: 10/06/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Esophageal cancer remains a significant global health challenge. Several treatment modalities were explored in randomized controlled trials (RCTs) in recent decades. This study evaluates the robustness of RCTs focusing on esophageal cancer treatment using the fragility index (FI) and reverse fragility index (RFI). METHODS A systematic review of RCTs studying different treatment modalities for esophageal cancer from 2000 to 2023 was conducted. The FI and RFI were utilized to gauge the robustness of statistically significant and non-significant outcomes, respectively. The FI represents the minimal number of patient outcomes that would need to alter to overturn a trial's statistical significance, while RFI indicates the minimal changes required to achieve significance in non-significant results. RESULTS Out of 4028 studies retrieved, 21 RCTs were included for final analysis. The studies spanned 2001 to 2023 with a mean followup of 66 months (range, 29-108 months) and median number of patients of 194 (range, 45-802). The most common treatment modalities examined in these studies were neoadjuvant chemoradiotherapy (n = 7, 33.3%), neoadjuvant chemotherapy (n = 4, 19.0%), and neoadjuvant immunotherapy (n = 2, 9.5%). Only 5 studies (23.8%) had a statistically significant primary outcome result with a median FI of 6 (IQR, 2.5-8.5). Non-significant primary outcomes were seen in 16 studies (76.2%) with a median RFI of 4 (IQR 1-11) and lost to followup of 0 (IQR 0-4). In the study with the highest FI (10), the FI was lower than the number of patients lost to followup (13). CONCLUSION Our findings demonstrate that most RCTs on esophageal cancer treatments did not report significant primary outcomes. The few studies that reported significant results had a low fragility index, suggesting a vulnerability in their findings.
Collapse
Affiliation(s)
- Noam Kahana
- Department of General Surgery, Cleveland Clinic Florida, 2950 Cleveland Clinic Blvd., Weston, FL, 33331, USA
- Department of General Surgery Shaare Zedek Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Elad Boaz
- Department of General Surgery, Cleveland Clinic Florida, 2950 Cleveland Clinic Blvd., Weston, FL, 33331, USA
- Department of General Surgery Shaare Zedek Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nir Horesh
- Department of General Surgery, Cleveland Clinic Florida, 2950 Cleveland Clinic Blvd., Weston, FL, 33331, USA
- Department of Surgery and Transplantations, Sheba Medical Center, Ramat Gan, Israel, Tel Aviv University, Tel Aviv, Israel
| | - Sameh Hany Emile
- Department of General Surgery, Cleveland Clinic Florida, 2950 Cleveland Clinic Blvd., Weston, FL, 33331, USA
- Colorectal Surgery Unit, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Justin Dourado
- Department of General Surgery, Cleveland Clinic Florida, 2950 Cleveland Clinic Blvd., Weston, FL, 33331, USA
| | - Pauline Aeschbacher
- Department of General Surgery, Cleveland Clinic Florida, 2950 Cleveland Clinic Blvd., Weston, FL, 33331, USA
| | - Pete Rogers
- Department of General Surgery, Cleveland Clinic Florida, 2950 Cleveland Clinic Blvd., Weston, FL, 33331, USA
| | - Rachel Gefen
- Department of General Surgery, Cleveland Clinic Florida, 2950 Cleveland Clinic Blvd., Weston, FL, 33331, USA
- Department of General Surgery, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Emanuele Lo Menzo
- Department of General Surgery, Cleveland Clinic Florida, 2950 Cleveland Clinic Blvd., Weston, FL, 33331, USA
| | - Raul J Rosenthal
- Department of General Surgery, Cleveland Clinic Florida, 2950 Cleveland Clinic Blvd., Weston, FL, 33331, USA.
| |
Collapse
|
28
|
Yang M, Lin W, Huang J, Mannucci A, Luo H. Novel immunotherapeutic approaches in gastric cancer. PRECISION CLINICAL MEDICINE 2024; 7:pbae020. [PMID: 39397869 PMCID: PMC11467695 DOI: 10.1093/pcmedi/pbae020] [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: 06/25/2024] [Revised: 09/08/2024] [Accepted: 09/08/2024] [Indexed: 10/15/2024] Open
Abstract
Gastric cancer is a malignant tumor that ranks third in cancer-related deaths worldwide. Early-stage gastric cancer can often be effectively managed through surgical resection. However, the majority of cases are diagnosed in advanced stages, where outcomes with conventional radiotherapy and chemotherapy remain unsatisfactory. Immunotherapy offers a novel approach to treating molecularly heterogeneous gastric cancer by modifying the immunosuppressive tumor microenvironment. Immune checkpoint inhibitors and adoptive cell therapy are regarded as promising modalities in cancer immunotherapy. Food and Drug Administration-approved programmed death-receptor inhibitors, such as pembrolizumab, in combination with chemotherapy, have significantly extended overall survival in gastric cancer patients and is recommended as a first-line treatment. Despite challenges in solid tumor applications, adoptive cell therapy has demonstrated efficacy against various targets in gastric cancer treatment. Among these approaches, chimeric antigen receptor-T cell therapy research is the most widely explored and chimeric antigen receptor-T cell therapy targeting claudin18.2 has shown acceptable safety and robust anti-tumor capabilities. However, these advancements primarily remain in preclinical stages and further investigation should be made to promote their clinical application. This review summarizes the latest research on immune checkpoint inhibitors and adoptive cell therapy and their limitations, as well as the role of nanoparticles in enhancing immunotherapy.
Collapse
Affiliation(s)
- Meng Yang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China
| | - Wuhao Lin
- Department of Molecular Diagnostics, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Jiaqian Huang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China
| | - Alessandro Mannucci
- Gastroenterology and Gastrointestinal Emndoscopy Unit, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan 20132, Italy
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope; Monrovia, CA 91016, USA
| | - Huiyan Luo
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China
| |
Collapse
|
29
|
Obayashi Y, Hirata S, Kono Y, Abe M, Miyahara K, Nakagawa M, Ishida M, Choda Y, Hamada K, Iwamuro M, Kawano S, Kawahara Y, Otsuka M. Clinical Significance of Prior Ramucirumab Use on the Effectiveness of Nivolumab as the Third-Line Regimen in Gastric Cancer: A Multicenter Retrospective Study. Drugs Real World Outcomes 2024; 11:557-564. [PMID: 39425893 PMCID: PMC11589076 DOI: 10.1007/s40801-024-00460-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND AND OBJECTIVE Because vascular endothelial growth factor inhibition has been suggested to improve immune cell function in the cancer microenvironment, we examined whether using ramucirumab (RAM) before nivolumab usage is more effective in advanced gastric cancer. METHODS This was a multicenter retrospective observational study. We analyzed patients who received nivolumab monotherapy as the third-line regimen for unresectable advanced or recurrent gastric cancer between October 2017 and December 2022. They were divided into the RAM (RAM-treated) group and the non-RAM (non-treated) group according to the RAM usage in the second-line regimen. The primary outcome was to compare the overall survival after nivolumab administration in the third-line regimen between the RAM and non-RAM groups. RESULTS Fifty-two patients were included in the present study: 42 patients in the RAM group and ten patients in the non-RAM group. The median overall survival was significantly longer in the RAM group than in the non-RAM group (8.5 months vs 6.9 months, p < 0.05). In the RAM group, patients without peritoneal metastasis had significantly better median overall survival than those with peritoneal metastasis (23.8 months vs 7.7 months, p = 0.0033). Multivariate Cox-proportional hazards analyses showed that the presence of peritoneal metastasis (hazard ratio, 2.4; 95% confidence interval 1.0-5.7) alone was significantly associated with overall survival in the RAM group. CONCLUSIONS The use of RAM prior to nivolumab monotherapy may contribute to prolonged survival in patients with gastric cancer, especially those without peritoneal metastasis.
Collapse
Affiliation(s)
- Yuka Obayashi
- Department of Internal Medicine, Hiroshima City Hiroshima Citizens Hospital, Hiroshima, Japan
| | - Shoichiro Hirata
- Department of Gastroenterology and Hepatology, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Yoshiyasu Kono
- Department of Gastroenterology and Hepatology, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan.
