201
|
Jia PF, Li YR, Wang LY, Lu XR, Guo X. Radiomics in esophagogastric junction cancer: A scoping review of current status and advances. Eur J Radiol 2024; 177:111577. [PMID: 38905802 DOI: 10.1016/j.ejrad.2024.111577] [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: 08/01/2023] [Revised: 06/03/2024] [Accepted: 06/14/2024] [Indexed: 06/23/2024]
Abstract
PURPOSE This scoping review aimed to understand the advances in radiomics in esophagogastric junction (EGJ) cancer and assess the current status of radiomics in EGJ cancer. METHODS We conducted systematic searches of PubMed, Embase, and Web of Science databases from January 18, 2012, to January 15, 2023, to identify radiomics articles related to EGJ cancer. Two researchers independently screened the literature, extracted data, and assessed the quality of the studies using the Radiomics Quality Score (RQS) and the METhodological RadiomICs Score (METRICS) tool, respectively. RESULTS A total of 120 articles were retrieved from the three databases, and after screening, only six papers met the inclusion criteria. These studies investigated the role of radiomics in differentiating adenocarcinoma from squamous carcinoma, diagnosing T-stage, evaluating HER2 overexpression, predicting response to neoadjuvant therapy, and prognosis in EGJ cancer. The median score percentage of RQS was 34.7% (range from 22.2% to 38.9%). The median score percentage of METRICS was 71.2% (range from 58.2% to 84.9%). CONCLUSION Although there is a considerable difference between the RQS and METRICS scores of the included literature, we believe that the research value of radiomics in EGJ cancer has been revealed. In the future, while actively exploring more diagnostic, prognostic, and biological correlation studies in EGJ cancer, greater emphasis should be placed on the standardization and clinical application of radiomics.
Collapse
Affiliation(s)
- Ping-Fan Jia
- Department of Medical Imaging, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Yu-Ru Li
- Department of Medical Imaging, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Lu-Yao Wang
- Department of Medical Imaging, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Xiao-Rui Lu
- Department of Medical Imaging, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Xing Guo
- Department of Medical Imaging, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, China.
| |
Collapse
|
202
|
Gao Z, Xu G, Wang S, Guo N, Yu Y, Wang X. Unusual presentation of PD-1 inhibitors in people living with HIV with advanced gastric cancer: Case report. Int J STD AIDS 2024; 35:733-738. [PMID: 38644514 DOI: 10.1177/09564624241248676] [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] [Indexed: 04/23/2024]
Abstract
This paper seeks to determine the effect of combination anti-PD-1 and antiretroviral therapy (ART) on people living with HIV (PLWH) with advanced gastric cancer. In our case, a PLWH with recurrent locally advanced gastric cancer was treated with anti-PD-1 inhibitor and ART. A significant reduction in tumor lesions (as demonstrated by contrast-enhanced CT imaging) and a better quality of life were achieved following treatment. There have been limited studies on the treatment of PLWH with advanced gastric cancer. Chemotherapy is most often used, however, with unsatisfactory outcomes. to date, there have been no published reports on the use of PD-1 inhibitors in PLWH with advanced gastric cancer. Our report provides a valuable reference for future management of such patients.
Collapse
Affiliation(s)
- Zhidi Gao
- Department of Oncology, Qingdao Branch of Shandong Public Health Clinical Center, Qingdao, People's Republic of China
| | - Guangyong Xu
- Department of Infectious Diseases, Qingdao Branch of Shandong Public Health Clinical Center, Qingdao, People's Republic of China
| | - Su Wang
- Department of Oncology, Hiser Hospital Affiliated to Qingdao University, Shandong, People's Republic of China
| | - Na Guo
- Department of Oncology, Qingdao Branch of Shandong Public Health Clinical Center, Qingdao, People's Republic of China
| | - Yang Yu
- Department of Oncology, Qingdao Branch of Shandong Public Health Clinical Center, Qingdao, People's Republic of China
| | - Xiaoni Wang
- Imaging Department, Qingdao Branch of Shandong Public Health Clinical Center, Qingdao, People's Republic of China
| |
Collapse
|
203
|
Naher SK, Mercieca-Bebber R, Siu D, Stockler MR, Kiely BE, Grimison P. Estimating survival scenarios in advanced or metastatic gastric and oesophageal adenocarcinoma: a systematic review of randomized-controlled trials. Curr Med Res Opin 2024; 40:1357-1367. [PMID: 38961804 DOI: 10.1080/03007995.2024.2376129] [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: 02/20/2024] [Revised: 06/24/2024] [Accepted: 07/01/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND We aimed to summarize survival data from RCTs in patients with GO adenocarcinoma; estimate and explain worst-, typical-, and best-case-scenarios of survival time; and determine if simple multiples of median overall survival (mOS) could estimate these percentiles. METHODS We systematically searched RCTs of systemic therapies for GO adenocarcinoma published 2000-2022. The following key percentiles were extracted from overall survival curves: 90th (worst-case), 75th (lower-typical), 25th (upper-typical), and 10th (best-case). We tested if these percentiles could be estimated by simple multiples of mOS: 0.25 of the median for the 90th percentile, 0.5 for the 75th, 2 for the 25th, and 3 for the 10th. RESULTS We identified 44 trials (22,447 participants). For first line chemotherapy and immunotherapy combined (CI) trials (n = 3) worst-to-best case survival time ranged from 4 months to not reached, compared to 3-30 months for other trials (n = 27) and 1-23 months for subsequent lines (n = 14). Simple multiples of mOS accurately estimated the following survival percentiles: 90th (n = 3/3 trials), 75th (n = 3/3), and 25th (n = 2/3) in first line CI trials. In other first line trials, the mOS accurately estimated the 90th survival percentile in n = 22/27 trials, 75th percentile in n = 26/27, 25th percentile in 27/27 trials, and 10th percentile in 22/27 trials. Simple multiples of the mOS accurately predicted the 90th, 75th, 25th, and 10th survival percentiles in the majority of trials of second and subsequent lines apart from chemotherapy and immunotherapy only trials. CONCLUSION We provide realistic, evidence-based prognostic information as scenarios for survival time which can inform clinical decision-making. Simple multiples of the mOS accurately estimated the percentiles for most groups.
Collapse
Affiliation(s)
- Sayeda K Naher
- National Health and Medical Research Council (NHMRC) Clinical Trials Centre (CTC), University of Sydney, Camperdown, NSW, Australia
- Illawarra and Shoalhaven Local Health District, Warrawong, NSW, Australia
| | - Rebecca Mercieca-Bebber
- National Health and Medical Research Council (NHMRC) Clinical Trials Centre (CTC), University of Sydney, Camperdown, NSW, Australia
| | - Derrick Siu
- National Health and Medical Research Council (NHMRC) Clinical Trials Centre (CTC), University of Sydney, Camperdown, NSW, Australia
| | - Martin R Stockler
- National Health and Medical Research Council (NHMRC) Clinical Trials Centre (CTC), University of Sydney, Camperdown, NSW, Australia
| | - Belinda E Kiely
- National Health and Medical Research Council (NHMRC) Clinical Trials Centre (CTC), University of Sydney, Camperdown, NSW, Australia
| | | |
Collapse
|
204
|
Becerra-Tomás N, Markozannes G, Cariolou M, Balducci K, Vieira R, Kiss S, Aune D, Greenwood DC, Dossus L, Copson E, Renehan AG, Bours M, Demark-Wahnefried W, Hudson MM, May AM, Odedina FT, Skinner R, Steindorf K, Tjønneland A, Velikova G, Baskin ML, Chowdhury R, Hill L, Lewis SJ, Seidell J, Weijenberg MP, Krebs J, Cross AJ, Tsilidis KK, Chan DSM. Post-diagnosis adiposity and colorectal cancer prognosis: A Global Cancer Update Programme (CUP Global) systematic literature review and meta-analysis. Int J Cancer 2024; 155:400-425. [PMID: 38692659 DOI: 10.1002/ijc.34905] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 12/15/2023] [Accepted: 01/17/2024] [Indexed: 05/03/2024]
Abstract
The adiposity influence on colorectal cancer prognosis remains poorly characterised. We performed a systematic review and meta-analysis on post-diagnosis adiposity measures (body mass index [BMI], waist circumference, waist-to-hip ratio, weight) or their changes and colorectal cancer outcomes. PubMed and Embase were searched through 28 February 2022. Random-effects meta-analyses were conducted when at least three studies had sufficient information. The quality of evidence was interpreted and graded by the Global Cancer Update Programme (CUP Global) independent Expert Committee on Cancer Survivorship and Expert Panel. We reviewed 124 observational studies (85 publications). Meta-analyses were possible for BMI and all-cause mortality, colorectal cancer-specific mortality, and cancer recurrence/disease-free survival. Non-linear meta-analysis indicated a reverse J-shaped association between BMI and colorectal cancer outcomes (nadir at BMI 28 kg/m2). The highest risk, relative to the nadir, was observed at both ends of the BMI distribution (18 and 38 kg/m2), namely 60% and 23% higher risk for all-cause mortality; 95% and 26% for colorectal cancer-specific mortality; and 37% and 24% for cancer recurrence/disease-free survival, respectively. The higher risk with low BMI was attenuated in secondary analyses of RCTs (compared to cohort studies), among studies with longer follow-up, and in women suggesting potential methodological limitations and/or altered physiological state. Descriptively synthesised studies on other adiposity-outcome associations of interest were limited in number and methodological quality. All the associations were graded as limited (likelihood of causality: no conclusion) due to potential methodological limitations (reverse causation, confounding, selection bias). Additional well-designed observational studies and interventional trials are needed to provide further clarification.
Collapse
Affiliation(s)
- Nerea Becerra-Tomás
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Georgios Markozannes
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Margarita Cariolou
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Katia Balducci
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Rita Vieira
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Sonia Kiss
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Dagfinn Aune
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Department of Nutrition, Oslo New University College, Oslo, Norway
- Department of Research, The Cancer Registry of Norway, Oslo, Norway
| | - Darren C Greenwood
- Leeds Institute for Data Analytics, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Laure Dossus
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Ellen Copson
- Cancer Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Andrew G Renehan
- The Christie NHS Foundation Trust, Manchester Cancer Research Centre, NIHR Manchester Biomedical Research Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Martijn Bours
- Department of Epidemiology, GROW School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Wendy Demark-Wahnefried
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Melissa M Hudson
- Department of Oncology, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Anne M May
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | | | - Roderick Skinner
- Department of Paediatric and Adolescent Haematology/Oncology, Great North Children's Hospital and Translational and Clinical Research Institute, and Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK
| | - Karen Steindorf
- Division of Physical Activity, Prevention and Cancer, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Anne Tjønneland
- Danish Cancer Society Research Center, Diet, Cancer and Health, Copenhagen, Denmark
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Galina Velikova
- School of Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | | | - Rajiv Chowdhury
- Department of Global Health, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA
| | - Lynette Hill
- World Cancer Research Fund International, London, UK
| | - Sarah J Lewis
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Jaap Seidell
- Department of Health Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Matty P Weijenberg
- Department of Epidemiology, GROW School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - John Krebs
- Department of Biology, University of Oxford, Oxford, UK
| | - Amanda J Cross
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Konstantinos K Tsilidis
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Doris S M Chan
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| |
Collapse
|
205
|
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
|
206
|
Sun YQ, Zhong Q, Lv CB, Zhu JY, Lin GT, Zhang ZQ, Wu D, Weng CM, Chen QX, Lian MQ, Zeng WM, Zhang YB, Chen QY, Lin JX, Xie JW, Li P, Zheng CH, Lu J, Cai LS, Huang CM. The safety and efficacy of neoadjuvant immunochemotherapy following laparoscopic gastrectomy for gastric cancer: a multicentre real-world clinical study. Int J Surg 2024; 110:4830-4838. [PMID: 38652275 PMCID: PMC11326023 DOI: 10.1097/js9.0000000000001468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 03/31/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND The safety and efficacy of neoadjuvant immunochemotherapy (nICT) for locally advanced gastric cancer (LAGC) remain controversial. METHODS Patients with LAGC who received either nICT or neoadjuvant chemotherapy (nCT) at 3 tertiary referral teaching hospitals in China between January 2016 and October 2022 were analyzed. After propensity-score matching (PSM), comparing the radiological response, pathological response rate, perioperative outcomes, and early recurrence between the two groups. RESULTS After PSM, 585 patients were included, with 195 and 390 patients comprising the nICT and nCT groups, respectively. The nICT group exhibited a higher objective response rate (79.5% vs. 59.0%; P <0.001), pathological complete response rate (14.36% vs. 6.41%; P =0.002) and major pathological response rate (39.49% vs. 26.15%; P =0.001) compared with the nCT group. The incidence of surgical complications (17.44% vs. 16.15%, P =0.694) and the proportion of perioperative textbook outcomes (80.0% vs. 81.0%; P =0.767) were similar in both groups. The nICT group had a significantly lower proportion of early recurrence than the nCT group (29.7% vs. 40.8%; P =0.047). Furthermore, the multivariable logistic analysis revealed that immunotherapy was an independent protective factor against early recurrence [odds ratio 0.62 (95% CI 0.41-0.92); P =0.018]. No significant difference was found in neoadjuvant therapy drug toxicity between the two groups (51.79% vs. 45.38%; P =0.143). CONCLUSIONS Compared with nCT, nICT is safe and effective, which significantly enhanced objective and pathological response rates and reduced the risk for early recurrence among patients with LAGC. TRIAL REGISTRATION Clinical Trials.gov.
Collapse
Affiliation(s)
- Yu-Qin Sun
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Qing Zhong
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Chen-Bin Lv
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
| | - Ji-Yun Zhu
- Department of General Surgery, The First Affiliated Hospital of Ningbo University, Zhejiang, China
| | - Guang-Tan Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Zhi-Quan Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Dong Wu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Cai-Ming Weng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Qiu-Xian Chen
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
| | - Ming-Qiao Lian
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
| | - Wei-Ming Zeng
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
| | - Yong-Bin Zhang
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Li-Sheng Cai
- Department of Gastrointestinal Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| |
Collapse
|
207
|
Malla RR, Nellipudi HR, Srilatha M, Nagaraju GP. HER-2 positive gastric cancer: Current targeted treatments. Int J Biol Macromol 2024; 274:133247. [PMID: 38906351 DOI: 10.1016/j.ijbiomac.2024.133247] [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: 01/03/2024] [Revised: 06/09/2024] [Accepted: 06/16/2024] [Indexed: 06/23/2024]
Abstract
Gastric cancer (GC) is highly metastatic and characterized by HER2 amplification. Aberrant HER2 expression drives metastasis, therapy resistance, and tumor recurrence. HER2 amplification contributes to drug resistance by upregulating DNA repair enzymes and drug afflux proteins, reducing drug efficacy. HER2 modulates transcription factors critical for cancer stem cell properties, further impacting drug resistance. HER2 activity is influenced by HER-family ligands, promoting oncogenic signaling. These features point to HER2 as a targetable driver in GC. This review outlines recent advances in HER2-mediated mechanisms and their upstream and downstream signaling pathways in GC. Additionally, it discusses preclinical research investigation that comprehends trastuzumab-sensitizing phytochemicals, chemotherapeutics, and nanoparticles as adjunct therapies. These developments hold promise for improving outcomes and enhancing the management of HER2-positive GC.
Collapse
Affiliation(s)
- Rama Rao Malla
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, Institute of Science, Gandhi Institute of Technology and Management (Deemed to be University), Visakhapatnam, AP 530045, India
| | | | - Mundla Srilatha
- Department of Biotechnology, Sri Venkateswara University, Tirupati 517502, AP, India
| | | |
Collapse
|
208
|
Blaize C, Surtouque E, Font J, Dolladille C, Postel-Vinay S, Da Silva A, Alexandre J, Morice PM. Arterial hypertension associated with PARPi: A meta-analysis of 41 placebo randomized controlled trials combined with a World Health Organization's pharmacovigilance study. Fundam Clin Pharmacol 2024; 38:610-629. [PMID: 38268495 DOI: 10.1111/fcp.12984] [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: 08/10/2023] [Revised: 12/05/2023] [Accepted: 01/08/2024] [Indexed: 01/26/2024]
Abstract
BACKGROUND Arterial hypertension has been recently reported from randomized controlled trials (RCTs) assessing poly (ADP-ribose) polymerase inhibitor (PARPi). OBJECTIVE In a context of increasing use of PARPi, it is crucial to properly assess risk and incidence of this adverse event for clinical practice. METHODS We performed a systematic review and meta-analysis in MEDLINE, Cochrane CENTRAL and ClinicalTrials.gov up to January 4, 2023 with an ongoing surveillance up to June 7, 2023. RCTs comparing PARPi to placebo in adult patients with solid tumors were included if hypertension was reported. The primary outcome was the summary risk ratio (RR, with 95% CIs) of any hypertension of PARPi class in placebo RCTs. Secondary outcomes were the summary risk and incidence of hypertension of each individual PARPi. To provide clinical features of PARPi-associated hypertension, we independently queried the WHO's pharmacovigilance database, up to September 1, 2022. RESULTS In total, 41 placebo RCTs (n = 15 264 adult patients) were included. PARPi class was not associated with an increased risk of hypertension compared with placebo. In individual analyses, the risk of hypertension was lower with olaparib than placebo (RR 0.77 [95% CI: 0.68-0.86], P < 0.01; I2 = 19%, χ2 P = 0.26). Niraparib monotherapy increased the risk of any (RR 2.84 [95% CI: 1.76-4.57], P < 0.01; I2 = 66%, χ2 P = 0.01) hypertension with a summary incidence of 19.87% (95% CI: 15.23-25.50). In real-life setting, niraparib-associated hypertension occurs within 20 days and was serious in 66%. Co-prescription of at least one antihypertensive or therapy-induced hypertension was reported in 20.5% or 14.4% of cases, respectively. CONCLUSIONS In a context of extensive assessment of niraparib in combination, these data reinforce the need of a close monitoring of this adverse event to preserve its clinical benefit on patients' survival.
