1
|
He Y, Kong J, Wang Z, Zhang Y, Qing T, Xie F, Chen T, Han J. Development of a novel molecular probe for visualizing mesothelin on the tumor via positron emission tomography. Eur J Nucl Med Mol Imaging 2025; 52:2605-2616. [PMID: 39878895 DOI: 10.1007/s00259-025-07087-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 01/12/2025] [Indexed: 01/31/2025]
Abstract
OBJECTIVES Mesothelin (MSLN) is an antigen that is overexpressed in various cancers, and its interaction with tumor-associated cancer antigen 125 plays a multifaceted role in tumor metastasis. The serum MSLN expression level can be detected using enzyme-linked immunosorbent assay; however, non-invasive visualization of its expression at the tumor site is currently lacking. Therefore, the aim of this study was to develop a molecular probe for imaging MSLN expression through positron emission tomography (PET). METHODS VHH 269-H4 was obtained via immunization of llama using a fragment of MSLN from residue 360 to residue 597. S-2-(4-isothiocyanatobenzyl)-1,4,7-triazacyclononane-1,4,7-triacetic acid (p-SCN-Bn-NOTA) was conjugated to VHH 269-H4 to yield precursor NOTA 269-H4 for radiolabeling. The chelator-to-VHH ratio was determined by mass spectrometry. The binding kinetics of VHH 269-H4 and NOTA 269-H4 were measured by surface plasmon resonance. Flow cytometry was carried out using the anti-mesothelin monoclonal antibody Anetumab to select MSLN-positive and MSLN-negative cell lines. After radiolabeling, the radiochemical purity and in vitro stability were tested by radio-thin-layer chromatography and size exclusion chromatography, respectively. A saturation binding assay was conducted to measure the dissociation constant (Kd) of [68Ga]Ga-NOTA-269-H4. By mircoPET/CT imaging and biodistribution studies, the in vivo performances of the novel tracer were investigated in NCG mice bearing OVCAR-8, SKOV-3, or patient-derived xenografts. RESULTS VHH 269-H4 targeting MSLN was obtained with a Kd value of 0.3 nM. After conjugation, approximately 27% and 3.2% of VHH were coupled to one and two NOTA chelators, respectively. This yielded precursor NOTA 269-H4 with a Kd value of 1.1 nM. The radiochemistry was accomplished with moderate radiochemical yields (34 ± 14%, n = 9, decay-corrected). [68Ga]Ga-NOTA-269-H4 was obtained with high radiochemical purity (> 99%), and was stable after 90 min incubation at room temperature. The binding affinity of the radioligand towards MSLN was kept in the nanomolar range. Flow cytometry revealed that OVCAR-8 cells possess a high level of MSLN expression, while MSLN expression on SKOV-3 cells was negligible. Consistently, in microPET/CT imaging, [68Ga]Ga-NOTA-269-H4 demonstrated clear tumor visualization using NCG mice bearing OVCAR-8 xenografts, but no radioactivity accumulation was observed in SKOV-3 xenografts, suggesting a high specificity of the tracer in vivo. In biodistribution studies, [68Ga]Ga-NOTA-269-H4 displayed radioactivity accumulation of 2.93 ± 0.39%ID/g in OVCAR-8 xenografts at 30 min post-injection, and the highest tumor-to-blood ratio (~ 3) was achieved at 90 min post-injection. In NCG mice bearing patient-derived xenografts, [68Ga]Ga-NOTA-269-H4 was able to noninvasively detect MSLN expression via microPET/CT imaging. CONCLUSIONS To our knowledge, our studies achieved the first-time to non-invasively detect MSLN expression clearly using a single domain antibody fragment. To sum up, [68Ga]Ga-NOTA-269-H4 is a highly promising PET probe to visualize MSLN expression in vivo and holds great potential to monitor MSLN expression during tumor development.
Collapse
Affiliation(s)
- Yingfang He
- Institute of Radiation Medicine, Fudan University, Xietu Road 2094, Shanghai, 200032, China
| | - Jinping Kong
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, 550009, China
| | - Ze Wang
- Institute of Radiation Medicine, Fudan University, Xietu Road 2094, Shanghai, 200032, China
| | - Yu Zhang
- Department of Translational Medicine, Zentera Therapeutics, 183 Hongqiao Road, Shanghai, China
| | - Tingting Qing
- Department of Translational Medicine, Zentera Therapeutics, 183 Hongqiao Road, Shanghai, China
| | - Fang Xie
- Department of Nuclear Medicine & PET center, Huashan Hospital, Fudan University, 200233, Shanghai, China
| | - Tengxiang Chen
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, 550009, China.
| | - Junbin Han
- Institute of Radiation Medicine, Fudan University, Xietu Road 2094, Shanghai, 200032, China.
| |
Collapse
|
2
|
Yue L, Kajino K, Kobayashi T, Sugitani Y, Sugihara M, Kakuta S, Harada N, Sasano H, Kojima M, Abe M, Lu R, Otsuji N, Orimo A, Hino O. ERC/Mesothelin Is Associated with the Formation of Microvilli on the Mesothelium and Has Limited Functional Relevance Under Physiological Conditions. Int J Mol Sci 2025; 26:4330. [PMID: 40362566 PMCID: PMC12072949 DOI: 10.3390/ijms26094330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 04/24/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
In adults, expressed in renal cancer (ERC)/mesothelin is exclusively expressed in the mesothelial cells lining the pleural, pericardial, and peritoneal cavities, yet its function under physiological conditions is unknown. To explore this, we studied ERC expression in wild-type (WT) mice at different developmental stages by immunohistochemistry and analyzed the ultrastructure of the mesothelium in WT and Erc-knockout (KO) mice via electron microscopy. Additionally, cardiopulmonary function in adult WT and Erc-KO mice was assessed using echocardiography and the forced oscillation technique (FOT). During embryonic development in WT mice, ERC expression was detected in the epicardium as early as embryonic day (E)12.5 but was absent in the pleura until E18.5. The timing of expression appeared to coincide with the active maturation of these organs, which implied a potential role in cardiopulmonary development. Electron microscopy revealed that microvilli on the mesothelium of Erc-KO mice were immature compared to those of WT mice. Based on these findings, we hypothesized that ERC might contribute to cardiopulmonary function; however, echocardiography and FOT did not reveal any functional differences between WT and Erc-KO mice. This suggests that ERC has limited functional relevance under physiological conditions.
Collapse
Affiliation(s)
- Liang Yue
- Department of Molecular Pathology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (L.Y.); (T.K.); (Y.S.); (M.A.); (N.O.); (A.O.); (O.H.)
| | - Kazunori Kajino
- Department of Molecular Pathology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (L.Y.); (T.K.); (Y.S.); (M.A.); (N.O.); (A.O.); (O.H.)