| | - Makoto Abe
- Department of Internal Medicine, Hiroshima City Hiroshima Citizens Hospital, Hiroshima, Japan
| | - Koji Miyahara
- Department of Internal Medicine, Hiroshima City Hiroshima Citizens Hospital, Hiroshima, Japan
| | - Masahiro Nakagawa
- Department of Endoscopy, Hiroshima City Hiroshima Citizens Hospital, Hiroshima, Japan
| | - Michihiro Ishida
- Department of Surgery, Hiroshima City Hiroshima Citizens Hospital, Hiroshima, Japan
| | - Yasuhiro Choda
- Department of Surgery, Hiroshima City Hiroshima Citizens Hospital, Hiroshima, Japan
| | - Kenta Hamada
- Department of Practical Gastrointestinal Endoscopy, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Masaya Iwamuro
- Department of Gastroenterology, Okayama University Hospital, Okayama, Japan
| | - Seiji Kawano
- Department of Gastroenterology, Okayama University Hospital, Okayama, Japan
| | - Yoshiro Kawahara
- Department of Practical Gastrointestinal Endoscopy, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Motoyuki Otsuka
- Department of Gastroenterology and Hepatology, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| |
Collapse
|
30
|
Qian M, Fang Y, Xiang Z, Zhang Y, Zhan H, Chen X, Chen Y, Xu T. The efficacy of neoadjuvant immunotherapy in gastric cancer, adenocarcinoma of the esophagogastric junction, and esophageal cancer: a meta-analysis. Front Oncol 2024; 14:1502611. [PMID: 39650059 PMCID: PMC11621004 DOI: 10.3389/fonc.2024.1502611] [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: 09/27/2024] [Accepted: 11/07/2024] [Indexed: 12/11/2024] Open
Abstract
Background Neoadjuvant immunotherapy holds promise in managing resectable locally advanced gastric cancer (GC), adenocarcinoma of the esophagogastric junction (AEG), and esophageal cancer (EC). However, consensus is lacking regarding the efficacy of programmed death-1 (PD-1) and programmed death ligand 1 (PD-L1) inhibitors in neoadjuvant immunochemotherapy (NICT). This study aims to assess the added benefit of PD-1/PD-L1 inhibitors in neoadjuvant chemotherapy (NCT) for these malignancies. Methods Up to October 2024, randomized controlled trials, case-control studies, and cohort studies that evaluated the addition of PD-1/PD-L1 inhibitors to NCT were systematically retrieved from electronic databases. The primary endpoints included pathologic complete response (pCR), major pathological response (MPR), overall survival (OS), and progression-free survival (PFS). Results Thirteen studies published between 2021 and 2024 were analyzed. Statistical analyses revealed significantly higher pCR rates (OR: 2.73, P < 0.001) and MPR rates (OR: 2.99, P < 0.001) in the NICT group compared to NCT group. The PFS was also higher in the NICT group, although the difference did not reach statistical significance (HR: 0.50, P = 0.072). Conclusion This meta-analysis demonstrates that NICT enhances pathological response rates in patients with resectable locally advanced GC, AEG, and EC. However, no significant long-term prognostic benefits were associated with NICT. Systematic review registration https://www.crd.york.ac.uk/prospero, identifier CRD42024545725.
Collapse
Affiliation(s)
- Mengyi Qian
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yingying Fang
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhiyi Xiang
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yueming Zhang
- Intensive Care Unit, Hospital of Zhejiang People’s Armed Police, Hangzhou, China
| | - Hujie Zhan
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Xiaotong Chen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yihang Chen
- School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, China
| | - Tinghui Xu
- Department of Cardiothoracic Surgery, Ningbo Yinzhou No. 2 Hospital, Ningbo, China
| |
Collapse
|
31
|
Formica V, Morelli C, Fornaro L, Riondino S, Rofei M, Fontana E, Smyth EC, Roselli M, Arkenau HT. PD-L1 thresholds predict efficacy of immune checkpoint inhibition in first-line treatment of advanced gastroesophageal adenocarcinoma. A systematic review and meta-analysis of seven phase III randomized trials. ESMO Open 2024; 9:103967. [PMID: 39541621 PMCID: PMC11613432 DOI: 10.1016/j.esmoop.2024.103967] [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: 06/03/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND High expression of programmed death-ligand 1 (PD-L1) has been recognized as a marker of improved efficacy of immunotherapy in gastroesophageal adenocarcinoma (GEA); however, the optimal PD-L1 cut-off is still debated. The aim of the present review was to analyze available phase III trials and to identify the appropriate PD-L1 expression cut-off for GEA. METHODS Phase III trials investigating the efficacy of anti-programmed cell death protein 1 (PD-1) therapies in addition to standard chemotherapy versus standard chemotherapy in the first-line setting were selected. Progression-free survival (PFS), overall survival (OS) and objective response rate (ORR) were the analyzed outcome measures. Pooled treatment effects were assessed in the unselected population and in subpopulations with different levels of PD-L1 expression. RESULTS PD-1 blockade efficacy was found to consistently increase in a linear manner with higher combined positive score (CPS) of PD-L1 expression: pooled hazard ratio (HR) for OS and PFS and pooled odds ratio (OR) for ORR of 0.80, 0.75 and 1.51, respectively, in the unselected population versus 0.67, 0.63 and 1.90, respectively, in the CPS ≥10 population (all P values < 0.0001). In the PD-L1-negative population (CPS <1) a significant benefit of anti-PD-1 agents could not be demonstrated in terms of OS and PFS (P = 0.28 and 0.12, respectively), but it was seen in terms of ORR (P = 0.03). PD-1 blockade was effective in the CPS <10 population (P value for pooled OS HR, PFS HR and response OR are all 0.01), while in the CPS <5 population the effect was of borderline significance for OS (P = 0.07) and significant for PFS and ORR (P = 0.02 and 0.03, respectively). CONCLUSION The present meta-analysis confirmed that the benefit of PD-1 blockade in GEA patients is related to PD-L1 CPS, with increased benefit observed for higher CPS cut-offs and no OS benefit in the CPS <1 subset. Overall, data indicate that PD-L1 CPS ≥5 could represent an acceptable cut-off to optimize the risk/benefit ratio of such agents. Our data suggest a potential clinical benefit of immunotherapy in selected patients within the CPS 1-4 population which needs further investigation.
Collapse
Affiliation(s)
- V Formica
- Medical Oncology Unit, Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy.
| | - C Morelli
- Medical Oncology Unit, Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - L Fornaro
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - S Riondino
- Medical Oncology Unit, Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - M Rofei
- Medical Oncology Unit, Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - E Fontana
- Sarah Cannon Research Institute UK, London, UK
| | - E C Smyth
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK
| | - M Roselli
- Medical Oncology Unit, Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | | |
Collapse
|
32
|
Fassan M, Kuwata T, Matkowskyj KA, Röcken C, Rüschoff J. Claudin-18.2 Immunohistochemical Evaluation in Gastric and Gastroesophageal Junction Adenocarcinomas to Direct Targeted Therapy: A Practical Approach. Mod Pathol 2024; 37:100589. [PMID: 39098518 DOI: 10.1016/j.modpat.2024.100589] [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: 04/24/2024] [Revised: 06/14/2024] [Accepted: 07/29/2024] [Indexed: 08/06/2024]
Abstract
Claudin-18.2 (CLDN18.2) expression evaluated by immunohistochemistry is a new biomarker for gastric and gastroesophageal junction adenocarcinomas that will soon have market authorization for implementation into routine clinical practice. Despite successful testing in the setting of clinical trials, no specific practical testing guidelines have been proposed. Several preanalytical and analytical variables may interfere with adequate CLDN18.2 staining interpretation; thus, this article provides practical guidance on CLDN18.2 testing and scoring in gastric and gastroesophageal junction adenocarcinomas to identify patients who may respond to targeted therapy with monoclonal antibodies directed against CLDN18.2. Based on available data, moderate to strong (2+/3+) membrane staining in ≥75% of adenocarcinoma cells is the proposed cutoff for clinical use of monoclonal antibody anti-CLDN18.2 (zolbetuximab).
Collapse
Affiliation(s)
- Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology Unit, University of Padua, Padua, Italy; Veneto Institute of Oncology, IOV-IRCCS, Padua, Italy.
| | - Takeshi Kuwata
- Department of Genetic Medicine and Services, National Cancer Center Hospital East, Chiba, Japan
| | | | - Christoph Röcken
- Department of Pathology, University-Hospital Schleswig-Holstein (UKSH), Kiel, Germany
| | - Josef Rüschoff
- Discovery Life Sciences Biomarker Services, Kassel, Germany
| |
Collapse
|
33
|
West NP, Mansoor W, Taniere P, Smyth E, Rodriguez-Justo M, Oniscu A, Carter P. Best-Practice Biomarker Testing of Oesophago-Gastric Cancer in the UK: Expert Consensus Recommendations Developed Using a Modified Delphi. Clin Oncol (R Coll Radiol) 2024; 36:701-709. [PMID: 39183086 DOI: 10.1016/j.clon.2024.08.002] [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: 06/17/2024] [Accepted: 08/06/2024] [Indexed: 08/27/2024]
Abstract
AIMS Oesophago-gastric cancers (OGCs) are amongst the most commonly diagnosed malignancies worldwide and are associated with high disease-related mortality. Predictive biomarkers are molecules that can be objectively measured and used to indicate a likely response to therapeutic intervention, thus facilitating individualised cancer therapy. However, there remains variation in uptake and implementation of biomarker testing across the UK. MATERIALS AND METHODS We conducted a modified Delphi study to formulate consensus recommendations for best-practice biomarker testing of OGC in the UK. We employed two rounds of online questionnaires followed by a virtual consensus meeting. Biomarkers for discussion included HER2, MSI/MMR, and PD-L1. Topics comprised the overall biomarker pathway, pre-analytical, analytical, and post-analytical considerations, including challenges in current practice. RESULTS Twenty-six and eighteen participants completed the first and second round Delphi questionnaire, respectively, with an even split of pathologists and oncologists from across the UK. There was consensus (>80% agreement) across several topics, including the requirements for standardisation of the pathway, which must include coordination throughout the tissue journey, requirements for a quality-assured process to ensure accuracy and validity of testing, plus the need for clear, detailed information on the pathology report to support treatment decisions. There was consensus amongst oncologists regarding reflex testing of all biomarkers depending on histology; however, concerns over capacity in relation to workload and availability of pathologists were evident among the pathologists. Overall, participants were in the opinion that reflex testing improves the speed of treatment decisions and improves patient care. CONCLUSION The recommendations reflect best-practices and should be implemented to support rapid multidisciplinary team decision-making within oesophago-gastric cancer. Results reflect the need for standardisation and demonstrate the challenges faced in clinical practice by those requesting and testing biomarkers for oesophago-gastric cancer, suggesting significant concerns relating to pathologist capacity.
