1
|
Akinbolade S, Fairbairn R, Inskip A, Potter R, Oliver A, Craig D. Repurposed Medicines: A Scan of the Non-commercial Clinical Research Landscape. Pharmacol Res Perspect 2025; 13:e70049. [PMID: 39686549 DOI: 10.1002/prp2.70049] [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: 06/20/2024] [Revised: 10/10/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Medicine repurposing is a strategy to identify new uses for the existing medicines for the purpose of addressing areas of unmet medical need. This paper aims to provide horizon scanning intelligence on repurposed medicines that are evaluated by non-commercial organizations such as academia and highlights opportunities for further research to improve patient health outcomes. A scan of the clinical landscape of non-commercially sponsored repurposed medicines is routinely conducted by the NIHR Innovation Observatory (IO). This ongoing project involves a horizon scan of clinical trial registries and the IO's internal horizon scanning Medicines Innovation Database to identify potential candidate medicines used as monotherapy or in combination to treat new indications outside the scope of their licensed indication. In addition to making these data publicly available, the output also supports the NHS England Medicines Repurposing Programme. The snapshot scan reported here (trials completing April 2020-March 2023) identified a total of 528 technologies (meaning, a single product or combination of medicinal products targeting a specific indication in one or more related trials). The technologies were classified according to their characteristics and targeted therapeutic indications as well as revealing the least treated disease conditions. The candidate medicines identified in this scan could potentially receive tailored support toward adoption into practice and policy. The NIHR IO regularly provides this scan as a source of intelligence on repurposed medicines. This provides valuable insights into innovation trends, gaps, and areas of unmet clinical need.
Collapse
Affiliation(s)
- Sola Akinbolade
- National Institute for Health and Care Research (NIHR) Innovation Observatory, Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Ross Fairbairn
- National Institute for Health and Care Research (NIHR) Innovation Observatory, Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Alex Inskip
- National Institute for Health and Care Research (NIHR) Innovation Observatory, Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Rhiannon Potter
- National Institute for Health and Care Research (NIHR) Innovation Observatory, Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Aoife Oliver
- National Institute for Health and Care Research (NIHR) Innovation Observatory, Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Dawn Craig
- National Institute for Health and Care Research (NIHR) Innovation Observatory, Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| |
Collapse
|
2
|
Wang B, Liu W, Song B, Li Y, Wang Y, Tan B. Targeting LINC00665/miR-199b-5p/SERPINE1 axis to inhibit trastuzumab resistance and tumorigenesis of gastric cancer via PI3K/AKt pathway. Noncoding RNA Res 2025; 10:153-162. [PMID: 39399377 PMCID: PMC11467570 DOI: 10.1016/j.ncrna.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 10/15/2024] Open
Abstract
Long noncoding RNAs (lncRNAs) serve as critical mediators of tumor progression and drug resistance in cancer. Herein, we identified a lncRNA, LINC00665, associated with trastuzumab resistance and development in gastric cancer (GC). LINC00665 was highly expressed in GC tissues and high expression of LINC00665 was correlated with poor prognosis. LINC00665 knockdown was verified to suppress migration, invasion, and resistance to trastuzumab in GC. Furthermore, we found that LINC00665 participates in the infiltration of naive B cells, mast cells, and T follicular helper (Tfh) cells. Mechanistically, LINC00665 was confirmed to regulate tumorigenesis and trastuzumab resistance by activating PI3K/AKt pathway. LINC00665 sponged miR-199b-5p to interact with SERPINE1 expression, resulting in the increase of phosphorylation of AKt, thus participating in the PI3K/AKt pathway. To summarize, LINC00665 facilitated the tumorigenesis and trastuzumab resistance of GC by sponging miR-199b-5p and promoting SERPINE1 expression, which further activated PI3K/AKt signaling; this finding reveals a new mechanism by which LINC00665 modulates tumor development and drug resistance in GC.
Collapse
Affiliation(s)
- Bingyu Wang
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, 050017, Shijiazhuang, China
| | - Wenbo Liu
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, 050017, Shijiazhuang, China
| | - Buyun Song
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, 050017, Shijiazhuang, China
| | - Yong Li
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, 050017, Shijiazhuang, China
| | - Yingying Wang
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, 050017, Shijiazhuang, China
| | - Bibo Tan
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, 050017, Shijiazhuang, China
| |
Collapse
|
3
|
YektaKooshali MH, SobhZahedi M, Razavi Tousi SMT, Hamidi M, Modiri L. Astaxanthin-loaded PLGA nanoparticles inhibit survival of MKN-45 gastric cancer cell line by modulating JAK2/STAT3/mTOR/PI3K pathway. BMC Cancer 2025; 25:44. [PMID: 39780129 PMCID: PMC11715247 DOI: 10.1186/s12885-024-13401-4] [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: 06/09/2024] [Accepted: 12/26/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/AIMS Gastric cancer (GC) is a significant global health issue with high incidence rates and poor prognoses, ranking among the top prevalent cancers worldwide. Due to undesirable side effects and drug resistance, there is a pressing need for the development of novel therapeutic strategies. Understanding the interconnectedness of the JAK2/STAT3/mTOR/PI3K pathway in tumorigenesis and the role of Astaxanthin (ASX), a red ketocarotenoid member of xanthophylls and potent antioxidant and anti-tumor activity, can be effective for cancer treatments. This study aimed to investigate the effect of ASX-loaded nanoparticles on the survival of MKN-45 GC cells and the expression of JAK2/STAT3/mTOR/PI3K, offering insights into potential targeted therapies for GC. METHODS The growth status and survival rate of MKN-45 GC cell lines were determined using the 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide(MTT) assay, and the optimal IC50 concentration of ASX, PLGA, and ASX + PLGA was estimated. Also, the clonogenic assay was performed to determine the reproductive power and colony formation of under-treatment cells. Apoptosis and necroptosis of cells were evaluated using acridine orange (AO) staining. The western blot assessed the protein's level of expression and intensity (JAK2/STAT3/mTOR/PI3K). SPSS version 16 software was used for statistical analysis, P-value was considered lower than 0.05. RESULTS Based on the results, increasing concentrations of ASX and ASX + PLGA led to a decrease in the viability of MKN-45 cells compared to the control group (P < 0.001). This value was lower for cells treated with ASX + PLGA (P = 0.003). The IC50 values for each of the studied groups (ASX, ASX + PLGA, and PLGA) were 81.45 µg/ml, 51.45 µg/ml, and 3.383 mg/ml, respectively. The levels of expression and intensity of JAK2, STAT3, and mTOR proteins in the Western blotting analysis under ASX + PLGA treatment increased compared to the control group. Conversely, the levels of expression and intensity of P-JAK2, P-STAT3, and P-mTOR proteins in the ASX + PLGA treatment group decreased by 41%, 34%, 37%, and 43%, respectively, compared to the control group. Protein expression levels and intensities of JAK2, STAT3, and mTOR significantly increased when treated with PLGA, ASX, and ASX + PLGA compared to the control group (P < 0.001). CONCLUSIONS The encapsulation of ASX in PLGA nanoparticles enhances drug stability, enables targeted delivery, and allows for sustained release. This study highlights the therapeutic potential of ASX-loaded nanoparticles in targeting JAK2/STAT3/mTOR/PI3K pathways in GC treatment. Further research is needed to understand the mechanisms and clinical applications of this novel immunotherapy strategy.
Collapse
Affiliation(s)
- Mohammad Hossein YektaKooshali
- Medical Biotechnology Research Center, School of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of Cellular and Molecular Biology, Lahijan Branch, Islamic Azad University, Lahijan, Iran
| | - Mahdieh SobhZahedi
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran.
| | | | - Masoud Hamidi
- Université libre de Bruxelles (ULB), École polytechnique de Bruxelles - 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, Brussels, 1050, Belgium
| | - Leila Modiri
- Department of Cellular and Molecular Biology, Lahijan Branch, Islamic Azad University, Lahijan, Iran.
| |
Collapse
|
4
|
Guo M, Zhao W, Chen Y, Zou D, Peng W, Sha H, Zhou G, Fang Y, Shen B. Efficacy of rechallenge after first-line immunotherapy for advanced gastric cancer: A retrospective real-world study. Hum Vaccin Immunother 2024; 20:2423479. [PMID: 39494935 PMCID: PMC11540071 DOI: 10.1080/21645515.2024.2423479] [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: 07/19/2024] [Revised: 10/15/2024] [Accepted: 10/28/2024] [Indexed: 11/05/2024] Open
Abstract
We aimed to explore the efficacy of rechallenge after first-line immunotherapy in advanced gastric cancer (AGC) and to analyze the factors affecting prognosis based on clinical characteristics. Eighty-five AGC patients who underwent rechallenged after the failure of first-line treatment with immune checkpoint inhibitors (ICIs) were retrospectively collected from July 2019 to December 2022 in Jiangsu Cancer Hospital. Potential factors affecting prognosis were analyzed by univariate and multivariate Cox analysis. Survival analysis was performed by Kaplan-Meier method and Log rank test. Stratified factors included human epidermal growth factor receptor 2 (HER-2) and programmed cell death-ligand 1 combined positive score (PD-L1 CPS). The objective response rate (ORR) was 15.3%, and the disease control rate (DCR) was 74.1%. The median progression-free survival (PFS) was 4.8 months. Results showed that patients in the I + C group had the best response. The ORR was 20.0% VS 8.7% in the I + C group and I + C + AAD group. The DCR was 78.0% VS 65.2%, and the median PFS was 6.7 VS 4.7 months [hazard ratio (HR): 0.55, 95% confidence interval (CI): 0.30-1.00, p = .022]. The ORR was 20.0% VS 8.3% in the I + C group and I + C + ADC group. The DCR was 78.0% VS 75.0%, and the median PFS was 6.7 VS 4.4 months (HR: 0.59, 95%CI: 0.26-1.30, p = .112). The median PFS was 4.7 VS 4.4 months in the I + C + AAD group and I + C + ADC group (HR: 1.21, 95%CI: 0.60-2.47, p = .580). Adverse events (AEs) were found in 34 patients, mainly including leukopenia 9 (10.6%), and neutropenia 8 (9.4%). The incidence of grade 3-4 AEs was 8.2%. There were no drug-related deaths and all AEs were manageable. Rechallenge after first-line immunotherapy showed good survival benefit and acceptable safety in the therapy of AGC. Especially for patients with HER-2-positive and PD-L1 CPS ≥ 1%, rechallenge may be an effective treatment modality.
Collapse
Affiliation(s)
- Mengya Guo
- Department of Oncology, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wenhui Zhao
- Department of Oncology, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yue Chen
- Department of Oncology, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dan Zou
- Department of Oncology, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weiwei Peng
- Department of Oncology, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Huanhuan Sha
- Department of Oncology, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Guoren Zhou
- Department of Oncology, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | | | - Bo Shen
- Department of Oncology, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
5
|
Pan Y, Ma Y, Guan H, Dai G. Pre-treatment of hyponatremia as a biomarker for poor immune prognosis in advanced or metastatic gastric cancer: A retrospective case analysis. Hum Vaccin Immunother 2024; 20:2414546. [PMID: 39411929 PMCID: PMC11486141 DOI: 10.1080/21645515.2024.2414546] [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: 05/17/2024] [Revised: 09/22/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
Hyponatremia, a prevalent electrolyte imbalance among tumor patients, has often been overlooked regarding its prognostic significance for immunotherapy. In this study, we delved into the prognostic ramifications of hyponatremia in advanced gastric cancer (AGC) patients undergoing immunotherapy. Enrolling AGC patients diagnosed between December 2014 and May 2021, we extracted pertinent data from electronic medical records, with a median follow-up of 35.8 months. Kaplan-Meier curves illuminated patients' progression-free survival (PFS) and overall survival (OS), while survival disparities were tested using the Mantel-Haenszel log rank test. COX and logistic regressions were employed to scrutinize the correlation between serum sodium levels and prognosis in 268 AGC patients, both at baseline and during treatment. Notably, patients with hyponatremia exhibited shorter PFS (4.7 vs 2.1 months, p = .001*) and OS (12.5 vs 3.9 months, p < .001*). Serum sodium emerged as an independent prognostic factor for both PFS (HR = 1.773; 95% CI 1.067-2.945; p = .001*) and OS (HR = 1.773; 95% CI 1.067-2.945; p = .003*). Subgroup analysis revealed that AGC patients with hyponatremia derived no benefit from immunotherapy in terms of PFS and OS. Strikingly, a decrease in serum sodium during immunotherapy was associated with early relapse and mortality. Based on these findings, we hypothesize that hyponatremia portends poor prognostic outcomes in AGC patients treated with immunotherapy and may serve as a valuable prognostic biomarker. However, further large-scale prospective studies are warranted to validate these observations.
Collapse
Affiliation(s)
- Yuting Pan
- Department of Medical Oncology, Medical School of Chinese PLA, Beijing, China
- Department of Medical Oncology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yue Ma
- Department of Medical Oncology, Medical School of Chinese PLA, Beijing, China
- Department of Medical Oncology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Huafang Guan
- External Relations Office, Yingtan City People’s Hospital, Yingtan, China
| | - Guanghai Dai
- Department of Medical Oncology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
6
|
Sheng LP, Huang YL, Wang Z, Zhang HF, Zhang JY, Lei XY. A pathological complete response to capecitabine plus oxaliplatin regimen combined with tislelizumab in advanced gastric cancer with liver metastases: A case report. Hum Vaccin Immunother 2024; 20:2406061. [PMID: 39400029 PMCID: PMC11485698 DOI: 10.1080/21645515.2024.2406061] [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: 08/28/2024] [Accepted: 09/14/2024] [Indexed: 10/15/2024] Open
Abstract
A 66-year-old female patient presenting with dysphagia was diagnosed with stage IV unresectable gastric cancer (cTxN+M1). Multiple liver metastases were identified. The patient subsequently underwent five courses of chemotherapy and immunotherapy, including the capecitabine plus oxaliplatin (XELOX) regimen combined with tislelizumab. After fifth course treatment, it was confirmed that the liver metastases had completely disappeared and the primary tumor had significantly reduced in size. Consequently, a laparoscopy was performed, revealing a retraction-like response in the primary tumor and no obvious metastases in the abdominal cavity. Subsequently, a radical total gastrectomy was carried out through open abdominal surgery. Pathological analysis showed no remaining cancer or lymph node metastases, and the tumor regression was classified as grade 0. The patient has been now receiving additional chemotherapy and immunotherapy to manage any potential residual metastases. This case illustrated the rare and significant impact of combining chemotherapy with tislelizumab, transitioning the treatment approach from palliative to curative. It highlighted the critical role of immunotherapy in managing advanced gastric cancer with liver metastases.
Collapse
Affiliation(s)
- Li-Ping Sheng
- Department of Gastroenterology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yun-Lin Huang
- Department of Gastroenterology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Zhi Wang
- Department of Anesthesiology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Hai-Fang Zhang
- Department of Pathology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jin-Yan Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xiao-Yi Lei
- Department of Gastroenterology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
7
|
Wei J, Xiang W, Wei H, Hu X, Lu Y, Dong X. Impact of nutrition risk index, prognostic nutritional index and skeletal muscle index on early myelosuppression of first-line chemotherapy in stage IV gastric cancer patients. BMC Gastroenterol 2024; 24:452. [PMID: 39695992 DOI: 10.1186/s12876-024-03548-6] [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: 04/07/2024] [Accepted: 12/02/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND In gastric cancer (GC) patients, malnutrition is common and has a negative impact on treatment tolerance, survival, and prognosis. The purpose of this study was to explore the relationship between prechemotherapy nutritional state and early myelosuppression in stage IV GC patients treated with first-line chemotherapy. METHODS This retrospective study included patients with stage IV GC who received first-line chemotherapy between July 2012 and December 2021. Clinical and laboratory data were collected within 1 week before chemotherapy to calculate nutrition risk index, prognostic nutritional index. Pretreatment abdominal computed tomography scans were used to quantify skeletal muscle index (SMI). The main measurable outcome was the incidence of grade ≥ 2 early myelosuppression after chemotherapy. RESULTS Among 102 patients eligible for analysis, 50% were malnourished, 50% were poor prognoses and 49% were sarcopenic at baseline.The side effects were generally well managed, with a 26.5% occurrence of grade 3/4 side effects. Pre-chemotherapy patients with low Nutrition Risk Index (NRI) (p = 0.002), low prognostic nutritional index (PNI) (p = 0.001), and low SMI (p = 0.001) had significantly higher incidences of grade ≥ 2 myelosuppression occurred after the first cycle of chemotherapy. Moreover, the high level of PNI was associated with higher completion rate of chemotherapy (p = 0.01). Multivariate logistic regression analysis revealed that SMI at baseline (p = 0.006) and hemoglobin level (p = < 0.001) were prognostic factors for grade ≥ 2 early myelosuppression. CONCLUSION Stage IV GC patients with low NRI, low PNI and low SMI experienced significantly more grade ≥ 2 early myelosuppression during the first line of chemotherapy.
Collapse
Affiliation(s)
- Juan Wei
- Department of Medical Oncology, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - WeiFeng Xiang
- Department of Medical Oncology, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - HangPing Wei
- Department of Medical Oncology, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - XiaoYan Hu
- Department of Medical Oncology, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - YiFang Lu
- Department of Medical Oncology, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - XiaoFang Dong
- Department of Medical Oncology, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China.
| |
Collapse
|
8
|
Norén N, Rouvelas I, Lundell L, Nilsson M, Sunde B, Szabo E, Edholm D, Hedberg J, Smedh U, Hermansson M, Lindblad M, Klevebro F. Curative treatment for oligometastatic gastroesophageal cancer- results of a prospective multicenter study. Langenbecks Arch Surg 2024; 410:10. [PMID: 39680192 DOI: 10.1007/s00423-024-03575-7] [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: 08/22/2024] [Accepted: 12/09/2024] [Indexed: 12/17/2024]
Abstract
PURPOSE Oligometastatic gastroesophageal cancer is a clinical entity with no standard treatment recommendation. Treatment with curative intent has recently emerged as an option for selected patients in contrast to the traditional palliative treatment strategy. This prospective study aimed to assess the safety and efficacy of combined systemic and local treatment with curative intent for patients with oligometastatic gastroesophageal cancer. METHODS In a multicenter study, consecutive patients with gastroesophageal cancer and metastases in the liver and/or extra-regional lymph nodes were screened for inclusion. Eligible patients were offered curatively intended perioperative chemotherapy followed by surgical resection or liver ablation. Primary endpoints were treatment safety and feasibility. Secondary outcomes included postoperative mortality, treatment response, progression-free survival, and overall survival. Subgroup analyses were stratified based on oligometastatic location. RESULTS A total of 29 (82.9%) patients completed treatment with surgical resection (93.1%), liver ablation (3.4%), or definitive chemoradiotherapy (3.4%). Postoperative complications were found in 19 (73.1%) patients, whereas postoperative mortality was 0%. The most common complications included infection (34.6%) and respiratory complications (34.6%). Median overall survival was 20.9 months (interquartile range 11.2-42.6) from diagnosis and 17.0 months (interquartile range 6.4-35.9) from surgery in patients who were treated with neoadjuvant chemotherapy followed by surgery. Median progression-free survival was 5.8 months (interquartile range 3.1-11.3). CONCLUSION This study found curative treatment to be a relatively safe option, with an overall survival of 20.8 months and no postoperative mortality.