- Department of Human Pathology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan;
- Department of Pathology, Juntendo University Tokyo Koto Geriatric Medical Center, 3-3-20 Shinsuna, Koto-ku, Tokyo 136-0075, Japan
| | - Toshiyuki Kobayashi
- Department of Molecular Pathology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (L.Y.); (T.K.); (Y.S.); (M.A.); (N.O.); (A.O.); (O.H.)
| | - Yoshinobu Sugitani
- Department of Molecular Pathology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (L.Y.); (T.K.); (Y.S.); (M.A.); (N.O.); (A.O.); (O.H.)
| | - Masami Sugihara
- Department of Clinical Laboratory Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan;
- Department of Clinical Laboratory, Juntendo University Tokyo Koto Geriatric Medical Center, 3-3-20 Shinsuna, Koto-ku, Tokyo 136-0075, Japan
| | - Soichiro Kakuta
- Laboratory of Morphology and Image Analysis, Biomedical Research Core Facilities, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan;
| | - Norihiro Harada
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (N.H.); (H.S.)
| | - Hitoshi Sasano
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (N.H.); (H.S.)
| | - Masataka Kojima
- Department of Otorhinolaryngology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan;
| | - Masaaki Abe
- Department of Molecular Pathology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (L.Y.); (T.K.); (Y.S.); (M.A.); (N.O.); (A.O.); (O.H.)
| | - Rong Lu
- Department of Human Pathology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan;
| | - Naomi Otsuji
- Department of Molecular Pathology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (L.Y.); (T.K.); (Y.S.); (M.A.); (N.O.); (A.O.); (O.H.)
- Department of Human Pathology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan;
| | - Akira Orimo
- Department of Molecular Pathology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (L.Y.); (T.K.); (Y.S.); (M.A.); (N.O.); (A.O.); (O.H.)
| | - Okio Hino
- Department of Molecular Pathology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (L.Y.); (T.K.); (Y.S.); (M.A.); (N.O.); (A.O.); (O.H.)
| |
Collapse
|
3
|
Kos M, Mertowska P, Mertowski S, Roliński J, Krasińska-Płachta A, Urbanowicz T, Gogacz M, Grywalska E. From Defense to Disease: How the Immune System Fuels Epithelial-Mesenchymal Transition in Ovarian Cancer. Int J Mol Sci 2025; 26:4041. [PMID: 40362280 PMCID: PMC12072087 DOI: 10.3390/ijms26094041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/11/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Ovarian cancer is one of the most deadly gynecological cancers, with over 300 thousand new cases per year, most of which are diagnosed in advanced stages. The limited availability of effective biomarkers and lack of characteristic symptoms make early diagnosis difficult, resulting in a five-year survival rate of 30-40%. Mutations in the BRCA1 and BRCA2 genes and abnormalities of signaling pathways such as PI3K/AKT and TP53 play a key role in the progression of ovarian cancer. The immune system, which can act against tumors, often supports tumor development in the ovarian cancer microenvironment through immunoevasion, which is influenced by cytokines such as IL-6, IL-10, and TGF-β. Epithelial-to-mesenchymal transition (EMT) allows cancer cells to acquire mesenchymal characteristics, increasing their invasiveness and metastatic capacity. Immunological factors, including pro-inflammatory cytokines and signals from the tumor microenvironment regulate the EMT process. This review aims to present the role of EMT in ovarian cancer progression, its interactions with the immune system, and potential biomarkers and therapeutic targets. Modulation of the immune response and inhibition of EMT may constitute the basis for personalized therapies, which opens new possibilities for improving the prognosis and efficacy of treatment in patients with ovarian cancer.
Collapse
Affiliation(s)
- Michał Kos
- II Clinic of Gynecology, Medical University of Lublin, 20-093 Lublin, Poland; (M.K.); (M.G.)
| | - Paulina Mertowska
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (S.M.); (E.G.)
| | - Sebastian Mertowski
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (S.M.); (E.G.)
| | - Jacek Roliński
- Department of Clinical Immunology, Medical University of Lublin, 4a Chodźki Street, 20-093 Lublin, Poland;
| | | | - Tomasz Urbanowicz
- Cardiac Surgery and Transplantology Department, Poznan University of Medical Sciences, 61-848 Poznan, Poland
| | - Marek Gogacz
- II Clinic of Gynecology, Medical University of Lublin, 20-093 Lublin, Poland; (M.K.); (M.G.)
| | - Ewelina Grywalska
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (S.M.); (E.G.)
| |
Collapse
|
4
|
Abul Rub F, Moursy N, Alhedeithy N, Mohamed J, Ifthikar Z, Elahi MA, Mir TA, Rehman MU, Tariq S, Alabudahash M, Chinnappan R, Yaqinuddin A. Modern Emerging Biosensing Methodologies for the Early Diagnosis and Screening of Ovarian Cancer. BIOSENSORS 2025; 15:203. [PMID: 40277517 PMCID: PMC12024575 DOI: 10.3390/bios15040203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/26/2025]
Abstract
Ovarian cancer (OC) is one of the leading causes of gynecological cancer-related death worldwide. Late diagnosis at advanced stages of OC is the reason for a higher mortality rate. Earlier diagnosis and proper treatment are important for improving the prognosis of OC patients. Biosensors offer accurate, low-cost, rapid, and user-friendly devices that can be employed for the detection of OC-specific biomarkers in the early stage. Therefore, it is important to consider the potential biomarkers in the biological fluids to confirm the OC prognosis. Out of many biomarkers, the most commonly tested clinically is cancer antigen 125 (CA-125). However, CA-125 is considered to be a poor biomarker for OC diagnosis. Several biosensing methods were developed for the sensitive and quantitative detection of each biomarker. In abnormal expression in OC patients, nucleic acids, enzymes, cells, and exosomes are used as target biomarkers for the construction of biosensors. This review focuses on the development for the detection of various biomarkers using multiple biosensing methods. Here, we describe the origin and the significance of OC-associated biomarkers, the working principle of biosensors, and the classification of biosensors based on their recognition elements and signal transducers. The modes of detection and sensitivity of the sensors are discussed. Finally, the challenges in the fabrication, obstacles in the clinical application, and future prospects are discussed.
Collapse
Affiliation(s)
- Farah Abul Rub
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (F.A.R.); (N.M.); (N.A.); (Z.I.); (M.A.E.); (T.A.M.)
| | - Naseel Moursy
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (F.A.R.); (N.M.); (N.A.); (Z.I.); (M.A.E.); (T.A.M.)
| | - Nouf Alhedeithy
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (F.A.R.); (N.M.); (N.A.); (Z.I.); (M.A.E.); (T.A.M.)
| | - Juraij Mohamed
- Faculty of Medicine, University of Colombo, Colombo 00800, Sri Lanka;
| | - Zainab Ifthikar
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (F.A.R.); (N.M.); (N.A.); (Z.I.); (M.A.E.); (T.A.M.)
| | - Muhammad Affan Elahi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (F.A.R.); (N.M.); (N.A.); (Z.I.); (M.A.E.); (T.A.M.)
| | - Tanveer Ahmed Mir
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (F.A.R.); (N.M.); (N.A.); (Z.I.); (M.A.E.); (T.A.M.)