Collapse
Affiliation(s)
- N P West
- Pathology & Data Analytics, Leeds Institute of Medical Research, University of Leeds and Department of Histopathology, Leeds Teaching Hospitals NHS Trust, Leeds, UK.
| | - W Mansoor
- The Christie NHS Foundation Trust, Manchester, UK.
| | - P Taniere
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.
| | - E Smyth
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| | - M Rodriguez-Justo
- University College London Hospitals NHS Foundation Trust, London, UK.
| | - A Oniscu
- Royal Infirmary of Edinburgh, NHS Lothian Edinburgh, UK; Karolinska University Hospital, Stockholm, Sweden.
| | - P Carter
- Health Economics & Outcomes Research Ltd, Rhymney House, Unit A Copse Walk, Cardiff Gate Business Park, Cardiff, UK.
| |
Collapse
|
34
|
Ren R, Zhang Z, Zhai S, Yang J, Tusong B, Wang J. Efficacy and safety of ramucirumab for gastric or gastro-esophageal junction adenocarcinoma: a systematic review and meta-analysis. Eur J Clin Pharmacol 2024; 80:1697-1714. [PMID: 39102039 DOI: 10.1007/s00228-024-03734-1] [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: 04/15/2024] [Accepted: 07/20/2024] [Indexed: 08/06/2024]
Abstract
PURPOSE Based on the comparison of ramucirumab monoclonal antibody with control treatments in randomized controlled trials, this study aims to elucidate the role of ramucirumab monoclonal antibody in cancer therapy and its potential side effects, providing scientific evidence for clinical treatment. METHODS PubMed, Embase, Cochrane, and Web of Science were searched systematically to obtain the trials on ramucirumab in the treatment of gastric or gastroesophageal junction (GEJ) adenocarcinoma up to April 13, 2023. We included randomized controlled trials (RCTs) evaluating the efficacy and safety of ramucirumab as monotherapy and in combination with other chemotherapy agents as interventions for treating gastric or gastroesophageal junction (GEJ) adenocarcinoma. RESULTS After screening 2200 studies, we finally included 8 eligible studies (involving a total of 3,283 participants). Meta-analysis results showed that compared to the control group, ramucirumab monotherapy significantly improved overall survival (OS) (hazard ratio [HR] = 0.77, 95% confidence interval [CI] [0.67, 0.89]) and progression-free survival (PFS) (HR = 0.48, 95% CI [0.40, 0.58]). Similar results were obtained for ramucirumab combined with paclitaxel. In the treatment combining ramucirumab with paclitaxel, compared to monotherapy, three severe adverse reactions (grade ≥ 3) were observed with significantly increased risks (OR > 2). These include proteinuria (OR = 5.37, 95% CI [1.22, 23.54]), hypertension (OR = 4.02, 95% CI [2.63, 6.14]), and gastrointestinal perforation (OR = 4.64, 95% CI [1.00, 21.60]). Subgroup analysis further indicated that ramucirumab is effective in both non-East Asian and East Asian populations, with East Asian patients more prone to developing proteinuria, while having a lower incidence of hypertension. Additionally, ramucirumab demonstrated comparable efficacy between first-line and second-line treatments, with a higher incidence of proteinuria observed in second-line therapy. CONCLUSION Ramucirumab significantly improves the prognosis of patients with gastric or gastroesophageal junction adenocarcinoma. When used in combination with paclitaxel, close monitoring of adverse reactions such as proteinuria (especially in East Asian populations), hypertension (especially in non-East Asian populations), and gastrointestinal perforation is essential.
Collapse
Affiliation(s)
- Ruiqi Ren
- Shihezi University School of Medicine, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China
| | - Zhewei Zhang
- Shihezi University School of Medicine, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China
| | - Shaokun Zhai
- Shihezi University School of Medicine, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China
| | - Jiahui Yang
- Shihezi University School of Medicine, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China
| | - BaihaiTihan Tusong
- Shihezi University School of Medicine, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China
| | - Jingzhou Wang
- Shihezi University School of Medicine, Bei-Er-Lu, Shihezi, 832000, Xinjiang, China.
| |
Collapse
|
35
|
Li J, Ding F, Zhang S, Jia Y, Zhang T, Wang S, Liu Q, Guo Z. Continuation of same programmed death-1 inhibitor regime beyond progression is a novel option for advanced gastric cancer. BMC Cancer 2024; 24:1292. [PMID: 39425079 PMCID: PMC11490043 DOI: 10.1186/s12885-024-13063-2] [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: 05/19/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND Gastric cancer is a significant global malignancy with poor prognosis. Although the emergence of immune checkpoint inhibitors (ICIs) prolonged the duration of survival, resistance and progression are inevitable. We aim to evaluate the effectiveness of programmed death-1 (PD-1) inhibitors in immunotherapy beyond progression (IBP). METHOD We divided the advanced gastric cancer patients who received two lines immunotherapy into same regimen group (with same PD-1 inhibitor regime after IBP) and different regimen group (with different PD-1 inhibitor regime after IBP). Statistical analysis conducted to compare patient characteristics and evaluate survival differences between groups. RESULT The clinical outcome analysis showed that the same PD-1 inhibitor regime seemed to exhibit a higher disease control rate (DCR) (51.8% vs. 29.2%, P = 0.062), significantly prolonged progression-free survival 2 (PFS2) (162 vs. 75 days, P = 0.001) and overall survival (OS) (312 vs. 166 days, P = 0.022) when compared with those of cross line. In the multivariate analysis, when using different regimen group as reference, the same regimen group was found to be independently associated with improved PFS2 [hazard ratio (HR) = 0.467, 95% confidence interval (CI): 0.267-0.816, P = 0.008] and OS (HR = 0.508, 95%CI: 0.278-0.927, P = 0.027). CONCLUSION Continuation of the same type of PD-1 inhibitor regime in IBP shows clinical benefits and represents a promising therapeutic approach.
Collapse
Affiliation(s)
- Jiasong Li
- Department of Rheumatology and Immunology, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, P.R. China
- Department of Gastroenterology and Hepatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Fang Ding
- Department of Clinical Medicine, Hebei Medical University, Zhongshan Campus, Shijiazhuang, P.R. China
| | - Shasha Zhang
- Department of Rheumatology and Immunology, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, P.R. China
| | - Yuanyuan Jia
- Department of Rheumatology and Immunology, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, P.R. China
| | - Tianhang Zhang
- Department of Rheumatology and Immunology, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, P.R. China
- Department of Gastroenterology and Hepatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Siqi Wang
- Department of Rheumatology and Immunology, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, P.R. China
- Department of Gastroenterology and Hepatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Qingyi Liu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, P.R. China.
| | - Zhanjun Guo
- Department of Rheumatology and Immunology, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, P.R. China.
| |
Collapse
|
36
|
Tong X, Jin M, Wang L, Zhang D, Yin Y, Shen Q. Prognostic biomarkers for immunotherapy in esophageal cancer. Front Immunol 2024; 15:1420399. [PMID: 39403382 PMCID: PMC11471503 DOI: 10.3389/fimmu.2024.1420399] [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: 04/20/2024] [Accepted: 09/05/2024] [Indexed: 03/22/2025] Open
Abstract
Esophageal cancer (EC), a common type of malignant tumor, ranks as the sixth highest contributor to cancer-related mortality worldwide. Due to the condition that most patients with EC are diagnosed at advanced or metastatic status, the efficacy of conventional treatments including surgery, chemotherapy and radiotherapy is limited, resulting in a dismal 5-year overall survival rate. In recent years, the application of immune checkpoint inhibitors (ICIs) has presented a novel therapeutic avenue for EC patients. Both ICIs monotherapy and immunotherapy combined with chemotherapy or chemoradiotherapy (CRT) have demonstrated marked benefits for patients with advanced EC. Adjuvant or neoadjuvant therapy incorporating immunotherapy has also demonstrated promising prospects in the context of perioperative treatment. Nonetheless, due to the variable response observed among patients undergoing immunotherapy, it is of vital importance to identify predictive biomarkers for patient stratification, to facilitate identification of subgroups who may derive greater benefits from immunotherapy. In this review, we summarize validated or potential biomarkers for immunotherapy in EC in three dimensions: tumor-cell-associated biomarkers, tumor-immune microenvironment (TIME)-associated factors, and host-associated biomarkers, so as to provide a theoretical foundation to inform tailored therapy for individuals diagnosed with EC.