Collapse
Affiliation(s)
- N Norén
- Department of Surgery and Oncology, CLINTEC, Karolinska Institutet, Dep. of Upper Gastrointestinal Diseases, Karolinska University Hospital, Stockholm, Sweden.
| | - I Rouvelas
- Department of Surgery and Oncology, CLINTEC, Karolinska Institutet, Dep. of Upper Gastrointestinal Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - L Lundell
- Department of Surgery and Oncology, CLINTEC, Karolinska Institutet, Dep. of Upper Gastrointestinal Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Surgery, Odense University Hospital, Odense, Denmark
| | - M Nilsson
- Department of Surgery and Oncology, CLINTEC, Karolinska Institutet, Dep. of Upper Gastrointestinal Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - B Sunde
- Department of Surgery and Oncology, CLINTEC, Karolinska Institutet, Dep. of Upper Gastrointestinal Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - E Szabo
- Örebro University Hospital, Örebro, Sweden
| | - D Edholm
- Department of Surgery, Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - J Hedberg
- Department of Surgical Sciences, Uppsala University, Uppsala University Hospital, Uppsala, Sweden
| | - U Smedh
- Sahlgrenska University Hospital, Gothenburg, Sweden
| | - M Hermansson
- Department of Surgery, Skåne University Hospital and Department of Clinical Sciences, Lund University, Lund, Sweden
| | - M Lindblad
- Department of Surgery and Oncology, CLINTEC, Karolinska Institutet, Dep. of Upper Gastrointestinal Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - F Klevebro
- Department of Surgery and Oncology, CLINTEC, Karolinska Institutet, Dep. of Upper Gastrointestinal Diseases, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
9
|
Noronha V, Patil VM, Menon N, Goud S, Singh A, Shah M, More S, Shah S, Yadav A, Sonawane S, Nawale K, Chowdhury OR, Kaushal RK, Ghosh-Laskar S, Agarwal JP, Yadav S, Pai T, Janu A, Mahajan A, Purandare N, Banavali S, Badwe R, Prabhash K. Phase III randomized trial comparing palliative systemic therapy to best supportive care in advanced esophageal/GEJ cancer. Int J Cancer 2024; 155:2232-2245. [PMID: 39093841 DOI: 10.1002/ijc.35111] [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: 01/16/2024] [Revised: 05/20/2024] [Accepted: 06/20/2024] [Indexed: 08/04/2024]
Abstract
No study has unequivocally proven that chemotherapy prolongs overall survival (OS) in advanced esophageal cancer. We conducted a Phase III randomized study in first-line advanced unresectable/metastatic esophageal/GEJ cancer. Patients aged 18-70 years, with performance status 0-2, were randomized to best supportive care (BSC) alone, or BSC with weekly paclitaxel 80 mg/m2. BSC comprised, as indicated, education, counselling, radiation, stenting, feeding tube placement, nutritional supplementation, medications like analgesics, and referral to a support group and palliative care. The primary endpoint was OS; secondary endpoints included progression free survival (PFS), response, toxicity, and QoL. Between May 2016-December 2020, we recruited 281 patients: 143 to chemotherapy and 138 to BSC. Histopathology was squamous in 269 (95.7%) patients. Median number of paclitaxel doses was 12 (IQR, 7-23). Median OS was 4.2 months (95% CI, 3.42-5.32) in BSC, and 9.2 months (95% CI, 8.02-10.48) in chemotherapy; HR, 0.49 (95% CI, 0.39-0.64); p < .001. As compared to BSC, chemotherapy increased response (2.9% to 39%), median PFS (2.1 to 4.2 months), 1-year OS (11% to 32%), 2-year OS (0 to 9%), median dysphagia-free survival (2.9 to 14.8 months), and global and esophagus-specific QoL, without significantly increasing all-grade or grade ≥3 toxicities. Using ESMO clinical benefit scale and ASCO Value Framework, palliative chemotherapy scored as having "substantial value." Our study provides the first level 1 evidence that chemotherapy prolongs survival in advanced esophageal/GEJ carcinoma. BSC alone is no longer appropriate. Weekly paclitaxel is an attractive option, especially in LMICs with limited access to immunotherapy.
Collapse
Affiliation(s)
- Vanita Noronha
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Vijay Maruti Patil
- Department of Medical Oncology, P D Hinduja Hospital & Medical Research Centre, Mumbai, India
| | - Nandini Menon
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Supriya Goud
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Ajaykumar Singh
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Minit Shah
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Sucheta More
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Srushti Shah
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Akanksha Yadav
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Sonali Sonawane
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Kavita Nawale
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Oindrila Roy Chowdhury
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Rajiv Kumar Kaushal
- Department of Pathology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Sarbani Ghosh-Laskar
- Department of Radiation Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Jai Prakash Agarwal
- Department of Radiation Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Subhash Yadav
- Department of Pathology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Trupti Pai
- Department of Pathology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Amit Janu
- Department of Radiodiagnosis, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Abhishek Mahajan
- The Clatterbridge Cancer Centre NHS Foundation Trust, University of Liverpool, Liverpool, UK
| | - Nilendu Purandare
- Department of Nuclear Medicine, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Shripad Banavali
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Rajendra Badwe
- Department of Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Kumar Prabhash
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
10
|
Qin X, Qiu B, Ge L, Wu S, Ma Y, Li W. Applying machine learning techniques to predict the risk of distant metastasis from gastric cancer: a real world retrospective study. Front Oncol 2024; 14:1455914. [PMID: 39703842 PMCID: PMC11655338 DOI: 10.3389/fonc.2024.1455914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 11/15/2024] [Indexed: 12/21/2024] Open
Abstract
Background Distant metastasis of gastric cancer can seriously affect the treatment strategy of gastric cancer patients, so it is essential to identify patients at high risk of distant metastasis of gastric cancer earlier. Method In this study, we retrospectively collected research data from 18,472 gastric cancer patients from the SEER database. We applied six machine learning algorithms to construct a model that can predict distant metastasis of gastric cancer. We constructed the machine learning model using 10-fold cross-validation. We evaluated the model using the area under the receiver operating characteristic curves (AUC), the area under the precision-recall curve (AUPRC), decision curve analysis, and calibration curves. In addition, we used Shapley's addition interpretation (SHAP) to interpret the machine learning model. We used data from 1595 gastric cancer patients in the First Hospital of Jilin University for external validation. We plotted the correlation heat maps of the predictor variables. We selected an optimal model and constructed a web-based online calculator for predicting the risk of distant metastasis of gastric cancer. Result The study included 18,472 patients with gastric cancer from the SEER database, including 4,202 (22.75%) patients with distant metastases. The results of multivariate logistic regression analysis showed that age, race, grade of differentiation, tumor size, T stage, radiotherapy, and chemotherapy were independent risk factors for distant metastasis of gastric cancer. In the ten-fold cross-validation of the training set, the average AUC value of the random forest (RF) model was 0.80. The RF model performed best in the internal test set and external validation set. The RF model had an AUC of 0.80, an AUPRC of 0.555, an accuracy of 0.81, and a precision of 0.78 in the internal test set. The RF model had a metric AUC of 0.76 in the external validation set, an AUPRC of 0.496, an accuracy of 0.82, and a precision of 0.81. Finally, we constructed a network calculator for distant metastasis of gastric cancer using the RF model. Conclusion With the help of pathological and clinical indicators, we constructed a well-performing RF model for predicting the risk of distant metastasis in gastric cancer patients to help clinicians make clinical decisions.
Collapse
Affiliation(s)
- Xinxin Qin
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Binxu Qiu
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Litao Ge
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Song Wu
- Nanjing Luhe People’s Hostipal, General Surgery, Nanjing, China
| | - Yuye Ma
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Wei Li
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
11
|
Wang Y, Wang S, Ma C, Qi W, Lv J, Zhang M, Wang S, Wang R, Lu Y, Qiu W. Nrf2 depletion enhanced curcumin therapy effect in gastric cancer by inducing the excessive accumulation of ROS. Sci Rep 2024; 14:30165. [PMID: 39627516 PMCID: PMC11615379 DOI: 10.1038/s41598-024-81375-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 11/26/2024] [Indexed: 12/06/2024] Open
Abstract
Gastric cancer (GC) is the most common malignant tumor of the gastrointestinal tract and currently has a poor clinical outcome. Turmeric's rhizome contains a polyphenolic component called curcumin (Cur), which has been demonstrated to inhibit a variety of tumor cells, such as pancreatic, colon, lung and gastric cancers. However, it remains to be elucidated how Cur functions in GC and what molecular processes underlie it. Here, Cur showed a stronger inhibitory effect on GC cells AGS and HGC27. In addition, Cur's inhibition of GC cells growth was accompanied by increased ROS production, triggering of the Keap1-Nrf2 signaling pathway, and increased transcription of its downstream antioxidant genes HO-1, GCLM, and NQO1. However, when a ROS scavenger NAC was used, the inhibitory effect of Cur on GC cells was reversed. Nuclear factor erythroid 2-related factor 2 (Nrf2) is overexpressed or activated in cancers to shield cancer cells from oxidative damage by responding to oxidative stress (OS). Cur has been found to act as an activator of Nrf2. Notably, compared with Nrf2 knockdown and Cur alone, the combination of the two dramatically increased Cur-induced ROS overaccumulation and inhibition of GC cells proliferation, migration, and invasive abilities. Consistent with in vitro experiments, Cur combined with Nrf2 knockdown significantly inhibited tumor growth in nude mice transplanted with AGS cells. Therefore, we concluded that Nrf2 depletion enhanced Cur therapy effect in GC by inducing the excessive accumulation of ROS, indicating that this is a promising treatment strategy.
Collapse
Affiliation(s)
- Yan Wang
- Department of oncology, Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao University, Qingdao, China
| | - Shasha Wang
- Department of oncology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chenchen Ma
- Spine Surgery, Qingdao Municipal Hospital, Qingdao, China
| | - Weiwei Qi
- Department of oncology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jing Lv
- Department of oncology, Affiliated Hospital of Qingdao University, Qingdao, China
| | | | | | - Rui Wang
- Qingdao University, Qingdao, China
| | | | - Wensheng Qiu
- Department of oncology, Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
12
|
Wang X, Zhang Y. Multi-omics joint screening of biomarkers related to M2 macrophages in gastric cancer. Discov Oncol 2024; 15:738. [PMID: 39623254 PMCID: PMC11612128 DOI: 10.1007/s12672-024-01623-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 11/25/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Due to high mortality rate and limited treatments in gastric cancer (GC), call for deeper exploration of M2 macrophages as biomarkers is needed. METHODS The data for this study were obtained from the Gene Expression Omnibus (GEO) and Genomic Data Commons (GDC). The Seurat package was utilized for single-cell RNA sequencing (scRNA-seq) analysis. FindAllMarkers was used to identify genes highly expressed among different cell subsets. DESeq2 package was leveraged to screen differentially expressed genes (DEGs), while limma package was utilized for identifying differentially expressed proteins (DEPs). Enrichment analyses of the genes were conducted using KOBAS-i database. MultipleROC was applied to evaluate the diagnostic potential of biomarkers, and rms package was utilized to construct diagnostic models. hTFtarget database was utilized to predict potential transcription factors (TFs). Finally, cell-based assays were performed to validate the expression and potential biological functions of the screened key markers. RESULTS This study found that M2 macrophages were enriched in protein, endoplasmic reticulum, and virus-related pathways. A total of 4146 DEGs and 1946 DEPs were obtained through screening, with 254 common DEGs/DEPs. The results of gene function enrichment analysis suggested that it may affect the occurrence and development of GC through DNA replication and cell cycle. This study identified three biomarkers, HSPH1, HSPD1, and IFI30, and constructed a diagnostic model based on these three genes. The AUC value greater than 0.8 proved the reliability of the model. Through screening TFs, SPI1 and KLF5 were found to be the common TFs for the three biomarkers. The expression of the three genes IFI30, HSPD1 and HSPH1 was up-regulated in GC cells, and IFI30 may play a facilitating role in the migration and invasion of GC cells. CONCLUSION This study identified three biomarkers and constructed a diagnostic model, providing a new perspective for the research and treatment of GC.
Collapse
Affiliation(s)
- Xilong Wang
- Tumor Hematology Department, Liaoyang Central Hospital, Liaoyang, 111000, China
| | - Ying Zhang
- General Surgery Department, Liaoyang Central Hospital, Liaoyang, 111000, China.
| |
Collapse
|
13
|
Leylek O, Honeywell ME, Lee MJ, Hemann MT, Ozcan G. Functional genomics reveals an off-target dependency of drug synergy in gastric cancer therapy. Gastric Cancer 2024; 27:1201-1219. [PMID: 39033209 PMCID: PMC11513712 DOI: 10.1007/s10120-024-01537-y] [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: 04/04/2024] [Accepted: 07/08/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Integrating molecular-targeted agents into combination chemotherapy is transformative for enhancing treatment outcomes in cancer. However, realizing the full potential of this approach requires a clear comprehension of the genetic dependencies underlying drug synergy. While the interactions between conventional chemotherapeutics are well-explored, the interplay of molecular-targeted agents with conventional chemotherapeutics remains a frontier in cancer treatment. Hence, we leveraged a powerful functional genomics approach to decode genomic dependencies that drive synergy in molecular-targeted agent/chemotherapeutic combinations in gastric adenocarcinoma, addressing a critical need in gastric cancer therapy. METHODS We screened pharmacological interactions between fifteen molecular-targeted agent/conventional chemotherapeutic pairs in gastric adenocarcinoma cells, and examined the genome-scale genetic dependencies of synergy integrating genome-wide CRISPR screening with the shRNA-based signature assay. We validated the synergy in cell death using fluorescence-based and lysis-dependent inference of cell death kinetics assay, and validated the genetic dependencies by single-gene knockout experiments. RESULTS Our combination screen identified SN-38/erlotinib as the drug pair with the strongest synergism. Functional genomics assays unveiled a genetic dependency signature of SN-38/erlotinib identical to SN-38. Remarkably, the enhanced cell death with improved kinetics induced by SN-38/erlotinib was attributed to erlotinib's off-target effect, inhibiting ABCG2, rather than its on-target effect on EGFR. CONCLUSION In the era of precision medicine, where emphasis on primary drug targets prevails, our research challenges this paradigm by showcasing a robust synergy underpinned by an off-target dependency. Further dissection of the intricate genetic dependencies that underlie synergy can pave the way to developing more effective combination strategies in gastric cancer therapy.
Collapse
Affiliation(s)
- Ozen Leylek
- Koç University Research Center for Translational Medicine, 34450, Istanbul, Turkey
| | - Megan E Honeywell
- Department of Systems Biology, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Michael J Lee
- Department of Systems Biology, UMass Chan Medical School, Worcester, MA, 01605, USA.
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA, 01605, USA.
| | - Michael T Hemann
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
- MIT Koch Institute for Integrative Cancer Research, Cambridge, MA, 02139, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, 02139, USA.
| | - Gulnihal Ozcan
- Koç University Research Center for Translational Medicine, 34450, Istanbul, Turkey.
- Department of Medical Pharmacology, Koç University School of Medicine, 34450, Istanbul, Turkey.
| |
Collapse
|
14
|
Wong R, Anderson B, Bashir B, Bateman J, Chalchal H, Davies J, Dehmoobed A, Geller G, Ghose A, Gill S, Gordon V, Green S, Hebbard P, Iqbal M, Ji S, Karachiwala H, Kidane B, Kim C, Kosyachkova E, Krahn M, Krishnan T, Kristjanson M, Lee S, Lee-Ying R, Lelond S, Liu HW, Meyers D, Mulder K, Paul J, Planincic E. Report from the 25th Annual Western Canadian Gastrointestinal Cancer Consensus Conference on Gastric and Gastroesophageal Cancers, Winnipeg, Manitoba, 26-27 October 2023. Curr Oncol 2024; 31:5987-6006. [PMID: 39451751 PMCID: PMC11505746 DOI: 10.3390/curroncol31100447] [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: 08/30/2024] [Revised: 09/20/2024] [Accepted: 09/21/2024] [Indexed: 10/26/2024] Open
Abstract
The 25th Annual Western Canadian Gastrointestinal Cancer Consensus Conference (WCGCCC) was held in Winnipeg, Manitoba, on 26-27 October 2023. The WCGCCC is an interactive multidisciplinary conference that was attended by healthcare professionals from across Western Canada (British Columbia, Alberta, and Manitoba) who are involved in the care of patients with gastrointestinal cancer. Surgical, medical, and radiation oncologists; pathologists; oncology nurses; pharmacists; and a family physician in oncology (FPO) participated in presentation and discussion sessions for the purpose of developing the recommendations presented here. This consensus statement addresses current issues in the management of gastroesophageal cancers.