- Laboratory of Tissue/Organ Bioengineering & BioMEMS, Organ Transplant Centre of Excellence (TR&I-Dpt), King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia
| | - Mati Ur Rehman
- Department of Biological and Biomedical Sciences, The Aga Khan University, Stadium Road, P.O. Box 3500, Karachi 74800, Pakistan;
| | - Saima Tariq
- Department of Obstetrics and Gynecology, Al Iman General Hospital, Ministry of Health, Riyadh 12684, Saudi Arabia;
| | - Mubark Alabudahash
- Strathclyde Institute of Pharmacy and Biomedical Sciences (SIPBS), Glasgow G4 0RE, UK;
| | - Raja Chinnappan
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (F.A.R.); (N.M.); (N.A.); (Z.I.); (M.A.E.); (T.A.M.)
- Laboratory of Tissue/Organ Bioengineering & BioMEMS, Organ Transplant Centre of Excellence (TR&I-Dpt), King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia
| | - Ahmed Yaqinuddin
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (F.A.R.); (N.M.); (N.A.); (Z.I.); (M.A.E.); (T.A.M.)
| |
Collapse
|
5
|
Chang MR, Matnurov EM, Wu C, Arakelyan J, Choe HJ, Kushnarev V, Yap JY, Soo XX, Chow MJ, Berger W, Ang WH, Babak MV. Leveraging Immunogenic Cell Death to Enhance the Immune Response against Malignant Pleural Mesothelioma Tumors. J Am Chem Soc 2025; 147:7908-7920. [PMID: 39992709 PMCID: PMC11887451 DOI: 10.1021/jacs.4c17966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/26/2025]
Abstract
Although various metal-based compounds have exhibited excellent immunogenic cell death (ICD)-inducing properties both in vitro and in vivo, the majority of these compounds have been discovered serendipitously. In this work, we have successfully synthesized and characterized 35 cyclometalated Au(III) complexes containing dithiocarbamate ligands, with 25 of these complexes being previously unreported. Their ability to induce phagocytosis in vitro against immunologically "cold" malignant pleural mesothelioma (MPM) cells was strongly dependent on the cyclometalated scaffold and the overall lipophilicity of the complexes. We elucidated the role of cell death mechanisms in the observed ICD effects and identified correlations between the ability of the complexes to induce necrotic cell death and ICD, both in vitro and in vivo. Complex 2G, with its high phagocytosis rates and low necrosis rates, was recognized as a bona fide ICD inducer, demonstrating a remarkably long-lasting immune response in vaccinated mice. In contrast, complex 1C, characterized by high phagocytosis rates and high necrosis rates, failed to elicit a sustained immune response upon following vaccination; however, it triggered selective activation of calreticulin in tumors upon direct in vivo administration. Overall, this study offers a framework for predicting ICD effects in vivo for structurally similar Au(III) complexes, with the potential for extension to other series of metal complexes.
Collapse
Affiliation(s)
- Meng Rui Chang
- Department
of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117543, Singapore
| | - Egor M. Matnurov
- Drug Discovery
Lab, Department of Chemistry, City University
of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, People’s Republic
of China
| | - Chengnan Wu
- Drug Discovery
Lab, Department of Chemistry, City University
of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, People’s Republic
of China
| | - Jemma Arakelyan
- Drug Discovery
Lab, Department of Chemistry, City University
of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, People’s Republic
of China
| | - Ho-Jung Choe
- Drug Discovery
Lab, Department of Chemistry, City University
of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, People’s Republic
of China
| | - Vladimir Kushnarev
- Drug Discovery
Lab, Department of Chemistry, City University
of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, People’s Republic
of China
| | - Jian Yu Yap
- Department
of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117543, Singapore
- NUS Graduate
School - Integrated Science and Engineering Programme (ISEP), National University of Singapore, Singapore 119077, Singapore
| | - Xiu Xuan Soo
- Department
of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117543, Singapore
| | - Mun Juinn Chow
- Department
of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117543, Singapore
| | - Walter Berger
- Center for
Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8A, Vienna 1090, Austria
| | - Wee Han Ang
- Department
of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117543, Singapore
- NUS Graduate
School - Integrated Science and Engineering Programme (ISEP), National University of Singapore, Singapore 119077, Singapore
| | - Maria V. Babak
- Drug Discovery
Lab, Department of Chemistry, City University
of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, People’s Republic
of China
| |
Collapse
|
6
|
Affὸ S, Sererols-Viñas L, Garcia-Vicién G, Cadamuro M, Chakraborty S, Sirica AE. Cancer-Associated Fibroblasts in Intrahepatic Cholangiocarcinoma: Insights into Origins, Heterogeneity, Lymphangiogenesis, and Peritoneal Metastasis. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:378-396. [PMID: 39117110 DOI: 10.1016/j.ajpath.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/11/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024]
Abstract
Intrahepatic cholangiocarcinoma (iCCA) denotes a rare, highly malignant, and heterogeneous class of primary liver adenocarcinomas exhibiting phenotypic characteristics of cholangiocyte differentiation. Among the distinctive pathological features of iCCA, one that differentiates the most common macroscopic subtype (eg, mass-forming type) of this hepatic tumor from conventional hepatocellular carcinoma is a prominent desmoplastic reaction manifested as a dense fibro-collagenous-enriched tumor stroma. Cancer-associated fibroblasts (CAFs) represent the most abundant mesenchymal cell type in the desmoplastic reaction. Although the protumor effects of CAFs in iCCA have been increasingly recognized, more recent cell lineage tracing studies, advanced single-cell RNA sequencing, and expanded biomarker analyses have provided new awareness into their ontogeny, as well as underscored their biological complexity as reflected by the presence of multiple subtypes. In addition, evidence supports CAFs' potential to display cancer-restrictive roles, including immunosuppression. However, CAFs also play important roles in facilitating metastasis, as exemplified by lymph node metastasis and peritoneal carcinomatosis, which are common in iCCA. Herein, the authors provide a timely appraisal of the origins and phenotypic and functional complexity of CAFs in iCCA, together with providing mechanistic insights into lymphangiogenesis and peritoneal metastasis relevant to this lethal human cancer.
Collapse
Affiliation(s)
- Silvia Affὸ
- Tumor Microenvironment Plasticity and Heterogeneity Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Laura Sererols-Viñas
- Tumor Microenvironment Plasticity and Heterogeneity Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Gemma Garcia-Vicién
- Tumor Microenvironment Plasticity and Heterogeneity Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | - Sanjukta Chakraborty
- Department of Medical Physiology, School of Medicine, Texas A&M Health Science Center, Bryan, Texas
| | - Alphonse E Sirica
- Department of Pathology (Emeritus), Virginia Commonwealth University School of Medicine, Richmond, Virginia.
| |
Collapse
|
7
|
Li D, Andaloori L, Crowe M, Lin S, Hong J, Zaidi N, Ho M. Development of CAR-T Therapies and Personalized Vaccines for the Treatment of Cholangiocarcinoma: Current Progress, Mechanisms of Action, and Challenges. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:453-469. [PMID: 39675505 PMCID: PMC11983698 DOI: 10.1016/j.ajpath.2024.10.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 10/15/2024] [Accepted: 10/24/2024] [Indexed: 12/17/2024]
Abstract
Cholangiocarcinoma (CCA) is a highly fatal malignancy with an increasing prevalence, a high mortality rate, poor overall survival, and limited responsiveness to conventional chemoradiotherapy. Targeted therapies addressing specific gene mutations have expanded treatment options for some patient populations. The introduction of chimeric antigen receptor-modified T-cell (CAR-T) immunotherapy and personalized vaccines have opened up a new avenue for managing various cancers. Considerable efforts have been dedicated to preclinical research and ongoing clinical trials of immunotherapeutic approaches including CAR-T therapy, vaccines, and antibody-based therapies such as antibody drug conjugates. However, the potential of CAR-T therapy and vaccines in treating advanced unresectable/metastatic cholangiocarcinoma remains largely unexplored. This article offers an overview of the current landscape of antibody-based immunotherapy, particularly CAR-T therapy and vaccines in the context of cholangiocarcinoma treatment. It outlines a framework for selecting CAR-T and vaccine targets and delves into the biology of promising targetable antigens, as well as potential future therapeutic targets.