Collapse
Affiliation(s)
- Clémence Blaize
- Department of Pharmacology, Caen-Normandy University Hospital, Caen, France
| | - Ellina Surtouque
- Department of Pharmacology, Caen-Normandy University Hospital, Caen, France
| | - Jonaz Font
- Normandie Univ, UNICAEN, INSERM U1086 'Interdisciplinary Research Unit for Cancers Prevention and Treatment' (ANTICIPE), Caen, France
- Department of Cardiology, Caen-Normandy University Hospital, Caen, France
| | - Charles Dolladille
- Normandie Univ, UNICAEN, INSERM U1086 'Interdisciplinary Research Unit for Cancers Prevention and Treatment' (ANTICIPE), Caen, France
- PICARO Cardio-Oncology Program, Department of Pharmacology, Caen-Normandy University Hospital, Caen, France
| | - Sophie Postel-Vinay
- European Research Council (ERC) StG Team, Inserm Unit U981, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
- Drug Development Department (DITEP), Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médicine, Université Paris-Sud, Université Paris-Saclay, Le Kremlin Bicêtre, France
| | - Angélique Da Silva
- Department of Pharmacology, Caen-Normandy University Hospital, Caen, France
- Department of medical oncology, Caen-Normandy University Hospital, Caen, France
| | - Joachim Alexandre
- Normandie Univ, UNICAEN, INSERM U1086 'Interdisciplinary Research Unit for Cancers Prevention and Treatment' (ANTICIPE), Caen, France
- PICARO Cardio-Oncology Program, Department of Pharmacology, Caen-Normandy University Hospital, Caen, France
| | - Pierre-Marie Morice
- Department of Pharmacology, Caen-Normandy University Hospital, Caen, France
- Normandie Univ, UNICAEN, INSERM U1086 'Interdisciplinary Research Unit for Cancers Prevention and Treatment' (ANTICIPE), Caen, France
| |
Collapse
|
209
|
Qureshi Z, Jamil A, Fatima E, Altaf F, Siddique R, Shah S. Pembrolizumab in combination with trastuzumab for treatment of HER2-positive advanced gastric or gastro-esophageal junction cancer. Ann Med Surg (Lond) 2024; 86:4647-4656. [PMID: 39118760 PMCID: PMC11305801 DOI: 10.1097/ms9.0000000000002305] [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: 04/15/2024] [Accepted: 06/14/2024] [Indexed: 08/10/2024] Open
Abstract
Introduction Gastric cancer remains a challenging malignancy with a high global mortality rate. Recent advances in targeted therapy and immunotherapy have shown promise in improving patient outcomes. This paper reviews the impact of incorporating targeted agents such as trastuzumab and immunotherapeutic agents like pembrolizumab into standard chemotherapy regimens for gastric cancer treatment. Methods A comprehensive analysis was conducted on pivotal clinical trials, including KEYNOTE-590, KEYNOTE-811, and ToGA, focusing on their methodologies, patient populations, treatment regimens, and outcome measures. The review also explored emerging research avenues in precision medicine, particularly genomic sequencing and biomarker identification. Aim To assess the efficacy and survival benefits of adding trastuzumab and pembrolizumab to standard chemotherapy in the treatment of gastric cancer and to outline future directions in gastric cancer research. Results Including trastuzumab and pembrolizumab in treatment regimens for human epidermal growth factor receptor 2 (HER2)-positive and PD-L1-expressing gastric cancers significantly improved progression-free and overall survival rates compared to chemotherapy alone. These findings highlight the potential of personalized therapy in enhancing treatment outcomes. Furthermore, ongoing research into the gastric cancer microenvironment and the role of the microbiome suggests novel targets for future therapeutic interventions. Conclusion The integration of targeted and immunotherapeutic agents with traditional chemotherapy represents a pivotal shift in gastric cancer treatment, moving towards more personalized and effective regimens.
Collapse
Affiliation(s)
- Zaheer Qureshi
- The Frank H. Netter M.D. School of Medicine at Quinnipiac University, Bridgeport, CT
| | - Abdur Jamil
- Department of Medicine, Samaritan Medical Centre, Watertown, NY
| | - Eeshal Fatima
- Department of Medicine, Services Institute of Medical Sciences, Lahore, Pakistan
| | - Faryal Altaf
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai/BronxCare Health System, New York, NY, USA
| | | | - Shivendra Shah
- Department of Medicine, Nepalgunj Medical College, Chisapani, Nepal
| |
Collapse
|
210
|
Shaik R, Venkata Renuka I, Ramamoorthy S, Potti R, Pranathi L. Human Epidermal Growth Factor 2 (Her-2) Expression in Gastric and Gastroesophageal Carcinomas: A Clinicopathological Evaluation in a Tertiary Care Institute. Cureus 2024; 16:e67895. [PMID: 39193056 PMCID: PMC11348447 DOI: 10.7759/cureus.67895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2024] [Indexed: 08/29/2024] Open
Abstract
Introduction Gastric carcinoma is a significant global health concern, known for its high mortality rate. HER-2 overexpression is observed in a notable proportion of gastric carcinoma and is associated with a worse prognosis. However, HER-2 expression enables targeting the protein by monoclonal antibodies that improve overall survival in HER-2-positive gastric cancers. This study aims to evaluate the HER-2 expression in gastric and gastroesophageal carcinomas. Materials and methods This observational study was conducted in the Department of Pathology, involving 60 endoscopic biopsy and resection specimens of gastric and gastroesophageal carcinomas. HER-2 expression was assessed by immunohistochemistry (IHC) based on the Trastuzumab for GAstric Cancer (ToGA) trial's scoring system. The primary outcome was HER-2 status, with statistical analysis performed to evaluate associations with various clinicopathological parameters. Results Among 60 cases, 26 (43.3%) showed HER-2 positivity. HER-2 positivity was significantly (p=0.004) associated with age, being higher in 20-39 years and ≥80 years age groups. Gender and tumor location were not significantly associated with HER-2 positivity. Moderate and poorly differentiated carcinomas exhibited higher HER-2 positivity. Histological types, tubular adenocarcinoma, and papillary adenocarcinoma showed significant (p=0.01) association with HER-2 positivity compared to other types. Conclusion HER-2 status assessment is crucial in managing gastric and gastroesophageal carcinomas. HER-2 positivity is notably higher in certain age groups and histological types particularly tubular and papillary adenocarcinoma, and in moderately to poorly differentiated carcinomas. These insights can aid in selecting appropriate gastric and gastroesophageal carcinomas that warrant HER-2 testing on IHC. Identifying gastric and gastroesophageal carcinomas that show HER2 expression may highlight potential candidates for targeted therapy.
Collapse
|
211
|
Lordick F, Van Cutsem E, Shitara K, Xu RH, Ajani JA, Shah MA, Oh M, Ganguli A, Chang L, Rhoten S, Bhattacharya P, Matsangou M, Park JW, Pophale R, Ranganath R, Kang YK. Health-related quality of life in patients with CLDN18.2-positive, locally advanced unresectable or metastatic gastric or gastroesophageal junction adenocarcinoma: results from the SPOTLIGHT and GLOW clinical trials. ESMO Open 2024; 9:103663. [PMID: 39146670 PMCID: PMC11374961 DOI: 10.1016/j.esmoop.2024.103663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 08/17/2024] Open
Abstract
BACKGROUND First-line zolbetuximab plus chemotherapy (SPOTLIGHT, mFOLFOX6; GLOW, CAPOX) significantly improved progression-free survival (PFS) and overall survival (OS) versus placebo plus chemotherapy in patients with human epidermal growth factor receptor 2-negative, locally advanced unresectable or metastatic gastric or gastroesophageal junction adenocarcinoma whose tumors were claudin 18 isoform 2-positive in the phase III SPOTLIGHT (NCT03504397) and GLOW (NCT03653507) studies. We present patient-reported outcomes (PROs) from these studies. MATERIALS AND METHODS Health-related quality of life (HRQoL) was measured in the full analysis sets using the European Organisation for Research and Treatment of Cancer Quality of Life of Cancer Patients Core Questionnaire (QLQ-C30) and Oesophago-Gastric Module (QLQ-OG25), Global Pain, and 5-level EQ-5D (EQ-5D-5L) questionnaires. Analyses focused on key PRO domains: global health status (GHS)/QoL, physical functioning, abdominal pain and discomfort, and nausea/vomiting. Least squares mean (LSM) changes from baseline and time to first definitive deterioration (TTDD) were evaluated combined across SPOTLIGHT and GLOW and for individual studies. Time to confirmed deterioration (TTCD) was evaluated independently for SPOTLIGHT and GLOW. RESULTS The combined analysis set included 1072 patients (zolbetuximab plus chemotherapy, 537; placebo plus chemotherapy, 535). Compliance rates were similar between treatment arms. Similar trends were observed in the zolbetuximab versus placebo arms for LSM changes from baseline in key PRO domains, with no clinically meaningful deterioration. Nausea/vomiting worsened during the first few zolbetuximab cycles but later returned to baseline levels. Overall TTCD and TTDD results were similar between arms in both studies. CONCLUSIONS Patients in SPOTLIGHT and GLOW maintained measured HRQoL relative to baseline when treated with first-line zolbetuximab added to chemotherapy. Zolbetuximab plus chemotherapy improved PFS and OS without negatively affecting HRQoL in key PRO domains compared with placebo plus chemotherapy.
Collapse
Affiliation(s)
- F Lordick
- Department of Medicine and University Cancer Center Leipzig, University of Leipzig Medical Center, Leipzig, Germany
| | - E Van Cutsem
- Digestive Oncology, University Hospitals Gasthuisberg, Leuven, and KULeuven, Leuven, Belgium
| | - K Shitara
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa City, Chiba, Japan
| | - R-H Xu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou; Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - J A Ajani
- The University of Texas, MD Anderson Cancer Center, Houston
| | - M A Shah
- Weill Cornell Medical College, New York City
| | - M Oh
- Astellas Pharma Global Development, Inc., Northbrook. https://twitter.com/AstellasUS
| | - A Ganguli
- Astellas Pharma Global Development, Inc., Northbrook. https://twitter.com/AstellasUS
| | - L Chang
- Astellas Pharma Global Development, Inc., Northbrook. https://twitter.com/AstellasUS
| | | | - P Bhattacharya
- Astellas Pharma Global Development, Inc., Northbrook. https://twitter.com/AstellasUS
| | - M Matsangou
- Astellas Pharma Global Development, Inc., Northbrook. https://twitter.com/AstellasUS
| | - J W Park
- Astellas Pharma Global Development, Inc., Northbrook. https://twitter.com/AstellasUS
| | - R Pophale
- Astellas Pharma Global Development, Inc., Northbrook. https://twitter.com/AstellasUS
| | - R Ranganath
- Astellas Pharma Global Development, Inc., Northbrook. https://twitter.com/AstellasUS
| | - Y-K Kang
- Department of Oncology, Asan Medical Center, University of Ulsan, Seoul, Republic of Korea.
| |
Collapse
|
212
|
Sukrithan V, Perez K, Pandit-Taskar N, Jimenez C. Management of metastatic pheochromocytomas and paragangliomas: when and what. Curr Probl Cancer 2024; 51:101116. [PMID: 39024846 DOI: 10.1016/j.currproblcancer.2024.101116] [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: 03/25/2024] [Accepted: 05/22/2024] [Indexed: 07/20/2024]
Abstract
Recently, the treatment landscape for metastatic pheochromocytomas and paragangliomas (MPPGL) has seen both progress and setbacks. We provide an up-to-date review of the multimodality management of MPPGL and discuss novel opportunities and current challenges in the treatment landscape. Given the unique clinical presentation of MPPGL, we discuss the management of hormone-related clinical sequelae and traditional modalities of therapy. Advances in the understanding of the molecular biology of these diverse tumors have enabled novel strategies such as augmenting DNA damage by targeted delivery of radionuclides such as 131I and 177Lu, abrogating tumor angiogenesis, hypoxia resistance, and DNA damage repair. Despite progress, we address the significant challenges still faced by patients and researchers engaged in efforts to improve outcomes in these rare cancers.
Collapse
Affiliation(s)
- Vineeth Sukrithan
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States.
| | - Kimberly Perez
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Neeta Pandit-Taskar
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Camilo Jimenez
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
213
|
Mondal J, Chakraborty K, Bunggulawa EJ, An JM, Revuri V, Nurunnabi M, Lee YK. Recent advancements of hydrogels in immunotherapy: Breast cancer treatment. J Control Release 2024; 372:1-30. [PMID: 38849092 DOI: 10.1016/j.jconrel.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/21/2024] [Accepted: 06/01/2024] [Indexed: 06/09/2024]
Abstract
Breast cancer is the most prevalent cancer among women and the leading cause of cancer-related deaths in this population. Recent advances in Immunotherapy, or combined immunotherapy, offering a more targeted and less toxic approach, expand the survival rate of patients more than conventional treatment. Notably, hydrogels, a versatile platform provided promising avenues to combat breast cancer in preclinical studies and extended to clinical practices. With advantages such as the alternation of tumor microenvironment, immunomodulation, targeted delivery of therapeutic agents, and their sustained release at specific sites of interest, hydrogels can potentially be used for the treatment of breast cancer. This review highlights the advantages, mechanisms of action, stimuli-responsiveness properties, and recent advancements of hydrogels for treating breast cancer immunotherapy. Moreover, post-treatment and its clinical translations are discussed in this review. The integration of hydrogels in immunotherapy strategies may pave the way for more effective, personalized, and patient-friendly approaches to combat breast cancer, ultimately contributing to a brighter future for breast cancer patients.
Collapse
Affiliation(s)
- Jagannath Mondal
- 4D Convergence Technology Institute, Korea National University of Transportation, Jeungpyeong 27909, Republic of Korea; Department of Green Bioengineering, Korea National University of Transportation, Chungju 27470, Republic of Korea; Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, USA
| | - Kushal Chakraborty
- Department of IT and Energy Convergence (BK21 FOUR), Korea National University of Transportation, Chungju 27469, Republic of Korea
| | - Edwin J Bunggulawa
- Department of Green Bioengineering, Korea National University of Transportation, Chungju 27470, Republic of Korea
| | - Jeong Man An
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Vishnu Revuri
- Department of Green Bioengineering, Korea National University of Transportation, Chungju 27470, Republic of Korea
| | - Md Nurunnabi
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79902, United States; Biomedical Engineering Program, College of Engineering, University of Texas at El Paso, El Paso, TX 79968, United States.
| | - Yong-Kyu Lee
- 4D Convergence Technology Institute, Korea National University of Transportation, Jeungpyeong 27909, Republic of Korea; Department of Green Bioengineering, Korea National University of Transportation, Chungju 27470, Republic of Korea; Department of Chemical & Biological Engineering, Korea National University of Transportation, Chungju 27470, Republic of Korea.
| |
Collapse
|
214
|
Masetti M, Lorenzen S. [What's new in gastric cancer?]. Dtsch Med Wochenschr 2024; 149:1015-1020. [PMID: 39146748 DOI: 10.1055/a-2179-0830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
In the locally advanced stage, multimodal therapies such as perioperative chemotherapy with FLOT or neoadjuvant radiochemotherapy are recommended. The integration of immunotherapy into these concepts could improve the prognosis. Phase II/III trials such as DANTE, KEYNOTE-585 and MATTERHORN show promising results in terms of pathological remissions but data on survival time extension for unselected patients are so far sobering. Immunotherapies and new targeted therapies offer hope in the palliative treatment of metastatic gastric cancer. Studies such as CheckMate-649 and KEYNOTE-859 show an improvement in survival and response rates. Currently, both pembrolizumab and nivolumab have been approved for the first-line treatment of tumors with positive PD-L1 expression. In HER2-positive tumors, the KEYNOTE-811 study showed that patients benefit from combination therapies with immune checkpoint inhibition and anti-HER2 therapies. The antibody-drug conjugate trastuzumab-deruxtecan is a promising second-line treatment option for HER2-positive tumors after treatment failure with trastuzumab.In addition, the bispecific antibody zanidatamab shows promising results in first-line treatment. New targeted therapies against CLDN18.2 and FGFR2b are showing promising results. The anti-claudin 18.2 (CLDN18.2) antibody zolbetuximab leads to improved survival compared to chemotherapy alone in patients with CLDN18.2 positive disease in first-line therapy, with approval expected in 2024.