Collapse
Affiliation(s)
- Ralph Wong
- CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada; (B.B.); (A.D.); (V.G.); (S.G.); (P.H.); (M.K.); (M.K.); (S.L.); (D.M.); (E.P.); (J.P.)
| | - Brady Anderson
- Western Manitoba Cancer Center, Brandon, MB R7A 5M8, Canada;
| | - Bashir Bashir
- CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada; (B.B.); (A.D.); (V.G.); (S.G.); (P.H.); (M.K.); (M.K.); (S.L.); (D.M.); (E.P.); (J.P.)
| | | | - Haji Chalchal
- Allan Blair Cancer Centre, Regina, SK S4T 7T1, Canada;
| | - Janine Davies
- BC Cancer Centre, Vancouver, BC V5Z 4E6, Canada; (J.D.); (S.G.); (T.K.)
| | - Anahita Dehmoobed
- CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada; (B.B.); (A.D.); (V.G.); (S.G.); (P.H.); (M.K.); (M.K.); (S.L.); (D.M.); (E.P.); (J.P.)
| | | | - Abhijit Ghose
- Chinook Regional Hospital, Lethbridge, AB T1J 1W5, Canada
| | - Sharlene Gill
- BC Cancer Centre, Vancouver, BC V5Z 4E6, Canada; (J.D.); (S.G.); (T.K.)
| | - Vallerie Gordon
- CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada; (B.B.); (A.D.); (V.G.); (S.G.); (P.H.); (M.K.); (M.K.); (S.L.); (D.M.); (E.P.); (J.P.)
| | - Susan Green
- CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada; (B.B.); (A.D.); (V.G.); (S.G.); (P.H.); (M.K.); (M.K.); (S.L.); (D.M.); (E.P.); (J.P.)
| | - Pamela Hebbard
- CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada; (B.B.); (A.D.); (V.G.); (S.G.); (P.H.); (M.K.); (M.K.); (S.L.); (D.M.); (E.P.); (J.P.)
| | | | - Shuying Ji
- Shared Health Manitoba, Winnipeg, MB R2N 0E2, Canada;
| | - Hatim Karachiwala
- Tom Baker Cancer Centre, Calgary, AB T2N 4N2, Canada; (H.K.); (R.L.-Y.)
| | - Biniam Kidane
- Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3A 1R9, Canada;
| | - Christina Kim
- CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada; (B.B.); (A.D.); (V.G.); (S.G.); (P.H.); (M.K.); (M.K.); (S.L.); (D.M.); (E.P.); (J.P.)
| | | | - Marianne Krahn
- CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada; (B.B.); (A.D.); (V.G.); (S.G.); (P.H.); (M.K.); (M.K.); (S.L.); (D.M.); (E.P.); (J.P.)
| | - Tharani Krishnan
- BC Cancer Centre, Vancouver, BC V5Z 4E6, Canada; (J.D.); (S.G.); (T.K.)
| | - Mark Kristjanson
- CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada; (B.B.); (A.D.); (V.G.); (S.G.); (P.H.); (M.K.); (M.K.); (S.L.); (D.M.); (E.P.); (J.P.)
| | - Sangjune Lee
- Department of Oncology, University of Calgary, Calgary, AB T2N 4N2, Canada;
| | - Richard Lee-Ying
- Tom Baker Cancer Centre, Calgary, AB T2N 4N2, Canada; (H.K.); (R.L.-Y.)
| | - Stephanie Lelond
- CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada; (B.B.); (A.D.); (V.G.); (S.G.); (P.H.); (M.K.); (M.K.); (S.L.); (D.M.); (E.P.); (J.P.)
| | - Hong-Wei Liu
- Central Alberta Cancer Center, Red Deer, AB T4N 6R2, Canada;
| | - Daniel Meyers
- CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada; (B.B.); (A.D.); (V.G.); (S.G.); (P.H.); (M.K.); (M.K.); (S.L.); (D.M.); (E.P.); (J.P.)
| | - Karen Mulder
- Cross Cancer Institute, Edmonton, AB T6G 1Z2, Canada;
| | - James Paul
- CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada; (B.B.); (A.D.); (V.G.); (S.G.); (P.H.); (M.K.); (M.K.); (S.L.); (D.M.); (E.P.); (J.P.)
| | - Elvira Planincic
- CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada; (B.B.); (A.D.); (V.G.); (S.G.); (P.H.); (M.K.); (M.K.); (S.L.); (D.M.); (E.P.); (J.P.)
| |
Collapse
|
15
|
Sharma S, Carey N, McConnell D, Lowery M, O'Sullivan J, McCullagh L. Systematic Review of Economic Evaluations of Systemic Treatments for Advanced and Metastatic Gastric Cancer. PHARMACOECONOMICS 2024; 42:1091-1110. [PMID: 39060831 PMCID: PMC11405472 DOI: 10.1007/s40273-024-01413-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/02/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND Recent advances in the development of biomarker-directed therapy and immunotherapy, for advanced and metastatic gastric cancers, have the potential to improve survival and quality of life. Much attention has been directed towards second- and later-line treatments, and the landscape here is evolving rapidly. However, uncertainty in relative effectiveness, high costs and uncertainty in cost effectiveness represent challenges for decision makers. OBJECTIVE To identify economic evaluations for the second-line or later-line treatment of advanced and metastatic gastric cancer. Also, to assess key criteria (including model assumptions, inputs and outcomes), reporting completeness and methodological quality to inform future cost-effectiveness evaluations. METHODS A systematic literature search (from database inception to 5 March 2023) of EconLit via EBSCOhost, Cochrane Library (restricted to National Health Service [NHS] Economic Evaluation Database and Health Technology Assessment [HTA] Database), Embase, MEDLINE and of grey literature was conducted. This aimed to identify systemic treatments that align with National Comprehensive Cancer Network (NCCN) and European Society for Medical Oncology (ESMO) Clinical Practice Guidelines. Data were collected on key criteria and on reporting completeness and methodological quality. A narrative synthesis focussed on cost-effectiveness and cost-of-illness studies. Outcomes of interest included total and incremental costs and outcomes (life-years and quality-adjusted life-years), ratios of incremental costs per unit outcome and other summary cost and outcome measures. Also, for cost-effectiveness studies, reporting completeness and the methodological quality were assessed using the Consolidated Health Economic Evaluation Reporting Standards (CHEERS) and the Philips Checklist, respectively. RESULTS A total of 19 eligible economic evaluations were identified (cost-effectiveness studies [n = 15] and cost-of-illness studies [n = 4]). There was a general lack of consistency in the methodological approaches taken across studies. In the main, the cost-effectiveness studies indicated that the intervention under consideration was more effective and more costly than the comparator(s). However, most interventions were not cost effective. No studies were fully compliant with reporting-completeness and methodological-quality requirements. Given the lack of consistency in the approaches taken across cost-of-illness studies, outcomes could not be directly compared. CONCLUSIONS To our knowledge, this is the first published systematic literature review that has qualitatively synthesised economic evaluations for advanced and metastatic gastric cancer. There were differences in the approaches taken across the cost-effectiveness studies and the cost-of-illness studies. The conclusions of most of the cost-effectiveness studies were consistent despite identified differences in approaches. In the main, the interventions under consideration were not cost effective, presenting challenges to sustainability and affordability. We highlight a requirement for cost-effectiveness evaluations and for second-line or later-line treatments of advanced and metastatic gastric cancer that consider all relevant comparators and that are compliant with reporting-completeness and methodological-quality requirements. By addressing the methodological gaps identified here, future healthcare decision-making, within the context of this rapidly changing treatment landscape, would be better informed. PROSPERO REGISTRATION NUMBER CRD42023405951.
Collapse
Affiliation(s)
- Shikha Sharma
- School of Medicine, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences,Trinity College Dublin, Dublin, D08 NHY1, Ireland.
- National Centre for Pharmacoeconomics, Dublin, Ireland.
| | - Niamh Carey
- National Centre for Pharmacoeconomics, Dublin, Ireland
- Department of Pharmacology and Therapeutics, Trinity Centre for Health Sciences, Trinity College Dublin, Dublin, Ireland
| | - David McConnell
- National Centre for Pharmacoeconomics, Dublin, Ireland
- Department of Pharmacology and Therapeutics, Trinity Centre for Health Sciences, Trinity College Dublin, Dublin, Ireland
| | - Maeve Lowery
- School of Medicine, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences,Trinity College Dublin, Dublin, D08 NHY1, Ireland
| | - Jacintha O'Sullivan
- School of Medicine, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences,Trinity College Dublin, Dublin, D08 NHY1, Ireland
| | - Laura McCullagh
- National Centre for Pharmacoeconomics, Dublin, Ireland
- Department of Pharmacology and Therapeutics, Trinity Centre for Health Sciences, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
16
|
Loew A, Schneider C, Pflüger M, Mantke R, Weylandt KH, Gretschel S. [Treatment reality of esophageal cancer in the Federal State of Brandenburg : Comparison between squamous cell carcinoma and adenocarcinoma]. CHIRURGIE (HEIDELBERG, GERMANY) 2024; 95:825-832. [PMID: 38861172 DOI: 10.1007/s00104-024-02110-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/14/2024] [Indexed: 06/12/2024]
Abstract
BACKGROUND Clinical cancer registries are intended to reflect the reality of care through differentiated data analysis and, if necessary, to offer approaches for improving care. METHODS For the years 2000-2018, the data of the Clinical Epidemiological Cancer Registry Brandenburg-Berlin were examined separately for adenocarcinoma and squamous cell carcinoma with respect to epidemiology and health care reality. RESULTS Between 2000 and 2018 a total of 3207 esophageal cancers were documented in the cancer registry, of which 2182 were squamous cell carcinomas (ESCC), 843 adenocarcinomas (EAC) and 182 various others or missing histology. During the observation period there was a clear dominance of ESCC but with a significant increase in EAC in both sexes. Overall, the rate of new cases was 5 times higher for men than for women. The relative 5‑year survival probability of all esophageal cancers was 17.4% in men and 22.5% in women. Patients with EAC survived significantly longer than those with ESCC. Radiotherapy and chemotherapy, individually or in combination, were mainly used as treatment methods. Surgery was performed on 19% of ESCC and 42% of EAC. CONCLUSION The proportion of ESCC in Brandenburg is still significantly higher than EAC, with a significant increase for the latter, especially in men. Although locally advanced tumors have been significantly more common, modern neoadjuvant concepts have rarely been documented, and although the quality of the surgery is comparable to the international standard, surgery is carried out in relatively few patients.
Collapse
Affiliation(s)
- Andreas Loew
- Medizinische Klinik B, Universitätsklinikum Ruppin-Brandenburg (ukrb), Medizinische Hochschule Brandenburg, 16816, Neuruppin, Deutschland.
| | - Constanze Schneider
- Klinisch-epidemiologisches Krebsregister Brandenburg-Berlin (KKRBB), 03044, Cottbus, Deutschland
| | - Maren Pflüger
- Klinisch-epidemiologisches Krebsregister Brandenburg-Berlin (KKRBB), 03044, Cottbus, Deutschland
| | - René Mantke
- Klinik für Allgemein- und Viszeralchirurgie, Universitätsklinikum Brandenburg, Medizinische Hochschule Brandenburg, 14770, Brandenburg an der Havel, Deutschland
- Fakultät für Gesundheitswissenschaften, gemeinsame Fakultät der Brandenburgischen Technischen Universität Cottbus-Senftenberg, der Brandenburgischen Medizinischen Hochschule Theodor Fontane und der Universität Potsdam, Potsdam, Deutschland
| | - Karsten H Weylandt
- Medizinische Klinik B, Universitätsklinikum Ruppin-Brandenburg (ukrb), Medizinische Hochschule Brandenburg, 16816, Neuruppin, Deutschland
- Fakultät für Gesundheitswissenschaften, gemeinsame Fakultät der Brandenburgischen Technischen Universität Cottbus-Senftenberg, der Brandenburgischen Medizinischen Hochschule Theodor Fontane und der Universität Potsdam, Potsdam, Deutschland
| | - Stephan Gretschel
- Fakultät für Gesundheitswissenschaften, gemeinsame Fakultät der Brandenburgischen Technischen Universität Cottbus-Senftenberg, der Brandenburgischen Medizinischen Hochschule Theodor Fontane und der Universität Potsdam, Potsdam, Deutschland
- Klinik für Allgemein‑, Viszeral‑, Thorax- und Gefäßchirurgie, Universitätsklinikum Ruppin-Brandenburg (ukrb), Medizinische Hochschule Brandenburg, 16816, Neuruppin, Deutschland
| |
Collapse
|
17
|
Lordick F, Rha SY, Muro K, Yong WP, Lordick Obermannová R. Systemic Therapy of Gastric Cancer-State of the Art and Future Perspectives. Cancers (Basel) 2024; 16:3337. [PMID: 39409957 PMCID: PMC11475804 DOI: 10.3390/cancers16193337] [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: 09/04/2024] [Revised: 09/27/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Background: The prognosis of patients diagnosed with locally advanced and metastatic gastric and esophago-gastric junction cancer is critical. The optimal choice of systemic therapy is essential to optimize survival outcomes. Methods: A comprehensive literature review via PubMed and analysis of major oncology congresses (European Society for Medical Oncology and American Society of Clinical Oncology websites) were conducted to ascertain the current status and latest developments in the systemic treatment of patients with localized or advanced gastric and esophago-gastric junction adenocarcinoma. Results: While neoadjuvant and perioperative chemotherapy for localized tumor stages is the preferred approach in the Western Hemisphere, adjuvant chemotherapy remains the preferred course of action in East Asia. The administration of chemotherapy, typically in the form of combinations comprising platinum and fluoropyrimidine compounds in combination with docetaxel, represents a standard of care. Investigations are underway into the potential of immunotherapy and other biologically targeted agents in the perioperative setting. To select the most appropriate therapy for advanced gastric cancer, including adenocarcinoma of the esophago-gastric junction, it is essential to determine biomarkers such as HER2 expression, PD-L1 combined positive score (CPS) (combined positive score), Claudin 18.2, and microsatellite instability (MSI). In the present clinical context, the standard first-line therapy is a combination of fluoropyrimidine and a platinum derivative. The selection of chemotherapy in combination with antibodies is contingent upon the specific biomarker under consideration. Conclusions: This article reviews the current state of the art based on recent clinical trial results and provides an outlook on the future of systemic therapy.
Collapse
Affiliation(s)
- Florian Lordick
- Department of Medicine (Oncology, Gastroenterology, Hepatology, Pulmonology), University of Leipzig Medical Center, Cancer Center Central Germany, 04103 Leipzig, Germany
| | - Sun Young Rha
- Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Kei Muro
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya 464-8681, Japan
| | - Wei Peng Yong
- Department of Haematology-Oncology, National University Cancer Institute, Singapore 119074, Singapore
| | - Radka Lordick Obermannová
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Faculty of Medicine, Masaryk University, 656 53 Brno, Czech Republic
| |
Collapse
|
18
|
Xiang Z, Ma L, Fu Y, Pan Y. Cost-effectiveness analysis of first-line sintilimab plus chemotherapy vs. chemotherapy alone for unresectable advanced or metastatic gastric or gastroesophageal junction cancer in China. Front Pharmacol 2024; 15:1411571. [PMID: 39295936 PMCID: PMC11408219 DOI: 10.3389/fphar.2024.1411571] [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: 04/03/2024] [Accepted: 08/26/2024] [Indexed: 09/21/2024] Open
Abstract
Background The Chinese Society of Clinical Oncology (CSCO) has recommended sintilimab plus chemotherapy (SINT + Chemo) as a standard first-line therapy for advanced gastric cancer or gastroesophageal junction cancer (GC/GEJC), based on the proven effectiveness and safety in the ORINT-16 trail. Its cost-effectiveness, however, remains to be evaluated. Methods We established a partitioned survival approach (PartSA) model with a 10-year time horizon to determine whether SINT + Chemo (vs. chemotherapy) was more cost-effective as a first-line treatment for unresectable advanced or metastatic GC/GEJC. Survival data was generated from the ORIENT-16 trail. Cost calculation was limited to direct medical costs. Database of Hunan Public Resources Trading Service Platform was used as the source for obtaining drug prices. Other cost and utility values were gathered from established literature. Incremental cost-effectiveness ratio (ICER) was the primary output. Additionally, we conducted sensitivity analysis, subgroup analysis, and scenario analysis. Results In the base-case analysis, group SINT + Chemo showed an increase in utility value by 0.32 quality-adjusted life-years (QALYs) at an extra cost of $7988.43, resulting in an ICER of $25239.29/QALY, below the Chinese cost-effective willingness-to-pay (WTP) threshold of $38223.34. Upon further subgroup analysis according to patients' programmed death 1 ligand (PD-L1) combined positive score (CPS), the ICERs were $26341.01/QALY for patients highly expressing PD-L1 (CPS ≥5) and $17658.26/QALY for patients lowly expressing PD-L1 (CPS <5). Based on the sensitivity analysis, we found the PFS utility was the parameter that had the most significant impact on the model's outcomes. Moreover, in scenario analysis, the results remained consistent despite variations in the model's time frame. Conclusion In China, SINT + Chemo is a more cost-effective option (vs. chemotherapy) as a first-line therapy for unresectable advanced or metastatic GC/GEJC, irrespective of PD-L1 expression levels.
Collapse
Affiliation(s)
- Zuojuan Xiang
- Department of Pharmacy, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
| | - Ling Ma
- Department of Clinical pharmacy, the First People's Hospital of Yunnan Province, the Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Yingzhou Fu
- Department of Pharmacy, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
| | - Yong Pan
- Department of Pharmacy, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
| |
Collapse
|
19
|
Wang FY, Huang XM, Cao YQ, Cao J, Ni J, Li K, Lu M, Huang XE. Nanoparticle Polymeric Micellar Paclitaxel Versus Paclitaxel for Patients with Advanced Gastric Cancer. J Gastrointest Cancer 2024; 55:1105-1110. [PMID: 38668776 PMCID: PMC11347489 DOI: 10.1007/s12029-024-01058-y] [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] [Accepted: 04/20/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND Nanoparticle polymeric micellar paclitaxel (NPMP) is a novel Cremophor EL (CrEL)-free nanoparticle micellar formulation of paclitaxel. This study evaluated the efficacy and toxicity of NPMP in the treatment of patients with advanced gastric cancer (AGC). METHODS Patients with histologically confirmed AGC in Jiangsu Cancer Hospital were retrospectively collected and divided into two groups. Patients in group A received NPMP at a total dose of 360 mg/m2 each cycle, and patients in group B were given paclitaxel at a dose of 210 mg/m2 each cycle. In addition, all patients received 5-fluorouracil at a dose of 0.75 g/m2 on days 1-4 and leucovorin at a dose of 200 mg/m2 on days 1-4 for at least 2 cycles. RESULTS From January 2021 to May 2023, 63 patients (32 in group A and 31 in group B) could be evaluated for treatment response. A marked disparity in the overall response was observed between groups A and B, indicating statistical significance. The overall response rate was 31% in group A (10/32) and 10% in group B (3/31) (P = 0.034). Disease control rate was 91% in group A (29/32) and 81% in group B (25/31) (P = 0.440). No statistically significant difference in adverse reactions was observed between the two groups. However, the incidence of anemia, leucopenia, nausea, vomiting, diarrhea, liver dysfunction, and allergy in group A was notably lower than that in group B. CONCLUSIONS NPMP combined chemotherapy offers a new, active, and safe treatment for patients with AGC.