Collapse
Affiliation(s)
- Dan Li
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Lalitya Andaloori
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Matthew Crowe
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Shaoli Lin
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Jessica Hong
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Neeha Zaidi
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland.
| | - Mitchell Ho
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.
| |
Collapse
|
8
|
Kutle I, Polten R, Stalp JL, Hachenberg J, Todzey F, Hass R, Zimmermann K, von der Ohe J, von Kaisenberg C, Neubert L, Kamp JC, Schaudien D, Seyda AK, Hillemanns P, Klapdor R, Morgan MA, Schambach A. Anti-Mesothelin CAR-NK cells as a novel targeted therapy against cervical cancer. Front Immunol 2024; 15:1485461. [PMID: 39781381 PMCID: PMC11707549 DOI: 10.3389/fimmu.2024.1485461] [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: 08/23/2024] [Accepted: 11/18/2024] [Indexed: 01/12/2025] Open
Abstract
Resistance to the currently available treatment paradigms is one of the main factors that contributes to poor outcomes in patients with advanced cervical cancer. Novel targeted therapy approaches might enhance the patient's treatment outcome and are urgently needed for this malignancy. While chimeric-antigen receptor (CAR)-based adoptive immunotherapy displays a promising treatment strategy for liquid cancers, their use against cervical cancer is largely unexplored. This study used alpharetroviral SIN vectors to equip natural killer (NK) cells with a third-generation CAR (including CD28 and 4-1BB co-stimulatory domains) targeting Mesothelin, which was identified to be highly expressed on primary human cervical cancer tissues and cervical cancer cell lines in this and other studies. Anti-Mesothelin CAR-NK cells demonstrated high cytotoxicity against cervical cancer cells in 2D and 3D culture models, which corresponded to increased degranulation of CAR-NK-92 cells upon exposure to Mesothelin+ target cells. Mesothelin- cervical cancer cells were generated by CRISPR-Cas9-mediated knockout and used to show target antigen specificity of anti-Mesothelin CAR-NK-92 cells and primary NK cells derived from different healthy donors in co-culture experiments. Combination of anti-Mesothelin CAR-NK-92 cells with chemotherapy revealed increased elimination of cancer cells as compared to monotherapy settings. Our findings indicate the promise of anti-Mesothelin CAR-NK cells as a potential treatment option against cervical cancer, as well as other Mesothelin+ malignancies.
Collapse
Affiliation(s)
- Ivana Kutle
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Robert Polten
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Jan Lennart Stalp
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- Department of Gynecology and Obstetrics, Hannover Medical School, Hannover, Germany
| | - Jens Hachenberg
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- Department of Gynecology and Obstetrics, Hannover Medical School, Hannover, Germany
| | - Felix Todzey
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Ralf Hass
- Department of Gynecology and Obstetrics, Hannover Medical School, Hannover, Germany
| | - Katharina Zimmermann
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Juliane von der Ohe
- Department of Gynecology and Obstetrics, Hannover Medical School, Hannover, Germany
| | | | - Lavinia Neubert
- Institute of Pathology, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - Jan C. Kamp
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
- Department of Respiratory Medicine and Infectious Diseases, Hannover Medical School, Hannover, Germany
| | - Dirk Schaudien
- Fraunhofer Institute for Toxicology and Experimental Medicine, ITEM, Hannover, Germany
| | - Ann-Kathrin Seyda
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Peter Hillemanns
- Department of Gynecology and Obstetrics, Hannover Medical School, Hannover, Germany
| | - Rüdiger Klapdor
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- Department of Gynecology and Obstetrics, Hannover Medical School, Hannover, Germany
- Department of Gynecology and Obstetrics, Albertinen Hospital Hamburg, Hamburg, Germany
| | | | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
9
|
Ewa T, Panchwagh N, Tai C, Avula LR, Joseph S, Rudloff MW, Malik N, Zhang X, Alewine C. Excess shed mesothelin disrupts pancreatic cancer cell clustering to impair peritoneal colonization. FASEB J 2024; 38:e70247. [PMID: 39673668 PMCID: PMC11646052 DOI: 10.1096/fj.202400446r] [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: 10/18/2024] [Accepted: 12/04/2024] [Indexed: 12/16/2024]
Abstract
Peritoneum is the second most common site of metastasis in patients with pancreatic ductal adenocarcinoma (PDAC). Peritoneal colonization is impaired in PDAC cells with knockout (KO) of the cancer surface antigen mesothelin (MSLN) or by introducing Y318A mutation in MSLN to prevent binding to mucin-16 (MUC-16). MSLN has a membrane-bound form but is also shed to release soluble MSLN (sMSLN). Their individual roles in peritoneal metastasis are unknown. Here, a C-terminal truncated MSLN mutant (∆591) incapable of cell membrane insertion but proficient in secretion was engineered. Expression of ∆591 MSLN failed to rescue peritoneal metastasis in MSLN KO cells and inhibited peritoneal colonization when overexpressed in WT PDAC cells. Exposing PDAC cells to conditioned medium (CM) containing excess sMSLN impaired cancer cell clustering in vitro and in peritoneal fluid in vivo, while CM containing only Y318A sMSLN did not. These data demonstrate that interaction of membrane-bound MSLN with MUC-16 promotes cell clustering that is critical for efficient peritoneal metastasis. However, peritoneal colonization by MSLN KO cells was rescued by expression of ∆591 mutant MSLN bearing Y318A mutation, suggesting that sMSLN also has a MUC-16-independent role in peritoneal spread. Alterations in inflammatory signaling pathways occurred following KO cell exposure to CM containing sMSLN, and CM from cancer cells with intact peritoneal metastasis provoked increased KO cell secretion of IL-1α. While excess sMSLN inhibits cell clustering and peritoneal colonization, sMSLN may also promote PDAC peritoneal metastasis independent of MUC-16.