Collapse
Affiliation(s)
- Xu Tong
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meiyuan Jin
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lulu Wang
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Dongli Zhang
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Yuping Yin
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Shen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
37
|
Shitara K, Xu RH, Ajani JA, Moran D, Guerrero A, Li R, Pavese J, Matsangou M, Bhattacharya P, Ueno Y, Wang X, Shah MA. Global prevalence of claudin 18 isoform 2 in tumors of patients with locally advanced unresectable or metastatic gastric or gastroesophageal junction adenocarcinoma. Gastric Cancer 2024; 27:1058-1068. [PMID: 38954176 PMCID: PMC11335819 DOI: 10.1007/s10120-024-01518-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 05/26/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND Limited data exist for global prevalence of claudin 18 isoform 2 (CLDN18.2) positivity and association of CLDN18.2 status with clinical and tumor characteristics in patients with locally advanced (LA) unresectable or metastatic gastric or gastroesophageal junction (mG/GEJ) adenocarcinoma. We report prevalence of CLDN18.2 positivity (phase 3; SPOTLIGHT, NCT03504397; GLOW, NCT03653507) and concordance of CLDN18.2 status between a subset of pair-matched tumor samples (phase 2, ILUSTRO, NCT03505320; phase 1, NCT03528629) from clinical studies of zolbetuximab. METHODS Tumor samples from patients with LA unresectable or mG/GEJ adenocarcinoma were tested for CLDN18.2 status by immunohistochemistry. Human epidermal growth factor receptor 2 (HER2) expression was tested per central or local assessment. RESULTS Across SPOTLIGHT and GLOW, the prevalence of CLDN18.2 positivity (≥ 75% of tumor cells demonstrating moderate-to-strong membranous CLDN18 staining) was 38.4%. Prevalence was similar in gastric versus GEJ adenocarcinoma samples and regardless of collection method (biopsy versus resection) or collection site (primary versus metastatic). CLDN18.2 positivity was most prevalent in patients with diffuse-type tumors. In ILUSTRO and the phase 1 study, concordance of CLDN18.2 positivity was 61.1% between archival (i.e., any time before treatment) and baseline (i.e., ≤ 3 months before first treatment) samples, and concordance of any CLDN18 staining (≥ 1% of tumor cells demonstrating moderate-to-strong membranous CLDN18 staining) was 88.9%. CONCLUSIONS CLDN18.2 was a highly prevalent biomarker in patients with HER2-negative, LA unresectable or mG/GEJ adenocarcinoma. CLDN18.2 positivity remained relatively stable over time in many patients. Biomarker testing for CLDN18.2 should be considered in standard clinical practice in these patients.
Collapse
Affiliation(s)
- Kohei Shitara
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa City, Chiba, Japan
| | - Rui-Hua Xu
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Diarmuid Moran
- Astellas Pharma Global Development, Inc., Northbrook, IL, USA
| | | | - Ran Li
- Astellas Pharma Global Development, Inc., Northbrook, IL, USA
| | - Janet Pavese
- Astellas Pharma Global Development, Inc., Northbrook, IL, USA
| | - Maria Matsangou
- Astellas Pharma Global Development, Inc., Northbrook, IL, USA
| | | | | | - Xuewei Wang
- Astellas Pharma Global Development, Inc., Northbrook, IL, USA
| | - Manish A Shah
- Division of Hematology and Medical Oncology, Weill Cornell Medical College, New York City, NY, USA.
| |
Collapse
|
38
|
Nakayama I, Shitara K. The current status of immunotherapy and future horizon in the treatment of metastatic and locally advanced gastroesophageal adenocarcinoma. Expert Opin Biol Ther 2024; 24:903-915. [PMID: 39171531 DOI: 10.1080/14712598.2024.2395921] [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: 05/01/2024] [Revised: 07/23/2024] [Accepted: 08/20/2024] [Indexed: 08/23/2024]
Abstract
INTRODUCTION Immunochemotherapy with PD-1 blockade has been established as the current standard first-line therapy for patients with mGEA. Reviewing the history of clinical trials offers valuable insight into the evolution of immune oncology in mGEA, paving the way for future advancements in this field. AREAS COVERED This review summarizes the findings of previous clinical trials related to immunotherapy for patients with GEA in the metastatic and locally advanced setting. We also introduce ongoing clinical trials to address the current challenging issues in clinical practice. EXPERT OPINION In general, GEA exhibits intermediate immunogenic characteristics with heterogeneous expressions, and responders to anti-PD-(L)1 therapy are mostly enriched to patients with specific genomic profiles such as MSI-H, high PD-L1 expression, high TMB, and EBV-associated type. Co-administration with anti-angiogenic agents or simultaneous blockade of immune checkpoint molecules is being explored to offer benefit of immunotherapy for more patients. We hope that CLDN18.2 and upcoming targets like FGFR2b will complement the treatment niche of immunotherapy in the field of mGEA. Bispecific antibodies, antibody drug conjugates, CAR-T, and vaccine are anticipated to enhance efficacy and expand the scope of immunotherapy.
Collapse
Affiliation(s)
- Izuma Nakayama
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kohei Shitara
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
39
|
Yuan W, Han J, Chen C, Qiu Y, Xu Y, Huang Y, Chen Z, Xu A, Sun M. UBR1 is a prognostic biomarker and therapeutic target associated with immune cell infiltration in gastric cancer. Aging (Albany NY) 2024; 16:12029-12049. [PMID: 39181686 PMCID: PMC11386912 DOI: 10.18632/aging.206079] [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: 12/19/2023] [Accepted: 07/15/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND Ubiquitination is a targeted protein modification process mediated by intracellular molecules. UBR1 encodes a protein that binds to unstable N-terminal residues of substrate proteins and contributes to the formation of substrate-linked polyubiquitin chains. However, the function and cellular pathways of UBR1 in tumors have received inadequate attention. This study aimed to investigate the potential of UBR1 as a prognostic biomarker and immunotherapy target for stomach adenocarcinoma (STAD) as well as its biological function and molecular mechanism in relation to the disease. METHODS Differential expression and pan-cancer gene set enrichment analysis (GSEA) were conducted using The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), and Genotype-Tissue Expression (GTEx) datasets. The Human Protein Atlas (HPA) database was utilized to identify UBR1-enriched pathways in AGS cells and to compare immunohistochemical differences between cancerous and adjacent non-cancerous tissues in gastric cancer. Quantitative Polymerase Chain Reaction (QPCR) and Western blot (WB) analyses were employed to validate these findings in both cancerous and adjacent non-cancerous tissues of gastric cancer. UBR1 expression in GES-1 and four gastric cancer cell lines was assessed using QPCR and WB. Kaplan-Meier curves, univariate and multivariate Cox regression analyses, and receiver operating characteristic (ROC) curve analyses were performed to evaluate the prognostic and diagnostic roles of UBR1. Additionally, the correlation between UBR1 expression and clinical parameters was analyzed using TCGA and GEO databases. UBR1 mutation data were obtained from the cBioPortal database. The mutation landscape, mutation-associated genes, protein structure, tumor mutation burden (TMB), and microsatellite instability (MSI) correlations were analyzed and illustrated. The biological functions of UBR1 were investigated using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. The correlation between UBR1 and immune infiltration was assessed using TIMER and EPIC computational methods. Protein expression levels of UBR1 in gastric cancer cell lines were determined by immunohistochemistry (IHC) and WB analysis. Quantitative real-time PCR (qRT-PCR) was employed to analyze mRNA expression. Immunoprecipitation (IP) assays were conducted to detect protein-protein interactions between UBR1 and PDL1, while cellular immunofluorescence was used to observe the co-localization of these proteins. Cell proliferation was evaluated using CCK8 and colony formation assays. Cell migration was assessed using Transwell and wound healing assays. Finally, apoptosis was analyzed using flow cytometry, and WB was used to detect changes in apoptotic proteins and NF-κB P65 pathway proteins. RESULTS UBR1 was upregulated in 28 cancer types, including STAD, and its overexpression was validated in gastric cancer cell lines and tissues. UBR1 expression was associated with advanced pathological characteristics. High UBR1 expression was linked to poor prognostic outcomes, including overall survival (OS), progression-free interval (PFI), disease-specific survival (DSS), as well as responses to surgery, chemotherapy, and HER2 expression. UBR1 expression showed significant correlations with clinical parameters such as age, gender, TNM stage, pathological stage, tumor resection, and anti-reflux therapy. Amplifications and deletions were the most frequent genetic alterations associated with UBR1. According to KEGG and GSEA analyses, UBR1 was significantly associated with several cancer pathways, oxidative phosphorylation, and the TNF-NFκB pathway. UBR1 also exhibited a significant correlation with immune cell infiltration and immunotherapy, including a direct interaction with PDL1. Knockdown of UBR1 inhibited the proliferation, migration, and invasion of STAD cells and promoted apoptosis. CONCLUSIONS UBR1 is overexpressed in STAD, promoting its progression and positively correlating with immune cell infiltration and immunotherapeutic responses. Therefore, UBR1 could be a promising biomarker for the prognosis and immunotherapy of STAD.
Collapse
Affiliation(s)
- Weiwei Yuan
- Department of Thyroid Surgery, Baoshan Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201999, China
| | - Jianye Han
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Chen Chen
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Anhui Public Health Clinical Center, Hefei 230012, China
| | - Yue Qiu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Yuanmin Xu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Yang Huang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Anhui Public Health Clinical Center, Hefei 230012, China
| | - Zhangming Chen
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Aman Xu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Minzhi Sun
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Anhui Public Health Clinical Center, Hefei 230012, China
| |
Collapse
|
40
|
Amilo D, Izuchukwu C, Sadri K, Yao HR, Hincal E, Shehu Y. A fractional-order model for optimizing combination therapy in heterogeneous lung cancer: integrating immunotherapy and targeted therapy to minimize side effects. Sci Rep 2024; 14:18484. [PMID: 39122747 PMCID: PMC11395867 DOI: 10.1038/s41598-024-66531-x] [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: 03/20/2024] [Accepted: 07/02/2024] [Indexed: 08/12/2024] Open
Abstract
This research presents a novel approach to address the complexities of heterogeneous lung cancer dynamics through the development of a Fractional-Order Model. Focusing on the optimization of combination therapy, the model integrates immunotherapy and targeted therapy with the specific aim of minimizing side effects. Notably, our approach incorporates a clever fusion of Proportional-Integral-Derivative (PID) feedback controls alongside the optimization process. Unlike previous studies, our model incorporates essential equations accounting for the interaction between regular and mutated cancer cells, delineates the dynamics between immune cells and mutated cancer cells, enhances immune cell cytotoxic activity, and elucidates the influence of genetic mutations on the spread of cancer cells. This refined model offers a comprehensive understanding of lung cancer progression, providing a valuable tool for the development of personalized and effective treatment strategies. the findings underscore the potential of the optimized treatment strategy in achieving key therapeutic goals, including primary tumor control, metastasis limitation, immune response enhancement, and controlled genetic mutations. The dynamic and adaptive nature of the treatment approach, coupled with economic considerations and memory effects, positions the research at the forefront of advancing precision and personalized cancer therapeutics.