Collapse
Affiliation(s)
- Michael Masetti
- Klinik und Poliklinik für Innere Medizin III, Klinikum rechts der Isar der TU München, München, Deutschland
| | - Sylvie Lorenzen
- Klinik und Poliklinik für Innere Medizin III, Klinikum rechts der Isar der TU München, München, Deutschland
| |
Collapse
|
215
|
Sekmek S, Karahan I, Ucar G, Ceylan F, Bayram D, Seven I, Bölek H, Ürün Y, Yücel KB, Yazici O, Kadioglu A, Karacin C, Canaslan K, Atag E, Demirer S, Erdem GU, Ergun Y, Atak M, Koksal B, Kiran MM, Turkay DO, Civelek B, Yalcin B, Uncu D. Effect of HER2/CEP17 ratio on survival in metastatic HER2-positive gastric cancer, multicenter study. Clin Transl Oncol 2024; 26:1878-1885. [PMID: 38451412 DOI: 10.1007/s12094-024-03410-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 02/12/2024] [Indexed: 03/08/2024]
Abstract
AIM HER2-positive metastatic gastric cancer is still a highly fatal disease despite advances. We aimed to investigate the relationship between HER2/CEP17 ratio and survival in patients with HER2-positive metastatic gastric cancer. METHODS A total of 99 patients from 8 different centers in Turkey were included in the study. Patients with HER2-positive metastatic gastric cancer and whose HER2/CEP17 ratio was examined were included in the study. Patients were divided into two groups according to HER2/CEP17 values, and survival analysis was performed. RESULTS The median age was 64 (24-83) years. There were 74 (74.8%) male and 25 (25.2%) female patients. OS in the high HER2/CEP17 ratio group was 21.97 months (95% CI: 16.36-27.58), and in the low ratio group was 16.17 months (95% CI: 10.95-21.38) (p = 0.015). OS was 17.7 months (95% CI: 7.02-28.37) in the high HER2 gene copy number group and 10.13 months (5.55-14.71) in the group with low copy number (p = 0.03). PFS was 10.94 months (95% CI: 7.55-14.33) in the group with high HER2 gene copy number and 7.56 months (4.62-10.49) in the low copy number group (p = 0.06). CONCLUSION Patients with both high HER2 gene amplification and high HER2/CEP17 ratio had better OS. The PFS of the group with high HER2 gene amplification was also better. To our knowledge, this is the first study in the literature showing that the HER2/CEP17 ratio affects survival in patients with metastatic gastric cancer.
Collapse
Affiliation(s)
- Serhat Sekmek
- Department of Medical Oncology, Ankara City Hospital, Ankara, Turkey.
| | - Irfan Karahan
- Department of Medical Oncology, Ankara City Hospital, Ankara, Turkey
| | - Gokhan Ucar
- Department of Medical Oncology, Ankara City Hospital, Ankara, Turkey
| | - Furkan Ceylan
- Department of Medical Oncology, Ankara City Hospital, Ankara, Turkey
| | - Dogan Bayram
- Department of Medical Oncology, Ankara City Hospital, Ankara, Turkey
| | - Ismet Seven
- Department of Medical Oncology, Ankara City Hospital, Ankara, Turkey
| | - Hatice Bölek
- Department of Medical Oncology, Ankara University, Ankara, Turkey
| | - Yüksel Ürün
- Department of Medical Oncology, Ankara University, Ankara, Turkey
| | | | - Ozan Yazici
- Department of Medical Oncology, Gazi University, Ankara, Turkey
| | - Ahmet Kadioglu
- Department of Medical Oncology, UHS Dr Abdurrahman Yurtaslan Ankara Oncology Training and Research Hospital, Ankara, Turkey
| | - Cengiz Karacin
- Department of Medical Oncology, UHS Dr Abdurrahman Yurtaslan Ankara Oncology Training and Research Hospital, Ankara, Turkey
| | - Kubra Canaslan
- Department of Medical Oncology, Dokuz Eylul University, Izmir, Turkey
| | - Elif Atag
- Department of Medical Oncology, Dokuz Eylul University, Izmir, Turkey
| | - Serhat Demirer
- Department of Medical Oncology, İstanbul Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| | - Gokmen Umut Erdem
- Department of Medical Oncology, İstanbul Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| | - Yakup Ergun
- Department of Medical Oncology, Antalya City Hospital, Antalya, Turkey
| | - Mehmetcan Atak
- Department of Medical Oncology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Baris Koksal
- Department of Medical Oncology, Hacettepe University, Ankara, Turkey
| | | | | | - Burak Civelek
- Department of Medical Oncology, Ankara City Hospital, Ankara, Turkey
| | - Bulent Yalcin
- Department of Medical Oncology, Ankara City Hospital, Ankara, Turkey
| | - Dogan Uncu
- Department of Medical Oncology, Ankara City Hospital, Ankara, Turkey
| |
Collapse
|
216
|
Choi E, Shin J, Ryu MH, Kim HD, Park YS. Heterogeneity of claudin 18.2 expression in metastatic gastric cancer. Sci Rep 2024; 14:17648. [PMID: 39085339 PMCID: PMC11291723 DOI: 10.1038/s41598-024-68411-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: 04/15/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024] Open
Abstract
Claudin 18.2 has emerged as a viable therapeutic target in gastric cancer (GC), but little is known about the heterogeneity of its expression in GC. This study investigated the heterogeneity of claudin 18.2 expression in 166 patients with metastatic GC whose surgical or paired primary-metastatic specimens were available. The prevalence of claudin 18.2 positivity (moderate-to-strong expression in ≥ 75% by the 43-14A clone) was 47.0%. Claudin 18.2-positive tumors exhibited more frequent peritoneal metastasis and a lower incidence of hepatic and distant lymph node involvement. Survival outcomes were comparable between patients with claudin 18.2-positive and -negative tumors. Intratumoral heterogeneity was noted in 38.5% of surgical specimens. Paired primary-metastatic site analysis revealed that 25.2% of patients had discordant results for claudin 18.2 positivity. Across different metastatic organ categories, peritoneal lesions showed the highest positivity rate (44.3%) and positive concordance rate (31.4%), whereas liver lesions had the lowest positivity rate (17.9%) and concordance rate (12.8%). In conclusion, claudin 18.2 expression exhibits intratumoral and intrapatient spatial heterogeneity in metastatic GC. Claudin 18.2 positivity is associated with more frequent peritoneal metastasis, and peritoneal lesions are more likely to have positively concordant claudin 18.2 results with the primary site than other metastatic sites.
Collapse
Affiliation(s)
- Eugene Choi
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Jinho Shin
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Min-Hee Ryu
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Hyung-Don Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| | - Young Soo Park
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| |
Collapse
|
217
|
Zhang Y, Rui J, Liang J, Lin L. Primary mucinous adenocarcinoma of the anterior mediastinum with HER-2 mutation: A rare case report. Heliyon 2024; 10:e34346. [PMID: 39100453 PMCID: PMC11295853 DOI: 10.1016/j.heliyon.2024.e34346] [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: 01/05/2024] [Revised: 07/08/2024] [Accepted: 07/08/2024] [Indexed: 08/06/2024] Open
Abstract
We report the case of a 68-year-old male whose Computed Tomography (CT) scan presented a mass (68*62*54 mm) of the right anterior mediastinal and pathologically diagnosis was mucinous adenocarcinoma(MA). The peripheral vessels are surrounded by the big mass in the anterior mediastinum which was associated with multiple metastases, thus we performed palliative chemoradiotherapy and we tried Human Epidermal Growth Factor Receptor-2 (HER-2) inhibitors based on the Next Generation Sequencing. The patient passed away 16 months after the onset of the disease. In this report, we review the rare case of anterior mediastinum MA as well as perspectives for potential future treatments.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Oncology Center, Peking University International Hospital, Life Park Road, Zhong Guancun Life Science Park, Changping District, Beijing, 102206, China
| | - Jinqiu Rui
- Department of Medicine, Geneplus-Beijing, Beijing, 102206, China
| | - Jun Liang
- Department of Oncology Center, Peking University International Hospital, Life Park Road, Zhong Guancun Life Science Park, Changping District, Beijing, 102206, China
| | - Li Lin
- Department of Oncology Center, Peking University International Hospital, Life Park Road, Zhong Guancun Life Science Park, Changping District, Beijing, 102206, China
| |
Collapse
|
218
|
Uçar G, Sekmek S, Karahan İ, Ergün Y, İsak ÖA, Tunç S, Doğan M, Gürler F, Bayram D, Açıkgöz Y, Esen SA, Civelek B, Köş FT, Bal Ö, Algın E, Eren T, İmamoğlu Gİ, Urakçı Z, Yazıcı O, Özdemir N, Uncu D. The Comparison of FLOT and DCF Regimens as Perioperative Treatment for Gastric Cancer. Oncology 2024:1-6. [PMID: 39079501 DOI: 10.1159/000540517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 07/15/2024] [Indexed: 09/06/2024]
Abstract
INTRODUCTION Locoregional gastric cancer is a still serious problem and perioperative treatments may improve the success of management. Different regimens were examined. The present study purposed to compare the efficacy of fluorouracil-leucovorin-oxaliplatin-docetaxel (FLOT) and docetaxel-cisplatin-fluorouracil (DCF) regimens. METHODS A retrospective multicenter study assessed the patients with locoregional gastric cancer. There are 240 patients (137 DCF, 103 FLOT). Survival rates were compared. RESULTS Demographic features were similar between the two groups, but the time period was different. The FLOT group had 7.8% pathological complete response, while the DCF group did not. Disease-free survival was longer in the FLOT than in the DCF group (median not reached - 13.94 months, respectively). Median overall survival was similar (30.9 vs. 37.8 months), but median follow-up affected the analysis. Survival for 36 months was 63% for the FLOT group and 40% for the DCF group (log-rank; p = 0.015). CONCLUSION FLOT regimen was superior to DCF regimen for response and survival rates. DCF is a historical approach. Long-term follow-up period is needed for FLOT treatment.
Collapse
Affiliation(s)
- Gökhan Uçar
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Serhat Sekmek
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara, Turkey
| | - İrfan Karahan
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Yakup Ergün
- Department of Medical Oncology, Antalya City Hospital, Ankara, Turkey
| | - Özlem Aydın İsak
- Department of Medical Oncology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Sezai Tunç
- Department of Medicak Oncology, Dicle University Faculty of Medicine, Diyarbakır, Turkey
| | - Mutlu Doğan
- Department of Medical Oncology, Ankara Dr. AY Oncology Training and Research Hospital, Ankara, Turkey
| | - Fatih Gürler
- Department of Medical Oncology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Doğan Bayram
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Yusuf Açıkgöz
- Medical Oncology, Kastamonu Education and Research Hospital, Kastamonu, Turkey
| | - Selin Aktürk Esen
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Burak Civelek
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Fahriye Tuğba Köş
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Öznur Bal
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Efnan Algın
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Tülay Eren
- Department of Medical Oncology, Ankara Etlik City Hospital, Ankara, Turkey
| | | | - Zuhat Urakçı
- Department of Medicak Oncology, Dicle University Faculty of Medicine, Diyarbakır, Turkey
| | - Ozan Yazıcı
- Department of Medical Oncology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Nuriye Özdemir
- Department of Medical Oncology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Doğan Uncu
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara, Turkey
| |
Collapse
|
219
|
Darwish IA, Zhang D, Alsalhi MS. A novel highly sensitive inner filter effect-based fluorescence immunoassay with quantum dots for bioanalysis of zolbetuximab, a monoclonal antibody used for immunotherapy of gastric and gastroesophageal junction adenocarcinoma. Heliyon 2024; 10:e34611. [PMID: 39114008 PMCID: PMC11305320 DOI: 10.1016/j.heliyon.2024.e34611] [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: 02/05/2024] [Revised: 06/11/2024] [Accepted: 07/12/2024] [Indexed: 08/10/2024] Open
Abstract
Zolbetuximab (ZOL) is a groundbreaking monoclonal antibody targeting CLDN 18.2, a cancer cell surface protein. It is a first-in-class therapy for gastric and gastroesophageal junction adenocarcinoma. However, there is currently any immunoassay available for bioanalysis of ZOL, hindering its pharmacokinetic studies, therapeutic monitoring, and safety profile refinement. To address this gap, this study presents the development and validation of a novel highly sensitive inner filter effect-based fluorescence immunoassay (IFE-FIA) with quantum dots (QDs) as a probe. This assay enables the quantitative determination of ZOL in plasma samples. The assay involved non-competitive capturing of ZOL from the samples using a specific antigen (CLDN 18.2 protein) immobilized on assay plate microwells. A horseradish peroxidase (HRP)-labelled anti-human IgG was used to measure the immune complex. The assay's detection system relies on the formation of a light-absorbing colored product through an HRP-catalyzed oxidative reaction with the substrate 3,3',5,5'-tetramethylbenzidine. This light absorption efficiently quenched the fluorescence of QDs via the IFE. The measured fluorescence signals corresponded to the concentrations of ZOL in the samples. The conditions of the IFE-FIA and its detection system were refined, and the optimum procedures were established. Following the guidelines of immunoassay validation for bioanalysis, the assay was validated, and all the validation criteria were acceptable. The assay demonstrates high sensitivity, accurately quantifying ZOL at concentrations as low as 10 ng/mL in plasma samples, with acceptable precision. Importantly, it avoids interferences from endogenous substances and plasma matrix. The recoveries in spiked human plasma ranged from 96.8 % to 104.5 %, with relative standard deviations of 4.1 %-6.5 %. The proposed IFE-FIA represents a valuable tool for quantifying ZOL in clinical settings, enabling assessment of its pharmacokinetics, therapeutic drug monitoring, and safety profile refinement.
Collapse
Affiliation(s)
- Ibrahim A. Darwish
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Saudi Arabia
| | - Daohong Zhang
- College of Food Engineering, Bio-Nanotechnology Research Institute, Ludong University, Yantai, 264025, Shandong, China
| | - Mohammed S. Alsalhi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
220
|
Alrouji M, Yasmin S, Alhumaydhi FA, Sharaf SE, Shahwan M, Shamsi A. ROS1 kinase inhibition reimagined: identifying repurposed drug via virtual screening and molecular dynamics simulations for cancer therapeutics. Front Chem 2024; 12:1392650. [PMID: 39136033 PMCID: PMC11317403 DOI: 10.3389/fchem.2024.1392650] [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: 02/27/2024] [Accepted: 07/01/2024] [Indexed: 08/15/2024] Open
Abstract
Precision medicine has revolutionized modern cancer therapeutic management by targeting specific molecular aberrations responsible for the onset and progression of tumorigenesis. ROS proto-oncogene 1 (ROS1) is a receptor tyrosine kinase (RTK) that can induce tumorigenesis through various signaling pathways, such as cell proliferation, survival, migration, and metastasis. It has emerged as a promising therapeutic target in various cancer types. However, there is very limited availability of specific ROS1 inhibitors for therapeutic purposes. Exploring repurposed drugs for rapid and effective treatment is a useful approach. In this study, we utilized an integrated approach of virtual screening and molecular dynamics (MD) simulations of repurposing existing drugs for ROS1 kinase inhibition. Using a curated library of 3648 FDA-approved drugs, virtual screening identified drugs capable of binding to ROS1 kinase domain. The results unveil two hits, Midostaurin and Alectinib with favorable binding profiles and stable interactions with the active site residues of ROS1. These hits were subjected to stability assessment through all-atom MD simulations for 200 ns. MD results showed that Midostaurin and Alectinib were stable with ROS1. Taken together, the study showed a rational framework for the selection of repurposed Midostaurin and Alectinib with ROS1 inhibitory potential for therapeutic management after further validation.
Collapse
Affiliation(s)
- Mohammed Alrouji
- Department of Medical Laboratories, College of Applied Medical Sciences, Shaqra University, Shaqra, Saudi Arabia
| | - Sabina Yasmin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Fahad A Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Sharaf E. Sharaf
- Pharmaceutical Sciences Department, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Moyad Shahwan
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman, United Arab Emirates
- Center of Medical and Bio-Allied Health Sciences Research (CMBHSR), Ajman University, Ajman, United Arab Emirates
| | - Anas Shamsi
- Center of Medical and Bio-Allied Health Sciences Research (CMBHSR), Ajman University, Ajman, United Arab Emirates
| |
Collapse
|
221
|
Ratti M, Orlandi E, Toscani I, Vecchia S, Anselmi E, Hahne JC, Ghidini M, Citterio C. Emerging Therapeutic Targets and Future Directions in Advanced Gastric Cancer: A Comprehensive Review. Cancers (Basel) 2024; 16:2692. [PMID: 39123420 PMCID: PMC11311890 DOI: 10.3390/cancers16152692] [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: 06/28/2024] [Revised: 07/19/2024] [Accepted: 07/21/2024] [Indexed: 08/12/2024] Open
Abstract
Metastatic gastric cancer (GC) still represents a critical clinical challenge, with limited treatment options and a poor prognosis. Most patients are diagnosed at advanced stages, limiting the chances of surgery and cure. The identification of molecular targets and the possibility of combining immune checkpoint inhibitors with chemotherapy have recently reshaped the therapeutic landscape of metastatic gastric cancer. The new classification of gastric cancer, mainly based on immunologic and molecular criteria such as programmed cell death 1 (PD-1), microsatellite instability (MSI), and human epidermal growth factor receptor 2 (HER2), has made it possible to identify and differentiate patients who may benefit from immunotherapy, targeted therapy, or chemotherapy alone. All relevant and available molecular and immunological targets in clinical practice for the systemic treatment of this disease are presented. Particular attention is given to possible future approaches, including circulating tumor DNA (ctDNA) for therapeutic monitoring, new targeting agents against molecular pathways such as fibroblast growth factor receptor (FGFR) and MET, chimeric antigen receptor (CAR)-T cells, and cancer vaccines. This review aims to provide a comprehensive understanding of current targets in advanced gastric cancer and to offer valuable insights into future directions of research and clinical practice in this challenging disease.