Collapse
Affiliation(s)
- Fei-Yu Wang
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University& Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, No. 42 Baiziting , Jiangsu, 210009, China
| | - Xiang-Ming Huang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yu-Qing Cao
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University& Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, No. 42 Baiziting , Jiangsu, 210009, China
| | - Jie Cao
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University& Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, No. 42 Baiziting , Jiangsu, 210009, China
| | - Jie Ni
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University& Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, No. 42 Baiziting , Jiangsu, 210009, China
| | - Ke Li
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University& Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, No. 42 Baiziting , Jiangsu, 210009, China
| | - Min Lu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xin-En Huang
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University& Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, No. 42 Baiziting , Jiangsu, 210009, China.
| |
Collapse
|
20
|
Liao Z, Xie Z. Construction of a disulfidptosis-related glycolysis gene risk model to predict the prognosis and immune infiltration analysis of gastric adenocarcinoma. Clin Transl Oncol 2024; 26:2309-2322. [PMID: 38587603 DOI: 10.1007/s12094-024-03457-w] [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: 01/02/2024] [Accepted: 03/11/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND The pattern of cell death known as disulfidptosis was recently discovered. Disulfidptosis, which may affect the growth of tumor cells, represents a potential new approach to treating tumors. Glycolysis affects tumor proliferation, invasion, chemotherapy resistance, the tumor microenvironment (TME), and immune evasion. However, the efficacy and therapeutic significance of disulfidptosis-related glycolysis genes (DRGGs) in stomach adenocarcinoma (STAD) remain uncertain. METHODS STAD clinical data and RNA sequencing data were downloaded from the TCGA database. DRGGs were screened using Cox regression and Lasso regression analysis to construct a prognostic risk model. The accuracy of the model was verified using survival studies, receiver operating characteristic (ROC) curves, column plots, and calibration curves. Additionally, our study investigated the relationships between the risk scores and immune cell infiltration, tumor mutational burden (TMB), and anticancer drug sensitivity. RESULTS We have successfully developed a prognosis risk model with 4 DRGGs (NT5E, ALG1, ANKZF1, and VCAN). The model showed excellent performance in predicting the overall survival of STAD patients. The DRGGs prognostic model significantly correlated with the TME, immune infiltrating cells, and treatment sensitivity. CONCLUSIONS The risk model developed in this work has significant clinical value in predicting the impact of immunotherapy in STAD patients and assisting in the choice of chemotherapeutic medicines. It can correctly estimate the prognosis of STAD patients.
Collapse
Affiliation(s)
- Zhaohui Liao
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, Nanchang, 330006, China
| | - Zhengyuan Xie
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, Nanchang, 330006, China.
| |
Collapse
|
21
|
Li X, Du Y. Lactate Metabolism Subtypes Analysis Reveals CCDC80 as a Novel Prognostic Biomarker in Gastric Cancer. J Cancer 2024; 15:5557-5576. [PMID: 39308689 PMCID: PMC11414610 DOI: 10.7150/jca.97640] [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: 04/22/2024] [Accepted: 07/16/2024] [Indexed: 09/25/2024] Open
Abstract
Lactate metabolism plays a vital role in tumor progression. Currently, gastric cancer (GC) has a poor prognosis. Therefore, our research aimed to investigate novel biomarkers related to lactate metabolism in patients. Patient data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) database were divided into subtypes based on the expression of lactate metabolism-related genes (LMRGs). Based on the subtypes, we identified coiled-coil domain containing 80 (CCDC80) for further investigation. Univariate and multivariate Cox regression models were constructed to determine the prognostic value of CCDC80 in GC. We further explored the mechanism by which CCDC80 affects GC prognosis using gene set enrichment analysis (GSEA). Immune infiltration and drug sensitivity analyses were also performed. Finally, immunohistochemical staining was used to evaluate CCDC80 expression in normal and tumor tissues. We observed that CCDC80 was overexpressed in GC samples and was significantly associated with T and pathological stages. Multivariate Cox analysis identified high CCDC80 expression as an independent prognostic marker. GSEA indicated that the oxidative phosphorylation pathway was highly enriched in the low CCDC80 expression group. Moreover, CCDC80 was associated with immune cell infiltration, especially that of M2 macrophages. Patients with higher CCDC80 expression exhibited lower sensitivity to paclitaxel. In conclusion, our findings demonstrate that CCDC80 is a critical regulator in GC progression and immune response and is associated with lactate metabolism, and it could be used as a novel biomarker for prognostic and chemotherapy treatment purposes.
Collapse
Affiliation(s)
- Xiang Li
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110000, China
| | - Yaqi Du
- Department of Gastroenterology, The First Hospital of China Medical University, Shenyang 110000, China
| |
Collapse
|
22
|
Ryu TY, Tae IH, Han TS, Lee J, Kim K, Kang Y, Kim S, Lee HJ, Jung CR, Lim JH, Kim DS, Son MY, Cho HS. Epigenetic alterations of TP53INP1 by EHMT2 regulate the cell cycle in gastric cancer. Exp Hematol Oncol 2024; 13:86. [PMID: 39160629 PMCID: PMC11334499 DOI: 10.1186/s40164-024-00554-y] [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: 03/30/2024] [Accepted: 08/07/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is a type of cancer with high incidence and mortality rates. Although various chemical interventions are being developed to treat gastric cancer, there is a constant demand for research into new GC treatment targets and modes of action (MOAs) because of the low effectiveness and side effects of current treatments. METHODS Using the TCGA data portal, we identified EHMT2 overexpression in GC samples. Using RNA-seq and EHMT2-specific siRNA, we investigated the role of EHMT2 in GC cell proliferation and validated its function with two EHMT2-specific inhibitors. Through the application of 3D spheroid culture, patient-derived gastric cancer organoids (PDOs), and an in vivo model, we confirmed the role of EHMT2 in GC cell proliferation. RESULTS In this study, we found that EHMT2, a histone 3 lysine 9 (H3K9) methyltransferase, is significantly overexpressed in GC patients compared with healthy individuals. Knockdown of EHMT2 with siRNA induced G1 cell cycle arrest and attenuated GC cell proliferation. Furthermore, we confirmed that TP53INP1 induction by EHMT2 knockdown induced cell cycle arrest and inhibited GC cell proliferation. Moreover, specific EHMT2 inhibitors, BIX01294 and UNC0638, induced cell cycle arrest in GC cell lines through TP53INP1 upregulation. The efficacy of EHMT2 inhibition was further confirmed in a 3D spheroid culture system, PDOs, and a xenograft model. CONCLUSIONS Our findings suggest that EHMT2 is an attractive therapeutic target for GC treatment.
Collapse
Affiliation(s)
- Tae Young Ryu
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - In Hwan Tae
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Tae-Su Han
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
- Korea University of Science and Technology, Daejeon, 34316, Republic of Korea
- Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Jinkwon Lee
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
- Korea University of Science and Technology, Daejeon, 34316, Republic of Korea
| | - Kwangho Kim
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Yunsang Kang
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
- Korea University of Science and Technology, Daejeon, 34316, Republic of Korea
| | - Solbi Kim
- Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Hyo Jin Lee
- Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Cho-Rok Jung
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
- Korea University of Science and Technology, Daejeon, 34316, Republic of Korea
| | - Jung Hwa Lim
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
- Korea University of Science and Technology, Daejeon, 34316, Republic of Korea
| | - Dae-Soo Kim
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea.
- Korea University of Science and Technology, Daejeon, 34316, Republic of Korea.
| | - Mi-Young Son
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea.
- Korea University of Science and Technology, Daejeon, 34316, Republic of Korea.
- Department of Biological Science, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Hyun-Soo Cho
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea.
- Korea University of Science and Technology, Daejeon, 34316, Republic of Korea.
- Department of Biological Science, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
23
|
Chong X, Madeti Y, Cai J, Li W, Cong L, Lu J, Mo L, Liu H, He S, Yu C, Zhou Z, Wang B, Cao Y, Wang Z, Shen L, Wang Y, Zhang X. Recent developments in immunotherapy for gastrointestinal tract cancers. J Hematol Oncol 2024; 17:65. [PMID: 39123202 PMCID: PMC11316403 DOI: 10.1186/s13045-024-01578-x] [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: 05/22/2024] [Accepted: 07/18/2024] [Indexed: 08/12/2024] Open
Abstract
The past few decades have witnessed the rise of immunotherapy for Gastrointestinal (GI) tract cancers. The role of immune checkpoint inhibitors (ICIs), particularly programmed death protein 1 (PD-1) and PD ligand-1 antibodies, has become increasingly pivotal in the treatment of advanced and perioperative GI tract cancers. Currently, anti-PD-1 plus chemotherapy is considered as first-line regimen for unselected advanced gastric/gastroesophageal junction adenocarcinoma (G/GEJC), mismatch repair deficient (dMMR)/microsatellite instability-high (MSI-H) colorectal cancer (CRC), and advanced esophageal cancer (EC). In addition, the encouraging performance of claudin18.2-redirected chimeric antigen receptor T-cell (CAR-T) therapy in later-line GI tract cancers brings new hope for cell therapy in solid tumour treatment. Nevertheless, immunotherapy for GI tumour remains yet precise, and researchers are dedicated to further maximising and optimising the efficacy. This review summarises the important research, latest progress, and future directions of immunotherapy for GI tract cancers including EC, G/GEJC, and CRC.
Collapse
Affiliation(s)
- Xiaoyi Chong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Yelizhati Madeti
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Jieyuan Cai
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Wenfei Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Lin Cong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Jialin Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Liyang Mo
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Huizhen Liu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Siyi He
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Chao Yu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Zhiruo Zhou
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Boya Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Yanshuo Cao
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Zhenghang Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Yakun Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China.
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China.
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| |
Collapse
|
24
|
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 DOI: 10.1080/17460441.2024.2370332] [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: 03/31/2024] [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
|
25
|
Xing Y, Zhang Z, Gao W, Song W, Li T. Immune infiltration and prognosis in gastric cancer: role of NAD+ metabolism-related markers. PeerJ 2024; 12:e17833. [PMID: 39099656 PMCID: PMC11297443 DOI: 10.7717/peerj.17833] [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: 04/05/2024] [Accepted: 07/09/2024] [Indexed: 08/06/2024] Open
Abstract
Background This study endeavored to develop a nicotinamide adenine dinucleotide (NAD+) metabolism-related biomarkers in gastric cancer (GC), which could provide a theoretical foundation for prognosis and therapy of GC patients. Methods In this study, differentially expressed genes (DEGs1) between GC and paraneoplastic tissues were overlapped with NAD+ metabolism-related genes (NMRGs) to identify differentially expressed NMRGs (DE-NMRGs). Then, GC patients were divided into high and low score groups by gene set variation analysis (GSVA) algorithm for differential expression analysis to obtain DEGs2, which was overlapped with DEGs1 for identification of intersection genes. These genes were further analyzed using univariate Cox and least absolute shrinkage and selection operator (LASSO) regression analyses to obtain prognostic genes for constructing a risk model. Enrichment and immune infiltration analyses further investigated investigate the different risk groups, and qRT-PCR validated the prognostic genes. Results Initially, we identified DE-NMRGs involved in NAD biosynthesis, with seven (DNAJB13, CST2, THPO, CIDEA, ONECUT1, UPK1B and SNCG) showing prognostic significance in GC. Subsequent, a prognostic model was constructed in which the risk score, derived from the expression profiles of these genes, along with gender, emerged as robust independent predictors of patient outcomes in GC. Enrichment analysis linked high-risk patients to synaptic membrane pathways and low-risk to the CMG complex pathway. Tumor immune infiltration analysis revealed correlations between risk scores and immune cell abundance, suggesting a relationship between NAD+ metabolism and immune response in GC. The prognostic significance of our identified genes was validated by qRT-PCR, which confirmed their upregulated expression in GC tissue samples. Conclusion In this study, seven NAD+ metabolism-related markers were established, which is of great significance for the development of prognostic molecular biomarkers and clinical prognosis prediction for gastric cancer patients.
Collapse
Affiliation(s)
- Yu Xing
- The Third Central Clinical College of Tianjin Medical University, Tianjin, China
- The Third Central Hospital of Tianjin, Tianjin, China
- Tianjin Institute of Hepatobiliary Disease, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
| | - Zili Zhang
- The Third Central Clinical College of Tianjin Medical University, Tianjin, China
- The Third Central Hospital of Tianjin, Tianjin, China
- Tianjin Institute of Hepatobiliary Disease, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
| | - Wenqing Gao
- The Third Central Hospital of Tianjin, Tianjin, China
- Tianjin Institute of Hepatobiliary Disease, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
| | - Weiliang Song
- The Third Central Hospital of Tianjin, Tianjin, China
- Tianjin Institute of Hepatobiliary Disease, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
| | - Tong Li
- The Third Central Clinical College of Tianjin Medical University, Tianjin, China
- The Third Central Hospital of Tianjin, Tianjin, China
- Tianjin Institute of Hepatobiliary Disease, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
| |
Collapse
|
26
|
Su R, Narenmandula, Qiao X, Hu Q. PDE4B promotes the progression of gastric cancer via the PI3K/AKT/MYC pathway and immune infiltration. Am J Cancer Res 2024; 14:3451-3467. [PMID: 39113853 PMCID: PMC11301292 DOI: 10.62347/tyos8160] [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: 01/27/2024] [Accepted: 06/25/2024] [Indexed: 08/10/2024] Open
Abstract
Phosphodiesterase 4B (PDE4B) is a key enzyme involved in regulating intracellular cyclic adenosine monophosphate levels and plays a significant role in the diagnosis, classification, treatment, and prognosis of various cancers. However, the role of PDE4B in gastric cancer (GC) remains unclear. We used the GEPIA2 (Gene Expression Profiling Interactive Analysis 2) database to analyze the differential expression level of PDE4B across tumor samples and verified our findings via qPCR and immunohistochemical analysis. We also analyzed the correlation between PDE4B expression levels and clinical pathological parameters, and prognosis, in the database. The effects of PDE4B on GC proliferation, migration, and invasion were evaluated through in vitro and in vivo experiments. Enrichment analysis was performed using bioinformatic tools, and results were validated by western blot analysis. The correlation between PDE4B expression and immune cell infiltration was investigated using bioinformatics tools. PDE4B is highly expressed in GC and is significantly associated with deep infiltration, distant metastasis, tumor, node, metastasis (TNM) stage, and preoperative CA199 levels. Over-expression of PDE4B promotes proliferation, clonal formation, migration, and invasion of GC cells and is associated with poor prognosis. PDE4B promotes the infiltration of immune cells into the tumor microenvironment (TME) and the phosphorylation of PI3K/AKT pathway, increasing MYC expression. PDE4B can serve as an independent prognostic biomarker for GC. We found that PDE4B can promote immune cell infiltration of the TME and mediate malignancy in gastric cancer through the PI3K/AKT/MYC pathway.
Collapse
Affiliation(s)
- Riya Su
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen UniversityNo. 74 Zhongshan Second Road, Yuexiu District, Guangzhou 510080, Guangdong, China
| | - Narenmandula
- School of Traditional Mongolian Medicine, Inner Mongolia Medical UniversityJinshan Development Zone, Hohhot 010110, Inner Mongolia, China
| | - Xiaojuan Qiao
- Department of Oncology, Affiliated Hospital of Inner Mongolia Medical UniversityNo. 1 Tongdao North Road, Hohhot 010050, Inner Mongolia, China
| | - Qun Hu
- Department of Oncology, Affiliated Hospital of Inner Mongolia Medical UniversityNo. 1 Tongdao North Road, Hohhot 010050, Inner Mongolia, China
| |
Collapse
|
27
|
Ramaswamy A, Bhargava P, Dubashi B, Gupta A, Kapoor A, Srinivas S, Shetty O, Jadhav P, Desai V, Noronha V, Joshi A, Menon N, Patil VM, Mishra BK, Sansar B, Singh A, Patel S, Singh SN, Dhal I, Vinayak KR, Pal V, Mandavkar S, Kannan S, Chaugule D, Patil R, Parulekar M, Nashikkar C, Ankathi SK, Kaushal RK, Shah A, Ganesan P, Kayal S, Ananthakrishnan R, Syed N, Samaddar D, Kapu V, Shah A, Kaaviya D, Suganiya R, Srinivasan ND, Prabhash K, Ostwal V. Docetaxel-oxaliplatin-capecitabine/5-fluorouracil (DOX/F) followed by docetaxel versus oxaliplatin-capecitabine/5-fluorouracil (CAPOX/FOLFOX) in HER2-negative advanced gastric cancers. JNCI Cancer Spectr 2024; 8:pkae054. [PMID: 39067037 PMCID: PMC11316615 DOI: 10.1093/jncics/pkae054] [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: 02/16/2024] [Revised: 04/29/2024] [Accepted: 06/26/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND We evaluated whether the addition of docetaxel (D) to a combination comprising 5-fluorouracil/leucovorin (5-FU/LV) or capecitabine (C) plus oxaliplatin (O) (DOF/DOX) improved overall survival (OS) compared with 6 months of 5-fluorouracil (5-FU) or capecitabine in combination with oxaliplatin (FOLFOX/CAPOX) alone in advanced HER2-negative gastroesophageal junction and gastric adenocarcinomas (G/GEJ). METHODS This study was an investigator-initiated, open-label, multi-institutional, randomized phase III trial in adult patients with HER2-negative advanced G/GEJs. The primary endpoint of the study was a comparison of median OS by Kaplan-Meier method. Next-generation sequencing was performed on tissue. RESULTS Of the 324 patients randomly assigned between July 2020 and November 2022, 305 patients were evaluable for analysis (FOLFOX/CAPOX: 156; DOF/DOX: 149). With a median follow-up time of 19.2 months (95% Confidence Interval [CI] = 16.5 months to 21.9 months) for the entire cohort, the median OS was 10.1 months (95% CI = 9.2 to 10.9) for FOLFOX/CAPOX and 8.9 months (95% CI = 7.3 to 10.5) for DOF/DOX, and this difference was not statistically significant (P = .70). An increased proportion of grade 3 or grade 4 neutropenia (21% vs 3%; P < .001) and grade 2/3 neuropathy (17% vs 7%; P = .005) was seen in patients receiving DOF/DOX. Genomic profiling revealed a low incidence of microsatellite instability (1%) and a high incidence of BRCA1 (8.4%) and BRCA2 (7.5%) somatic alterations. CONCLUSION FOLFOX or CAPOX chemotherapy for 6 months remains one of the standards of care in advanced HER2-negative gastroesophageal junction and gastric adenocarcinomas, with no additional survival benefit seen with the addition of docetaxel. Genomic profiling of patients revealed a higher than previously known incidence of somatic BRCA alterations, which requires further evaluation.CTRI (Clinical Trial Registry of India: CTRI/2020/03/023944).