Collapse
Affiliation(s)
- David Amilo
- Mathematics Research Center, Near East University TRNC, Mersin 10, 99138, Nicosia, Turkey
- Department of Mathematics, Near East University TRNC, Mersin 10, 99138, Nicosia, Turkey
- Faculty of Art and Science, University of Kyrenia, Kyrenia, TRNC, Mersin 10, Kyrenia, Turkey
| | - Chinedu Izuchukwu
- School of Mathematics, University of the Witwatersrand, Private Bag 3, Johannesburg, 2050, South Africa.
| | - Khadijeh Sadri
- Mathematics Research Center, Near East University TRNC, Mersin 10, 99138, Nicosia, Turkey
- Department of Mathematics, Near East University TRNC, Mersin 10, 99138, Nicosia, Turkey
- Faculty of Art and Science, University of Kyrenia, Kyrenia, TRNC, Mersin 10, Kyrenia, Turkey
| | - Hao-Ren Yao
- National Institutes of Health, Bethesda, MD, USA
| | - Evren Hincal
- Mathematics Research Center, Near East University TRNC, Mersin 10, 99138, Nicosia, Turkey
- Department of Mathematics, Near East University TRNC, Mersin 10, 99138, Nicosia, Turkey
- Faculty of Art and Science, University of Kyrenia, Kyrenia, TRNC, Mersin 10, Kyrenia, Turkey
| | - Yekini Shehu
- School of Mathematical Sciences, Zhejiang Normal University, Jinhua, 321004, People's Republic of China
| |
Collapse
|
41
|
Ketch PW, Zaharias RS, Leath CA. Pharmacotherapy for cervical cancer: current standard of care and new perspectives. Expert Opin Pharmacother 2024; 25:1591-1603. [PMID: 39164924 PMCID: PMC11453679 DOI: 10.1080/14656566.2024.2395379] [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: 06/17/2024] [Accepted: 08/19/2024] [Indexed: 08/22/2024]
Abstract
INTRODUCTION Cervical cancer, while highly preventable, remains an international public health challenge especially in under resourced regions. Although early-stage cervix confined cancers are often amenable to surgical resection, larger tumors deemed locally advanced cervical cancer (LACC) necessitate systemic therapy as part of chemoradiation therapy. Moreover, systemic therapy is the standard therapeutic approach for those presenting with primary metastasis or recurrence. AREAS COVERED While several agents have been approved to treat recurrent cervical cancer including checkpoint inhibitors as well as both biomarker agnostic and specific antibody drug conjugates, the development of agents added to chemoradiation has been less fruitful. Until recently, the addition of novel therapies to chemoradiation has been negative in terms of improving outcomes; however, results of a recent Phase III clinical trial (NCT04221945) in LACC demonstrated that the addition of pembrolizumab to standard of care chemoradiation was associated with an improvement in progression-free survival and resulted in an FDA approval for this therapy. This observation led to the first change in treating LACC since the early 2000s. EXPERT OPINION Improvements in systemic therapy both alone and in combination with chemoradiation for cervical cancer have been realized. Ongoing research is needed for therapeutic options following immunotherapy.
Collapse
Affiliation(s)
- Peter W. Ketch
- Division of Gynecologic Oncology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Rennan S. Zaharias
- Division of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Charles A. Leath
- Division of Gynecologic Oncology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
42
|
Sun YQ, Zhong Q, Lv CB, Zhu JY, Lin GT, Zhang ZQ, Wu D, Weng CM, Chen QX, Lian MQ, Zeng WM, Zhang YB, Chen QY, Lin JX, Xie JW, Li P, Zheng CH, Lu J, Cai LS, Huang CM. The safety and efficacy of neoadjuvant immunochemotherapy following laparoscopic gastrectomy for gastric cancer: a multicentre real-world clinical study. Int J Surg 2024; 110:4830-4838. [PMID: 38652275 PMCID: PMC11326023 DOI: 10.1097/js9.0000000000001468] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 03/31/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND The safety and efficacy of neoadjuvant immunochemotherapy (nICT) for locally advanced gastric cancer (LAGC) remain controversial. METHODS Patients with LAGC who received either nICT or neoadjuvant chemotherapy (nCT) at 3 tertiary referral teaching hospitals in China between January 2016 and October 2022 were analyzed. After propensity-score matching (PSM), comparing the radiological response, pathological response rate, perioperative outcomes, and early recurrence between the two groups. RESULTS After PSM, 585 patients were included, with 195 and 390 patients comprising the nICT and nCT groups, respectively. The nICT group exhibited a higher objective response rate (79.5% vs. 59.0%; P <0.001), pathological complete response rate (14.36% vs. 6.41%; P =0.002) and major pathological response rate (39.49% vs. 26.15%; P =0.001) compared with the nCT group. The incidence of surgical complications (17.44% vs. 16.15%, P =0.694) and the proportion of perioperative textbook outcomes (80.0% vs. 81.0%; P =0.767) were similar in both groups. The nICT group had a significantly lower proportion of early recurrence than the nCT group (29.7% vs. 40.8%; P =0.047). Furthermore, the multivariable logistic analysis revealed that immunotherapy was an independent protective factor against early recurrence [odds ratio 0.62 (95% CI 0.41-0.92); P =0.018]. No significant difference was found in neoadjuvant therapy drug toxicity between the two groups (51.79% vs. 45.38%; P =0.143). CONCLUSIONS Compared with nCT, nICT is safe and effective, which significantly enhanced objective and pathological response rates and reduced the risk for early recurrence among patients with LAGC. TRIAL REGISTRATION Clinical Trials.gov.
Collapse
Affiliation(s)
- Yu-Qin Sun
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Qing Zhong
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Chen-Bin Lv
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
| | - Ji-Yun Zhu
- Department of General Surgery, The First Affiliated Hospital of Ningbo University, Zhejiang, China
| | - Guang-Tan Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Zhi-Quan Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Dong Wu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Cai-Ming Weng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Qiu-Xian Chen
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
| | - Ming-Qiao Lian
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
| | - Wei-Ming Zeng
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
| | - Yong-Bin Zhang
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Li-Sheng Cai
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| |
Collapse
|
43
|
Zeng Y, Lockhart AC, Jin RU. The preclinical discovery and development of zolbetuximab for the treatment of gastric cancer. Expert Opin Drug Discov 2024; 19:873-886. [PMID: 38919123 PMCID: PMC11938084 DOI: 10.1080/17460441.2024.2370332] [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: 03/31/2024] [Revised: 06/04/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024]
Abstract
INTRODUCTION Gastric cancer remains a formidable challenge in oncology with high mortality rates and few advancements in treatment. Claudin-18.2 (CLDN18.2) is a tight junction protein primarily expressed in the stomach and is frequently overexpressed in certain subsets of gastric cancers. Targeting CLDN18.2 with monoclonal antibodies, such as zolbetuximab (IMAB362), has shown promising efficacy results in combination with chemotherapy. AREAS COVERED The molecular cell biology of CLDN18.2 is discussed along with studies demonstrating the utility of CLDN18.2 expression as a biomarker and therapeutic target. Important clinical studies are reviewed, including Phase III trials, SPOTLIGHT and GLOW, which demonstrate the efficacy of zolbetuximab in combination with chemotherapy in patients with CLDN18.2-positive advanced gastric cancer. EXPERT OPINION CLDN18.2 is involved in gastric differentiation through maintenance of epithelial barrier function and coordination of signaling pathways, and its expression in gastric cancers reflects a 'gastric differentiation' program. Targeting Claudin-18.2 represents the first gastric cancer specific 'targeted' treatment. Further studies are needed to determine its role within current gastric cancer treatment sequencing, including HER2-targeted therapies and immunotherapies. Management strategies will also be needed to better mitigate zolbetuximab-related treatment side effects, including gastrointestinal (GI) toxicities.