Collapse
Affiliation(s)
- Margherita Ratti
- Oncology and Hematology Department, Piacenza General Hospital, Via Taverna 49, 29121 Piacenza, Italy; (E.O.); (I.T.); (S.V.); (E.A.); (C.C.)
| | - Elena Orlandi
- Oncology and Hematology Department, Piacenza General Hospital, Via Taverna 49, 29121 Piacenza, Italy; (E.O.); (I.T.); (S.V.); (E.A.); (C.C.)
| | - Ilaria Toscani
- Oncology and Hematology Department, Piacenza General Hospital, Via Taverna 49, 29121 Piacenza, Italy; (E.O.); (I.T.); (S.V.); (E.A.); (C.C.)
| | - Stefano Vecchia
- Oncology and Hematology Department, Piacenza General Hospital, Via Taverna 49, 29121 Piacenza, Italy; (E.O.); (I.T.); (S.V.); (E.A.); (C.C.)
| | - Elisa Anselmi
- Oncology and Hematology Department, Piacenza General Hospital, Via Taverna 49, 29121 Piacenza, Italy; (E.O.); (I.T.); (S.V.); (E.A.); (C.C.)
| | - Jens Claus Hahne
- Division of Molecular Pathology, The Institute of Cancer Research, Sutton, London SM2 5NG, UK;
| | - Michele Ghidini
- Medical Oncology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Chiara Citterio
- Oncology and Hematology Department, Piacenza General Hospital, Via Taverna 49, 29121 Piacenza, Italy; (E.O.); (I.T.); (S.V.); (E.A.); (C.C.)
| |
Collapse
|
222
|
Tojjari A, Nagdas S, Saeed A, Saeed A. Deciphering the FGFR2 Code: Innovative Targets in Gastric Cancer Therapy. Curr Oncol 2024; 31:4305-4317. [PMID: 39195304 DOI: 10.3390/curroncol31080321] [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: 06/19/2024] [Revised: 07/26/2024] [Accepted: 07/27/2024] [Indexed: 08/29/2024] Open
Abstract
Gastric cancer (GC) represents a major global health challenge as a highly prevalent disease with high mortality whose global incidence and mortality are predicted to worsen over the coming years. To date, our standard of care for advanced gastric cancer of combination chemotherapy and immunotherapy has a 1-year overall survival rate of 55%. Significant efforts have gone into identifying targetable alterations in gastric cancer, ultimately yielding the Fibroblast Growth Factor Receptors (FGFRs) family, specifically FGFR2 as a promising target. FGFR2 is overexpressed in GC, particularly diffuse-type GC, and is associated with poor prognostic outcomes. In recent years, there has been an increasing number of small molecule inhibitors and monoclonal antibodies targeting FGFR2 that have entered into clinical trials. Specifically for GC, these agents are currently being trialed in various phases as monotherapies or with standard-of-care treatments to make a clinically meaningful impact on what appears to be an important biological axis of GC. In this review, we outline the underlying biology of FGFR2, its putative role in GC, and the various FGFR2-targeted agents currently in clinical trials for gastric cancer patients as well as postulate some challenges in adopting these therapeutics for clinically meaningful benefit.
Collapse
Affiliation(s)
- Alireza Tojjari
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA 15261, USA
| | | | - Ali Saeed
- Department of Medicine, Ochsner Lafayette General Medical Center, Lafayette, LA 70503, USA
| | - Anwaar Saeed
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA 15261, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| |
Collapse
|
223
|
Peng W, Zhang H, Yin M, Kong D, Kang L, Teng X, Wang J, Chu Z, Sun Y, Long P, Cui C, Lyu B, Zhang J, Xiao H, Wu M, Wang Y, Li Y. Combined Inhibition of PI3K and STAT3 signaling effectively inhibits bladder cancer growth. Oncogenesis 2024; 13:29. [PMID: 39068158 PMCID: PMC11283499 DOI: 10.1038/s41389-024-00529-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: 04/12/2024] [Revised: 07/07/2024] [Accepted: 07/11/2024] [Indexed: 07/30/2024] Open
Abstract
Bladder cancer is characterized by aberrant activation of the phosphatidylinositol-3-OH kinase (PI3K) signaling, underscoring the significance of directing therapeutic efforts toward the PI3K pathway as a promising strategy. In this study, we discovered that PI3K serves as a potent therapeutic target for bladder cancer through a high-throughput screening of inhibitory molecules. The PI3K inhibitor demonstrated a robust anti-tumor efficacy, validated both in vitro and in vivo settings. Nevertheless, the feedback activation of JAK1-STAT3 signaling reinstated cell and organoid survival, leading to resistance against the PI3K inhibitor. Mechanistically, the PI3K inhibitor suppresses PTPN11 expression, a negative regulator of the JAK-STAT pathway, thereby activating STAT3. Conversely, restoration of PTPN11 enhances the sensitivity of cancer cells to the PI3K inhibitor. Simultaneous inhibition of both PI3K and STAT3 with small-molecule inhibitors resulted in sustained tumor regression in patient-derived bladder cancer xenografts. These findings advocate for a combinational therapeutic approach targeting both PI3K and STAT3 pathways to achieve enduring cancer eradication in vitro and in vivo, underscoring their promising therapeutic efficacy for treating bladder cancer.
Collapse
Affiliation(s)
- Weidong Peng
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, China
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Haojie Zhang
- Department of Urology, Huadong Hospital, Fudan University, Shanghai, China
| | - Mingwei Yin
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Dejie Kong
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Liping Kang
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Xinkun Teng
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Jingjing Wang
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Zhimin Chu
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Yating Sun
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Pengpeng Long
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Chengying Cui
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Bin Lyu
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Jinzhi Zhang
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Han Xiao
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Mingqing Wu
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China.
| | - Yongqiang Wang
- Department of Urology, South China Hospital, Medical School, Shenzhen University, Shenzhen, China.
| | - Yang Li
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, China.
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China.
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, China.
| |
Collapse
|
224
|
Qian YF, Chen HP, Ren GF. Acute therapy-related myelodysplastic syndromes following capecitabine and oxaliplatin therapy in gastric malignant tumor: A case report. Medicine (Baltimore) 2024; 103:e39049. [PMID: 39058884 PMCID: PMC11272321 DOI: 10.1097/md.0000000000039049] [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: 04/17/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
RATIONAL Patients with gastric cancer show a relatively low incidence of developing secondary myelodysplastic syndrome (MDS). PATIENT CONCERNS A 60-year-old man was admitted because of pain and discomfort in the upper abdomen and intermittent abdominal pain. DIAGNOSES Ulcerative moderately poorly differentiated adenocarcinoma (pT2N2M0G3, stage IIB) and MDS. INTERVENTIONS The patient underwent chemotherapy with oxaliplatin (OXP, intravenously guttae on day 1) plus capecitabine (CAP, bis in die orally on day 1-14). The patient developed degree III myelosuppression after OXP plus CAP chemotherapy and MDS was subsequently confirmed by diagnosis of the bone marrow biopsy. Temporary but significant hematological improvements were observed after the patient received corresponding treatment, which helped achieve remission and improve pancytopenia. OUTCOMES The patient presented partial remission after corresponding treatment and no other complications have been recorded. LESSONS Acute MDS is an unusual adverse effect induced by OXP plus CAP chemotherapy. It is urgent to suggest implementing a supplementary assessment or examination for patients receiving these therapies in future cases.
Collapse
Affiliation(s)
- Yi-fan Qian
- Department of Pharmacy, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Hang-ping Chen
- Department of Pharmacy, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Guo-fei Ren
- Department of Pharmacy, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
225
|
Kaleem M, Thool M, Dumore NG, Abdulrahman AO, Ahmad W, Almostadi A, Alhashmi MH, Kamal MA, Tabrez S. Management of triple-negative breast cancer by natural compounds through different mechanistic pathways. Front Genet 2024; 15:1440430. [PMID: 39130753 PMCID: PMC11310065 DOI: 10.3389/fgene.2024.1440430] [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: 05/29/2024] [Accepted: 07/09/2024] [Indexed: 08/13/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is the most severe form of breast cancer, characterized by the loss of estrogen, progesterone, and human epidermal growth factor receptors. It is caused by various genetic and epigenetic factors, resulting in poor prognosis. Epigenetic changes, such as DNA methylation and histone modification, are the leading mechanisms responsible for TNBC progression and metastasis. This review comprehensively covers the various subtypes of TNBC and their epigenetic causes. In addition, the genetic association of TNBC with all significant genes and signaling pathways linked to the progression of this form of cancer has been enlisted. Furthermore, the possible uses of natural compounds through different mechanistic pathways have also been discussed in detail for the successful management of TNBC.
Collapse
Affiliation(s)
- Mohammed Kaleem
- Department of Pharmacology, Dadasaheb Balpande College of Pharmacy, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, Maharashtra, India
| | - Mandar Thool
- Department of Pharmaceutics, Dadasaheb Balpande College of Pharmacy, Nagpur, Maharashtra, India
| | - Nitin G. Dumore
- Department of Pharmacology, Dadasaheb Balpande College of Pharmacy, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, Maharashtra, India
| | | | - Wasim Ahmad
- Department of KuliyateTib, National Institute of Unani Medicine, Bengaluru, India
| | - Amal Almostadi
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammad Hassan Alhashmi
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammad Amjad Kamal
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Center for High Altitude Medicine, Institutes for Systems Genetics, West China School of Nursing, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Birulia, Bangladesh
- Centre for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
- Enzymoics, Hebersham, NSW, Australia; Novel Global Community Educational Foundation, Hebersham, NSW, Australia
| | - Shams Tabrez
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
226
|
Farzeen Z, Khan RRM, Chaudhry AR, Pervaiz M, Saeed Z, Rasheed S, Shehzad B, Adnan A, Summer M. Dostarlimab: A promising new PD-1 inhibitor for cancer immunotherapy. J Oncol Pharm Pract 2024:10781552241265058. [PMID: 39056234 DOI: 10.1177/10781552241265058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
OBJECTIVE Dostarlimab, a humanized monoclonal PD-1 blocking antibody, is being tested as a cancer therapy in this review. Specifically, it addresses mismatch repair failure in endometrial cancer and locally progressed rectal cancer patients. DATA SOURCES A thorough database search found Dostarlimab clinical trials and studies. Published publications and ongoing clinical trials on Dostarlimab's efficacy as a single therapy and in conjunction with other medicines across cancer types were searched. DATA SUMMARY The review recommends Dostarlimab for endometrial cancer mismatch repair failure, as supported by GARNET studies. The analysis also highlights locally advanced rectal cancer findings. In the evolving area of cancer therapy, immune checkpoint inhibitors including pembrolizumab, avelumab, atezolizumab, nivolumab, and durvalumab were discussed. CONCLUSIONS Locally advanced rectal cancer patients responded 100% to Dostarlimab. Many clinical trials, including ROSCAN, AMBER, IOLite, CITRINO, JASPER, OPAL, PRIME, PERLA, and others, are investigating Dostarlimab in combination treatment. This research sheds light on Dostarlimab's current and future possibilities, in improving cancer immunotherapy understanding.
Collapse
Affiliation(s)
- Zubaria Farzeen
- Department of Chemistry, Government College University Lahore, Lahore, Punjab, Pakistan
| | | | - Ayoub Rashid Chaudhry
- Department of Chemistry, Government College University Lahore, Lahore, Punjab, Pakistan
| | - Muhammad Pervaiz
- Department of Chemistry, Government College University Lahore, Lahore, Punjab, Pakistan
| | - Zohaib Saeed
- Department of Chemistry, Government College University Lahore, Lahore, Punjab, Pakistan
| | - Shahzad Rasheed
- Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Behram Shehzad
- Department of Chemistry, Government College University Lahore, Lahore, Punjab, Pakistan
| | - Ahmad Adnan
- Department of Chemistry, Government College University Lahore, Lahore, Punjab, Pakistan
| | - Muhammad Summer
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University Lahore, Lahore, Pakistan
| |
Collapse
|
227
|
Zhou W, Xu Z, Liu S, Lou X, Liu P, Xie H, Zhang S, Liu X, Zhuo B, Huang H. Landscape of clinical drug development of ADCs used for the pharmacotherapy of cancers: an overview of clinical trial registry data from 2002 to 2022. BMC Cancer 2024; 24:898. [PMID: 39060958 PMCID: PMC11282866 DOI: 10.1186/s12885-024-12652-5] [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: 01/12/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND To provide reference for clinical development of ADCs in the industry, we analyzed the landscape and characteristics of clinical trials about antibody-drug conjugates (ADCs). METHOD Clinical trials to study ADCs used for the pharmacotherapy of cancers initiated by the sponsor were searched in the Cite line Pharma Intelligence (Trialtrove database), and the landscape and characteristics of these clinical trials were analyzed from multiple perspectives, such as the number, phases, status, indications, and targets of the clinical trials. RESULT As of December 31, 2022, a total of 431 clinical trials have been initiated to study ADCs used for the pharmacotherapy of cancers, and the number of the last 10 years was 5.5 times as large as the first 11 years. These clinical trials involved 47 indications, including breast cancer, lymphoma (lymphoma, non-Hodgkin's and lymphoma, Hodgkin's), unspecified solid tumor, bladder cancer and lung cancer (lung, non-small cell cancer and lung, small cell cancer). As for each of these five indications, 50 + clinical trials have been carried out, accounting for as high as 48.50% (454/936). ADCs involve 38 targets, which are relatively concentrated. Among them, ERBB2 (HER2) and TNFRSF8 (CD30) involve in 100 + registered clinical trials, and TNFRSF17 (BCMA), NECTIN4 and CD19 in 10 + trials. The clinical trials for these five targets account for 79.02% (354/448) of the total number. Up to 93.97% (405/431) of these clinical trials explored the correlation between biomarkers and efficacy. Up to 45.91% (292/636) of Lots (lines of treatment) applied in the clinical trials were the second line. Until December 31, 2022, 54.52% (235/431) of the clinical trials have been completed or terminated. CONCLUSION ADCs are a hotspot of research and development in oncology clinical trials, but the indications, targets, phases, and Lot that have been registered are seemingly relatively concentrated at present. This study provides a comprehensive analysis which can assist researchers/developer quickly grasp relevant knowledge to assess a product and also providing new clues and ideas for future research.
Collapse
Affiliation(s)
- Wenjing Zhou
- Department of Clinical Trials Center, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, 518000, P. R. China.
| | - Zhiyuan Xu
- Department of Clinical Oncology Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518000, P. R. China
| | - Shu Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Xiaohuan Lou
- Department of Clinical Trials Center, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, 518000, P. R. China
| | - Pengcheng Liu
- School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Huali Xie
- Department of Pharmacy, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518000, P. R. China
| | - Shuiyan Zhang
- Department of Chinese Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518000, P. R. China
| | - Xi Liu
- Department of Clinical Trials Center, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, 518000, P. R. China
| | - Baoshan Zhuo
- Department of Clinical Trials Center, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, 518000, P. R. China
| | - Hongbing Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| |
Collapse
|
228
|
Wang X, Allen MJ, Espin-Garcia O, Suzuiki C, Bach Y, Panov E, Ma LX, Jang RW, Chen EX, Darling GE, Yeung J, Swallow CJ, Brar SS, Kalimuthu S, Wong R, Veit-Haibach P, Elimova E. Outcomes in older adults with metastatic esophageal and gastric carcinoma treated with palliative chemotherapy. Oncologist 2024:oyae190. [PMID: 39046894 DOI: 10.1093/oncolo/oyae190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/21/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND The incidence of esophageal and gastric carcinoma (GEC) in elderly patients is increasing, yet patients ≥75 years have historically been underrepresented in clinical trials. We sought to investigate palliative chemotherapy administration patterns and survival outcomes in older adults. MATERIALS AND METHODS A retrospective analysis identified patients aged 65-74 (young-old) and ≥75 years (older-old) diagnosed with advanced GEC. Patient and tumor characteristics were recorded, with descriptive analysis, time-to-event data analysis using Kaplan-Meier curves and multivariate Cox proportional hazards regression analysis performed. RESULTS One hundred and ninety-eight "young-old" and 109 'older-old' patients were identified. Patient characteristics were similar between groups except for Charlson Co-morbidity Index (CCI), with lower co-morbidities in the "young-old" compared to "older-old" cohort (P < .001; CCI = 0 in 103 (52%) "young-old" vs 31 (28%) "older-old"). The primary diagnosis in both groups was adenocarcinoma. 119 (60%) "young-old" and 25 (23%) "older-old" patients received chemotherapy (P < .001). Performance status was the primary explanation for chemotherapy non-receipt in both cohorts; age was the explanation in 21 (25%) "older-old" patients and none in the "young-old" patients. PFS for first-line systemic therapy in "young-old" patients was 6.4 (95% CI 5.9-7.6) versus 7.5 months (95% CI 5.1-11.3) in "older-old" patients (P = .69) whilst respective OS was 12.3 (95% CI 10.1-15.5) and 10.4 months (95% CI 9.0-14.6) (P = .0816). Toxicity prompted chemotherapy cessation in 17 (15%) "young-old" and 3 (13%) "older-old" patients (P = .97). Multivariate analysis identified CCI and ECOG performance status as predictive for PFS and OS, respectively. No causative relationship was identified with other variables. CONCLUSION Our study of real-world older-adults show that significant number of "older-old" patients with GEC do not receive chemotherapy. Among "older-old" adults who do receive systemic therapy, outcomes are comparable; this underscores the importance of geriatric assessment-guided care and suggests that age alone should not be a barrier to receipt of chemotherapy in patients with advanced GEC.