Collapse
Affiliation(s)
- Anant Ramaswamy
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Prabhat Bhargava
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Biswajit Dubashi
- Department of Medical Oncology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Anuj Gupta
- Department of Medical Oncology, Homi Bhabha Cancer Hospital, Varanasi, India
| | - Akhil Kapoor
- Department of Medical Oncology, Homi Bhabha Cancer Hospital, Varanasi, India
| | - Sujay Srinivas
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Omshree Shetty
- Department of Molecular Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Poonam Jadhav
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Veena Desai
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Vanita Noronha
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Amit Joshi
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Nandini Menon
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Vijay M Patil
- Consultant Medical Oncologist, P.D. Hinduja Hospital & Medical Research Centre, Mumbai, India
| | - Bal Krishna Mishra
- Department of Medical Oncology, Homi Bhabha Cancer Hospital, Varanasi, India
| | - Bipinesh Sansar
- Department of Medical Oncology, Homi Bhabha Cancer Hospital, Varanasi, India
| | - Arpita Singh
- Department of Medical Oncology, Homi Bhabha Cancer Hospital, Varanasi, India
| | - Swapnil Patel
- Department of Surgical Oncology, Homi Bhabha Cancer Hospital, Varanasi, India
| | | | - Ipsita Dhal
- Department of Pathology, Homi Bhabha Cancer Hospital, Varanasi, India
| | | | - Vikash Pal
- Department of Medical Oncology, Homi Bhabha Cancer Hospital, Varanasi, India
| | - Sarika Mandavkar
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Sadhana Kannan
- Department of Statistics, Advanced Centre for Treatment, Research and Education in Cancer, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Deepali Chaugule
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Rajshree Patil
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Manali Parulekar
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Chaitali Nashikkar
- Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Suman Kumar Ankathi
- Department of Radiodiagnosis, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Rajiv Kumar Kaushal
- Department of Pathology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Aekta Shah
- Department of Pathology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Prasanth Ganesan
- Department of Medical Oncology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Smita Kayal
- Department of Medical Oncology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Ramesh Ananthakrishnan
- Department of Radiodiagnosis, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Noorzia Syed
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Debdeep Samaddar
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Venkatesh Kapu
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Anokhi Shah
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - D Kaaviya
- Department of Medical Oncology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - R Suganiya
- Department of Medical Oncology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Nirmala Devi Srinivasan
- Department of Medical Oncology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Kumar Prabhash
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Vikas Ostwal
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| |
Collapse
|
28
|
Huang Y, Yang X, Wei M, Yang X, Yuan Z, Huang J, Wei J, Tian L. FUT11 expression in gastric cancer: its prognostic significance and role in immune regulation. Discov Oncol 2024; 15:250. [PMID: 38941002 PMCID: PMC11213843 DOI: 10.1007/s12672-024-01120-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 06/21/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is a malignant digestive tract tumor with a high recurrence rate and poor prognosis. Fucosylation is important in tumor glycosylation, in which the key enzyme is fucosyltransferase (FUT). FUT11 is a member of the fucosyltransferase family and has been closely associated with the development of multiple cancers. However, the specific relationship between FUT11 and GC prognosis and its molecular mechanism has not been fully studied. This study explored FUT11 expression, clinical correlation, and its role in GC occurrence and development to deepen understanding of its function. METHODS FUT11 expression in 33 cancers was preliminarily analyzed using the Tumor Immunoassay Resource (TIMER2.0) database. FUT11 expression in GC was evaluated using The Cancer Genome Atlas stomach adenocarcinoma (TCGA-STAD) and Gene Expression Profiling Interactive Analysis (GEPIA2) data and verified using the Gene Expression Omnibus (GEO) GSE65801 dataset. Furthermore, we studied the survival prognosis of FUT11 in GC and analyzed its effect on the survival rate of patients with GC using the KM-plotter. We also performed COX regression analysis on TCGA GC clinical data and analyzed FUT11 expression in the pathway using the STRING and LinkedOmics databases. Moreover, the relationship between FUT11 and GC immune infiltration level was examined, and the Kaplan-Meier survival analysis diagram was constructed. The FUT11 genetic variation information was retrieved using cBioPortal, and its drug sensitivity was analyzed using CellMiner. Finally, differential FUT11 expression in GC tissues was verified using immunohistochemistry. RESULTS The data mining and analysis demonstrated that FUT11 expression was abnormally elevated in GC tissues and correlated with poor patient prognosis. The FUT11 expression level was an independent prognostic factor for GC. The difference in FUT11 expression level resulted in different degrees of immune cell infiltration in the patients with GC, which might regulate the tumor microenvironment. FUT11 affected GC development by participating in cancer pathways such as PI3K-AKT, neuroactive ligand-receptor, and MAPK. Immunohistochemical staining revealed that FUT11 was highly expressed in GC. CONCLUSIONS This study revealed that FUT11 expression is significantly increased in GC tissues. This increase is associated with poor prognosis and might affect immune regulation. FUT11 might have immunological and targeted therapeutic value, providing a new approach to GC treatment.
Collapse
Affiliation(s)
- Yanqing Huang
- The First Clinical Medical College, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Gastrointestine & Gland Surgery Division I, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xiaoying Yang
- The First Clinical Medical College, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Gastrointestine & Gland Surgery Division I, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Mengda Wei
- The First Clinical Medical College, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Gastrointestine & Gland Surgery Division I, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xi Yang
- The First Clinical Medical College, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Gastrointestine & Gland Surgery Division I, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Zhenmin Yuan
- The First Clinical Medical College, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Gastrointestine & Gland Surgery Division I, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Junjie Huang
- The First Clinical Medical College, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Gastrointestine & Gland Surgery Division I, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Junren Wei
- The First Clinical Medical College, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Gastrointestine & Gland Surgery Division I, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Lei Tian
- The First Clinical Medical College, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Gastrointestine & Gland Surgery Division I, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
29
|
Zhao Z, Dai E, Jin B, Deng P, Salehebieke Z, Han B, Wu R, Yu Z, Ren J. A prognostic nomogram to predict the cancer-specific survival of patients with initially diagnosed metastatic gastric cancer: a validation study in a Chinese cohort. Clin Transl Oncol 2024:10.1007/s12094-024-03576-4. [PMID: 38918302 DOI: 10.1007/s12094-024-03576-4] [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: 05/09/2024] [Accepted: 06/15/2024] [Indexed: 06/27/2024]
Abstract
BACKGROUND Few studies have been designed to predict the survival of Chinese patients initially diagnosed with metastatic gastric cancer (mGC). Therefore, the objective of this study was to construct and validate a new nomogram model to predict cancer-specific survival (CSS) in Chinese patients. METHODS We collected 328 patients with mGC from Northern Jiangsu People's Hospital as the training cohort and 60 patients from Xinyuan County People's Hospital as the external validation cohort. Multivariate Cox regression was used to identify risk factors, and a nomogram was created to predict CSS. The predictive performance of the nomogram was evaluated using the consistency index (C-index), the calibration curve, and the decision curve analysis (DCA) in the training cohort and the validation cohort. RESULTS Multivariate Cox regression identified differentiation grade (P < 0.001), T-stage (P < 0.05), N-stage (P < 0.001), surgery (P < 0.05), and chemotherapy (P < 0.001) as independent predictors of CSS. Nomogram of chemotherapy regimens and cycles was also designed by us for the prediction of mGC. Thus, these factors are integrated into the nomogram model: the C-index value was 0.72 (95% CI 0.70-0.85) for the nomogram model and 0.82 (95% CI 0.79-0.89) and 0.73 (95% CI 0.70-0.86) for the internal and external validation cohorts, respectively. Calibration curves and DCA also demonstrated adequate fit and ideal net benefit in prediction and clinical applications. CONCLUSIONS We established a practical nomogram to predict CSS in Chinese patients initially diagnosed with mGC. Nomograms can be used to individualize survival predictions and guide clinicians in making therapeutic decisions.
Collapse
Affiliation(s)
- Ziming Zhao
- Department of General Surgery, Northern Jiangsu People's Hospital, Clinical Medical School, Yangzhou University, Yangzhou, People's Republic of China
- Department of General Surgery, General Surgery Institute of Yangzhou, Northern Jiangsu People's Hospital, Yangzhou, People's Republic of China
| | - Erxun Dai
- Department of Oncology, Northern Jiangsu People's Hospital, Clinical Medical School, Yangzhou University, Yangzhou, People's Republic of China
| | - Bao Jin
- Department of General Surgery, Xinyuan County People's Hospital, Ili Kazak Autonomous Prefecture, People's Republic of China
| | - Ping Deng
- Department of General Surgery, Xinyuan County People's Hospital, Ili Kazak Autonomous Prefecture, People's Republic of China
| | - Zulihaer Salehebieke
- Department of General Surgery, Xinyuan County People's Hospital, Ili Kazak Autonomous Prefecture, People's Republic of China
| | - Bin Han
- Department of General Surgery, Xinyuan County People's Hospital, Ili Kazak Autonomous Prefecture, People's Republic of China
| | - Rongfan Wu
- Department of General Surgery, Northern Jiangsu People's Hospital, Clinical Medical School, Yangzhou University, Yangzhou, People's Republic of China
- Department of General Surgery, General Surgery Institute of Yangzhou, Northern Jiangsu People's Hospital, Yangzhou, People's Republic of China
| | - Zhaowu Yu
- Department of General Surgery, Xinyuan County People's Hospital, Ili Kazak Autonomous Prefecture, People's Republic of China.
| | - Jun Ren
- Department of General Surgery, Northern Jiangsu People's Hospital, Clinical Medical School, Yangzhou University, Yangzhou, People's Republic of China.
- Department of General Surgery, Xinyuan County People's Hospital, Ili Kazak Autonomous Prefecture, People's Republic of China.
- Department of General Surgery, General Surgery Institute of Yangzhou, Northern Jiangsu People's Hospital, Yangzhou, People's Republic of China.
- Department of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, People's Republic of China.
| |
Collapse
|
30
|
Xu Y, Jin H, Chen Y, Yang Z, Xu D, Zhang X, Yang J, Wang Y. Comprehensive analysis of the expression, prognostic, and immune infiltration for COL4s in stomach adenocarcinoma. BMC Med Genomics 2024; 17:168. [PMID: 38907304 PMCID: PMC11191235 DOI: 10.1186/s12920-024-01934-3] [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: 06/08/2023] [Accepted: 06/14/2024] [Indexed: 06/23/2024] Open
Abstract
BACKGROUND Collagen (COL) genes, play a key role in tumor invasion and metastasis, are involved in tumor extracellular matrix (ECM)-receptor interactions and focal adhesion pathways. However, studies focusing on the diagnostic value of the COL4 family in stomach adenocarcinoma (STAD) are currently lacking. METHODS The TCGA database was employed to retrieve the clinical features and RNA sequencing expression profiles of patients with STAD. We conducted an investigation to examine the expression disparities between STAD and adjacent normal tissues. Kaplan-Meier survival analysis was utilized to assess their prognostic significance, while Spearman correlation analysis was employed to determine their association with immune checkpoint genes and immunomodulatory molecules. Furthermore, GO and KEGG analyses were performed on the COL4s-related genes, revealing potential biological pathways through gene set enrichment analysis (GSEA). Subsequently, we explored the extent of immune infiltration of the COL4 family in STAD using the TIMER database. Lastly, the expression levels of the COL4 family in STAD were further validated through quantitative PCR (qPCR) and western blot techniques. RESULTS The expression levels of COL4A1/2 were significantly upregulated, while COL4A5/6 were conspicuously downregulated in STAD. The survival analysis revealed that the upregulated COL4s indicated poorer overall survival, first progression and post-progression survival outcomes. Additionally, our findings demonstrated a positive correlation between the expressions of COL4A1/2/3/4 and the infiltration of immune cells, including CD8 + T cells, dendritic cells, macrophages, neutrophils and CD4 + T cells. Further correlation analysis uncovered a favorable association between the expression of COL4A1/2/3/4 and various crucial immunomodulatory molecules, immunological checkpoint molecules, and chemokines. Quantitative PCR analysis confirmed that the expression patterns of COL4A1/3/4/6 genes aligned with the finding from the TCGA database. However, gastric cancer cells exhibited downregulation of COL4A2. Consistently, the protein level of COL4A1 was elevated, whereas the protein level of COL4A2 was reduced in the gastric cancer cell lines. CONCLUSION COL4s could potentially serve as biomarkers for diagnosing and predicting the prognosis of STAD.
Collapse
Affiliation(s)
- Ying Xu
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, China
- Hangzhou Institute of Digestive Diseases, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| | - Hangbin Jin
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, China
- Hangzhou Institute of Digestive Diseases, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yan Chen
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, China
- Hangzhou Institute of Digestive Diseases, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| | - Zhen Yang
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Dongchao Xu
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, China
- Hangzhou Institute of Digestive Diseases, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| | - Xiaofeng Zhang
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, China.
- Hangzhou Institute of Digestive Diseases, Hangzhou, China.
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, China.
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou, Zhejiang, China.
| | - Jianfeng Yang
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, China.
- Hangzhou Institute of Digestive Diseases, Hangzhou, China.
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, China.
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou, Zhejiang, China.
| | - Yu Wang
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, China.
- Hangzhou Institute of Digestive Diseases, Hangzhou, China.
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
31
|
Duan Y, Li J, Zhou S, Bi F. Effectiveness of PD-1 inhibitor-based first-line therapy in Chinese patients with metastatic gastric cancer: a retrospective real-world study. Front Immunol 2024; 15:1370860. [PMID: 38933261 PMCID: PMC11199409 DOI: 10.3389/fimmu.2024.1370860] [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: 01/15/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
Objective Programmed cell death protein-1 (PD-1) inhibitor-based therapy has demonstrated promising results in metastatic gastric cancer (MGC). However, the previous researches are mostly clinical trials and have reached various conclusions. Our objective is to investigate the efficacy of PD-1 inhibitor-based treatment as first-line therapy for MGC, utilizing real-world data from China, and further analyze predictive biomarkers for efficacy. Methods This retrospective study comprised 105 patients diagnosed with MGC who underwent various PD-1 inhibitor-based treatments as first-line therapy at West China Hospital of Sichuan University from January 2018 to December 2022. Patient characteristics, treatment regimens, and tumor responses were extracted. We also conducted univariate and multivariate analyses to assess the relationship between clinical features and treatment outcomes. Additionally, we evaluated the predictive efficacy of several commonly used biomarkers for PD-1 inhibitor treatments. Results Overall, after 28.0 months of follow-up among the 105 patients included in our study, the objective response rate (ORR) was 30.5%, and the disease control rate (DCR) was 89.5% post-treatment, with two individuals (1.9%) achieving complete response (CR). The median progression-free survival (mPFS) was 9.0 months, and the median overall survival (mOS) was 22.0 months. According to both univariate and multivariate analyses, favorable OS was associated with patients having Eastern Cooperative Oncology Group performance status (ECOG PS) of 0-1. Additionally, normal baseline levels of carcinoembryonic antigen (CEA), as well as the combination of PD-1 inhibitors with chemotherapy and trastuzumab in patients with human epidermal growth factor receptor 2 (HER2)-positive MGC, independently predicted longer PFS and OS. However, microsatellite instability/mismatch repair (MSI/MMR) status and Epstein-Barr virus (EBV) infection status were not significantly correlated with PFS or OS extension. Conclusion As the first-line treatment, PD-1 inhibitors, either as monotherapy or in combination therapy, are promising to prolong survival for patients with metastatic gastric cancer. Additionally, baseline level of CEA is a potential predictive biomarker for identifying patients mostly responsive to PD-1 inhibitors.
Collapse
Affiliation(s)
| | | | | | - 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, Sichuan, China
| |
Collapse
|
32
|
Maratta MG, Vitale A, Basso M, Vivolo R, Di Monte E, Biondi A, Di Giorgio A, Rosa F, Tondolo V, Agnes A, Tortora G, Strippoli A, Pozzo C. Benefit of a multimodal approach combining chemotherapy and surgery in oligometastatic gastric cancer: experience from a tertiary referral center. Front Oncol 2024; 14:1343596. [PMID: 38912067 PMCID: PMC11190071 DOI: 10.3389/fonc.2024.1343596] [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: 11/23/2023] [Accepted: 05/17/2024] [Indexed: 06/25/2024] Open
Abstract
Introduction Gastric cancer (GC) is the fourth leading cause of cancer-related death worldwide with limited therapeutic options. The aim of this study was to analyze the value of adding surgery to the first-line treatment in patients with oligometastatic GC (OGC). Methods This retrospective study included patients with OGC who underwent induction chemotherapy followed by surgery of both primary tumor and synchronous metastasis between April 2012 and April 2022. Endpoints were overall survival (OS) and relapse-free survival (RFS) analyzed by the Kaplan-Meier method. Prognostic factors were assessed with the Cox model. Results Data from 39 patients were collected. All cases were referred to our multidisciplinary tumor board (MTB) to evaluate the feasibility of radical surgery. After a median follow-up of 33.6 months (mo.), median OS was 26.6 mo. (95% CI 23.8-29.4) and median RFS was 10.6 mo. (95% CI 6.3-14.8). Pathologic response according to the Mandard criteria (TRG 1-3, not reached versus 20.5 mo. for TRG 4-5; HR 0.23, p=0.019), PS ECOG ≤ 1 (26.7 mo. for PS ≤ 1 versus 11.2 mo. for PS >1; HR 0.3, p=0.022) and a low metastatic burden (26.7 mo. for single site versus 12.9 mo. for ≥2 sites; HR 0.34, p=0.039) were related to good prognosis. No major intraoperative complications nor surgery-related deaths occurred in our series. Discussion A sequential strategy of preoperative chemotherapy and radical surgical excision of both primary tumor and metastases was demonstrated to significantly improve OS and RFS. Multidisciplinary evaluation is mandatory to identify patients who could benefit from this strategy.