Collapse
Affiliation(s)
- Theressa Ewa
- Laboratory of Molecular Biology, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Neel Panchwagh
- Laboratory of Molecular Biology, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
- Present address:
NYU Grossman School of MedicineNew YorkNew York10016USA
| | - Chin‐Hsien Tai
- Laboratory of Molecular Biology, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Leela Rani Avula
- Laboratory of Molecular Biology, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Sarah Joseph
- Laboratory of Molecular Biology, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
- Present address:
Department of MedicineUniversity of VirginiaCharlottesvilleVirginia22903USA
| | - Michael W. Rudloff
- Laboratory of Molecular Biology, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
- Present address:
Department of MedicineUniversity of North CarolinaChapel HillNorth Carolina27599USA
| | - Nargis Malik
- Laboratory of Molecular Biology, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Xianyu Zhang
- Laboratory of Molecular Biology, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Christine Alewine
- Laboratory of Molecular Biology, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
10
|
d’Alessandro M, Cameli P, Cotton CV, Lamb JA, Bergantini L, Gangi S, Sugden S, Spencer LG, Frediani B, New RP, Chinoy H, Bargagli E, Conticini E. Panel of serum biomarkers for differential diagnosis of idiopathic interstitial lung disease and interstitial lung disease-secondary to systemic autoimmune rheumatic disease. PLoS One 2024; 19:e0311357. [PMID: 39361584 PMCID: PMC11449321 DOI: 10.1371/journal.pone.0311357] [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: 07/23/2024] [Accepted: 09/17/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Interstitial lung disease (ILD) may complicate the course of systemic autoimmune rheumatic disease (SARD) and diagnostic biomarkers are needed. Krebs von den Lungen-6 (KL-6), ferritin (FER) and interleukin 6 (IL-6) have been involved in the ILD development. Our study aimed to compare KL-6, FER, IL-6 and soluble mesothelin-related peptide (SMRP) concentrations in a cohort of idiopathic and SARD-ILD. METHODS 3169 patients were enrolled in the "UK Biomarkers in Interstitial Lung Disease (UK-BILD) Study". We selected patients affected by SARD-ILD and idiopathic ILD (usual interstitial pneumonia-idiopathic pulmonary fibrosis and fibrotic non-specific interstitial pneumonia). Serum marker concentrations were measured through chemiluminescent assays (Fujirebio Europe, Ghent, Belgium). RESULTS 1013 patients were selected for the study: 520 (51.3%) had idiopathic ILD and 493 (48.7%) SARD-ILD. Idiopathic ILD patients displayed higher KL-6 values than SARD-ILD (p = 0.0002). FER and SMRP, though within normal ranges, were significantly higher in idiopathic ILD (p<0.0001). Logistic regression showed good sensitivity (69.4%) and specificity (80.4%) selecting the variables FER and KL-6 concentrations, age and gender-male correlated with a diagnosis of idiopathic ILD. CONCLUSION Our study showed the excellent diagnostic value of KL-6 for detecting ILD, which irrespective of the final diagnosis and extent of disease, is always elevated and is a reliable biomarker of lung fibrosis in various diseases, ranging from idiopathic to autoimmune forms. Our study proposed an ILD differentiation model including clinical background. In this context, combination of serum markers and clinical data, as seen in our cohort, may lead to a further improvement in diagnostic accuracy for ILD.
Collapse
Affiliation(s)
- Miriana d’Alessandro
- Department of Medical and Surgical Sciences & Neurosciences, Respiratory Diseases Unit, University of Siena, Siena, Italy
| | - Paolo Cameli
- Department of Medical and Surgical Sciences & Neurosciences, Respiratory Diseases Unit, University of Siena, Siena, Italy
| | - Caroline V. Cotton
- Department of Rheumatology, Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| | - Janine A. Lamb
- Epidemiology and Public Health Group, School of Health Sciences, University of Manchester, Manchester, United Kingdom
| | - Laura Bergantini
- Department of Medical and Surgical Sciences & Neurosciences, Respiratory Diseases Unit, University of Siena, Siena, Italy
| | - Sara Gangi
- Department of Medical and Surgical Sciences & Neurosciences, Respiratory Diseases Unit, University of Siena, Siena, Italy
| | - Sarah Sugden
- Epidemiology and Public Health Group, School of Health Sciences, University of Manchester, Manchester, United Kingdom
| | - Lisa G. Spencer
- Aintree Chest Centre, Aintree Hospital, Liverpool, United Kingdom
| | - Bruno Frediani
- Department of Medicine, Surgery & Neurosciences, Rheumatology Unit, University of Siena, Siena, Italy
| | - Robert P. New
- Faculty of Biology, Medicine and Health, Division of Musculoskeletal and Dermatological Sciences, The University of Manchester, Manchester, United Kingdom
| | - Hector Chinoy
- Faculty of Biology, Medicine and Health, Division of Musculoskeletal and Dermatological Sciences, The University of Manchester, Manchester, United Kingdom
- Department of Rheumatology, Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Manchester Academic Health Science Centre, Salford, United Kingdom
| | - Elena Bargagli
- Department of Medical and Surgical Sciences & Neurosciences, Respiratory Diseases Unit, University of Siena, Siena, Italy
| | - Edoardo Conticini
- Department of Medicine, Surgery & Neurosciences, Rheumatology Unit, University of Siena, Siena, Italy
| |
Collapse
|
11
|
She Y, Liu X, Liu H, Yang H, Zhang W, Han Y, Zhou J. Combination of clinical and spectral-CT iodine concentration for predicting liver metastasis in gastric cancer: a preliminary study. Abdom Radiol (NY) 2024; 49:3438-3449. [PMID: 38744700 DOI: 10.1007/s00261-024-04346-0] [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: 01/20/2024] [Revised: 04/13/2024] [Accepted: 04/16/2024] [Indexed: 05/16/2024]
Abstract
PURPOSE This study aimed to determine the diagnostic efficacy of various indicators and models for the prediction of gastric cancer with liver metastasis. METHODS Clinical and spectral computed tomography (CT) data from 80 patients with gastric adenocarcinoma who underwent surgical resection were retrospectively analyzed. Patients were divided into metastatic and non-metastatic groups based on whether or not to occur liver metastasis, and the region of interest (ROI) was measured manually on each phase iodine map at the largest level of the tumor. Iodine concentration (IC), normalized iodine concentration (nIC), and clinical data of the primary gastric lesions were analyzed. Logistic regression analysis was used to construct the clinical indicator (CI) and clinical indicator-spectral CT iodine concentration (CI-Spectral CT-IC) Models, which contained all of the parameters with statistically significant differences between the groups. Receiver operating characteristic (ROC) curves were constructed to evaluate the accuracy of the models. RESULTS The metastatic group showed significantly higher levels of Cancer antigen125 (CA125), carcinoembryonic antigen (CEA), IC, and nIC in the arterial phase, venous phase, and delayed phase than the non-metastatic group (all p < 0.05). Normalized iodine concentration Venous Phase (nICVP) exhibited a favorable performance among all IC and nIC parameters for forecasting gastric cancer with liver metastasis (area under the curve (AUC), 0.846). The combination model of clinical data with significant differences and nICVP showed the best diagnostic accuracy for predicting liver metastasis from gastric cancer, with an AUC of 0.897. CONCLUSION nICVP showed the best diagnostic efficacy for predicting gastric cancer with liver metastasis. Clinical Indicators-normalized ICVP model can improve the prediction accuracy for this condition.