Collapse
Affiliation(s)
- Xin Wang
- Division of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto ON, Canada
| | - Michael J Allen
- Division of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto ON, Canada
| | - Osvaldo Espin-Garcia
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Chihiro Suzuiki
- Division of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto ON, Canada
| | - Yvonne Bach
- Division of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto ON, Canada
| | - Elan Panov
- Division of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto ON, Canada
| | - Lucy X Ma
- Division of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto ON, Canada
| | - Raymond W Jang
- Division of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto ON, Canada
| | - Eric X Chen
- Division of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto ON, Canada
| | - Gail E Darling
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto ON, Canada
| | - Jonathan Yeung
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto ON, Canada
| | - Carol J Swallow
- Department of Surgical Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto ON, Canada
- Department of Surgery, Mount Sinai Hospital, Toronto, ON, Canada
| | | | - Sangeetha Kalimuthu
- Division of Pathology, Toronto General Hospital, University Health Network, Toronto ON, Canada
| | - Rebecca Wong
- Division of Radiation Oncology, Princess Margaret Hospital, University Health Network, Toronto ON, Canada
| | - Patrick Veit-Haibach
- Joint Department of Medical Imaging, Toronto General Hospital, University Health Network, Toronto ON, Canada
| | - Elena Elimova
- Division of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto ON, Canada
| |
Collapse
|
229
|
Liu Z, Jiang H, Lee SY, Kong N, Chan YW. FANCM promotes PARP inhibitor resistance by minimizing ssDNA gap formation and counteracting resection inhibition. Cell Rep 2024; 43:114464. [PMID: 38985669 DOI: 10.1016/j.celrep.2024.114464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 05/22/2024] [Accepted: 06/24/2024] [Indexed: 07/12/2024] Open
Abstract
Poly(ADP-ribose) polymerase inhibitors (PARPis) exhibit remarkable anticancer activity in tumors with homologous recombination (HR) gene mutations. However, the role of other DNA repair proteins in PARPi-induced lethality remains elusive. Here, we reveal that FANCM promotes PARPi resistance independent of the core Fanconi anemia (FA) complex. FANCM-depleted cells retain HR proficiency, acting independently of BRCA1 in response to PARPis. FANCM depletion leads to increased DNA damage in the second S phase after PARPi exposure, driven by elevated single-strand DNA (ssDNA) gap formation behind replication forks in the first S phase. These gaps arise from both 53BP1- and primase and DNA directed polymerase (PRIMPOL)-dependent mechanisms. Notably, FANCM-depleted cells also exhibit reduced resection of collapsed forks, while 53BP1 deletion restores resection and mitigates PARPi sensitivity. Our results suggest that FANCM counteracts 53BP1 to repair PARPi-induced DNA damage. Furthermore, FANCM depletion leads to increased chromatin bridges and micronuclei formation after PARPi treatment, elucidating the mechanism underlying extensive cell death in FANCM-depleted cells.
Collapse
Affiliation(s)
- Zeyuan Liu
- School of Biological Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Huadong Jiang
- School of Biological Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Sze Yuen Lee
- School of Biological Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Nannan Kong
- School of Biological Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Ying Wai Chan
- School of Biological Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.
| |
Collapse
|
230
|
Yi M, Li T, Niu M, Zhang H, Wu Y, Wu K, Dai Z. Targeting cytokine and chemokine signaling pathways for cancer therapy. Signal Transduct Target Ther 2024; 9:176. [PMID: 39034318 PMCID: PMC11275440 DOI: 10.1038/s41392-024-01868-3] [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: 02/28/2024] [Revised: 04/30/2024] [Accepted: 05/11/2024] [Indexed: 07/23/2024] Open
Abstract
Cytokines are critical in regulating immune responses and cellular behavior, playing dual roles in both normal physiology and the pathology of diseases such as cancer. These molecules, including interleukins, interferons, tumor necrosis factors, chemokines, and growth factors like TGF-β, VEGF, and EGF, can promote or inhibit tumor growth, influence the tumor microenvironment, and impact the efficacy of cancer treatments. Recent advances in targeting these pathways have shown promising therapeutic potential, offering new strategies to modulate the immune system, inhibit tumor progression, and overcome resistance to conventional therapies. In this review, we summarized the current understanding and therapeutic implications of targeting cytokine and chemokine signaling pathways in cancer. By exploring the roles of these molecules in tumor biology and the immune response, we highlighted the development of novel therapeutic agents aimed at modulating these pathways to combat cancer. The review elaborated on the dual nature of cytokines as both promoters and suppressors of tumorigenesis, depending on the context, and discussed the challenges and opportunities this presents for therapeutic intervention. We also examined the latest advancements in targeted therapies, including monoclonal antibodies, bispecific antibodies, receptor inhibitors, fusion proteins, engineered cytokine variants, and their impact on tumor growth, metastasis, and the tumor microenvironment. Additionally, we evaluated the potential of combining these targeted therapies with other treatment modalities to overcome resistance and improve patient outcomes. Besides, we also focused on the ongoing research and clinical trials that are pivotal in advancing our understanding and application of cytokine- and chemokine-targeted therapies for cancer patients.
Collapse
Affiliation(s)
- Ming Yi
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, People's Republic of China
| | - Tianye Li
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310000, People's Republic of China
| | - Mengke Niu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Haoxiang Zhang
- Department of Hepatopancreatobiliary Surgery, Fujian Provincial Hospital, Fuzhou, 350001, People's Republic of China
| | - Yuze Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| | - Zhijun Dai
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, People's Republic of China.
| |
Collapse
|
231
|
Hu L, Zhang S, Sienkiewicz J, Zhou H, Berahovich R, Sun J, Li M, Ocampo A, Liu X, Huang Y, Harto H, Xu S, Golubovskaya V, Wu L. HER2-CD3-Fc Bispecific Antibody-Encoding mRNA Delivered by Lipid Nanoparticles Suppresses HER2-Positive Tumor Growth. Vaccines (Basel) 2024; 12:808. [PMID: 39066446 PMCID: PMC11281407 DOI: 10.3390/vaccines12070808] [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: 05/31/2024] [Revised: 07/10/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
The human epidermal growth factor receptor 2 (HER2) is a transmembrane tyrosine kinase receptor and tumor-associated antigen abnormally expressed in various types of cancer, including breast, ovarian, and gastric cancer. HER2 overexpression is highly correlated with increased tumor aggressiveness, poorer prognosis, and shorter overall survival. Consequently, multiple HER2-targeted therapies have been developed and approved; however, only a subset of patients benefit from these treatments, and relapses are common. More potent and durable HER2-targeted therapies are desperately needed for patients with HER2-positive cancers. In this study, we developed a lipid nanoparticle (LNP)-based therapy formulated with mRNA encoding a novel HER2-CD3-Fc bispecific antibody (bsAb) for HER2-positive cancers. The LNPs efficiently transfected various types of cells, such as HEK293S, SKOV-3, and A1847, leading to robust and sustained secretion of the HER2-CD3-Fc bsAb with high binding affinity to both HER2 and CD3. The bsAb induced potent T-cell-directed cytotoxicity, along with secretion of IFN-λ, TNF-α, and granzyme B, against various types of HER2-positive tumor cells in vitro, including A549, NCI-H460, SKOV-3, A1847, SKBR3, and MDA-MB-231. The bsAb-mediated antitumor effect is highly specific and strictly dependent on its binding to HER2, as evidenced by the gained resistance of A549 and A1847 her2 knockout cells and the acquired sensitivity of mouse 4T1 cells overexpressing the human HER2 extracellular domain (ECD) or epitope-containing subdomain IV to the bsAb-induced T cell cytotoxicity. The bsAb also relies on its binding to CD3 for T-cell recruitment, as ablation of CD3 binding abolished the bsAb's ability to elicit antitumor activity. Importantly, intratumoral injection of the HER2-CD3-Fc mRNA-LNPs triggers a strong antitumor response and completely blocks HER2-positive tumor growth in a mouse xenograft model of human ovarian cancer. These results indicate that the novel HER2-CD3-Fc mRNA-LNP-based therapy has the potential to effectively treat HER2-positive cancer.
Collapse
Affiliation(s)
- Liang Hu
- Promab Biotechnologies, 2600 Hilltop Drive, Richmond, CA 94806, USA; (L.H.); (S.Z.); (J.S.); (H.Z.); (R.B.); (J.S.); (M.L.); (A.O.); (X.L.); (Y.H.); (H.H.); (S.X.)
| | - Shiming Zhang
- Promab Biotechnologies, 2600 Hilltop Drive, Richmond, CA 94806, USA; (L.H.); (S.Z.); (J.S.); (H.Z.); (R.B.); (J.S.); (M.L.); (A.O.); (X.L.); (Y.H.); (H.H.); (S.X.)
| | - John Sienkiewicz
- Promab Biotechnologies, 2600 Hilltop Drive, Richmond, CA 94806, USA; (L.H.); (S.Z.); (J.S.); (H.Z.); (R.B.); (J.S.); (M.L.); (A.O.); (X.L.); (Y.H.); (H.H.); (S.X.)
| | - Hua Zhou
- Promab Biotechnologies, 2600 Hilltop Drive, Richmond, CA 94806, USA; (L.H.); (S.Z.); (J.S.); (H.Z.); (R.B.); (J.S.); (M.L.); (A.O.); (X.L.); (Y.H.); (H.H.); (S.X.)
| | - Robert Berahovich
- Promab Biotechnologies, 2600 Hilltop Drive, Richmond, CA 94806, USA; (L.H.); (S.Z.); (J.S.); (H.Z.); (R.B.); (J.S.); (M.L.); (A.O.); (X.L.); (Y.H.); (H.H.); (S.X.)
| | - Jinying Sun
- Promab Biotechnologies, 2600 Hilltop Drive, Richmond, CA 94806, USA; (L.H.); (S.Z.); (J.S.); (H.Z.); (R.B.); (J.S.); (M.L.); (A.O.); (X.L.); (Y.H.); (H.H.); (S.X.)
| | - Michael Li
- Promab Biotechnologies, 2600 Hilltop Drive, Richmond, CA 94806, USA; (L.H.); (S.Z.); (J.S.); (H.Z.); (R.B.); (J.S.); (M.L.); (A.O.); (X.L.); (Y.H.); (H.H.); (S.X.)
| | - Adrian Ocampo
- Promab Biotechnologies, 2600 Hilltop Drive, Richmond, CA 94806, USA; (L.H.); (S.Z.); (J.S.); (H.Z.); (R.B.); (J.S.); (M.L.); (A.O.); (X.L.); (Y.H.); (H.H.); (S.X.)
| | - Xianghong Liu
- Promab Biotechnologies, 2600 Hilltop Drive, Richmond, CA 94806, USA; (L.H.); (S.Z.); (J.S.); (H.Z.); (R.B.); (J.S.); (M.L.); (A.O.); (X.L.); (Y.H.); (H.H.); (S.X.)
| | - Yanwei Huang
- Promab Biotechnologies, 2600 Hilltop Drive, Richmond, CA 94806, USA; (L.H.); (S.Z.); (J.S.); (H.Z.); (R.B.); (J.S.); (M.L.); (A.O.); (X.L.); (Y.H.); (H.H.); (S.X.)
| | - Hizkia Harto
- Promab Biotechnologies, 2600 Hilltop Drive, Richmond, CA 94806, USA; (L.H.); (S.Z.); (J.S.); (H.Z.); (R.B.); (J.S.); (M.L.); (A.O.); (X.L.); (Y.H.); (H.H.); (S.X.)
| | - Shirley Xu
- Promab Biotechnologies, 2600 Hilltop Drive, Richmond, CA 94806, USA; (L.H.); (S.Z.); (J.S.); (H.Z.); (R.B.); (J.S.); (M.L.); (A.O.); (X.L.); (Y.H.); (H.H.); (S.X.)
| | - Vita Golubovskaya
- Promab Biotechnologies, 2600 Hilltop Drive, Richmond, CA 94806, USA; (L.H.); (S.Z.); (J.S.); (H.Z.); (R.B.); (J.S.); (M.L.); (A.O.); (X.L.); (Y.H.); (H.H.); (S.X.)
| | - Lijun Wu
- Promab Biotechnologies, 2600 Hilltop Drive, Richmond, CA 94806, USA; (L.H.); (S.Z.); (J.S.); (H.Z.); (R.B.); (J.S.); (M.L.); (A.O.); (X.L.); (Y.H.); (H.H.); (S.X.)
- Forevertek Biotechnology, Janshan Road, Changsha Hi-Tech Industrial Development Zone, Changsha 410205, China
| |
Collapse
|
232
|
Santero M, Meade AG, Selva A, Savall-Esteve O, Bracchiglione J, Macías I, Leache L, Cerdà P, Bonfill Cosp X. Utilising systematic reviews to assess potential overtreatment and claim for better evidence-based research: an analysis of anticancer drugs versus supportive care in advanced esophageal cancer. Syst Rev 2024; 13:186. [PMID: 39026378 PMCID: PMC11256491 DOI: 10.1186/s13643-024-02594-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 06/20/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Highlighting the identified gaps in evidence-based research concerning advanced esophageal cancer (EC) treatment and care, this review evaluates the efficacy and safety of anticancer drugs compared to supportive care for advanced EC patients, aiming to assess the appropriateness of usual treatments and identify the gaps that need to be filled with primary research. METHODS We searched (May 2022) MEDLINE, EMBASE, the Cochrane Central Register of Controlled Trials (CENTRAL), Epistemonikos, and trial registries (ClinicalTrials.gov and PROSPERO) for randomised controlled trials (RCTs) comparing anticancer drugs (chemotherapy, immunotherapy, or biological/targeted therapy) with supportive care in advanced EC. The results were summarised using GRADE summary of finding tables. RESULTS We included 15 RCTs. Most studies did not have a special focus on EC, did not detail the treatment lines in all patients, and did not evaluate all outcomes. Anticancer drugs may result in a slight increase in overall survival (OS) (HR 0.78; 95% CI 0.71, 0.86; MD 0.83 months) and better progression-free survival (PFS) (HR 0.56 95% CI 0.49, 0.64, MD 0.68 months), but also may increase toxicity (RR 1.37; 95% CI 1.13, 1.65), without a significant improvement in quality of life. The certainty of evidence was low or very low due to indirectness of results and lack of specific focus on EC in some studies. CONCLUSION RCTs on advanced EC lack specificity, detailed treatment line information, and evaluation of all relevant outcomes. Moreover, when they find any benefit, this is negligible. Therefore, the certainty to justify anticancer drug treatments instead of supportive care in advanced EC is low or very low, and this information should be actively shared with affected patients. More and better RCTs should be conducted to assess whether any old or new proposed treatment for advanced EC patients provides a better balance of benefits and harms than the supportive care. SYSTEMATIC REVIEW REGISTRATION The study protocol was registered in OSF ( https://doi.org/10.17605/OSF.IO/7CHX6 ) on 2022-03-29.
Collapse
Affiliation(s)
- Marilina Santero
- Universitat Autònoma Barcelona, Barcelona, Spain.
- Iberoamerican Cochrane Centre, Institut de Recerca Sant Pau (IR, SANT PAU), Barcelona, Spain.
| | - Adriana-Gabriela Meade
- Iberoamerican Cochrane Centre, Institut de Recerca Sant Pau (IR, SANT PAU), Barcelona, Spain
| | - Anna Selva
- Universitat Autònoma Barcelona, Barcelona, Spain
- Iberoamerican Cochrane Centre, Institut de Recerca Sant Pau (IR, SANT PAU), Barcelona, Spain
- Clinical Epidemiology and Cancer Screening, Parc Taulí Hospital Universitari, Institut d'Investigació I Innovació Parc Taulí (I3PT_CERCA), Sabadell, Spain
- CIBER de Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Olga Savall-Esteve
- Universitat Autònoma Barcelona, Barcelona, Spain
- Iberoamerican Cochrane Centre, Institut de Recerca Sant Pau (IR, SANT PAU), Barcelona, Spain
| | - Javier Bracchiglione
- Universitat Autònoma Barcelona, Barcelona, Spain
- Iberoamerican Cochrane Centre, Institut de Recerca Sant Pau (IR, SANT PAU), Barcelona, Spain
- Interdisciplinary Centre for Health Studies (CIESAL), Universidad de Valparaíso, Viña del Mar, Chile
- CIBER de Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Ismael Macías
- Servicio Oncología Médica, Hospital de Sabadell-Corporació Sanitària Parc Taulí, Sabadell, Barcelona, Spain
| | - Leire Leache
- Unit of Innovation and Organization, Navarre Health Service, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Paula Cerdà
- Universitat Autònoma Barcelona, Barcelona, Spain
- Iberoamerican Cochrane Centre, Institut de Recerca Sant Pau (IR, SANT PAU), Barcelona, Spain
- Hospital de La Santa Creu I Sant Pau, Barcelona, Spain
| | - Xavier Bonfill Cosp
- Universitat Autònoma Barcelona, Barcelona, Spain
- Iberoamerican Cochrane Centre, Institut de Recerca Sant Pau (IR, SANT PAU), Barcelona, Spain
- CIBER de Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
- Hospital de La Santa Creu I Sant Pau, Barcelona, Spain
| |
Collapse
|
233
|
Li Z, Song Z, Hong W, Yang N, Wang Y, Jian H, Liang Z, Hu S, Peng M, Yu Y, Wang Y, Jiao Z, Zhao K, Song K, Li Y, Shi W, Lu S. SHR-A1811 (antibody-drug conjugate) in advanced HER2-mutant non-small cell lung cancer: a multicenter, open-label, phase 1/2 study. Signal Transduct Target Ther 2024; 9:182. [PMID: 39004647 PMCID: PMC11247081 DOI: 10.1038/s41392-024-01897-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/27/2024] [Accepted: 06/18/2024] [Indexed: 07/16/2024] Open
Abstract
A dose-escalation and expansion, phase 1/2 study (ClinicalTrials.gov, NCT04818333) was conducted to assess the novel antibody-drug conjugate SHR-A1811 in pretreated HER2-altered advanced non-small cell lung cancer (NSCLC). Here, we report results from the phase 1 portion. Patients who had previously failed or were intolerant to platinum-based chemotherapy were enrolled and received SHR-A1811 intravenously at doses of 3.2 to 8.0 mg/kg every 3 weeks. Dose escalation followed a Bayesian logistic regression model that included overdose control, with subsequent selection of tolerable levels for dose expansion. Overall, 63 patients were enrolled, including 43 receiving a recommended dose for expansion of 4.8 mg/kg. All patients had HER2-mutant disease. Dose-limiting toxicity occurred in one patient in the 8.0 mg/kg dose cohort. Grade ≥ 3 treatment-related adverse events occurred in 29 (46.0%) patients. One patient in the 6.4 mg/kg cohort died due to interstitial lung disease. As of April 11, 2023, the 4.8 mg/kg cohort showed an objective response rate of 41.9% (95% CI 27.0-57.9), and a disease control rate of 95.3% (95% CI 84.2-99.4). The median duration of response was 13.7 months, with 13 of 18 responses ongoing. The median progression-free survival was 8.4 months (95% CI 7.1-15.0). SHR-A1811 demonstrated favourable safety and clinically meaningful efficacy in pretreated advanced HER2-mutant NSCLC.