Collapse
Affiliation(s)
- Maria Grazia Maratta
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonio Vitale
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Michele Basso
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Raffaella Vivolo
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Elena Di Monte
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alberto Biondi
- Department of Surgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Andrea Di Giorgio
- Department of Surgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Fausto Rosa
- Department of Surgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Vincenzo Tondolo
- Department of Surgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Annamaria Agnes
- Department of Surgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giampaolo Tortora
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonia Strippoli
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Carmelo Pozzo
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
33
|
de Jongh C, van der Meulen MP, Gertsen EC, Brenkman HJF, van Sandick JW, van Berge Henegouwen MI, Gisbertz SS, Luyer MDP, Nieuwenhuijzen GAP, van Lanschot JJB, Lagarde SM, Wijnhoven BPL, de Steur WO, Hartgrink HH, Stoot JHMB, Hulsewe KWE, Spillenaar Bilgen EJ, van Det MJ, Kouwenhoven EA, Daams F, van der Peet DL, van Grieken NCT, Heisterkamp J, van Etten B, van den Berg JW, Pierie JP, Eker HH, Thijssen AY, Belt EJT, van Duijvendijk P, Wassenaar E, Wevers KP, Hol L, Wessels FJ, Haj Mohammad N, Frederix GWJ, van Hillegersberg R, Siersema PD, Vegt E, Ruurda JP. Impact of 18FFDG-PET/CT and Laparoscopy in Staging of Locally Advanced Gastric Cancer: A Cost Analysis in the Prospective Multicenter PLASTIC-Study. Ann Surg Oncol 2024; 31:4005-4017. [PMID: 38526832 PMCID: PMC11076388 DOI: 10.1245/s10434-024-15103-4] [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: 11/15/2023] [Accepted: 02/12/2024] [Indexed: 03/27/2024]
Abstract
BACKGROUND Unnecessary D2-gastrectomy and associated costs can be prevented after detecting non-curable gastric cancer, but impact of staging on treatment costs is unclear. This study determined the cost impact of 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18FFDG-PET/CT) and staging laparoscopy (SL) in gastric cancer staging. MATERIALS AND METHODS In this cost analysis, four staging strategies were modeled in a decision tree: (1) 18FFDG-PET/CT first, then SL, (2) SL only, (3) 18FFDG-PET/CT only, and (4) neither SL nor 18FFDG-PET/CT. Costs were assessed on the basis of the prospective PLASTIC-study, which evaluated adding 18FFDG-PET/CT and SL to staging advanced gastric cancer (cT3-4 and/or cN+) in 18 Dutch hospitals. The Dutch Healthcare Authority provided 18FFDG-PET/CT unit costs. SL unit costs were calculated bottom-up. Gastrectomy-associated costs were collected with hospital claim data until 30 days postoperatively. Uncertainty was assessed in a probabilistic sensitivity analysis (1000 iterations). RESULTS 18FFDG-PET/CT costs were €1104 including biopsy/cytology. Bottom-up calculations totaled €1537 per SL. D2-gastrectomy costs were €19,308. Total costs per patient were €18,137 for strategy 1, €17,079 for strategy 2, and €19,805 for strategy 3. If all patients undergo gastrectomy, total costs were €18,959 per patient (strategy 4). Performing SL only reduced costs by €1880 per patient. Adding 18FFDG-PET/CT to SL increased costs by €1058 per patient; IQR €870-1253 in the sensitivity analysis. CONCLUSIONS For advanced gastric cancer, performing SL resulted in substantial cost savings by reducing unnecessary gastrectomies. In contrast, routine 18FFDG-PET/CT increased costs without substantially reducing unnecessary gastrectomies, and is not recommended due to limited impact with major costs. TRIAL REGISTRATION NCT03208621. This trial was registered prospectively on 30-06-2017.
Collapse
Affiliation(s)
- Cas de Jongh
- Department of Surgery, Medical Oncology and Radiology, University Medical Center (UMC) Utrecht, Utrecht, The Netherlands
| | | | - Emma C Gertsen
- Department of Surgery, Medical Oncology and Radiology, University Medical Center (UMC) Utrecht, Utrecht, The Netherlands
| | - Hylke J F Brenkman
- Department of Surgery, Medical Oncology and Radiology, University Medical Center (UMC) Utrecht, Utrecht, The Netherlands
| | - Johanna W van Sandick
- Surgery and Nuclear Medicine Department, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Mark I van Berge Henegouwen
- Surgery Department, Amsterdam UMC, Location University of Amsterdam, Amsterdam, The Netherlands
- Surgery and Pathology Department, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Suzanne S Gisbertz
- Surgery Department, Amsterdam UMC, Location University of Amsterdam, Amsterdam, The Netherlands
- Surgery and Pathology Department, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Misha D P Luyer
- Surgery Department, Catharina Hospital Eindhoven, Eindhoven, The Netherlands
| | | | - Jan J B van Lanschot
- Surgery and Nuclear Medicine Department, Erasmus Medical Center UMC Rotterdam, Rotterdam, The Netherlands
| | - Sjoerd M Lagarde
- Surgery and Nuclear Medicine Department, Erasmus Medical Center UMC Rotterdam, Rotterdam, The Netherlands
| | - Bas P L Wijnhoven
- Surgery and Nuclear Medicine Department, Erasmus Medical Center UMC Rotterdam, Rotterdam, The Netherlands
| | | | | | - Jan H M B Stoot
- Surgery Department, Zuyderland MC, Sittard-Geleen, The Netherlands
| | | | | | - Marc J van Det
- Surgery Department, ZGT Hospital, Almelo, The Netherlands
| | | | - Freek Daams
- Surgery and Pathology Department, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Surgery and Pathology Department, Location Vrije University, Amsterdam UMC, Amsterdam, The Netherlands
| | - Donald L van der Peet
- Surgery and Pathology Department, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Surgery and Pathology Department, Location Vrije University, Amsterdam UMC, Amsterdam, The Netherlands
| | - Nicole C T van Grieken
- Surgery and Pathology Department, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Surgery and Pathology Department, Location Vrije University, Amsterdam UMC, Amsterdam, The Netherlands
| | - Joos Heisterkamp
- Surgery Department, Elisabeth Twee-Steden Hospital, Tilburg, The Netherlands
| | | | | | - Jean-Pierre Pierie
- Surgery Department, Medical Center Leeuwarden, Leeuwarden, The Netherlands
| | - Hasan H Eker
- Surgery Department, Medical Center Leeuwarden, Leeuwarden, The Netherlands
| | - Annemieke Y Thijssen
- Gastroenterology Department, Albert Schweitzer Hospital, Dordrecht, The Netherlands
| | - Eric J T Belt
- Gastroenterology Department, Albert Schweitzer Hospital, Dordrecht, The Netherlands
| | | | - Eelco Wassenaar
- Surgery Department, Gelre Hospitals, Apeldoorn, The Netherlands
| | - Kevin P Wevers
- Surgery Department, Isala Hospital, Zwolle, The Netherlands
| | - Lieke Hol
- Gastroenterology Department, Maasstad Hospital, Rotterdam, The Netherlands
| | - Frank J Wessels
- Department of Surgery, Medical Oncology and Radiology, University Medical Center (UMC) Utrecht, Utrecht, The Netherlands
| | - Nadia Haj Mohammad
- Department of Surgery, Medical Oncology and Radiology, University Medical Center (UMC) Utrecht, Utrecht, The Netherlands
| | - Geert W J Frederix
- Julius Center for Health Sciences and Primary Care, UMC Utrecht, Utrecht, The Netherlands
| | - Richard van Hillegersberg
- Department of Surgery, Medical Oncology and Radiology, University Medical Center (UMC) Utrecht, Utrecht, The Netherlands
| | - Peter D Siersema
- Gastroenterology and Hepatology Department, Erasmus MC - University Medical Center, Rotterdam, Rotterdam, The Netherlands
| | - Erik Vegt
- Surgery and Nuclear Medicine Department, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
- Surgery and Nuclear Medicine Department, Erasmus Medical Center UMC Rotterdam, Rotterdam, The Netherlands
| | - Jelle P Ruurda
- Department of Surgery, Medical Oncology and Radiology, University Medical Center (UMC) Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
34
|
Niu C, Ren D, Liu BL. Editorial: Novel therapeutic targets of gastric carcinogenesis: from basic research to drug development and clinical application. Front Oncol 2024; 14:1431520. [PMID: 38854722 PMCID: PMC11159047 DOI: 10.3389/fonc.2024.1431520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 05/15/2024] [Indexed: 06/11/2024] Open
Affiliation(s)
- Chenchen Niu
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, United States
| | - Dong Ren
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, United States
| | - Bella Lingjia Liu
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
35
|
Wang J, Lin J, Wang R, Tong T, Zhao Y. Immunotherapy combined with apatinib in the treatment of advanced or metastatic gastric/gastroesophageal tumors: a systematic review and meta-analysis. BMC Cancer 2024; 24:603. [PMID: 38760737 PMCID: PMC11102247 DOI: 10.1186/s12885-024-12340-4] [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: 02/28/2024] [Accepted: 05/06/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND Immunotherapy or apatinib alone has been used as third-line adjuvant therapy for advanced or metastatic gastric/gastroesophageal junction (G/GEJ) tumors, but the efficacy of combining them with each other for the treatment of patients with advanced or metastatic G/GEJ is unknown; therefore, we further evaluated the efficacy and safety of immunotherapy combined with apatinib in patients with advanced or metastatic G/GEJ. METHODS The main search was conducted on published databases: Embase, Cochrane library, PubMed.The search was conducted from the establishment of the database to December 2023.Clinical trials with patients with advanced or metastatic G/GEJ and immunotherapy combined with apatinib as the study variable were collected. Review Manager 5.4 software as well as stata 15.0 software were used for meta-analysis. RESULTS A total of 651 patients from 19 articles were included in this meta-analysis. In the included studies, immunotherapy combined with apatinib had a complete response (CR) of 0.03 (95% CI: 0.00 -0.06), partial response (PR) of 0.34 (95% CI: 0.19-0.49), stable disease (SD) of 0.43 (95% CI: 0.32-0.55), objective response rate (ORR) was 0.36 (95% CI: 0.23-0.48), disease control rate (DCR) was 0.80 (95% CI: 0.74-0.86), and median progression-free survival (PFS) was 4.29 (95% CI: 4.05-4.52), median Overall survival (OS) was 8.79 (95% CI: 7.92-9.66), and the incidence of grade ≥ 3 TRAEs was 0.34 (95% CI: 0:19-0.49). PR, ORR, DCR, median PFS and median OS were significantly higher in the immunotherapy and apatinib combination chemotherapy group (IAC) than in the immunotherapy combination apatinib group (IA). And the difference was not significant in the incidence of SD and grade ≥ 3 TRAEs. CONCLUSION This meta-analysis shows that immunotherapy combined with apatinib is safe and effective in the treatment of advanced or metastatic G/GEJ, where IAC can be a recommended adjuvant treatment option for patients with advanced or metastatic G/GEJ. However, more large multicenter randomized studies are urgently needed to reveal the long-term outcomes of immunotherapy combined with apatinib treatment.
Collapse
Affiliation(s)
- Jincheng Wang
- Department of Thoracic Surgery, the Second Hospital of Jilin University, Changchun City, China
| | - Jie Lin
- Department of Hepatobiliary and Pancreatic Surgery, the Second Hospital of Jilin University, Changchun City, 130000, Jilin, China
| | - Ruimin Wang
- Department of Operating Room, The Second Hospital of Jilin University, Changchun City, 130041, Jilin, China
| | - Ti Tong
- Department of Thoracic Surgery, the Second Hospital of Jilin University, Changchun City, China
| | - Yinghao Zhao
- Department of Thoracic Surgery, the Second Hospital of Jilin University, Changchun City, China.
| |
Collapse
|
36
|
Liu Y, Chen W, Ruan R, Zhang Z, Wang Z, Guan T, Lin Q, Tang W, Deng J, Wang Z, Li G. Deep learning based digital pathology for predicting treatment response to first-line PD-1 blockade in advanced gastric cancer. J Transl Med 2024; 22:438. [PMID: 38720336 PMCID: PMC11077733 DOI: 10.1186/s12967-024-05262-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Advanced unresectable gastric cancer (GC) patients were previously treated with chemotherapy alone as the first-line therapy. However, with the Food and Drug Administration's (FDA) 2022 approval of programmed cell death protein 1 (PD-1) inhibitor combined with chemotherapy as the first-li ne treatment for advanced unresectable GC, patients have significantly benefited. However, the significant costs and potential adverse effects necessitate precise patient selection. In recent years, the advent of deep learning (DL) has revolutionized the medical field, particularly in predicting tumor treatment responses. Our study utilizes DL to analyze pathological images, aiming to predict first-line PD-1 combined chemotherapy response for advanced-stage GC. METHODS In this multicenter retrospective analysis, Hematoxylin and Eosin (H&E)-stained slides were collected from advanced GC patients across four medical centers. Treatment response was evaluated according to iRECIST 1.1 criteria after a comprehensive first-line PD-1 immunotherapy combined with chemotherapy. Three DL models were employed in an ensemble approach to create the immune checkpoint inhibitors Response Score (ICIsRS) as a novel histopathological biomarker derived from Whole Slide Images (WSIs). RESULTS Analyzing 148,181 patches from 313 WSIs of 264 advanced GC patients, the ensemble model exhibited superior predictive accuracy, leading to the creation of ICIsNet. The model demonstrated robust performance across four testing datasets, achieving AUC values of 0.92, 0.95, 0.96, and 1 respectively. The boxplot, constructed from the ICIsRS, reveals statistically significant disparities between the well response and poor response (all p-values < = 0.001). CONCLUSION ICIsRS, a DL-derived biomarker from WSIs, effectively predicts advanced GC patients' responses to PD-1 combined chemotherapy, offering a novel approach for personalized treatment planning and allowing for more individualized and potentially effective treatment strategies based on a patient's unique response situations.
Collapse
Affiliation(s)
- Yifan Liu
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun Yat-sen University, Zhongshan 2nd Street, No. 58, Guangzhou, 510080, 86, Guangdong, China
| | - Wei Chen
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Ruiwen Ruan
- Department of Oncology, First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Zhimei Zhang
- Department of Pathology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhixiong Wang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun Yat-sen University, Zhongshan 2nd Street, No. 58, Guangzhou, 510080, 86, Guangdong, China
| | - Tianpei Guan
- Department of Gastrointestinal Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Qi Lin
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun Yat-sen University, Zhongshan 2nd Street, No. 58, Guangzhou, 510080, 86, Guangdong, China
| | - Wei Tang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun Yat-sen University, Zhongshan 2nd Street, No. 58, Guangzhou, 510080, 86, Guangdong, China
| | - Jun Deng
- Department of Oncology, First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China.
| | - Zhao Wang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun Yat-sen University, Zhongshan 2nd Street, No. 58, Guangzhou, 510080, 86, Guangdong, China.
| | - Guanghua Li
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun Yat-sen University, Zhongshan 2nd Street, No. 58, Guangzhou, 510080, 86, Guangdong, China.
| |
Collapse
|
37
|
Wainberg ZA, Kang YK, Lee KW, Qin S, Yamaguchi K, Kim IH, Saeed A, Oh SC, Li J, Turk HM, Teixeira A, Hitre E, Udrea AA, Cardellino GG, Sanchez RG, Zahlten-Kümeli A, Taylor K, Enzinger PC. Bemarituzumab as first-line treatment for locally advanced or metastatic gastric/gastroesophageal junction adenocarcinoma: final analysis of the randomized phase 2 FIGHT trial. Gastric Cancer 2024; 27:558-570. [PMID: 38308771 PMCID: PMC11016503 DOI: 10.1007/s10120-024-01466-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 01/04/2024] [Indexed: 02/05/2024]
Abstract
BACKGROUND We report the final results of the randomized phase 2 FIGHT trial that evaluated bemarituzumab, a humanized monoclonal antibody selective for fibroblast growth factor receptor 2b (FGFR2b), plus mFOLFOX6 in patients with FGFR2b-positive (2 + /3 + membranous staining by immunohistochemistry), HER-2-negative gastric or gastroesophageal junction cancer (GC). METHODS Patients received bemarituzumab (15 mg/kg) or placebo once every 2 weeks with an additional bemarituzumab (7.5 mg/kg) or placebo dose on cycle 1 day 8. All patients received mFOLFOX6. The primary endpoint was investigator-assessed progression-free survival (PFS). Secondary endpoints included overall survival (OS), objective response rate, and safety. Efficacy was evaluated after a minimum follow-up of 24 months. RESULTS In the bemarituzumab-mFOLFOX6 (N = 77) and placebo-mFOLFOX6 (N = 78) arms, respectively, 59.7% and 66.7% of patients were FGFR2b-positive in ≥ 10% of tumor cells. The median PFS (95% confidence interval [CI]) was 9.5 months (7.3-13.7) with bemarituzumab-mFOLFOX6 and 7.4 months (5.7-8.4) with placebo-mFOLFOX6 (hazard ratio [HR], 0.72; 95% CI 0.49-1.08); median OS (95% CI) was 19.2 (13.6-24.2) and 13.5 (9.3-15.9) months, respectively (HR 0.77; 95% CI 0.52-1.14). Observed efficacy in FGFR2b-positive GC in ≥ 10% of tumor cells was: PFS: HR 0.43 (95% CI 0.26-0.73); OS: HR 0.52 (95% CI 0.31-0.85). No new safety findings were reported. CONCLUSIONS In FGFR2b-positive advanced GC, the combination of bemarituzumab-mFOLFOX6 led to numerically longer median PFS and OS compared with mFOLFOX6 alone. Efficacy was more pronounced with FGFR2b overexpression in ≥ 10% of tumor cells. Confirmatory phase 3 trials are ongoing (NCT05052801, NCT05111626). CLINICAL TRIAL REGISTRATION NCT03694522.