Collapse
Affiliation(s)
- Yingxia She
- Radiology of Department, Lanzhou University Second Hospital, Cuiyingmen No.82, Chengguan District, Lanzhou, 730030, People's Republic of China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, People's Republic of China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, People's Republic of China
| | - Xianwang Liu
- Radiology of Department, Lanzhou University Second Hospital, Cuiyingmen No.82, Chengguan District, Lanzhou, 730030, People's Republic of China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, People's Republic of China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, People's Republic of China
| | - Hong Liu
- Radiology of Department, Lanzhou University Second Hospital, Cuiyingmen No.82, Chengguan District, Lanzhou, 730030, People's Republic of China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, People's Republic of China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, People's Republic of China
| | - Haiting Yang
- Radiology of Department, Lanzhou University Second Hospital, Cuiyingmen No.82, Chengguan District, Lanzhou, 730030, People's Republic of China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, People's Republic of China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, People's Republic of China
| | - Wenjuan Zhang
- Radiology of Department, Lanzhou University Second Hospital, Cuiyingmen No.82, Chengguan District, Lanzhou, 730030, People's Republic of China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, People's Republic of China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, People's Republic of China
| | - Yinping Han
- Radiology of Department, Lanzhou University Second Hospital, Cuiyingmen No.82, Chengguan District, Lanzhou, 730030, People's Republic of China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, People's Republic of China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, People's Republic of China
| | - Junlin Zhou
- Radiology of Department, Lanzhou University Second Hospital, Cuiyingmen No.82, Chengguan District, Lanzhou, 730030, People's Republic of China.
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, People's Republic of China.
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, People's Republic of China.
| |
Collapse
|
12
|
Nabeta R, Kanaya A, Shimada K, Matsuura K, Yoshimura A, Oyamada T, Azakami D, Furuya T, Uchide T. Characterization of mesothelin gene expression in dogs and overexpression in canine mesotheliomas. Front Vet Sci 2024; 11:1436621. [PMID: 39315086 PMCID: PMC11417096 DOI: 10.3389/fvets.2024.1436621] [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: 05/22/2024] [Accepted: 08/26/2024] [Indexed: 09/25/2024] Open
Abstract
Introduction Canine mesotheliomas are uncommon malignant tumors typically detected late. Minimally invasive diagnostic biomarkers would facilitate diagnosis at earlier stages, thereby improving clinical outcomes. We hypothesized that mesothelin could be used as a reliable diagnostic biomarker for canine mesotheliomas since it has been used as a cancer biomarker for human mesothelioma. We aimed to explore and characterize mesothelin gene expression in dogs and assess its use as a diagnostic biomarker for canine mesotheliomas. Materials and methods We quantified expressed canine mesothelin transcripts via reverse transcription polymerase chain reaction (RT-PCR) and sequenced them using ribonucleic acid (RNA) extracted from a canine mesothelioma cell line. After confirming mesothelin expression, we assessed its levels in major organ tissues and compared them with those in the mesothelioma tissues using quantitative PCR (qPCR). Mesothelin overexpression in mesotheliomas was detected, and we further compared its levels using qPCR between mesotheliomas and non-mesotheliomas using tumor tissues and clinical sample effusions, confirming its significance as a diagnostic biomarker for canine mesothelioma. Results Mesothelin complementary deoxyribonucleic acid (cDNA) was amplified via RT-PCR, yielding a single band of expected upon DNA electrophoresis. Sequence analyses confirmed it as a predicted canine mesothelin transcript from the genome sequence database. Comparative sequence analysis of the deduced amino acid sequence of the expressed canine mesothelin demonstrated molecular signature similarities with the human mesothelin. However, the pre-sequence of canine mesothelin lacks the mature megakaryocyte potentiating factor (MPF) portion, which is typically cleaved post-translationally with furin. Mesothelin expression was quantified via qPCR revealing low levels in the mesothelial and lung tissues, with negligible expression in the other major organs. Canine mesothelin exhibited significantly higher expression in the canine mesotheliomas than in the noncancerous tissues. Moreover, analysis of clinical samples using qPCR demonstrated markedly elevated mesothelin expression in canine mesotheliomas compared to non-mesothelioma cases. Discussion and conclusion Canine mesothelin exhibits molecular and biological characteristics akin to human mesothelin. It could serve as a vital biomarker for diagnosing canine mesotheliomas, applicable to both tissue- and effusion-based samples.
Collapse
Affiliation(s)
- Rina Nabeta
- Laboratory of Veterinary Molecular Pathology and Therapeutics, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Ami Kanaya
- Laboratory of Veterinary Molecular Pathology and Therapeutics, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Kazumi Shimada
- Laboratory of Veterinary Surgery, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Katsuhiro Matsuura
- Laboratory of Veterinary Surgery, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
- Department of Small Animal Clinical Science, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Aritada Yoshimura
- Animal Medical Center, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Tomohiro Oyamada
- Animal Medical Center, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Daigo Azakami
- Laboratory of Veterinary Clinical Oncoogy, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Tetsuya Furuya
- Laboratory of Veterinary Infectious Diseases, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| | - Tsuyoshi Uchide
- Laboratory of Veterinary Molecular Pathology and Therapeutics, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
| |
Collapse
|
13
|
Rupert PB, Buerger M, Friend DJ, Strong RK. Structural elucidation of the mesothelin-mucin-16/CA125 interaction. Structure 2024; 32:1049-1054.e2. [PMID: 38703776 PMCID: PMC11316652 DOI: 10.1016/j.str.2024.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/24/2024] [Accepted: 04/09/2024] [Indexed: 05/06/2024]
Abstract
Mesothelin (MSLN) is a cell-surface glycoprotein expressed at low levels on normal mesothelium but overexpressed in many cancers. Mesothelin has been implicated to play role/s in cell adhesion and multiple signaling pathways. Mucin-16/CA125 is an enormous cell-surface glycoprotein, also normally expressed on mesothelium and implicated in the progression and metastasis of several cancers, and directly binds mesothelin. However, the precise biological function/s of mesothelin and mucin-16/CA125 remain mysterious. We report protein engineering and recombinant production, qualitative and quantitative binding studies, and a crystal structure determination elucidating the molecular-level details governing recognition of mesothelin by mucin-16/CA125. The interface is small, consistent with the ∼micromolar binding constant and is free of glycan-mediated interactions. Sequence comparisons and modeling suggest that multiple mucin-16/CA125 modules can interact with mesothelin through comparable interactions, potentially generating a high degree of avidity at the cell surface to overcome the weak affinity, with implications for functioning and therapeutic interventions.