Collapse
Affiliation(s)
- Ziming Li
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200030, China
| | - Zhengbo Song
- Phase I Clinical Trial Ward, Zhejiang Cancer Hospital, Hangzhou, 310000, China
| | - Wei Hong
- Department of Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou, 310000, China
| | - Nong Yang
- Department of Lung & Gastrointestinal Oncology, Hunan Cancer Hospital, Changsha, 410031, China
| | - Yongsheng Wang
- Thoracic Oncology Ward/Cancer Center, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Hong Jian
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200030, China
| | - Zibin Liang
- Department of Thoracic Oncology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, China
| | - Sheng Hu
- Department of Medical Oncology, Hubei Cancer Hospital, Wuhan, 430000, China
| | - Min Peng
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430200, China
| | - Yan Yu
- Department of Thoracic Medicine, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Yan Wang
- Department of Medical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Zicong Jiao
- Geneplus-Beijing, Co., Ltd., Beijing, 102206, China
| | - Kaijing Zhao
- Jiangsu Hengrui Pharmaceuticals, Co., Ltd., Shanghai, 200120, China
| | - Ke Song
- Jiangsu Hengrui Pharmaceuticals, Co., Ltd., Shanghai, 200120, China
| | - You Li
- Jiangsu Hengrui Pharmaceuticals, Co., Ltd., Shanghai, 200120, China
| | - Wei Shi
- Jiangsu Hengrui Pharmaceuticals, Co., Ltd., Shanghai, 200120, China
| | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200030, China.
| |
Collapse
|
234
|
Hu J, Li X, Wang Y, Xu B, He P, Wang Z, He L, Chen H. SOX combined with apatinib and camrelizumab in the treatment of resectable locally advanced gastric cancer: a case report. Front Immunol 2024; 15:1410284. [PMID: 39072331 PMCID: PMC11272450 DOI: 10.3389/fimmu.2024.1410284] [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: 03/31/2024] [Accepted: 06/28/2024] [Indexed: 07/30/2024] Open
Abstract
Gastric cancer is highly prevalent in China, yet early diagnosis and overall survival rates are low. The primary treatment strategy is comprehensive therapy centered on surgery. Studies indicate that neoadjuvant chemotherapy can enhance radical resection rates and extend survival in locally advanced gastric cancer. Combining VEGFR inhibitors with chemotherapy improves efficacy in digestive system tumors, while PD-1/PD-L1 inhibitors combined with anti-angiogenesis agents or chemotherapy show synergistic effects. This report presents a case of gastric adenocarcinoma (cT3N1M0) treated with SOX, apatinib mesylate, and camrelizumab as neoadjuvant therapy, followed by D2 distal gastrectomy and postoperative adjuvant therapy with the same regimen. The patient completed all treatment cycles successfully. Post-neoadjuvant therapy, only focal residual cancer cells were found in the lamina propria (pT1a). During postoperative adjuvant therapy follow-up, gastroscopic biopsy indicated a pathological complete response with no recurrence or metastasis. The patient primarily experienced dyspepsia, oropharyngeal pain, capillary proliferation, mild bone marrow suppression, nausea, and vomiting as side effects. Therefore, SOX combined with apatinib mesylate and camrelizumab shows promise for treating resectable locally advanced gastric cancer.
Collapse
Affiliation(s)
- JiKe Hu
- The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Key Laboratory of Environmental Oncology, The Second Hospital and Clinical Medical School, Lanzhou, China
| | - Xuemei Li
- The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Key Laboratory of Environmental Oncology, The Second Hospital and Clinical Medical School, Lanzhou, China
| | - Yunpeng Wang
- The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Key Laboratory of Environmental Oncology, The Second Hospital and Clinical Medical School, Lanzhou, China
| | - Bo Xu
- The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Key Laboratory of Environmental Oncology, The Second Hospital and Clinical Medical School, Lanzhou, China
| | - Puyi He
- The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Key Laboratory of Environmental Oncology, The Second Hospital and Clinical Medical School, Lanzhou, China
| | - Zhuanfang Wang
- The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Key Laboratory of Environmental Oncology, The Second Hospital and Clinical Medical School, Lanzhou, China
| | - Lijuan He
- The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Key Laboratory of Environmental Oncology, The Second Hospital and Clinical Medical School, Lanzhou, China
| | - Hao Chen
- The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Provincial Key Laboratory of Environmental Oncology, The Second Hospital and Clinical Medical School, Lanzhou, China
- Department of Surgical Oncology, The Second Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
235
|
Rafnsdottir S, Jang K, Halldorsdottir ST, Vinod M, Tomasdottir A, Möller K, Halldorsdottir K, Reynisdottir T, Atladottir LH, Allison KE, Ostacolo K, He J, Zhang L, Northington FJ, Magnusdottir E, Chavez-Valdez R, Anderson KJ, Bjornsson HT. SMYD5 is a regulator of the mild hypothermia response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.11.540170. [PMID: 37333301 PMCID: PMC10274674 DOI: 10.1101/2023.05.11.540170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
The mild hypothermia response (MHR) maintains organismal homeostasis during cold exposure and is thought to be critical for the neuroprotection documented with therapeutic hypothermia. To date, little is known about the transcriptional regulation of the MHR. We utilize a forward CRISPR-Cas9 mutagenesis screen to identify the histone lysine methyltransferase SMYD5 as a regulator of the MHR. SMYD5 represses the key MHR gene SP1 at euthermia. This repression correlates with temperature-dependent levels of H3K36me3 at the SP1-locus and globally, indicating that the mammalian MHR is regulated at the level of histone modifications. We have identified 37 additional SMYD5 regulated temperature-dependent genes, suggesting a broader MHR-related role for SMYD5. Our study provides an example of how histone modifications integrate environmental cues into the genetic circuitry of mammalian cells and provides insights that may yield therapeutic avenues for neuroprotection after catastrophic events.
Collapse
Affiliation(s)
- Salvor Rafnsdottir
- Louma G. Laboratory of Epigenetic Research, Faculty of Medicine, University of Iceland; Reykjavik, Iceland
| | - Kijin Jang
- Louma G. Laboratory of Epigenetic Research, Faculty of Medicine, University of Iceland; Reykjavik, Iceland
| | - Sara Tholl Halldorsdottir
- Louma G. Laboratory of Epigenetic Research, Faculty of Medicine, University of Iceland; Reykjavik, Iceland
| | - Meghna Vinod
- Louma G. Laboratory of Epigenetic Research, Faculty of Medicine, University of Iceland; Reykjavik, Iceland
| | - Arnhildur Tomasdottir
- Louma G. Laboratory of Epigenetic Research, Faculty of Medicine, University of Iceland; Reykjavik, Iceland
| | - Katrin Möller
- Louma G. Laboratory of Epigenetic Research, Faculty of Medicine, University of Iceland; Reykjavik, Iceland
| | - Katrin Halldorsdottir
- Louma G. Laboratory of Epigenetic Research, Faculty of Medicine, University of Iceland; Reykjavik, Iceland
| | - Tinna Reynisdottir
- Louma G. Laboratory of Epigenetic Research, Faculty of Medicine, University of Iceland; Reykjavik, Iceland
| | - Laufey Halla Atladottir
- Louma G. Laboratory of Epigenetic Research, Faculty of Medicine, University of Iceland; Reykjavik, Iceland
| | | | - Kevin Ostacolo
- Louma G. Laboratory of Epigenetic Research, Faculty of Medicine, University of Iceland; Reykjavik, Iceland
- Department of Genetics and Molecular Medicine, Landspitali University Hospital; Reykjavik, Iceland
| | - Jin He
- Department of Biochemistry and Molecular Biology, College of Natural Science, Michigan State University; MI, USA
| | - Li Zhang
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine; Baltimore, MD, USA
| | - Frances J Northington
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine; Baltimore, MD, USA
- Neuroscience Intensive Care Nursery Program, Johns Hopkins University; Baltimore, MD, USA
| | - Erna Magnusdottir
- Department of Biomedical Science and Department of Anatomy, Faculty of Medicine, University of Iceland; Reykjavík, Iceland
| | - Raul Chavez-Valdez
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine; Baltimore, MD, USA
- Neuroscience Intensive Care Nursery Program, Johns Hopkins University; Baltimore, MD, USA
| | - Kimberley Jade Anderson
- Department of Genetics and Molecular Medicine, Landspitali University Hospital; Reykjavik, Iceland
| | - Hans Tomas Bjornsson
- Louma G. Laboratory of Epigenetic Research, Faculty of Medicine, University of Iceland; Reykjavik, Iceland
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine; Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University; Baltimore, MD, USA
- Department of Genetics and Molecular Medicine, Landspitali University Hospital; Reykjavik, Iceland
- Lead contact
| |
Collapse
|
236
|
Xu R, Zhang Y, Wang Z, Chen K, Zhao J. Construction and validation of a prognostic model for gastric cancer patients with tumor deposits. PeerJ 2024; 12:e17751. [PMID: 39006037 PMCID: PMC11246019 DOI: 10.7717/peerj.17751] [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: 04/23/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
Background Tumor deposits (TD) was a significant risk factor impacting the prognosis of patients diagnosed with gastric cancer (GC), yet it was not currently incorporated into TNM staging systems. The objective of this research was to develop a predictive model for assessing the prognosis of patients with TD-positive GC. Methods Retrospective analysis was performed on the data of 4,972 patients treated for GC with D2 radical gastrectomy at Wannan Medical College's Yijishan Hospital between January 2012 and December 2021. The patients were categorized based on the number of TD (L1: 1, L2: 2-3, L3: ≥4) and the anatomical location of TD (Q1: single area, Q2: multiple areas). In a 3:1 ratio, patients were randomly assigned to one of two groups: training or validation. Results The study included a total of 575 patients who were divided into the training group (n = 432) and validation group (n = 143). Survival analysis showed that the number and anatomical location of TD had a significant impact on the prognosis of patients with TD-positive GC. Univariate analysis of the training group data revealed that tumor size, T-stage, N-stage, histological grade, number and distribution of TD, neural invasion, and postoperative chemotherapy were associated with prognosis. Multivariate Cox regression analysis identified poor histological grade, T4 stage, N3 stage, number of TD, neural invasion, and postoperative chemotherapy as independent prognostic factors for GC patients with TD. A nomogram was developed using these variables, demonstrating well predictive ability for 1, 3, and 5-year overall survival (OS) in the validation set. The DCA curve shows that the constructed model shows a large positive net gain compared to the eighth edition Tumour, Node, Metastasis (TNM) staging system. Conclusion The prognostic model developed for patients with TD-positive GC has a higher clinical utility compared to the eighth edition of TNM staging.
Collapse
Affiliation(s)
- Ran Xu
- Department of General Surgery, The Yijishan Hospital of Wannan Medical College, Wuhu, China
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yisheng Zhang
- Department of General Surgery, The Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Zhengguang Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ke Chen
- Department of Vascular Surgery, Drum Tower Hospital, Jiangsu, China
| | - Jun Zhao
- Department of General Surgery, The Yijishan Hospital of Wannan Medical College, Wuhu, China
| |
Collapse
|
237
|
Oladeru O, Rajack F, Esnakula A, Naab TJ, Kanaan Y, Ricks-Santi L. Beyond Triple-Negative: High Prevalence of Quadruple-Negative Breast Cancer in African Americans. Biomedicines 2024; 12:1522. [PMID: 39062096 PMCID: PMC11275194 DOI: 10.3390/biomedicines12071522] [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: 05/06/2024] [Revised: 06/13/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
Quadruple-negative breast cancer (QNBC) is a triple-negative breast cancer (TNBC) subtype that lacks expression of the androgen (AR) receptor. Few studies have focused on this highly aggressive breast cancer, portending worse survival rates. We aimed to determine the following: (1) QNBC's molecular and clinical characteristics and compare them with other subtypes and (2) QNBC's association with clinicopathological factors and prognostic markers. We performed immunohistochemical evaluations of ARs on tissue tumor microarrays from FFPE tumor blocks of invasive ductal breast carcinomas in 202 African American women. Univariate analysis was performed using the chi-square test, with survival rates calculated using Kaplan-Meier curves. Overall, 75.8% of TNBCs were AR-negative. Compared to the luminal subtypes, TNBC and QNBC tumors were likely to be a higher grade (p < 0.001); HER2+/AR- and QNBCs were also larger than the other subtypes (p < 0.001). They also expressed increasing mean levels of proteins involved in invasion, such as CD44, fascin, and vimentin, as well as decreasing the expression of proteins involved in mammary differentiation, such as GATA3 and mammaglobin. We found no association between QNBC and stage, recurrence-free survival, or overall survival rates. The high prevalence of TNBC AR-negativity in these women could explain observed worse outcomes, supporting the existence of the unique QNBC subtype.
Collapse
Affiliation(s)
| | - Fareed Rajack
- Department of Pathology, Howard University Hospital, Washington, DC 20059, USA; (F.R.)
| | - Ashwini Esnakula
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Tammey J. Naab
- Department of Pathology, Howard University Hospital, Washington, DC 20059, USA; (F.R.)
| | - Yasmine Kanaan
- Department of Microbiology, Howard University College of Medicine, Washington, DC 20059, USA
| | - Luisel Ricks-Santi
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, FL 32610, USA
| |
Collapse
|
238
|
Mizoguchi C, Nishikawa T, Yoshida H, Yasuda M, Kato T, Hasegawa K, Yonemori K. HER2-negative or low expression as an unfavorable prognostic factor in patients with stage I/II uterine carcinosarcoma. J Gynecol Oncol 2024; 36:36.e14. [PMID: 38991945 DOI: 10.3802/jgo.2025.36.e14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/03/2024] [Accepted: 06/11/2024] [Indexed: 07/13/2024] Open
Abstract
OBJECTIVE Uterine carcinosarcoma (UCS) is uncommon high-grade endometrial cancer with limited treatment options. We evaluated the prognostic significance of human epidermal growth factor receptor 2 (HER2) expression and HER2 gene amplification within large cohorts of UCS, and clarify clinicopathologic characteristics of HER2-low UCS. METHODS We examined HER2 protein expression in 148 patients of UCS using in vivo diagnostic HER2 immunohistochemistry (IHC) kits and HER2 gene amplification using fluorescence in situ hybridization (FISH) in 72 patients. RESULTS HER2 IHC score was evaluated according to the latest American Society of Clinical Oncology/College of American Pathologists criteria for gastric cancer, which was negative in 41 patients, low expression of 1+ was observed in 57 patients, and HER2 high expression was observed in 50 patients (2+ in 38 and 3+ in 12 patients). There was no significant statistical difference in clinicopathological characteristics based on HER2 protein expression status. HER2 negative and low expression compared to high expression revealed poor overall survival in stage I/ II. The concordance between IHC and FISH results were relatively low compared to other cancer types (HER2 IHC score 1+, 2+, and 3+ were 5%, 15%, and 50%), and combining these results was not efficient as a prognostic factor in UCS. In contrast, the HER2 IHC score alone was a prognostic factor in stage I/II UCS. HER2 low group did not show specific clinicopathologic features. CONCLUSION Since the HER2 IHC score low in advanced UCS is a predictive factor, stratification of UCS using HER2 IHC score for HER2 IHC score low group and developing adjuvant therapy may be proposed in the near future.