Collapse
Affiliation(s)
- Zev A Wainberg
- Division of Hematology-Oncology, Department of Medicine, University of California Los Angeles Medical Center, David Geffen School of Medicine, 2825 Santa Monica Blvd., Suite 200, Santa Monica, Los Angeles, CA, 90404-2429, USA.
| | - Yoon-Koo Kang
- Asan Medical Centre, University of Ulsan College of Medicine, Seoul, South Korea
| | - Keun-Wook Lee
- Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Shukui Qin
- Nanjing Tianyinshan Hospital, The 1st Affiliated Hospital of China Pharmaceutical University, Nanjing, China
| | - Kensei Yamaguchi
- Gastroenterological Chemotherapy Department, The Cancer Institute Hospital of JFCR, Tokyo, Japan
| | - In-Ho Kim
- Department of Oncology, The Catholic University of Korea, Seoul St Mary's Hospital, Seoul, South Korea
| | - Anwaar Saeed
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sang Cheul Oh
- Department of Internal Medicine, Korea University Guro Hospital, Seoul, South Korea
| | - Jin Li
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haci Mehmet Turk
- Department of Medical Oncology, Bezmialem Vakif University, Istanbul, Turkey
| | - Alexandra Teixeira
- Gastroenterology Division, Hospital da Senhora da Oliveira, Guimarães, Portugal
| | - Erika Hitre
- Department of Medical Oncology and Clinical Pharmacology "B", National Institute of Oncology, Budapest, Hungary
| | - Adrian A Udrea
- Medical Oncology, Medisprof Cancer Center, Cluj-Napoca, Romania
| | | | | | | | | | - Peter C Enzinger
- Department of Medicine, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
38
|
Sun B, Zheng X, Zhang X, Zhang H, Jiang Y. Oxaliplatin-Loaded Mil-100(Fe) for Chemotherapy-Ferroptosis Combined Therapy for Gastric Cancer. ACS OMEGA 2024; 9:16676-16686. [PMID: 38617668 PMCID: PMC11007804 DOI: 10.1021/acsomega.4c00658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 02/26/2024] [Accepted: 03/19/2024] [Indexed: 04/16/2024]
Abstract
Oxaliplatin (Oxa) is a commonly used chemotherapy drug in the treatment of gastric cancer, but its toxic side effects and drug resistance after long-term use have seriously limited its efficacy. Loading chemotherapy drugs with nanomaterials and delivering them to the tumor site are common ways to overcome the above problems. However, nanomaterials as carriers do not have therapeutic functions on their own, and the effect of single chemotherapy is relatively limited, so there is still room for progress in related research. Herein, we construct Oxa@Mil-100(Fe) nanocomposites by loading Oxa with a metal-organic framework (MOF) Mil-100(Fe) with high biocompatibility and a large specific surface area. The pore structure of Mil-100(Fe) is conducive to a large amount of Oxa loading with a drug-loading rate of up to 27.2%. Oxa@Mil-100(Fe) is responsive to the tumor microenvironment (TME) and can release Oxa and Fe3+ under external stimulation. On the one hand, Oxa can inhibit the synthesis of DNA and induce the apoptosis of gastric cancer cells. On the other hand, Fe3+ can clear overexpressed glutathione (GSH) in TME and be reduced to Fe2+, inhibiting the activity of glutathione peroxidase 4 (GPX4), leading to the accumulation of intracellular lipid peroxides (LPO), and at the same time releasing a large number of reactive oxygen species (ROS) through the Fenton reaction, inducing ferroptosis in gastric cancer cells. With the combination of apoptosis and ferroptosis, Oxa@Mil-100(Fe) shows a good therapeutic effect, and the killing effect on gastric cancer cells is obvious. In a nude mouse model of subcutaneous tumor transplantation, Oxa@Mil-100(Fe) shows a significant inhibitory effect on tumor growth, with an inhibition rate of nearly 60%. In addition to its excellent antitumor activity, Oxa@Mil-100(Fe) has no obvious toxic or side effects. This study provides a new idea and method for the combined treatment of gastric cancer.
Collapse
Affiliation(s)
- Boyao Sun
- Department
of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130031, P. R. China
| | - Xuewei Zheng
- Department
of Radiology, China-Japan Union Hospital
of Jilin University, Changchun 130031, P. R. China
| | - Xiaoyu Zhang
- Department
of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130031, P. R. China
| | - Huaiyu Zhang
- Department
of Rehabilitation Medicine, China-Japan
Union Hospital of Jilin University, Changchun 130031, P. R. China
| | - Yang Jiang
- Department
of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130031, P. R. China
| |
Collapse
|
39
|
Reese M, Eichelmann AK, Nowacki TM, Pascher A, Sporn JC. The role of cytoreductive surgery and HIPEC for the treatment of primary and secondary peritoneal malignancies-experience from a tertiary care center in Germany. Langenbecks Arch Surg 2024; 409:113. [PMID: 38589714 DOI: 10.1007/s00423-024-03309-9] [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: 12/28/2023] [Accepted: 04/03/2024] [Indexed: 04/10/2024]
Abstract
PURPOSE Peritoneal surface malignancies (PSM) are commonly known to have a dismal prognosis. Over the past decades, novel techniques such as cytoreductive surgery (CRS), hyperthermic intraperitoneal chemotherapy (HIPEC), and pressurized intraperitoneal aerosol chemotherapy (PIPAC) have been introduced for the treatment of PSM which could improve the overall survival and quality of life of patients with PSM. The decision to proceed with CRS and HIPEC is often challenging due the complexity of the disease, the extent of the procedure, associated side effects, and potential risks. Here, we present our experience with CRS and HIPEC to add to the ongoing discussion about eligibility criteria, technical approach, and expected outcomes and contribute to the evolution of this powerful and promising tool in the multidisciplinary treatment of patients with primary and secondary PSM. METHODS A single-center retrospective chart review was conducted and included a total of 40 patients treated with CRS and HIPEC from April 2020 to September 2022 at the University Hospital Münster Department of Surgery. All patients had histologically confirmed primary or secondary peritoneal malignancies of various primary origins. RESULTS Our study included 22 patients with peritoneal metastases from gastric cancer (55%), 8 with pseudomyxoma peritonei (20%), 4 with mesothelioma of the peritoneum (10%), and 6 patients with PSM originating from other primary tumor locations. Median PCI at time of cytoreduction was 4 (0-25). Completeness of cytoreduction score was 0 in 37 patients (92.5%), 1 in two patients (5%), and 2 in one patient (2.5%). Median overall survival across all patients was 3.69 years. CONCLUSION Complete cytoreduction during CRS and HIPEC can be achieved for patients with low PCI, for patients with high PCI in low-grade malignancies, and even for patients with initially high PCI in high-grade malignancies following a significant reduction of cancer burden due to extensive preoperative treatment with PIPAC and systemic chemotherapy.
Collapse
Affiliation(s)
- Mikko Reese
- Department of General, Visceral and Transplant Surgery, University Hospital Münster, Waldeyerstraße 1, 48149, Münster, Germany
| | - Ann-Kathrin Eichelmann
- Department of General, Visceral and Transplant Surgery, University Hospital Münster, Waldeyerstraße 1, 48149, Münster, Germany
| | - Tobias M Nowacki
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clinical Infectiology, University Hospital Münster, Albert-Schweitzer-Campus 1, Münster, 48149, Germany
- Department of Gastroenterology, UKM Marienhospital Steinfurt, Mauritiusstr. 5, Steinfurt, 48565, Germany
| | - Andreas Pascher
- Department of General, Visceral and Transplant Surgery, University Hospital Münster, Waldeyerstraße 1, 48149, Münster, Germany
| | - Judith C Sporn
- Department of General, Visceral and Transplant Surgery, University Hospital Münster, Waldeyerstraße 1, 48149, Münster, Germany.
| |
Collapse
|
40
|
Chang L, Zhang X, Ma Q, Kong L, Yu Y, Tao J, Li Q. Safety and efficacy of apatinib in combination with chemotherapy with or without immunotherapy versus chemotherapy alone as first-line treatment for advanced gastric cancer. Invest New Drugs 2024; 42:161-170. [PMID: 38367168 PMCID: PMC10944401 DOI: 10.1007/s10637-024-01423-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 01/22/2024] [Indexed: 02/19/2024]
Abstract
The specific first-line regimen for advanced gastric cancer (GC) is still controversial. The benefit of apatinib for first-line treatment of advanced GC remains unknown and needs to be further explored. Eighty-two patients with advanced GC treated in our institution from October 2017 to March 2023 were retrospectively reviewed. All individuals had her-2 negative GC and had received at least two cycles of first-line treatment, including 44 patients in the combination treatment group (apatinib in combination with chemotherapy with or without immunotherapy) and 38 patients in the simple chemotherapy group. We evaluated the efficacy and safety of apatinib in combination with chemotherapy with or without immunotherapy in the first-line treatment of advanced GC by comparing the efficacy, progression-free survival (PFS), and adverse events in two groups of patients. The median PFS of the simple chemotherapy group was 9.25 months (95% confidence interval (CI), 6.1-11.2 months), and that of the combination treatment group was 10.9 months (95% CI, 7.9-15.8 months), which was 1.65 months longer than the simple chemotherapy group. Statistically significant differences are shown (P = 0.022). The objective response rate (ORR) of the combination treatment group was 65.9%, and 36.8% in the simple chemotherapy group. Statistically significant differences are shown (P = 0.014). No serious (Grade IV) adverse events occurred in either group. Our study indicates that apatinib in combination with chemotherapy with or without immunotherapy as first-line treatment for advanced GC exhibits good anti-tumor activity and is well tolerated by patients.
Collapse
Affiliation(s)
- Lele Chang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang Province 150001, China
| | - Xuemei Zhang
- Department of Radiation Oncology, Quzhou People's Hospital, Quzhou, China
| | - Qian Ma
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang Province 150001, China
| | - Lingyang Kong
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang Province 150001, China
| | - Yang Yu
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Ji Tao
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang Province 150001, China
| | - Qingwei Li
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang Province 150001, China.
| |
Collapse
|
41
|
Baeksgaard Jensen L, Yilmaz M, Nordsmark M, Möller S, Elle IC, Ladekarl M, Qvortrup C, Pfeiffer P. TRIFLURIDINE/TIPIRACIL (FTD/TPI) with or without bevacizumab in previously treated patients with esophago-gastric adenocarcinoma, a randomised phase III trial. EClinicalMedicine 2024; 70:102521. [PMID: 38495525 PMCID: PMC10940909 DOI: 10.1016/j.eclinm.2024.102521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 02/03/2024] [Accepted: 02/19/2024] [Indexed: 03/19/2024] Open
Abstract
Background Trifluridine-tipiracil has shown a survival benefit compared with placebo in patients with chemorefractory metastatic esophago-gastric adenocarcinoma. We aimed to compare the efficacy of trifluridine-tipiracil plus bevacizumab vs trifluridine-tipiracil monotherapy in pre-treated patients with metastatic esophago-gastric adenocarcinoma. Methods This investigator-initiated, open-label, randomized trial enrolled patients with metastatic esophago-gastric adenocarcinoma. The main inclusion criteria were patients with pre-treated metastatic esophago-gastric adenocarcinoma, and WHO performance status 0 or 1. Participants were randomly assigned (1:1) to receive oral trifluridine-tipiracil (35 mg/m2 twice daily on days 1-5 and 8-12 every 28 days) alone or combined with bevacizumab (5 mg/kg on days 1 and 15) until progression, unacceptable toxicity, or patient decision to withdraw. Randomisation was stratified by sex and treatment line. The primary endpoint was investigator-evaluated progression-free survival. All analyses were based on intention to treat. This trial is registered with EudraCT, 2018-004845-18. Findings From Oct 1, 2019, to Sept 30, 2021, 103 patients were enrolled and randomly assigned to trifluridine-tipiracil (n = 53) or trifluridine-tipiracil plus bevacizumab (n = 50). The clinical cut-off date was March 1st, 2023, after a median follow-up of 36.6 months. Median progression-free survival was 3.1 months (95% CI 2.0-4.3) in the trifluridine-tipiracil group vs 3.9 months (3.0-6.3) in the trifluridine-tipiracil plus bevacizumab group (hazard ratio 0.68, 95% CI 0.46-1.02; p = 0.058). The most frequent grade 3 or worse adverse event was neutropenia, observed in 26 (49%) patients in the trifluridine-tipiracil group vs 23 patients (46%) in the trifluridine-tipiracil plus bevacizumab group. At least one hospitalization was observed in 21 patients (40%) in the trifluridine-tipiracil group and 22 patients (44%) in the trifluridine-tipiracil plus bevacizumab group. No deaths were deemed treatment related. Interpretation In patients with pre-treated metastatic esophago-gastric cancer, trifluridine-tipiracil plus bevacizumab, compared to trifluridine-tipiracil monotherapy, did not significantly prolong progression-free survival. The combination of trifluridine-tipiracil with bevacizumab was well tolerated without increase in severe neutropenia and no new safety signals. Funding Servier, Roche.
Collapse
Affiliation(s)
| | - Mette Yilmaz
- Department of Oncology and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | | | - Sören Möller
- Odense Patient Data Explorative Network (OPEN), Odense University Hospital, Denmark
| | | | - Morten Ladekarl
- Department of Oncology and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | | | - Per Pfeiffer
- Department of Oncology, Odense University Hospital, Denmark
| |
Collapse
|
42
|
Ahmed S, Amir M, Adnan K, Zilong Z, Akbar A, Khan S, Jadoon SK, Saleem Khan M. A Clinical Study of Intraoperative Perfusion Chemotherapy in Gastric Cancer: A Prospective Cohort Study. Cureus 2024; 16:e58482. [PMID: 38644943 PMCID: PMC11031129 DOI: 10.7759/cureus.58482] [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] [Accepted: 04/16/2024] [Indexed: 04/23/2024] Open
Abstract
INTRODUCTION Gastric cancer (GC) is the third largest cause of cancer-related death worldwide, with major geographic disparities in incidence and outcomes. Sociodemographic indicators, food habits, and genetic predispositions all add to the load. Despite advances in systemic treatments, peritoneal metastasis remains a concern, with intraperitoneal chemotherapy (IPC) emerging as a promising treatment option. METHODS A prospective cohort research was done, with 30 GC patients receiving cytoreductive surgery (CRS) followed by lobaplatin-based intraoperative chemotherapy. The study evaluated postoperative complications, survival rates, and disease recurrence using Statistical Package for the Social Sciences (SPSS) version 25.0 (IBM SPSS Statistics, Armonk, NY) for data analysis. The purpose of this study is to assess the effectiveness, safety, and dependability of lobaplatin as an intraoperative chemotherapeutic agent in patients having gastric cancer surgery, with a particular emphasis on those patients who do not have distant metastases. RESULTS The study population had a balanced gender distribution, with an average age of 44.83 years. Most patients had advanced-stage cancer (T3 and T4), and lobaplatin treatment resulted in a low frequency of serious postoperative sequelae. Preliminary studies suggest that lobaplatin is a safe and potentially effective IPC drug for GC, with few side effects and adequate survival rates. CONCLUSION Lobaplatin shows promise as an intraoperative chemotherapeutic treatment for gastric cancer, necessitating more research in bigger, randomized controlled studies to determine its efficacy and safety profile. The study emphasizes the need for novel treatment strategies to enhance the prognosis of GC patients, particularly those with peritoneal involvement.
Collapse
Affiliation(s)
- Sohail Ahmed
- Gastrointestinal Surgery, Yangtze University, Jingzhou, CHN
| | - Muhammad Amir
- Emergency, Midland Doctors Medical Institute, Muzaffarabad, PAK
| | - Khan Adnan
- Gastrointestinal Surgery, Yangtze University, Jingzhou, CHN
| | - Zhang Zilong
- Oncology, Jingzhou Central Hospital, Jingzhou, CHN
| | - Amna Akbar
- Surgery, District Headquarter Hospital, Jhelum Valley, Muzaffarabad, PAK
| | - Sania Khan
- Oncology, Shaukat Khanum Memorial Cancer Hospital and Research Centre, Lahore, PAK
| | | | | |
Collapse
|
43
|
Bao Z, Du J, Zheng Y, Guo Q, Ji R. Deep learning or radiomics based on CT for predicting the response of gastric cancer to neoadjuvant chemotherapy: a meta-analysis and systematic review. Front Oncol 2024; 14:1363812. [PMID: 38601765 PMCID: PMC11004479 DOI: 10.3389/fonc.2024.1363812] [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: 12/31/2023] [Accepted: 03/18/2024] [Indexed: 04/12/2024] Open
Abstract
Background Artificial intelligence (AI) models, clinical models (CM), and the integrated model (IM) are utilized to evaluate the response to neoadjuvant chemotherapy (NACT) in patients diagnosed with gastric cancer. Objective The objective is to identify the diagnostic test of the AI model and to compare the accuracy of AI, CM, and IM through a comprehensive summary of head-to-head comparative studies. Methods PubMed, Web of Science, Cochrane Library, and Embase were systematically searched until September 5, 2023, to compile English language studies without regional restrictions. The quality of the included studies was evaluated using the Quality Assessment of Diagnostic Accuracy Studies-2 (QUADAS-2) criteria. Forest plots were utilized to illustrate the findings of diagnostic accuracy, while Hierarchical Summary Receiver Operating Characteristic curves were generated to estimate sensitivity (SEN) and specificity (SPE). Meta-regression was applied to analyze heterogeneity across the studies. To assess the presence of publication bias, Deeks' funnel plot and an asymmetry test were employed. Results A total of 9 studies, comprising 3313 patients, were included for the AI model, with 7 head-to-head comparative studies involving 2699 patients. Across the 9 studies, the pooled SEN for the AI model was 0.75 (95% confidence interval (CI): 0.66, 0.82), and SPE was 0.77 (95% CI: 0.69, 0.84). Meta-regression was conducted, revealing that the cut-off value, approach to predicting response, and gold standard might be sources of heterogeneity. In the head-to-head comparative studies, the pooled SEN for AI was 0.77 (95% CI: 0.69, 0.84) with SPE at 0.79 (95% CI: 0.70, 0.85). For CM, the pooled SEN was 0.67 (95% CI: 0.57, 0.77) with SPE at 0.59 (95% CI: 0.54, 0.64), while for IM, the pooled SEN was 0.83 (95% CI: 0.79, 0.86) with SPE at 0.69 (95% CI: 0.56, 0.79). Notably, there was no statistical difference, except that IM exhibited higher SEN than AI, while maintaining a similar level of SPE in pairwise comparisons. In the Receiver Operating Characteristic analysis subgroup, the CT-based Deep Learning (DL) subgroup, and the National Comprehensive Cancer Network (NCCN) guideline subgroup, the AI model exhibited higher SEN but lower SPE compared to the IM. Conversely, in the training cohort subgroup and the internal validation cohort subgroup, the AI model demonstrated lower SEN but higher SPE than the IM. The subgroup analysis underscored that factors such as the number of cohorts, cohort type, cut-off value, approach to predicting response, and choice of gold standard could impact the reliability and robustness of the results. Conclusion AI has demonstrated its viability as a tool for predicting the response of GC patients to NACT Furthermore, CT-based DL model in AI was sensitive to extract tumor features and predict the response. The results of subgroup analysis also supported the above conclusions. Large-scale rigorously designed diagnostic accuracy studies and head-to-head comparative studies are anticipated. Systematic review registration PROSPERO, CRD42022377030.