Collapse
Affiliation(s)
- Yongji Zeng
- Section of Gastroenterology, Department of Medicine, Baylor College of Medicine, Houston, USA
| | - A. Craig Lockhart
- Division of Hematology/Oncology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Ramon U. Jin
- Section of Hematology/Oncology, Department of Medicine, Baylor College of Medicine, Houston, USA
| |
Collapse
|
44
|
Nian Z, Deng M, Ye L, Tong X, Xu Y, Xu Y, Chen R, Wang Y, Mao F, Xu C, Lu R, Mao Y, Xu H, Shen X, Xue X, Guo G. RNA epigenetic modifications in digestive tract cancers: Friends or foes. Pharmacol Res 2024; 206:107280. [PMID: 38914382 DOI: 10.1016/j.phrs.2024.107280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 06/26/2024]
Abstract
Digestive tract cancers are among the most common malignancies worldwide and have high incidence and mortality rates. Thus, the discovery of more effective diagnostic and therapeutic targets is urgently required. The development of technologies to accurately detect RNA modification has led to the identification of numerous RNA chemical modifications in humans (epitranscriptomics) that are involved in the occurrence and development of digestive tract cancers. RNA modifications can cooperatively regulate gene expression to facilitate normal physiological functions of the digestive system. However, the dysfunction of relevant RNA-modifying enzymes ("writers," "erasers," and "readers") can lead to the development of digestive tract cancers. Consequently, targeting dysregulated enzyme activity could represent a potent therapeutic strategy for the treatment of digestive tract cancers. In this review, we summarize the most widely studied roles and mechanisms of RNA modifications (m6A, m1A, m5C, m7G, A-to-I editing, pseudouridine [Ψ]) in relation to digestive tract cancers, highlight the crosstalk between RNA modifications, and discuss their roles in the interactions between the digestive system and microbiota during carcinogenesis. The clinical significance of novel therapeutic methods based on RNA-modifying enzymes is also discussed. This review will help guide future research into digestive tract cancers that are resistant to current therapeutics.
Collapse
Affiliation(s)
- Zekai Nian
- Second Clinical College, Wenzhou Medical University, Wenzhou, China
| | - Ming Deng
- School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Lele Ye
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xinya Tong
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yixi Xu
- School of public administration, Hangzhou Normal University, Hangzhou, China
| | - Yiliu Xu
- Research Center of Fluid Machinery Engineering & Technology, Jiangsu University, Zhenjiang, China
| | - Ruoyao Chen
- Second Clinical College, Wenzhou Medical University, Wenzhou, China
| | - Yulin Wang
- School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Feiyang Mao
- Second Clinical College, Wenzhou Medical University, Wenzhou, China
| | - Chenyv Xu
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Ruonan Lu
- First Clinical College, Wenzhou Medical University, Wenzhou, China
| | - Yicheng Mao
- Ophthalmology College, Wenzhou Medical University, Wenzhou, China
| | - Hanlu Xu
- Ophthalmology College, Wenzhou Medical University, Wenzhou, China
| | - Xian Shen
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Xiangyang Xue
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China.
| | - Gangqiang Guo
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
45
|
Lordick F, Van Cutsem E, Shitara K, Xu RH, Ajani JA, Shah MA, Oh M, Ganguli A, Chang L, Rhoten S, Bhattacharya P, Matsangou M, Park JW, Pophale R, Ranganath R, Kang YK. Health-related quality of life in patients with CLDN18.2-positive, locally advanced unresectable or metastatic gastric or gastroesophageal junction adenocarcinoma: results from the SPOTLIGHT and GLOW clinical trials. ESMO Open 2024; 9:103663. [PMID: 39146670 PMCID: PMC11374961 DOI: 10.1016/j.esmoop.2024.103663] [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: 03/19/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 08/17/2024] Open
Abstract
BACKGROUND First-line zolbetuximab plus chemotherapy (SPOTLIGHT, mFOLFOX6; GLOW, CAPOX) significantly improved progression-free survival (PFS) and overall survival (OS) versus placebo plus chemotherapy in patients with human epidermal growth factor receptor 2-negative, locally advanced unresectable or metastatic gastric or gastroesophageal junction adenocarcinoma whose tumors were claudin 18 isoform 2-positive in the phase III SPOTLIGHT (NCT03504397) and GLOW (NCT03653507) studies. We present patient-reported outcomes (PROs) from these studies. MATERIALS AND METHODS Health-related quality of life (HRQoL) was measured in the full analysis sets using the European Organisation for Research and Treatment of Cancer Quality of Life of Cancer Patients Core Questionnaire (QLQ-C30) and Oesophago-Gastric Module (QLQ-OG25), Global Pain, and 5-level EQ-5D (EQ-5D-5L) questionnaires. Analyses focused on key PRO domains: global health status (GHS)/QoL, physical functioning, abdominal pain and discomfort, and nausea/vomiting. Least squares mean (LSM) changes from baseline and time to first definitive deterioration (TTDD) were evaluated combined across SPOTLIGHT and GLOW and for individual studies. Time to confirmed deterioration (TTCD) was evaluated independently for SPOTLIGHT and GLOW. RESULTS The combined analysis set included 1072 patients (zolbetuximab plus chemotherapy, 537; placebo plus chemotherapy, 535). Compliance rates were similar between treatment arms. Similar trends were observed in the zolbetuximab versus placebo arms for LSM changes from baseline in key PRO domains, with no clinically meaningful deterioration. Nausea/vomiting worsened during the first few zolbetuximab cycles but later returned to baseline levels. Overall TTCD and TTDD results were similar between arms in both studies. CONCLUSIONS Patients in SPOTLIGHT and GLOW maintained measured HRQoL relative to baseline when treated with first-line zolbetuximab added to chemotherapy. Zolbetuximab plus chemotherapy improved PFS and OS without negatively affecting HRQoL in key PRO domains compared with placebo plus chemotherapy.
Collapse
Affiliation(s)
- F Lordick
- Department of Medicine and University Cancer Center Leipzig, University of Leipzig Medical Center, Leipzig, Germany
| | - E Van Cutsem
- Digestive Oncology, University Hospitals Gasthuisberg, Leuven, and KULeuven, Leuven, Belgium
| | - K Shitara
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa City, Chiba, Japan
| | - R-H Xu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou; Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - J A Ajani
- The University of Texas, MD Anderson Cancer Center, Houston
| | - M A Shah
- Weill Cornell Medical College, New York City
| | - M Oh
- Astellas Pharma Global Development, Inc., Northbrook. https://twitter.com/AstellasUS
| | - A Ganguli
- Astellas Pharma Global Development, Inc., Northbrook. https://twitter.com/AstellasUS
| | - L Chang
- Astellas Pharma Global Development, Inc., Northbrook. https://twitter.com/AstellasUS
| | | | - P Bhattacharya
- Astellas Pharma Global Development, Inc., Northbrook. https://twitter.com/AstellasUS
| | - M Matsangou
- Astellas Pharma Global Development, Inc., Northbrook. https://twitter.com/AstellasUS
| | - J W Park
- Astellas Pharma Global Development, Inc., Northbrook. https://twitter.com/AstellasUS
| | - R Pophale
- Astellas Pharma Global Development, Inc., Northbrook. https://twitter.com/AstellasUS
| | - R Ranganath
- Astellas Pharma Global Development, Inc., Northbrook. https://twitter.com/AstellasUS
| | - Y-K Kang
- Department of Oncology, Asan Medical Center, University of Ulsan, Seoul, Republic of Korea.
| |
Collapse
|
46
|
Prenen H, Deva S, Keam B, Lindsay CR, Lugowska I, Yang JC, Longo F, de Miguel M, Ponz-Sarvise M, Ahn MJ, Gumus M, Champiat S, Italiano A, Salas S, Perets R, Arslan C, Cho BC, Evers S, Boetsch C, Marbach D, Dejardin D, Sleiman N, Ardeshir C, Richard M, Charo J, Kraxner A, Keshelava N, Teichgräber V, Moreno V. Phase II Study to Determine the Antitumor Activity and Safety of Simlukafusp Alfa (FAP-IL2v) Combined with Atezolizumab in Esophageal Cancer. Clin Cancer Res 2024; 30:2945-2953. [PMID: 38709220 DOI: 10.1158/1078-0432.ccr-23-2677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/29/2024] [Accepted: 05/01/2024] [Indexed: 05/07/2024]
Abstract
PURPOSE In this study, we report the results from the esophageal squamous cell carcinoma (SCC) cohort of a phase II, noncomparative, basket study evaluating the antitumor activity and safety of fibroblast activation protein-IL2 variant (FAP-IL2v) plus atezolizumab in patients with advanced/metastatic solid tumors (NCT03386721). PATIENTS AND METHODS Eligible patients had an Eastern Cooperative Oncology Group performance status of 0 to 1; measurable metastatic, persistent, or recurrent esophageal SCC; progression on ≥1 prior therapy; and were checkpoint inhibitor-naïve. Patients received FAP-IL2v 10 mg plus atezolizumab 1,200 mg intravenously every 3 weeks, or FAP-IL2v weekly for 4 weeks and then every 2 weeks plus atezolizumab 840 mg intravenously every 2 weeks. The primary endpoint was investigator-assessed objective response rate (ORR). RESULTS In the response-evaluable population (N = 34), the best confirmed ORR was 20.6% [95% confidence interval (CI), 10.4-36.8], with a complete response seen in 1 patient and partial responses in 6 patients. The disease control rate was 44.1% (complete response = 2.9%; partial response = 17.6%; stable disease = 23.5%), and the median duration of response was 10.1 mon/ths (95% CI, 5.6-26.7). The median progression-free survival was 1.9 months (95% CI, 1.8-3.7). Analysis of response by PDL1 expression (Ventana SP263) resulted in an ORR of 26.7% for patients with PDL1-positive tumors (tumor area positivity cutoff ≥1%; n = 15) and 7.1% for patients with PDL1-negative tumors (tumor area positivity cutoff <1%; n = 14). Overall, the treatment combination was tolerable, and adverse events were consistent with the known safety profiles of each drug. CONCLUSIONS FAP-IL2v plus atezolizumab demonstrated clinical activity and was tolerable in patients with previously treated esophageal SCC.