Collapse
Affiliation(s)
- Chiharu Mizoguchi
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Tadaaki Nishikawa
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan.
| | - Hiroshi Yoshida
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
| | - Masanori Yasuda
- Department of Diagnostic Pathology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Tomoyasu Kato
- Department of Gynecology, National Cancer Center Hospital, Tokyo, Japan
| | - Kosei Hasegawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Kan Yonemori
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
239
|
Li N, Wu H, Xu X, Wei Q, Ding Y, Liu S, Wu J, Zheng Y, Xu N, Gao Y, Jiang H. A retrospective study of adjuvant albumin-bound paclitaxel plus S-1 after D2 gastrectomy versus oxaliplatin plus S-1 in gastric cancer. Sci Rep 2024; 14:15150. [PMID: 38956232 PMCID: PMC11219745 DOI: 10.1038/s41598-024-65724-8] [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: 04/26/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024] Open
Abstract
Adjuvant oxaliplatin plus S-1 (SOX) chemotherapy for gastric cancer (GC) after D2 gastrectomy has been proven effective. There has yet to be a study that evaluates adjuvant nanoparticle albumin-bound paclitaxel (nab-paclitaxel) plus S-1. In this single-center, retrospective study, GC patients after D2 gastrectomy received either nab-paclitaxel plus S-1 (AS group) or SOX group were recruited between January 2018 and December 2020 in The First Affiliated Hospital of Zhejiang University. Intravenous nab-paclitaxel 120 mg/m2 or 260 mg/m2 and oxaliplatin 130 mg/m2 were administered as eight 3 week cycle, especially in the AS and SOX group. Patients received S-1 twice daily with a dose of 40 mg/m2 in the two groups on days 1-14 of each cycle. The end points were disease-free survival (DFS) rate at 3 years and adverse events (AEs). There were 56 eligible patients, 28 in the AS group and 35 in the SOX group. The 3 year DFS rate was 78.0% in AS group versus 70.7% in SOX group (p = 0.46). Subgroup analysis showed that the patients with signet-ring positive in the AS group had a prolonged DFS compared with the SOX group (40.0 vs. 13.8 m, p = 0.02). The diffuse-type GC or low differentiation in the AS group was associated with numerically prolonged DFS compared with the SOX group, but the association was not statistically significant (p = 0.27 and p = 0.15 especially). Leukopenia (14.3%) were the most prevalent AEs in the AS group, while thrombocytopenia (28.5%) in the SOX group. Neutropenia (7.1% in AS group) and thrombocytopenia (22.8% in SOX group) were the most common grade 3 or 4 AEs. In this study analyzing past data, a tendency towards a greater 3 year DFS was observed when using AS regimen in signet-ring positive patients. AS group had fewer thrombocytopenia compared to SOX group. More studies should be conducted with larger sample sizes.
Collapse
Affiliation(s)
- Ning Li
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310001, China
| | - Hui Wu
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310001, China
| | - Xin Xu
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310001, China
| | - Qinming Wei
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310001, China
| | - Yongfeng Ding
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310001, China
| | - Shan Liu
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310001, China
| | - Jinqiong Wu
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310001, China
| | - Yulong Zheng
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310001, China
| | - Nong Xu
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310001, China
| | - Yuan Gao
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310001, China
| | - Haiping Jiang
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310001, China.
| |
Collapse
|
240
|
Fong C, Patel B, Peckitt C, Bourmpaki E, Satchwell L, Cromarty S, Kidd S, von Loga K, Uhlik M, Begum R, Rana T, Waddell T, Darby S, Bradshaw A, Roques T, Morgan C, Rees C, Herbertson R, Das P, Thompson C, Hewish M, Petty R, Thistlethwaite F, Rao S, Starling N, Chau I, Cunningham D. Maintenance durvalumab after first-line chemotherapy in patients with HER2-negative advanced oesophago-gastric adenocarcinoma: results from the randomised PLATFORM study. ESMO Open 2024; 9:103622. [PMID: 39002179 PMCID: PMC11298822 DOI: 10.1016/j.esmoop.2024.103622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/26/2024] [Accepted: 05/31/2024] [Indexed: 07/15/2024] Open
Abstract
BACKGROUND PLAnning Treatment For Oesophago-gastric Cancer: a Randomised Maintenance Therapy Trial (PLATFORM) is an adaptive phase II study assessing the role of maintenance therapies in advanced oesophago-gastric (OG) adenocarcinoma. We evaluated the role of the anti-programmed death-ligand 1 (PD-L1) inhibitor durvalumab in these patients. PATIENTS AND METHODS Patients with human epidermal growth factor receptor 2-negative locally advanced or metastatic OG adenocarcinoma with disease control or response to 18 weeks of platinum-based first-line chemotherapy were randomised to active surveillance or maintenance durvalumab. The primary endpoint was progression-free survival (PFS). Safety was assessed in all patients who had commenced surveillance visits or received at least one dose of durvalumab. Exploratory survival analyses according to PD-L1 Combined Positive Score (CPS) and immune (biomarker-positive) or angiogenesis dominant (biomarker-negative) tumour microenvironment (TME) phenotypes were conducted. RESULTS Between March 2015 and April 2020, 205 patients were randomised to surveillance (n = 100) and durvalumab (n = 105). No significant differences were seen in PFS [hazard ratio (HR) 0.84, P = 0.13] and overall survival (OS; HR 0.98, P = 0.45) between surveillance and durvalumab. Five patients randomised to durvalumab demonstrated incremental radiological responses compared with none with surveillance. Treatment-related adverse events occurred in 77 (76.2%) durvalumab-assigned patients. A favourable effect in OS with durvalumab over surveillance in CPS ≥5 and immune biomarker-positive patients was observed compared with CPS <5 and biomarker-negative subgroups, respectively: CPS ≥5 versus <5: HR 0.63, 95% confidence interval (CI) 0.32-1.22 versus HR 0.93, 95% CI 0.44-1.96; biomarker-positive versus negative: HR 0.60, 95% CI 0.29-1.23 versus HR 0.84, 95% CI 0.42-1.65. CONCLUSION Maintenance durvalumab does not improve PFS in patients with OG adenocarcinoma who respond to first-line chemotherapy but induced incremental radiological responses in a subset of patients. TME characterisation could refine patient selection for anti-PD-L1 therapy above PD-L1 CPS alone.
Collapse
Affiliation(s)
- C Fong
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey
| | - B Patel
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey
| | - C Peckitt
- Research Data & Statistics Unit, Royal Marsden Clinical Trials Unit, Royal Marsden Hospital, London and Surrey
| | - E Bourmpaki
- Research Data & Statistics Unit, Royal Marsden Clinical Trials Unit, Royal Marsden Hospital, London and Surrey
| | - L Satchwell
- Research Data & Statistics Unit, Royal Marsden Clinical Trials Unit, Royal Marsden Hospital, London and Surrey
| | - S Cromarty
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey
| | - S Kidd
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey
| | - K von Loga
- Integrative Pathology Unit, Institute of Cancer Research, Sutton, UK
| | - M Uhlik
- Research and Biomarker Discovery, OncXerna Therapeutics, Inc., Waltham, USA
| | - R Begum
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey
| | - T Rana
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey
| | - T Waddell
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester
| | - S Darby
- Weston Park Cancer Centre, Sheffield
| | - A Bradshaw
- Cancer Services, The Freeman Hospital's Northern Centre for Cancer Care, Newcastle
| | - T Roques
- Department of Clinical Oncology, Norfolk and Norwich University Hospital, Norfolk & Norwich
| | - C Morgan
- Department of Clinical Oncology, Velindre Cancer Centre, Cardiff
| | - C Rees
- Cancer Care Division, Medical Oncology Department, University Hospital Southampton NHS Foundation Trust, Southampton
| | - R Herbertson
- Sussex Cancer Centre, Royal Sussex County Hospital, Brighton
| | - P Das
- Department of Oncology, University Hospitals of Derby and University of Nottingham School of Medicine
| | - C Thompson
- University Hospitals of Morecambe Bay NHS Foundation Trust, Kendal
| | - M Hewish
- Royal Surrey Cancer Centre, Royal Surrey Hospitals NHS Foundation Trust, Guildford
| | - R Petty
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee
| | - F Thistlethwaite
- Department of Medical Oncology, The Christie NHS Foundation Trust and Division of Cancer Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - S Rao
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey
| | - N Starling
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey
| | - I Chau
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey
| | - D Cunningham
- Gastrointestinal Unit, Department of Medicine, Royal Marsden Hospital, London and Surrey.
| |
Collapse
|
241
|
Dai MF, Wang X, Xin WX, Kong SS, Xu WB, Ding HY, Fang L. Safety and hematological toxicities of PARP inhibitors in patients with cancer: a systematic review of randomized controlled trials and a pharmacovigilance analysis. Expert Rev Anticancer Ther 2024; 24:613-622. [PMID: 38761169 DOI: 10.1080/14737140.2024.2357822] [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: 11/03/2023] [Accepted: 05/07/2024] [Indexed: 05/20/2024]
Abstract
INTRODUCTION This study aimed to estimate the toxicities of PARP inhibitors (PARPis), based on randomized controlled trials (RCTs) and the FDA Adverse Event Reporting System (FAERS) database. METHODS Four electronic databases were searched from inception to 16 April 2024, for RCTs of approved PARPis. The primary and secondary outcomes were grade 3-5 adverse events (AEs) and grade 3-5 hematological AE, respectively. We conducted network meta-analyses to calculate the relative risks (RRs) and 95% confidence intervals (CIs) of outcomes. A disproportionality analysis was conducted to estimate the signals of hematological AEs associated with PARPis from the FAERS database. RESULTS Overall, 27 RCTs involving 11,067 patients with cancer were included. Olaparib had the best safety profile for any grade 3-5 AEs and hematological AEs among four approved PARPis. Olaparib did not increase the risk of thrombocytopenia (RR: 1.48; 95%CI: 0.64-3.39), but other PARPis did. Furthermore 14,780 hematological AE reports associated with PARPis were identified in the FAERS database, and all PARPis were associated with strong hematological AE signals. Hematological AEs mainly occurred within the first 3 months (80.84%) after PARPi initiation. CONCLUSION Olaparib had the best safety profile among five PARPis. PARPi-associated hematological AEs mainly occurred within the first 3 months. REGISTRATION PROSPERO (CRD42022385274).
Collapse
Affiliation(s)
- Meng-Fei Dai
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Xin Wang
- Department of Pharmacy, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen-Xiu Xin
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Si-Si Kong
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Wei-Ben Xu
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Hai-Ying Ding
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Luo Fang
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| |
Collapse
|
242
|
Nishibata T, Weng J, Omori K, Sato Y, Nakazawa T, Suzuki T, Yamada T, Nakajo I, Kinugasa F, Türeci Ö, Şahin U, Yoshida T. Effect of anti-claudin 18.2 monoclonal antibody zolbetuximab alone or combined with chemotherapy or programmed cell death-1 blockade in syngeneic and xenograft gastric cancer models. J Pharmacol Sci 2024; 155:84-93. [PMID: 38797537 DOI: 10.1016/j.jphs.2024.04.004] [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: 12/12/2023] [Revised: 04/12/2024] [Accepted: 04/17/2024] [Indexed: 05/29/2024] Open
Abstract
The development of targeted cancer therapies based on monoclonal antibodies against tumor-associated antigens has progressed markedly over recent decades. This approach is dependent on the identification of tumor-specific, normal tissue-sparing antigenic targets. The transmembrane protein claudin-18 splice variant 2 (CLDN18.2) is frequently and preferentially displayed on the surface of primary gastric adenocarcinomas, making it a promising monoclonal antibody target. Phase 3 studies of zolbetuximab, a chimeric immunoglobulin G1 monoclonal antibody targeting CLDN18.2, combined with 5-fluorouracil/leucovorin plus oxaliplatin (modified FOLFOX6) or capecitabine plus oxaliplatin (CAPOX) in advanced or metastatic first-line gastric or gastroesophageal junction (G/GEJ) adenocarcinoma have demonstrated favorable clinical results with zolbetuximab. In studies using xenograft or syngeneic models with gastric cancer cell lines, zolbetuximab mediated death of CLDN18.2-positive human cancer cell lines via antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity in vitro and demonstrated anti-tumor efficacy as monotherapy and combined with chemotherapy in vivo. Mice treated with zolbetuximab plus chemotherapy displayed a significantly higher frequency of tumor-infiltrating CD8+ T cells versus vehicle/isotype control-treated mice. Furthermore, zolbetuximab combined with an anti-mouse programmed cell death-1 antibody more potently inhibited tumor growth compared with either agent alone. These results support the potential of zolbetuximab as a novel treatment option for G/GEJ adenocarcinoma.
Collapse
Affiliation(s)
| | - Jane Weng
- Astellas Pharma, Inc., Ibaraki, Japan
| | | | - Yuji Sato
- Astellas Pharma, Inc., Ibaraki, Japan
| | | | | | | | | | | | - Özlem Türeci
- Formerly of Ganymed Pharmaceuticals AG, Mainz, Germany; Biontech SE, Mainz, Germany; Helmholtz Institute for Translational Oncology (HI-TRON) By DKFZ, Johannes Gutenberg University, Mainz, Germany
| | - Uğur Şahin
- Formerly of Ganymed Pharmaceuticals AG, Mainz, Germany; Biontech SE, Mainz, Germany; Helmholtz Institute for Translational Oncology (HI-TRON) By DKFZ, Johannes Gutenberg University, Mainz, Germany
| | | |
Collapse
|
243
|
Seadawy MG, Lotfy MM, Saeed AA, Ageez AM. Novel HER2-based multi-epitope vaccine (HER2-MEV) against HER2-positive breast cancer: In silico design and validation. Hum Immunol 2024; 85:110832. [PMID: 38905717 DOI: 10.1016/j.humimm.2024.110832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/10/2024] [Accepted: 06/10/2024] [Indexed: 06/23/2024]
Abstract
Breast cancer (BC) continues to be the malignancy with the highest diagnosis rate worldwide. Between 15 % and 30 % of BC patients show overexpressed human epidermal growth factor receptor 2 (HER2), which is linked to poor clinical results in terms of invasiveness and recurrence risk. Passive immunity-based therapeutic approaches for treating HER2-enriched BC, are not effective and significant problems need to be tackled. Constructing multi-epitope vaccines is favored over single-epitope vaccines due to its ability to induce immunity against a variety of antigenic targets which will improve the efficacy of the vaccine. The current study describes a multi-epitope vaccine from HER2 protein against HER2-positive BC using several immunoinformatic techniques to achieve a potent and durable immune response. Nine Cytotoxic T lymphocytes (CTL) and five Helper T lymphocytes (HTL) epitopes were predicted and validated from HER2 protein using in silico tools. The expressed protein of the designed vaccine is predicted to be highly thermostable with better solubility. The predicted vaccine 3D structure was validated by ProSA servers and by the ERRAT server. Molecular docking analysis revealed a high binding affinity and stability of the designed vaccine with MHCI and TLR-2, 4, 7, and 9 receptors. The analysis of the C-ImmSim server revealed that the novel vaccine construct had the ability to elicit robust anti-cancerous innate, humoral, and cell-mediated immune responses. The vaccine can be a suitable option for HER2-positive BC patients and other patients with HER2-positive cancers to evoke immune responses. However, in vitro and in vivo experiments are needed to assess its effectiveness and safety.
Collapse
Affiliation(s)
- Mohamed G Seadawy
- Biodefense Center for Infectious and Emerging Diseases, Ministry of Defense, Cairo, Egypt.
| | - Mai M Lotfy
- Cancer Biology Department, National Cancer Institute, Cairo University, Giza 12613, Egypt.
| | - Aya A Saeed
- Cancer Biology Department, National Cancer Institute, Cairo University, Giza 12613, Egypt.
| | - Amr M Ageez
- Faculty of Biotechnology, October University for Modern Sciences and Arts, MSA University, 6 October City 12451, Giza, Egypt.
| |
Collapse
|
244
|
Jenke R, Oliinyk D, Zenz T, Körfer J, Schäker-Hübner L, Hansen FK, Lordick F, Meier-Rosar F, Aigner A, Büch T. HDAC inhibitors activate lipid peroxidation and ferroptosis in gastric cancer. Biochem Pharmacol 2024; 225:116257. [PMID: 38705532 DOI: 10.1016/j.bcp.2024.116257] [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: 01/18/2024] [Revised: 04/18/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
Gastric cancer remains among the deadliest neoplasms worldwide, with limited therapeutic options. Since efficacies of targeted therapies are unsatisfactory, drugs with broader mechanisms of action rather than a single oncogene inhibition are needed. Preclinical studies have identified histone deacetylases (HDAC) as potential therapeutic targets in gastric cancer. However, the mechanism(s) of action of HDAC inhibitors (HDACi) are only partially understood. This is particularly true with regard to ferroptosis as an emerging concept of cell death. In a panel of gastric cancer cell lines with different molecular characteristics, tumor cell inhibitory effects of different HDACi were studied. Lipid peroxidation levels were measured and proteome analysis was performed for the in-depth characterization of molecular alterations upon HDAC inhibition. HDACi effects on important ferroptosis genes were validated on the mRNA and protein level. Upon HDACi treatment, lipid peroxidation was found increased in all cell lines. Class I HDACi (VK1, entinostat) showed the same toxicity profile as the pan-HDACi vorinostat. Proteome analysis revealed significant and concordant alterations in the expression of proteins related to ferroptosis induction. Key enzymes like ACSL4, POR or SLC7A11 showed distinct alterations in their expression patterns, providing an explanation for the increased lipid peroxidation. Results were also confirmed in primary human gastric cancer tissue cultures as a relevant ex vivo model. We identify the induction of ferroptosis as new mechanism of action of class I HDACi in gastric cancer. Notably, these findings were independent of the genetic background of the cell lines, thus introducing HDAC inhibition as a more general therapeutic principle.