Collapse
Affiliation(s)
- Zhixian Bao
- Department of Gastroenterology, the First Hospital of Lanzhou University, Lanzhou, China
- Department of Gastroenterology, Xi’an NO.1 Hospital, Xi’an, Shaanxi, China
| | - Jie Du
- Department of Social Medicine and Health Management, School of Public Health, Lanzhou University, Lanzhou, China
| | - Ya Zheng
- Department of Gastroenterology, the First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Qinghong Guo
- Department of Gastroenterology, the First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Rui Ji
- Department of Gastroenterology, the First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
44
|
Yarahmadi G, Tavakoli Ataabadi S, Dashti Z, Dehghanian M. A review on expression and regulatory mechanisms of miR-337-3p in cancer. J Biomol Struct Dyn 2024:1-10. [PMID: 38500239 DOI: 10.1080/07391102.2024.2329294] [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: 09/04/2023] [Accepted: 03/06/2024] [Indexed: 03/20/2024]
Abstract
A group of diseases generally referred to as cancer represents a serious threat to people's health all over the world and has a significant negative influence on every aspect of the lives of patients. The development of cancer is influenced by several environmental, genetic, and epigenetic factors. MicroRNAs (miRNAs), a class of non-coding RNAs, can alter the expression of genes involved in cell proliferation, migration, metastasis, and apoptosis, lead to the pathogenesis of cancer. Additionally, several effectors modify miRNAs directly, including methylation, circular RNAs, and long non-coding RNAs (lncRNAs). In this review, we have explained the role of mir-337-3p in the pathways related to the pathogenesis of different cancers. Studying the functional role of miR-337-3p is necessary for detecting novel molecules as tumor markers and discovering novel targets for cancer treatment.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ghafour Yarahmadi
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Sadegh Tavakoli Ataabadi
- Department of Medical Genetics School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Dashti
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Genetics, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences Campus, Yazd, Iran
| | - Mehran Dehghanian
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
45
|
Ramachandran R, Grantham T, Parvataneni S, Budh D, Gollapalli S, Reddy M, Gaduputi V. Gastric Cancer: Clinical Features, Screening, Diagnosis, Treatment, and Prevention. J Community Hosp Intern Med Perspect 2024; 14:49-57. [PMID: 38966500 PMCID: PMC11221451 DOI: 10.55729/2000-9666.1304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 12/12/2023] [Accepted: 01/02/2024] [Indexed: 07/06/2024] Open
Abstract
The objective of this article is to highlight the clinical features, screening, diagnosis, treatment, and prevention of gastric cancer (GC). Early GC is often asymptomatic leading to frequent delays in diagnosis. Weight loss and persistent abdominal pain are the most common symptoms at initial diagnosis. The diagnosis of GC typically involves a combination of endoscopy, biopsy, and imaging studies. Endoscopic resection techniques are emerging as successful treatment options for early GC. Treatment options for advanced GC include surgery and chemotherapy. The first line chemotherapy for advanced GC consists of doublet therapy with a combination of platinum and fluoropyrimidines. Trastuzumab, a monoclonal antibody, is used in the treatment of human epidermal growth factor 2 positive GCs. Antiangiogenic agents and immunotherapy are also useful in the treatment of GC. Currently there are no GC screening guidelines in the United States, but they exist in other regions where there is increased prevalence of GC. Prevention strategies for GC include Helicobacter pylori eradication and adoption of a healthy diet consisting of fruits and vegetables.
Collapse
Affiliation(s)
| | - Tyler Grantham
- Department of Internal Medicine, Staten Island University Hospital, 475 Seaview Avenue, Staten Island, NY 10305,
USA
| | - Swetha Parvataneni
- Department of Internal Medicine, Geisinger Lewistown Hospital, 400 Highland Ave, Lewistown, PA 17044,
USA
| | - Deepa Budh
- Department of Internal Medicine, St. Barnabas Hospital, 4422 3rd Avenue, Bronx, NY 10457,
USA
| | - Sindhu Gollapalli
- Department of Internal Medicine, St. Barnabas Hospital, 4422 3rd Avenue, Bronx, NY 10457,
USA
| | - Madhavi Reddy
- Department of Gastroenterology, The Brooklyn Hospital Center, 121 Dekalb Avenue, Brooklyn, NY 11201,
USA
| | - Vinaya Gaduputi
- Department of Gastroenterology, Blanchard Valley Health System, 1900 S Main St, Findlay, OH 45840,
USA
| |
Collapse
|
46
|
Zhang X, Zhang Y, Zhang Q, Lu M, Chen Y, Zhang X, Zhang P. Role of AT-rich interaction domain 1A in gastric cancer immunotherapy: Preclinical and clinical perspectives. J Cell Mol Med 2024; 28:e18063. [PMID: 38041544 PMCID: PMC10902580 DOI: 10.1111/jcmm.18063] [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: 09/13/2023] [Revised: 10/31/2023] [Accepted: 11/14/2023] [Indexed: 12/03/2023] Open
Abstract
The application of immune checkpoint inhibitor (ICI) using monoclonal antibodies has brought about a profound transformation in the clinical outcomes for patients grappling with advanced gastric cancer (GC). Nonetheless, despite these achievements, the quest for effective functional biomarkers for ICI therapy remains constrained. Recent research endeavours have shed light on the critical involvement of modified epigenetic regulators in the pathogenesis of gastric tumorigenesis, thus providing a glimpse into potential biomarkers. Among these regulatory factors, AT-rich interaction domain 1A (ARID1A), a pivotal constituent of the switch/sucrose non-fermentable (SWI/SNF) complex, has emerged as a promising candidate. Investigations have unveiled the pivotal role of ARID1A in bridging the gap between genome instability and the reconfiguration of the tumour immune microenvironment, culminating in an enhanced response to ICI within the landscape of gastric cancer treatment. This all-encompassing review aims to dissect the potential of ARID1A as a valuable biomarker for immunotherapeutic approaches in gastric cancer, drawing from insights garnered from both preclinical experimentation and clinical observations.
Collapse
Affiliation(s)
- Xuemei Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Youzhi Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- School of PharmacyHubei University of Science and TechnologyXianningChina
| | - Qiaoyun Zhang
- School of PharmacyHubei University of Science and TechnologyXianningChina
| | - Mengyao Lu
- Department of Oncology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yuan Chen
- Department of Oncology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiaoyu Zhang
- Division of Gastrointestinal Surgery, Department of General Surgery, Huai'an Second People's Hospitalthe Affiliated Huai'an Hospital of Xuzhou Medical UniversityHuaianChina
| | - Peng Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
47
|
Matsunaga T, Satio H, Sakano Y, Makinoya M, Shimizu S, Shishido Y, Miyatani K, Hanaki T, Kihara K, Yamamoto M, Tokuyasu N, Takano S, Sakamoto T, Hasegawa T, Fujiwara Y. Prognostic significance of the cachexia index in patients with unresectable advanced gastric cancer receiving palliative chemotherapy: a retrospective single-center study. Surg Today 2024; 54:231-239. [PMID: 37526733 DOI: 10.1007/s00595-023-02721-w] [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: 04/21/2023] [Accepted: 06/13/2023] [Indexed: 08/02/2023]
Abstract
PURPOSE To investigate the prognostic utility of the cachexia index (CXI) in unresectable advanced gastric cancer (UAGC). METHODS The relationship between CXI and the outcomes was evaluated in 102 patients with UAGC who had received first-line palliative 5-fluorouracil-based chemotherapy between January 2012 and December 2021. RESULTS The median survival time (MST) from first-line chemotherapy initiation was 16.2 months, and the cohort included 60 and 42 patients with high and low CXIs, respectively, based on the optimal CXI cutoff. The rates of patients with a performance status score of 0, recurrence, third-line chemotherapy, and all grade 3-4 side effects, including febrile neutropenia (FN), were significantly higher in the CXIhigh group than in the CXIlow group. The prognosis based on MST was significantly better in the CXIhigh group than in the CXIlow group (22.5 vs. 11.6 months, p < 0.001). According to a multivariate analysis, a low CXI and performance status score of 1-2 were poor prognostic factors. CONCLUSIONS Patients with UAGC and a low CXI had poorer prognoses and more frequent grade 3-4 side effects, including FN, than those with a high CXI. Patients with UAGC and a low CXI should be carefully managed to control for side effects to receive subsequent treatment.
Collapse
Affiliation(s)
- Tomoyuki Matsunaga
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, 683-8504, Japan.
| | - Hiroaki Satio
- Department of Surgery, Japanese Red Cross Tottori Hospital, 117 Shotoku-Cho, Tottori, 680-8517, Japan
| | - Yu Sakano
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, 683-8504, Japan
| | - Masahiro Makinoya
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, 683-8504, Japan
| | - Shota Shimizu
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, 683-8504, Japan
| | - Yuji Shishido
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, 683-8504, Japan
| | - Kozo Miyatani
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, 683-8504, Japan
| | - Takehiko Hanaki
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, 683-8504, Japan
| | - Kyoichi Kihara
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, 683-8504, Japan
| | - Manabu Yamamoto
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, 683-8504, Japan
| | - Naruo Tokuyasu
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, 683-8504, Japan
| | - Shuichi Takano
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, 683-8504, Japan
| | - Teruhisa Sakamoto
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, 683-8504, Japan
| | - Toshimichi Hasegawa
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, 683-8504, Japan
| | - Yoshiyuki Fujiwara
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of Medicine, Tottori University Faculty of Medicine, 36-1 Nishi-Cho, Yonago, 683-8504, Japan
| |
Collapse
|
48
|
Deng GM, Song HB, Du ZZ, Xue YW, Song HJ, Li YZ. Evaluating the influence of sarcopenia and myosteatosis on clinical outcomes in gastric cancer patients undergoing immune checkpoint inhibitor. World J Gastroenterol 2024; 30:863-880. [PMID: 38516238 PMCID: PMC10950641 DOI: 10.3748/wjg.v30.i8.863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/16/2024] [Accepted: 02/01/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND The development and progression of gastric cancer (GC) are closely linked to the nutritional status of patients. Although immunotherapy has been demonstrated to be clinically effective, the relationships of sarcopenia and myosteatosis with the use of immune checkpoint inhibitors (ICIs) in patients with gastric cancer remain to be characterized. AIM To assess the effects of sarcopenia and myosteatosis on the clinical outcomes of patients with GC undergoing treatment with an ICI. METHODS We performed a retrospective study of patients who were undergoing immunotherapy for GC. For the evaluation of sarcopenia, the optimal cut-off value for the skeletal muscle index was established using receiver operating characteristic analysis of data obtained from pre-treatment computed tomography images at the L3 vertebral level. Myosteatosis was defined using the mean skeletal muscle density (SMD), with a threshold value of < 41 Hounsfield units (HU) for patients with a body mass index (BMI) < 25 kg/m² and < 33 HU for those with a BMI ≥ 25 kg/m². The log-rank test was used to compare progression-free survival (PFS) and overall survival (OS), and a Cox proportional hazard model was used to identify prognostic factors. Nomograms were developed to predict the PFS and OS of patients on the basis of the results of multivariate analyses. RESULTS We studied 115 patients who were undergoing ICI therapy for GC, of whom 27.4% had sarcopenia and 29.8% had myosteatosis. Patients with sarcopenia or myosteatosis had significantly shorter PFS and OS than those without these conditions. Furthermore, both sarcopenia and myosteatosis were found to be independent predictors of PFS and OS in patients with GC administering an ICI. The prediction models created for PFS and OS were associated with C-indexes of 0.758 and 0.781, respectively. CONCLUSION The presence of sarcopenia or myosteatosis is a reliable predictor of the clinical outcomes of patients with GC who are undergoing treatment with an ICI.
Collapse
Affiliation(s)
- Gui-Ming Deng
- Department of Gastrointestinal Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Hai-Bin Song
- Department of Gastrointestinal Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Zhong-Ze Du
- Department of Gastrointestinal Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Ying-Wei Xue
- Department of Gastrointestinal Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Hong-Jiang Song
- Department of Gastrointestinal Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Yuan-Zhou Li
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| |
Collapse
|
49
|
de Moraes FCA, Pasqualotto E, Chavez MP, Ferreira ROM, De Castria TB, Burbano RMR. Efficacy and safety of Zolbetuximab plus chemotherapy for advanced CLDN18.2-positive gastric or gastro-oesophageal adenocarcinoma: a meta-analysis of randomized clinical trials. BMC Cancer 2024; 24:240. [PMID: 38383390 PMCID: PMC10882870 DOI: 10.1186/s12885-024-11980-w] [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: 11/02/2023] [Accepted: 02/07/2024] [Indexed: 02/23/2024] Open
Abstract
BACKGROUND The benefit of adding Zolbetuximab to the treatment in patients with Claudin-18 isoform 2 (CLDN18.2)-positive, human epidermal growth factor receptor 2-negative, locally advanced unresectable or metastatic gastric or gastro-oesophageal junction adenocarcinoma (GC/GEJ) is not yet fully elucidated. METHODS We searched PubMed, Embase and Cochrane databases for randomized controlled trials (RCTs) that investigated Zolbetuximab plus chemotherapy versus chemotherapy alone for GC or GEJ adenocarcinoma. We computed hazard-ratios (HRs) or odds-ratios (ORs) for binary endpoints, with 95% confidence intervals (CIs). RESULTS Three studies and 1,233 patients were included. Comparing with Zolbetuximab plus chemotherapy versus chemotherapy alone, progression-free survival (PFS) rate (HR 0.64; 95% CI 0.49-0.84; p < 0.01) and overall survival (OS) rate (HR 0.72; 95% CI 0.62-0.83; p < 0.01) were significant in favor of the Zolbetuximab group. Regarding effectiveness, the Objective Response Rate (ORR) was (OR 1.15; 95% CI 0.87-1.53; p = 0.34). CONCLUSIONS In this comprehensive systematic review and meta-analysis of RCTs, the incorporation of Zolbetuximab alongside chemotherapy offers a promising prospect for reshaping the established treatment paradigms for patients diagnosed with advanced CLDN18.2-positive GC/GEJ cancer.
Collapse
Affiliation(s)
| | - Eric Pasqualotto
- Federal University of Santa Catarina, 88040-900, Florianópolis, Santa Catarina, Brazil
| | | | | | - Tiago Biachi De Castria
- Moffitt Cancer Center, 12902 USF Magnolia Drive, 33612, Tampa, FL, USA
- Morsani College of Medicine, University of South Florida, 12901 Bruce B. Downs Blvd., 33612, Tampa, FL, USA
| | | |
Collapse
|
50
|
Sharan S, Bansal S, Manaise HK, Jimenez PB, Raikot SR, Ahmed SH, Popp R, Popp K, Sukniam K, Kowkabany G, Mubarak F, Gabriel E. Time to treatment disparities in gastric cancer patients in the United States of America: a comprehensive retrospective analysis. Front Oncol 2024; 14:1292793. [PMID: 38406814 PMCID: PMC10889117 DOI: 10.3389/fonc.2024.1292793] [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: 10/24/2023] [Accepted: 01/19/2024] [Indexed: 02/27/2024] Open
Abstract
Introduction Gastric cancer ranks as the 5th most prevalent cancer and the 4th leading cause of cancer-related deaths worldwide. Various treatment modalities, including surgical resection, chemotherapy, and radiotherapy, are available for gastric cancer patients. However, disparities related to age, sex, race, socioeconomic factors, insurance status, and demographic factors often lead to delayed time to treatment. Methods In this retrospective study, conducted between 2004 and 2019, we utilized data from the National Cancer Database (NCDB) to investigate the factors contributing to disparities in the time to first treatment, surgery, chemotherapy, and radiotherapy among gastric cancer patients. Our analysis incorporated several variables, and statistical analysis was conducted to provide valuable insights into these disparities. Results We observed notable disparities in the timing of treatment for various demographic groups, including age, sex, race, insurance status, geographic location, and facility type. These disparities include longer time to treatment in males (32.67 vs 30.75), Native Americans (35.10 vs 31.09 in Asians), low-income patients (32 vs 31.15), patients getting treatment in an academic setting (36.11 vs 29.61 in community setting), significantly longer time to chemotherapy in 70+ age group (51.13 vs 40.38 in <40 y age group), black race (55.81 vs 47.05 in whites), low income people (49.64 vs 46.74), significantly longer time to radiotherapy in females (101.61 vs 79.75), blacks and Asians (109.68 and 113.96 respectively vs 92.68 in Native Americans) etc. There are various other disparities in time to surgery, chemotherapy, and radiotherapy. Conclusions Understanding these disparities is crucial in developing targeted strategies to improve timely access to appropriate treatments and enhance outcomes for gastric cancer patients. Future research with updated data and prospective study designs can provide a more comprehensive understanding of the factors influencing patient outcomes in gastric cancer.
Collapse
Affiliation(s)
- Seema Sharan
- Department of Surgery, Government Medical College and Hospital, Chandigarh, India
| | - Shivam Bansal
- Department of Surgery, Government Medical College and Hospital, Chandigarh, India
| | | | - Paola Berrios Jimenez
- Department of Surgery, University of Puerto Rico School of Medicine, San Juan, Puerto Rico
| | - Swathi R. Raikot
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
| | | | - Reed Popp
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Kyle Popp
- Department of Surgery, Florida State University, Tallahassee, FL, United States
| | - Kulkaew Sukniam
- Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | | | - Fatima Mubarak
- Department of Surgery, Aga Khan University, Karachi, Pakistan
| | - Emmanuel Gabriel
- Department of General Surgery, Mayo Clinic Florida, Jacksonville, FL, United States
| |
Collapse
|