Collapse
Affiliation(s)
- Hans Prenen
- Antwerp University Hospital, Edegem, Belgium
| | | | - Bhumsuk Keam
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Colin R Lindsay
- The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Iwona Lugowska
- Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - James C Yang
- National Taiwan University Hospital, National Taiwan University Cancer Center, Taipei City, Taiwan
| | - Federico Longo
- Ramón y Cajal University Hospital, IRYCIS, Universidad Alcala, CIBERONC, Madrid, Spain
| | - Maria de Miguel
- START-Madrid-FJD, Hospital Universitario HM Sanchinarro, Madrid, Spain
| | | | - Myung-Ju Ahn
- Samusung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | | - Stephane Champiat
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Villejuif, France
| | | | | | - Ruth Perets
- Division of Oncology, Clinical Research Institute at Rambam, Rambam Medical Center, Haifa, Israel
| | - Cagatay Arslan
- Izmir Economy University Medical Point Medical Hospital, Izmir, Turkey
| | - Byoung C Cho
- Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Stefan Evers
- Roche Pharma Research and Early Development, Discovery and Translational Area Oncology, Roche Innovation Center, Basel, Switzerland
| | | | - Daniel Marbach
- Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | - David Dejardin
- Biostatistics, Product Development, Roche Innovation Center, Basel, Switzerland
| | - Nassim Sleiman
- Biostatistics, Product Development, Roche Innovation Center, Basel, Switzerland
| | | | - Muriel Richard
- Roche Pharma Research and Early Development, Discovery and Translational Area Oncology, Roche Innovation Center, Basel, Switzerland
| | - Jehad Charo
- Roche Pharma Research and Early Development, Discovery and Translational Area Oncology, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Anton Kraxner
- Roche Pharma Research and Early Development, Discovery and Translational Area Oncology, Roche Innovation Center, Basel, Switzerland
| | - Nino Keshelava
- Roche Pharma Research and Early Development, Discovery and Translational Area Oncology, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Volker Teichgräber
- Roche Pharma Research and Early Development, Discovery and Translational Area Oncology, Roche Innovation Center, Basel, Switzerland
| | - Victor Moreno
- START-Madrid-FJD, Hospital Fundación Jimenez Diaz, Madrid, Spain
| |
Collapse
|
47
|
Sato S, Ssuzuki T, Chinen T, Yamaguchi H, Suzuki Y, Hokamura N, Saze Z, Kono K, Takahashi K, Yano F, Kunisaki C, Kosaka T, Endo I, Ichikawa Y, Miyawaki Y, Sato H, Shimada H. The real-world data of immune-checkpoint inhibitor combination therapy for unresectable or metastatic esophageal cancer: a multi-institutional cohort study. Int J Clin Oncol 2024; 29:994-1001. [PMID: 38679627 DOI: 10.1007/s10147-024-02532-0] [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: 01/10/2024] [Accepted: 04/06/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND The real-world efficacy, feasibility, and prognostic factors of immune-checkpoint inhibitor combination therapy for unresectable or metastatic esophageal cancer are not fully established. METHODS This multi-institutional retrospective cohort study evaluated 71 consecutive patients treated with immune-checkpoint inhibitor combination therapy for esophageal cancer between March 2021 and December 2022. We assessed tumor response, safety, and long-term survival. RESULTS In patients with measurable lesions, the response rate was 58%, and the disease control rate for all enrolled patients was 80%. Five patients (7.0%) underwent successful conversion surgery. Grade 3 or higher immune-related adverse events occurred in 13% of patients, and one patient (1.4%) died due to cholangitis. Median progression-free survival was 9.7 (95% confidence interval: 6.5-not reached). C-reactive protein levels and performance status were identified as significant predictors of progression-free survival through Cox proportional hazards analysis. CONCLUSIONS Immune-checkpoint inhibitor combination therapy for esophageal cancer demonstrated comparable tumor response, safety, and long-term survival to previous randomized clinical trials. Patients with good performance status and low C-reactive protein levels may be suitable candidates for this treatment.
Collapse
Affiliation(s)
- Sho Sato
- Department of Surgery, Yokohama City University Gastroenterological Center, Yokohama, Japan
| | - Takashi Ssuzuki
- Department of Surgery, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-Ku, Tokyo, 143-8541, Japan
| | - Takashi Chinen
- Department of Clinical Oncology, Jichi Medical University Hospital, Tochigi, Japan
| | - Hironori Yamaguchi
- Department of Clinical Oncology, Jichi Medical University Hospital, Tochigi, Japan
| | - Yusuke Suzuki
- Department of Surgery, Teikyo University School of Medicine, Tokyo, Japan
| | - Nobukazu Hokamura
- Department of Surgery, Teikyo University School of Medicine, Tokyo, Japan
| | - Zenichiro Saze
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University, Fukushima, Japan
| | - Koji Kono
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University, Fukushima, Japan
| | - Keita Takahashi
- Department of Surgery, Jikei University School of Medicine, Tokyo, Japan
| | - Fumiaki Yano
- Department of Surgery, Jikei University School of Medicine, Tokyo, Japan
| | - Chikara Kunisaki
- Department of Surgery, Yokohama City University Gastroenterological Center, Yokohama, Japan
| | - Takashi Kosaka
- Department of Gastroenterological Surgery, Yokohama City University School of Medicine, Yokohama, Japan
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University School of Medicine, Yokohama, Japan
| | - Yasushi Ichikawa
- Department of Clinical Oncology, Yokohama City University School of Medicine, Yokohama, Japan
| | - Yutaka Miyawaki
- Department of Gastroenterological Surgery, Saitama Medical University International Medical Center, Saitama, Japan
| | - Hiroshi Sato
- Department of Gastroenterological Surgery, Saitama Medical University International Medical Center, Saitama, Japan
| | - Hideaki Shimada
- Department of Surgery, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-Ku, Tokyo, 143-8541, Japan.
| |
Collapse
|
48
|
Qi F, Zhang J, Li J, Li D, Gao N, Qi Z, Kong X, Yu Z, Fang Y, Cui W, Xia J. Synergistic immunochemotherapy targeted SAMD4B-APOA2-PD-L1 axis potentiates antitumor immunity in hepatocellular carcinoma. Cell Death Dis 2024; 15:421. [PMID: 38886351 PMCID: PMC11183041 DOI: 10.1038/s41419-024-06699-2] [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: 02/26/2024] [Revised: 04/15/2024] [Accepted: 04/18/2024] [Indexed: 06/20/2024]
Abstract
Targeted and immunotherapy combined with interventional therapy can improve the prognosis of advanced cancer patients, and it has become a hot spot to find the new therapeutic schemes, but most of which are not satisfactory. Single-cell RNA sequencing was performed in PDX mouse models with or without TCC therapy. 2-'O-Methylation modification and multiplex immunofluorescence staining were used to explore the function and mechanism of SAMD4B in the immune context of HCC. Here, we propose for the first time a synergistic immunochemotherapy that exerts a potent antitumour effect for patients with advanced hepatocellular carcinoma (HCC) in clinical practice based on three common antitumour drugs and found that HCC patients with new synergistic immunochemotherapy had better three-year overall survival (p = 0.004) and significantly higher survival ratio (increased by 2.3 times) than the control group. We further reveal the immunoregulatory mechanism of synergistic immunochemotherapy through 2'-O-Methylation modification mediated by SAMD4B, a tumour suppressor gene. Mechanistically, SAMD4B, increased by the reduced mutations of upstream genes NOTCH1 and NOTCH2, affected the instability of APOA2 mRNA by 2-'O-Methylation modification of the C-terminus. The decreased APOA2 further attenuated programmed death ligand 1 (PD-L1) level with a direct interaction pattern. The high-SAMD4B tumour tissues contained fewer native CD29+CD8+ T cells, which improved immune microenvironment to achieve the effect of antitumour effect. Overall, we developed a potent synergistic immunochemotherapy strategy that exerts an efficient anti-HCC effect inducing SAMD4B-APOA2-PD-L1 axis to inhibit tumour immune evasion.