Collapse
Affiliation(s)
- Robert Jenke
- University Cancer Center Leipzig (UCCL), University Hospital Leipzig, Leipzig, Germany; Leipzig University, Medical Faculty, Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, Leipzig, Germany; Comprehensive Cancer Center Central Germany (CCCG), Leipzig and Jena, Germany
| | - Denys Oliinyk
- Jena University Hospital, Functional Proteomics, Research Center Lobeda, Jena, Germany
| | - Tamara Zenz
- Leipzig University, Medical Faculty, Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, Leipzig, Germany
| | - Justus Körfer
- University Cancer Center Leipzig (UCCL), University Hospital Leipzig, Leipzig, Germany; University Hospital Leipzig, Institute for Anatomy, Leipzig, Germany
| | - Linda Schäker-Hübner
- University of Bonn, Pharmaceutical Institute, Department of Pharmaceutical and Cell Biological Chemistry, Bonn, Germany
| | - Finn K Hansen
- University of Bonn, Pharmaceutical Institute, Department of Pharmaceutical and Cell Biological Chemistry, Bonn, Germany
| | - Florian Lordick
- University Cancer Center Leipzig (UCCL), University Hospital Leipzig, Leipzig, Germany; Comprehensive Cancer Center Central Germany (CCCG), Leipzig and Jena, Germany
| | - Florian Meier-Rosar
- Jena University Hospital, Functional Proteomics, Research Center Lobeda, Jena, Germany; Comprehensive Cancer Center Central Germany (CCCG), Leipzig and Jena, Germany
| | - Achim Aigner
- Leipzig University, Medical Faculty, Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, Leipzig, Germany; Comprehensive Cancer Center Central Germany (CCCG), Leipzig and Jena, Germany.
| | - Thomas Büch
- Leipzig University, Medical Faculty, Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, Leipzig, Germany; Comprehensive Cancer Center Central Germany (CCCG), Leipzig and Jena, Germany
| |
Collapse
|
245
|
Jiang L, Zhao X, Li Y, Hu Y, Sun Y, Liu S, Zhang Z, Li Y, Feng X, Yuan J, Li J, Zhang X, Chen Y, Shen L. The tumor immune microenvironment remodeling and response to HER2-targeted therapy in HER2-positive advanced gastric cancer. IUBMB Life 2024; 76:420-436. [PMID: 38126920 DOI: 10.1002/iub.2804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/26/2023] [Indexed: 12/23/2023]
Abstract
Combination therapy with anti-HER2 agents and immunotherapy has demonstrated significant clinical benefits in gastric cancer (GC), but the underlying mechanism remains unclear. In this study, we used multiplex immunohistochemistry to assess the changes of the tumor microenvironment in 47 advanced GC patients receiving anti-HER2 therapy. Additionally, we performed single-cell transcriptional sequencing to investigate potential cell-to-cell communication and molecular mechanisms in four HER2-positive GC baseline samples. We observed that post-treated the infiltration of NK cells, CD8+ T cells, and B lymphocytes were significantly higher in patients who benefited from anti-HER2 treatment than baseline. Further spatial distribution analysis demonstrated that the interaction scores between NK cells and CD8+ T cells, B lymphocytes and M2 macrophages, B lymphocytes and Tregs were also significantly higher in benefited patients. Cell-cell communication analysis from scRNA sequencing showed that NK cells utilized CCL3/CCL4-CCR5 to recruit CD8+ T cell infiltration. B lymphocytes employed CD74-APP/COPA/MIF to interact with M2 macrophages, and utilized TNF-FAS/ICOS/TNFRSR1B to interact with Tregs. These cell-cell interactions contribute to inhibit the immune resistance of M2 macrophages and Tregs. Our research provides potential guidance for the use of anti-HER2 therapy in combination with immune therapy.
Collapse
Affiliation(s)
- Lei Jiang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xingwang Zhao
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yilin Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yajie Hu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yu Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Shengde Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zizhen Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yanyan Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xujiao Feng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jiajia Yuan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jian Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaotian Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yang Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Lin Shen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
246
|
Khan IR, Sadida HQ, Hashem S, Singh M, Macha MA, Al-Shabeeb Akil AS, Khurshid I, Bhat AA. Therapeutic implications of signaling pathways and tumor microenvironment interactions in esophageal cancer. Biomed Pharmacother 2024; 176:116873. [PMID: 38843587 DOI: 10.1016/j.biopha.2024.116873] [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: 03/24/2024] [Revised: 05/21/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024] Open
Abstract
Esophageal cancer (EC) is significantly influenced by the tumor microenvironment (TME) and altered signaling pathways. Downregulating these pathways in EC is essential for suppressing tumor development, preventing metastasis, and enhancing therapeutic outcomes. This approach can increase tumor sensitivity to treatments, enhance patient outcomes, and inhibit cancer cell proliferation and spread. The TME, comprising cellular and non-cellular elements surrounding the tumor, significantly influences EC's development, course, and treatment responsiveness. Understanding the complex relationships within the TME is crucial for developing successful EC treatments. Immunotherapy is a vital TME treatment for EC. However, the heterogeneity within the TME limits the application of anticancer drugs outside clinical settings. Therefore, identifying reliable microenvironmental biomarkers that can detect therapeutic responses before initiating therapy is crucial. Combining approaches focusing on EC signaling pathways with TME can enhance treatment outcomes. This integrated strategy aims to interfere with essential signaling pathways promoting cancer spread while disrupting factors encouraging tumor development. Unraveling aberrant signaling pathways and TME components can lead to more focused and efficient treatment approaches, identifying specific cellular targets for treatments. Targeting the TME and signaling pathways may reduce metastasis risk by interfering with mechanisms facilitating cancer cell invasion and dissemination. In conclusion, this integrative strategy has significant potential for improving patient outcomes and advancing EC research and therapy. This review discusses the altered signaling pathways and TME in EC, focusing on potential future therapeutics.
Collapse
Affiliation(s)
- Inamu Rashid Khan
- Department of Zoology, Central University of Kashmir, Ganderbal, Jammu and Kashmir 191201, India
| | - Hana Q Sadida
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha 26999, Qatar
| | - Sheema Hashem
- Department of Human Genetics, Sidra Medicine Doha 26999, Qatar
| | - Mayank Singh
- Department of Medical Oncology (Lab), Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Muzafar A Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Awantipora, Jammu and Kashmir 192122, India
| | - Ammira S Al-Shabeeb Akil
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha 26999, Qatar
| | - Ibraq Khurshid
- Department of Zoology, Central University of Kashmir, Ganderbal, Jammu and Kashmir 191201, India.
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha 26999, Qatar.
| |
Collapse
|
247
|
Kono K, Nakajima S, Mimura K. Biomarker-oriented chemo-immunotherapy for advanced gastric cancer. Int J Clin Oncol 2024; 29:865-872. [PMID: 38647874 DOI: 10.1007/s10147-024-02525-z] [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: 03/18/2024] [Accepted: 04/01/2024] [Indexed: 04/25/2024]
Abstract
The biomarker-oriented chemo-immunotherapy is useful and promising in the development of new anticancer agents, since the responders can be enriched by selecting patients with biomarkers. Compared to colorectal and lung cancers, the development of biomarker-driven molecular-targeted therapeutics for gastric cancers has been straggled. However, several new biomarkers in gastric cancers have been discovered and clinical trials in enrichment design with certain biomarkers have been conducted. Therefore, there are currently several treatment options to treat gastric cancer patients based on individual biomarker-oriented strategies. In the present review, we describe the useful biomarkers in gastric cancer, with focusing on HER2, PD-L1, and Claudin18.2, in relation to their clinical significance and associated targeted agents.
Collapse
Affiliation(s)
- Koji Kono
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, 960-1295, Japan.
| | - Shotaro Nakajima
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, 960-1295, Japan
- Department of Multidisciplinary Treatment of Cancer and Regional Medical Support, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima City, Fukushima, 960-1295, Japan
| | - Kosaku Mimura
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, 960-1295, Japan
- Department of Blood Transfusion and Transplantation Immunology, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, 960-1295, Japan
| |
Collapse
|
248
|
Sert F, Bilkay Gorken I, Ozkok S, Colpan Oksuz D, Yucel B, Kaytan Saglam E, Aksu G, Cetin E, Aktan M, Canyilmaz E, Ozbek Okumus N, Yildirim B, Akyurek S, Serin M, Kurt M, Arican Alicikus Z, Erdis E, Yalman D. Who would be the winner? A prognostic nomogram for predicting the benefit of postoperative radiotherapy ± chemotherapy in patients with locally advanced gastric cancer: TROD-02-01 study. Asian J Surg 2024; 47:3056-3062. [PMID: 38443256 DOI: 10.1016/j.asjsur.2024.02.100] [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: 11/15/2023] [Revised: 01/24/2024] [Accepted: 02/16/2024] [Indexed: 03/07/2024] Open
Abstract
OBJECTIVES We aimed to develop a basic, easily applicable nomogram to improve the survival prediction of the patients with stage II/III gastric cancer (GC) and to select the best candidate for postoperative radiotherapy (RT). METHODS In this multicentric trial, we retrospectively evaluated the data of 1597 patients with stage II/III GC after curative gastrectomy followed by postoperative RT ± chemotherapy (CT). Patients were divided into a training set (n = 1307) and an external validation set (n = 290). Nomograms were created based on independent predictors identified by Cox regression analysis in the training set. The consistency index (C-index) and the calibration curve were used to evaluate the discriminative ability and accuracy of the nomogram. A nomogram was created based on the predictive model and the identified prognostic factors to predict 5-year cancer-specific survival (CSS) and progression-free survival (PFS). RESULTS The multivariate Cox model recognized lymph node (LN) involvement status, lymphatic dissection (LD) width, and metastatic LN ratio as covariates associated with CSS. Depth of invasion, LN involvement status, LD width, metastatic LN ratio, and lymphovascular invasion were the factors associated with PFS. Calibration of the nomogram predicted both CSS and PFS corresponding closely with the actual results. In our validation set, discrimination was good (C-index, 0.76), and the predicted survival was within a 10% margin of ideal nomogram. CONCLUSIONS In our relatively large cohort, we created and validated both CSS and PFS nomograms that could be useful for underdeveloped or developing countries rather than Korea and Japan, where the D2 gastrectomy is routinely performed. This could serve as a true map for oncologists who must make decisions without an experienced surgeon and a multidisciplinary tumor board.
Collapse
Affiliation(s)
- Fatma Sert
- Ege University, Faculty of Medicine, Department of Radiation Oncology, Izmir, Turkey.
| | - Ilknur Bilkay Gorken
- Dokuz Eylul University, Faculty of Medicine, Department of Radiation Oncology, Izmir, Turkey
| | - Serdar Ozkok
- Ege University, Faculty of Medicine, Department of Radiation Oncology, Izmir, Turkey
| | - Didem Colpan Oksuz
- Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Department of Radiation Oncology, Istanbul, Turkey
| | - Birsen Yucel
- Sivas Cumhuriyet University, Faculty of Medicine, Department of Radiation Oncology, Sivas, Turkey
| | | | - Gamze Aksu
- Akdeniz University, Faculty of Medicine, Department of Radiation Oncology, Antalya, Turkey
| | - Eren Cetin
- Gazi University, Faculty of Medicine, Department of Radiation Oncology, Ankara, Turkey
| | - Meryem Aktan
- Necmettin Erbakan University, Faculty of Medicine, Department of Radiation Oncology, Konya, Turkey
| | - Emine Canyilmaz
- Karadeniz Technical University, Faculty of Medicine, Department of Radiation Oncology, Trabzon, Turkey
| | - Nilgün Ozbek Okumus
- On Dokuz Mayıs University, Faculty of Medicine, Department of Radiation Oncology, Samsun, Turkey
| | - Berna Yildirim
- University of Health Sciences, Prof Dr Cemil Tascioglu City Hospital, Department of Radiation Oncology, Istanbul, Turkey
| | - Serap Akyurek
- Ankara University, Faculty of Medicine, Department of Radiation Oncology, Ankara, Turkey
| | - Meltem Serin
- Acıbadem Mehmet Ali Aydinlar University, Adana Hospital, Department of Radiation Oncology, Adana, Turkey
| | - Meral Kurt
- Bursa Uludag University, Faculty of Medicine, Department of Radiation Oncology, Bursa, Turkey
| | - Zumre Arican Alicikus
- Dokuz Eylul University, Faculty of Medicine, Department of Radiation Oncology, Izmir, Turkey
| | - Eda Erdis
- Sivas Cumhuriyet University, Faculty of Medicine, Department of Radiation Oncology, Sivas, Turkey
| | - Deniz Yalman
- Ege University, Faculty of Medicine, Department of Radiation Oncology, Izmir, Turkey
| |
Collapse
|
249
|
Qiao J, Feng M, Zhou W, Tan Y, Yang S, Liu Q, Wang Q, Feng W, Pan Y, Cui L. YAP inhibition overcomes adaptive resistance in HER2-positive gastric cancer treated with trastuzumab via the AKT/mTOR and ERK/mTOR axis. Gastric Cancer 2024; 27:785-801. [PMID: 38782859 PMCID: PMC11193831 DOI: 10.1007/s10120-024-01508-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Human epidermal growth factor receptor 2 (HER2)-positive gastric cancer (GC) is a heterogeneous GC subtype characterized by the overexpression of HER2. To date, few specific targeted therapies have demonstrated durable efficacy in HER2-positive GC patients, with resistance to trastuzumab typically emerging within 1 year. However, the mechanisms of resistance to trastuzumab remain incompletely understood, presenting a significant challenge to clinical practice. METHODS In this study, we integrated genetic screening and bulk transcriptome and epigenomic profiling to define the mechanisms mediating adaptive resistance to HER2 inhibitors and identify potential effective therapeutic strategies for treating HER2-positive GCs. RESULTS We revealed a potential association between adaptive resistance to trastuzumab in HER2-positive GC and the expression of YES-associated protein (YAP). Notably, our investigation revealed that long-term administration of trastuzumab triggers extensive chromatin remodeling and initiates YAP gene transcription in HER2-positive cells characterized by the initial inhibition and subsequent reactivation. Furthermore, treatment of HER2-positive GC cells and cell line-derived xenografts (CDX) models with YAP inhibitors in combination with trastuzumab was found to induce synergistic effects through the AKT/mTOR and ERK/mTOR pathways. CONCLUSION These findings underscore the pivotal role of reactivated YAP and mTOR signaling pathways in the development of adaptive resistance to trastuzumab and may serve as a promising joint target to overcome resistance to trastuzumab.
Collapse
Affiliation(s)
- Jiao Qiao
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Mei Feng
- Translational Cancer Research Center, Peking University First Hospital, Beijing, 100034, China
- Division of General Surgery, Peking University First Hospital, Peking University, No. 8 Xi Shiku Street, Beijing, 100034, China
| | - Wenyuan Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yuan Tan
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Shuo Yang
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Qi Liu
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Qingchen Wang
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Weimin Feng
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Yisheng Pan
- Division of General Surgery, Peking University First Hospital, Peking University, No. 8 Xi Shiku Street, Beijing, 100034, China
| | - Liyan Cui
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China.
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China.
- Core Unit of National Clinical Research Center for Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
250
|
Mohammed O, Gizaw ST, Degef M. Potential diagnostic, prognostic, and predictive biomarkers of gastric cancer. Health Sci Rep 2024; 7:e2261. [PMID: 39040881 PMCID: PMC11260885 DOI: 10.1002/hsr2.2261] [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: 12/13/2023] [Revised: 06/29/2024] [Accepted: 07/04/2024] [Indexed: 07/24/2024] Open
Abstract
Background Gastric cancer (GC), a malignant epithelial tumor, is the fourth leading cause of cancer-related death worldwide. Therapeutic strategies for GC, despite the biggest challenges, can significantly improve survival rates through early detection and effective screening methods. Aim To provide brief information on the necessity of multiple specific diagnostic, prognostic, and predictive markers for GC. Methods This review was conducted using a variety of search engines, including PubMed Central, Scopus, Web of Science, Google Scholar, and others. Results Some potential biomarkers that provide essential information include circulating tumor cells (CTCs), DNA methylation, claudin 18.2, fibroblast growth factor receptor 2 (FGFR2), long noncoding RNAs (lncRNAs), cell-free DNA (cfDNA), microRNAs, and serum pepsinogens. Conclusion Multiple tumor markers are essential for screening, tumor identification, staging, prognostic assessment, and monitoring recurrence after therapy due to the absence of a single tumor indicator for diagnosing, prognosticating, and predicting GC.
Collapse
Affiliation(s)
- Ousman Mohammed
- Department of Medical Laboratory SciencesCollege of Medicine and Health Sciences, Wollo UniversityDessieEthiopia
| | - Solomon Tebeje Gizaw
- Department of Medical BiochemistrySchool of Medicine, College of Health Sciences, AAUAddis AbabaEthiopia
| | - Maria Degef
- Department of Medical BiochemistrySchool of Medicine, College of Health Sciences, AAUAddis AbabaEthiopia
| |
Collapse
|