Collapse
Affiliation(s)
- Feng Qi
- National Medical Center & National Clinical Research Center for Interventional Medicine. Liver Cancer Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jian Zhang
- National Medical Center & National Clinical Research Center for Interventional Medicine. Liver Cancer Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Jia Li
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20025, China
| | - Donghe Li
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Na Gao
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Zhuoran Qi
- National Medical Center & National Clinical Research Center for Interventional Medicine. Liver Cancer Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Xiuyan Kong
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Zhijie Yu
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Yuan Fang
- Department of Liver Surgery, Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200000, China.
| | - Jinglin Xia
- National Medical Center & National Clinical Research Center for Interventional Medicine. Liver Cancer Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
49
|
Masetti M, Al-Batran SE, Goetze TO, Thuss-Patience P, Knorrenschild JR, Goekkurt E, Folprecht G, Ettrich TJ, Lindig U, Luley KB, Pink D, Dechow T, Sookthai D, Junge S, Loose M, Pauligk C, Lorenzen S. Efficacy of ramucirumab combination chemotherapy as second-line treatment in patients with advanced adenocarcinoma of the stomach or gastroesophageal junction after exposure to checkpoint inhibitors and chemotherapy as first-line therapy. Int J Cancer 2024; 154:2142-2150. [PMID: 38447003 DOI: 10.1002/ijc.34894] [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: 09/28/2023] [Revised: 12/20/2023] [Accepted: 01/12/2024] [Indexed: 03/08/2024]
Abstract
FOLFOX plus nivolumab represents a standard of care for first-line therapy of advanced gastroesophageal cancer (aGEC) with positive PD-L1 expression. The efficacy of second-line VEGFR-2 inhibition with ramucirumab (RAM) plus chemotherapy after progression to immunochemotherapy remains unclear. Medical records of patients with aGEC enrolled in the randomized phase II AIO-STO-0417 trial after treatment failure to first-line FOLFOX plus nivolumab and ipilimumab were retrospectively analyzed. Patients were divided into two groups based on second-line therapy: RAM plus chemotherapy (RAM group) or treatment without RAM (control group). Eighty three patients were included. In the overall population, progression-free survival (PFS) in the RAM group was superior to the control (4.5 vs 2.9 months). Responders (CR/PR) to first-line immunochemotherapy receiving RAM containing second-line therapy had prolonged OS from start of first-line therapy (28.9 vs 16.5 months), as well as second-line OS (9.6 vs 7.5 months), PFS (5.6 vs 2.9 months) and DCR (53% vs 29%) compared to the control. PD-L1 CPS ≥1 was 42% and 44% for the RAM and the control, respectively. Patients with CPS ≥1 in the RAM group showed better tumor control (ORR 25% vs 10%) and improved survival (total OS 11.5 vs 8.0 months; second-line OS 6.5 vs 3.9 months; PFS 4.5 vs 1.6 months) compared to the control. Prior exposure to first-line FOLFOX plus dual checkpoint inhibition followed by RAM plus chemotherapy shows favorable response and survival rates especially in patients with initial response and positive PD-L1 expression and has the potential to advance the treatment paradigm in aGEC.
Collapse
Affiliation(s)
- Michael Masetti
- Klinikum rechts der Isar, Technische Universität München, Klinik für Innere Medizin III, Munich, Germany
| | - Salah-Eddin Al-Batran
- Institut für Klinische Krebsforschung IKF am Krankenhaus Nordwest, Frankfurt, Germany and Krankenhaus Nordwest, University Cancer Center Frankfurt, Frankfurt, Germany
| | - Thorsten O Goetze
- Institut für Klinische Krebsforschung IKF am Krankenhaus Nordwest, Frankfurt, Germany and Krankenhaus Nordwest, University Cancer Center Frankfurt, Frankfurt, Germany
| | - Peter Thuss-Patience
- Medizinische Klinik mit Schwerpunkt Hämatologie, Onkologie und Tumorimmunologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Eray Goekkurt
- Haematologisch-Onkologische Praxis Eppendorf, Universitäres Cancer Center Hamburg (UCCH), Hamburg, Germany
| | - Gunnar Folprecht
- Medizinische Klinik I, Universitätsklinikum Carl Gustav Carus, Dresden, Germany
| | | | - Udo Lindig
- Klinik für Innere Medizin II, Universitätsklinikum Jena, Jena, Germany
| | - Kim Barbara Luley
- UKSH Campus Lübeck, Klinik für Hämatologie und Onkologie, Lübeck, Germany
| | - Daniel Pink
- Klinik und Poliklinik für Innere Medizin C, Hämatologie und Onkologie, Transplantationszentrum, Palliativmedizin, Universität Greifswald and Klinik für Hämatologie, Onkologie und Palliativmedizin-Sarkomzentrum, HELIOS Klinikum Bad Saarow, Bad Saarow, Germany
| | | | - Disorn Sookthai
- Institut für Klinische Krebsforschung IKF am Krankenhaus Nordwest, Frankfurt, Germany
| | - Sabine Junge
- Institut für Klinische Krebsforschung IKF am Krankenhaus Nordwest, Frankfurt, Germany
| | - Maria Loose
- Institut für Klinische Krebsforschung IKF am Krankenhaus Nordwest, Frankfurt, Germany
| | - Claudia Pauligk
- Institut für Klinische Krebsforschung IKF am Krankenhaus Nordwest, Frankfurt, Germany
| | - Sylvie Lorenzen
- Klinikum rechts der Isar, Technische Universität München, Klinik für Innere Medizin III, Munich, Germany
| |
Collapse
|
50
|
Shah NJ, Della Pia A, Wu T, Williams A, Weber M, Sinclaire B, Gourna Paleoudis E, Alaoui A, Lev-Ari S, Adams S, Kaufman J, Parikh SB, Tonti E, Muller E, Serzan M, Cheruku D, Lee A, Sridhar A, Hee B(TP, Ahn J, Pecora A, Ip A, Atkins MB. Clinical Outcomes of Immune Checkpoint Inhibitors in Unique Cohorts Underrepresented in Clinical Trials. Cancers (Basel) 2024; 16:2223. [PMID: 38927928 PMCID: PMC11202168 DOI: 10.3390/cancers16122223] [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: 04/08/2024] [Revised: 05/29/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Regulatory approval of immune checkpoint inhibitors (ICIs) was based on results of large, randomized clinical trials, resulting in limited outcomes data in patient cohorts typically underrepresented in such trials. The objective of this study was to evaluate the efficacy and safety of ICIs in these unique patient cohorts. This is a multicenter, retrospective analysis of real-world data at six academic and community clinics in the United States from 1 January 2011 to 1 April 2018. Patients were included if they had received at least one cycle of ICI treatment. Unique patient cohorts included age > 75 years, non-White race, positive smoking history, ECOG performance status (PS) ≥ 2, BMI ≥ 30 kg/m2, autoimmune diseases (AIDs), chronic viral infections (CVI), extensive prior lines of therapy (LOTs), or >three metastatic sites. Immune-related adverse events (irAEs), overall survival (OS), and time to treatment failure were evaluated in the entire cohort and in NSCLC patients treated with PD-(L)1 monotherapy. Outcomes and their association with unique patient cohorts were compared on univariate analysis and multivariate analysis to those without a particular characteristic in the entire NSCLC PD-(L)1 monotherapy cohorts. In total, 1453 patients were included: 56.5%-smokers, 30.4%-non-White, 22.8%-elderly, 20.8%-ECOG PS ≥ 2, 15.7%-history of AIDs, and 4.7%-history of CVI. The common ICIs were nivolumab (37.1%) and pembrolizumab (22.2%). Black patients, compared to White patients, experienced fewer irAEs (OR 0.54, p < 0.001). An ECOG PS of ≥2 (HR = 2.01, p < 0.001) and an increased number of previous LOTs were associated with poor OS (the median OS of 26.2 vs. 16.2 vs. 9.6 months for one vs. two vs. three prior LOTs, p < 0.001). The above results were confirmed in anti-PD-(L)1 monotherapy non-small cell lung cancer patients (n = 384). Overall, ICIs were safe and efficacious in these typically underrepresented patient cohorts. We noted ECOG PS ≥ 2 and an increased prior LOTs were associated with poor ICI efficacy, and Black patients, compared to White patients, experienced fewer irAEs.
Collapse
Affiliation(s)
- Neil J. Shah
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Medicine, Weill Cornell Medical Center, New York, NY 10065, USA
| | - Alexandra Della Pia
- John Theurer Cancer Center at Hackensack Meridian Health, Hackensack, NJ 07601, USA
| | - Tianmin Wu
- Department of Biostatistics, Georgetown University Medical Center, Washington, DC 20007, USA
| | - Aquino Williams
- Hackensack Meridian Health Mountainside Medical Center, Montclair, NJ 07042, USA
| | - Melinda Weber
- John Theurer Cancer Center at Hackensack Meridian Health, Hackensack, NJ 07601, USA
| | - Brittany Sinclaire
- John Theurer Cancer Center at Hackensack Meridian Health, Hackensack, NJ 07601, USA
| | | | - Adil Alaoui
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Washington, DC 20007, USA
| | - Shaked Lev-Ari
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Washington, DC 20007, USA
| | - Shari Adams
- John Theurer Cancer Center at Hackensack Meridian Health, Hackensack, NJ 07601, USA
| | - Jordan Kaufman
- John Theurer Cancer Center at Hackensack Meridian Health, Hackensack, NJ 07601, USA
| | - Sahil B. Parikh
- John Theurer Cancer Center at Hackensack Meridian Health, Hackensack, NJ 07601, USA
| | - Emily Tonti
- John Theurer Cancer Center at Hackensack Meridian Health, Hackensack, NJ 07601, USA
| | - Eric Muller
- John Theurer Cancer Center at Hackensack Meridian Health, Hackensack, NJ 07601, USA
| | - Michael Serzan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Divya Cheruku
- Hackensack Meridian Health Mountainside Medical Center, Montclair, NJ 07042, USA
| | - Albert Lee
- Hackensack Meridian School of Medicine, Nutley, NJ 07110, USA
| | | | | | - Jaeil Ahn
- Department of Biostatistics, Georgetown University Medical Center, Washington, DC 20007, USA
| | - Andrew Pecora
- John Theurer Cancer Center at Hackensack Meridian Health, Hackensack, NJ 07601, USA
- Hackensack Meridian School of Medicine, Nutley, NJ 07110, USA
| | - Andrew Ip
- John Theurer Cancer Center at Hackensack Meridian Health, Hackensack, NJ 07601, USA
- Hackensack Meridian School of Medicine, Nutley, NJ 07110, USA
| | - Michael B. Atkins
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Washington, DC 20007, USA
| |
Collapse
|