Collapse
Affiliation(s)
- Peter B Rupert
- Division of Basic Science, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Matthew Buerger
- Division of Basic Science, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Della J Friend
- Division of Basic Science, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Roland K Strong
- Division of Basic Science, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| |
Collapse
|
14
|
Solidoro R, Centonze A, Miciaccia M, Baldelli OM, Armenise D, Ferorelli S, Perrone MG, Scilimati A. Fluorescent imaging probes for in vivo ovarian cancer targeted detection and surgery. Med Res Rev 2024; 44:1800-1866. [PMID: 38367227 DOI: 10.1002/med.22027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 12/05/2023] [Accepted: 01/25/2024] [Indexed: 02/19/2024]
Abstract
Ovarian cancer is the most lethal gynecological cancer, with a survival rate of approximately 40% at five years from the diagno. The first-line treatment consists of cytoreductive surgery combined with chemotherapy (platinum- and taxane-based drugs). To date, the main prognostic factor is related to the complete surgical resection of tumor lesions, including occult micrometastases. The presence of minimal residual diseases not detected by visual inspection and palpation during surgery significantly increases the risk of disease relapse. Intraoperative fluorescence imaging systems have the potential to improve surgical outcomes. Fluorescent tracers administered to the patient may support surgeons for better real-time visualization of tumor lesions during cytoreductive procedures. In the last decade, consistent with the discovery of an increasing number of ovarian cancer-specific targets, a wide range of fluorescent agents were identified to be employed for intraoperatively detecting ovarian cancer. Here, we present a collection of fluorescent probes designed and developed for fluorescence-guided ovarian cancer surgery. Original articles published between 2011 and November 2022 focusing on fluorescent probes, currently under preclinical and clinical investigation, were searched in PubMed. The keywords used were targeted detection, ovarian cancer, fluorescent probe, near-infrared fluorescence, fluorescence-guided surgery, and intraoperative imaging. All identified papers were English-language full-text papers, and probes were classified based on the location of the biological target: intracellular, membrane, and extracellular.
Collapse
Affiliation(s)
- Roberta Solidoro
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari, Bari, Italy
| | - Antonella Centonze
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari, Bari, Italy
| | - Morena Miciaccia
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari, Bari, Italy
| | - Olga Maria Baldelli
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari, Bari, Italy
| | - Domenico Armenise
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari, Bari, Italy
| | - Savina Ferorelli
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari, Bari, Italy
| | | | - Antonio Scilimati
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari, Bari, Italy
| |
Collapse
|
15
|
Pan Y, Xuan Y, Hao P, Chen X, Yan R, Zhang C, Ke X, Qu Y, Zhang X. Time-dependent proteomics and drug response in expanding cancer cells. Pharmacol Res 2024; 204:107208. [PMID: 38729587 DOI: 10.1016/j.phrs.2024.107208] [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: 04/05/2024] [Revised: 04/26/2024] [Accepted: 05/06/2024] [Indexed: 05/12/2024]
Abstract
Cancer cell line is commonly used for discovery and development of anti-cancer drugs. It is generally considered that drug response remains constant for a certain cell line due to the identity of genetics thus protein patterns. Here, we demonstrated that cancer cells continued dividing even after reaching confluence, in that the proteomics was changed continuously and dramatically with strong relevance to cell division, cell adhesion and cell metabolism, indicating time-dependent intrinsically reprogramming of cells during expansion. Of note, the inhibition effect of most anti-cancer drugs was strikingly attenuated in culture cells along with cell expansion, with the strongest change at the third day when cells were still expanding. Profiling of an FDA-approved drug library revealed that attenuation of response with cell expansion is common for most drugs, an exception was TAK165 that was a selective inhibitor of mitochondrial respiratory chain complex I. Finally, we screened a panel of natural products and identified four pentacyclic triterpenes as selective inhibitors of cancer cells under prolonged growth. Taken together, our findings underscore that caution should be taken in evaluation of anti-cancer drugs using culture cells, and provide agents selectively targeting overgrowth cancer cells.
Collapse
Affiliation(s)
- Yuting Pan
- Center for Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Ying Xuan
- Center for Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Piliang Hao
- School of Life Science and Technology, ShanghaiTech University, Shanghai, PR China
| | - Xianzhi Chen
- Center for Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Rong Yan
- Center for Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Chengqian Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, PR China
| | - Xisong Ke
- Center for Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China.
| | - Yi Qu
- Center for Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China.
| | - Xue Zhang
- Center for Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China.
| |
Collapse
|
16
|
Bertoli E, De Carlo E, Bortolot M, Stanzione B, Del Conte A, Spina M, Bearz A. Targeted Therapy in Mesotheliomas: Uphill All the Way. Cancers (Basel) 2024; 16:1971. [PMID: 38893092 PMCID: PMC11171080 DOI: 10.3390/cancers16111971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 05/16/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Mesothelioma (MM) is an aggressive and lethal disease with few therapeutic opportunities. Platinum-pemetrexed chemotherapy is the backbone of first-line treatment for MM. The introduction of immunotherapy (IO) has been the only novelty of the last decades, allowing an increase in survival compared to standard chemotherapy (CT). However, IO is not approved for epithelioid histology in many countries. Therefore, therapy for relapsed MM remains an unmet clinical need, and the prognosis of MM remains poor, with an average survival of only 18 months. Increasing evidence reveals MM complexity and heterogeneity, of which histological classification fails to explain. Thus, scientific focus on possibly new molecular markers or cellular targets is increasing, together with the search for target therapies directed towards them. The molecular landscape of MM is characterized by inactivating tumor suppressor alterations, the most common of which is found in CDKN2A, BAP1, MTAP, and NF2. In addition, cellular targets such as mesothelin or metabolic enzymes such as ASS1 could be potentially amenable to specific therapies. This review examines the major targets and relative attempts of therapeutic approaches to provide an overview of the potential prospects for treating this rare neoplasm.
Collapse
Affiliation(s)
- Elisa Bertoli
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (E.B.); (E.D.C.); (M.B.); (B.S.); (A.D.C.); (M.S.)
| | - Elisa De Carlo
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (E.B.); (E.D.C.); (M.B.); (B.S.); (A.D.C.); (M.S.)
| | - Martina Bortolot
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (E.B.); (E.D.C.); (M.B.); (B.S.); (A.D.C.); (M.S.)
- Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Brigida Stanzione
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (E.B.); (E.D.C.); (M.B.); (B.S.); (A.D.C.); (M.S.)
| | - Alessandro Del Conte
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (E.B.); (E.D.C.); (M.B.); (B.S.); (A.D.C.); (M.S.)
| | - Michele Spina
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (E.B.); (E.D.C.); (M.B.); (B.S.); (A.D.C.); (M.S.)
| | - Alessandra Bearz
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (E.B.); (E.D.C.); (M.B.); (B.S.); (A.D.C.); (M.S.)
| |
Collapse
|
17
|
Ding T, Shang Z, Zhao H, Song R, Xiong J, He C, Liu D, Yi B. Anoikis-related gene signatures in colorectal cancer: implications for cell differentiation, immune infiltration, and prognostic prediction. Sci Rep 2024; 14:11525. [PMID: 38773226 PMCID: PMC11109202 DOI: 10.1038/s41598-024-62370-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 05/16/2024] [Indexed: 05/23/2024] Open
Abstract
Colorectal cancer (CRC) is a malignant tumor originating from epithelial cells of the colon or rectum, and its invasion and metastasis could be regulated by anoikis. However, the key genes and pathways regulating anoikis in CRC are still unclear and require further research. The single cell transcriptome dataset GSE221575 of GEO database was downloaded and applied to cell subpopulation type identification, intercellular communication, pseudo time cell trajectory analysis, and receptor ligand expression analysis of CRC. Meanwhile, the RNA transcriptome dataset of TCGA, the GSE39582, GSE17536, and GSE17537 datasets of GEO were downloaded and merged into one bulk transcriptome dataset. The differentially expressed genes (DEGs) related to anoikis were extracted from these data sets, and key marker genes were obtained after feature selection. A clinical prognosis prediction model was constructed based on the marker genes and the predictive effect was analyzed. Subsequently, gene pathway analysis, immune infiltration analysis, immunosuppressive point analysis, drug sensitivity analysis, and immunotherapy efficacy based on the key marker genes were conducted for the model. In this study, we used single cell datasets to determine the anoikis activity of cells and analyzed the DEGs of cells based on the score to identify the genes involved in anoikis and extracted DEGs related to the disease from the transcriptome dataset. After dimensionality reduction selection, 7 marker genes were obtained, including TIMP1, VEGFA, MYC, MSLN, EPHA2, ABHD2, and CD24. The prognostic risk model scoring system built by these 7 genes, along with patient clinical data (age, tumor stage, grade), were incorporated to create a nomogram, which predicted the 1-, 3-, and 5-years survival of CRC with accuracy of 0.818, 0.821, and 0.824. By using the scoring system, the CRC samples were divided into high/low anoikis-related prognosis risk groups, there are significant differences in immune infiltration, distribution of immune checkpoints, sensitivity to chemotherapy drugs, and efficacy of immunotherapy between these two risk groups. Anoikis genes participate in the differentiation of colorectal cancer tumor cells, promote tumor development, and could predict the prognosis of colorectal cancer.
Collapse
Affiliation(s)
- Taohui Ding
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
- 2nd Abdominal Surgery Department, Jiangxi Cancer Institute, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, 330029, Jiangxi, People's Republic of China
| | - Zhao Shang
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
| | - Hu Zhao
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
- 2nd Abdominal Surgery Department, Jiangxi Cancer Institute, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, 330029, Jiangxi, People's Republic of China
| | - Renfeng Song
- Department of Digestive Oncology, Jiangxi Cancer Institute, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, 330029, Jiangxi, People's Republic of China
| | - Jianyong Xiong
- 2nd Abdominal Surgery Department, Jiangxi Cancer Institute, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, 330029, Jiangxi, People's Republic of China
| | - Chuan He
- Department of Digestive Oncology, Jiangxi Cancer Institute, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, 330029, Jiangxi, People's Republic of China
| | - Dan Liu
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China.
| | - Bo Yi
- 2nd Abdominal Surgery Department, Jiangxi Cancer Institute, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, 330029, Jiangxi, People's Republic of China.
| |
Collapse
|
18
|
Cutri-French C, Nasioudis D, George E, Tanyi JL. CAR-T Cell Therapy in Ovarian Cancer: Where Are We Now? Diagnostics (Basel) 2024; 14:819. [PMID: 38667465 PMCID: PMC11049291 DOI: 10.3390/diagnostics14080819] [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: 02/12/2024] [Revised: 03/29/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
The success of chimeric antigen receptor T-cell (CAR-T) therapies in the treatment of hematologic malignancies has led to the investigation of their potential in the treatment of solid tumors, including ovarian cancer. While the immunosuppressive microenvironment of ovarian cancer has been a barrier in their implementation, several early phase clinical trials are currently evaluating CAR-T cell therapies targeting mesothelin, folate receptor a, HER2, MUC16, and B7H3. Ongoing challenges include cytokine-associated and "on-target, off-tumor" toxicities, while most common adverse events include cytokine release syndrome, hemophagocytic lymphohistiocytosis/macrophage activation-like syndrome (HLH/MAS), and neurotoxicity. In the present review, we summarize the current status of CAR-T therapy in ovarian cancer and discuss future directions.
Collapse
Affiliation(s)
- Clare Cutri-French
- Department of Obstetrics and Gynecology, University of Pennsylvania Health System, Philadelphia, PA 19104, USA;
| | - Dimitrios Nasioudis
- Division of Gynecologic Oncology, University of Pennsylvania Health System, Philadelphia, PA 19104, USA
| | - Erin George
- Moffitt Cancer Center, Richard M. Schulze Family Foundation Outpatient Center at McKinley Campus, 10920 McKinley Dr, Tampa, FL 33612, USA
| | - Janos L. Tanyi
- Division of Gynecologic Oncology, University of Pennsylvania Health System, Philadelphia, PA 19104, USA
| |
Collapse
|
19
|
Ferreira JP, Packer M, Sattar N, Butler J, Pocock SJ, Anker SD, Maldonado SG, Panova-Noeva M, Sumin M, Masson S, Zannad F, Januzzi JL. Carbohydrate antigen 125 concentrations across the ejection fraction spectrum in chronic heart failure: The EMPEROR programme. Eur J Heart Fail 2024; 26:788-802. [PMID: 38439582 DOI: 10.1002/ejhf.3166] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 03/06/2024] Open
Abstract
AIM Vascular congestion may lead to an increase of carbohydrate antigen 125 (CA-125). The role of CA-125 as a biomarker of congestion or for prognosis across the full ejection fraction (EF) spectrum of chronic heart failure (HF) remains unknown. METHODS AND RESULTS Serum CA-125 was measured in 1111 study participants from the EMPEROR-Reduced and EMPEROR-Preserved trials. Congestive signs and symptoms were evaluated across CA-125 tertiles. Cox regression was used to study the association with outcomes. The primary outcome was a composite of first HF hospitalization or cardiovascular (CV) death. No significant association was present between baseline CA-125 levels and congestive signs or symptoms. In the overall population, higher CA-125 levels were not associated with an increased risk of primary outcome (tertile 3 vs. tertile 1: hazard ratio [HR] 1.34; 95% confidence interval [CI] 0.91-1.96; p-trend = 0.11). However, higher CA-125 levels were associated with an increased risk of primary outcome in patients with HF and reduced EF (HFrEF; tertile 3 vs. tertile 1: HR 2.25 [95% CI 1.30-3.89]), but not among patients with preserved EF (HFpEF; tertile 3 vs. tertile 1: HR 0.68 [95% CI 0.38-1.21]); interaction-p = 0.02). Patients in the upper CA-125 tertile also showed the steepest estimated glomerular filtration rate decline over time (p-trend = 0.03). The effect of empagliflozin to reduce the risk of CV death or HF hospitalization appeared to be attenuated in those with lower baseline CA-125 levels (interaction-p-trend = 0.09). CONCLUSION Across the range of EF in patients with chronic HF enrolled in the EMPEROR trials, the majority of whom did not have clinical evidence of congestion, CA-125 concentrations were not significantly associated with congestive signs or symptoms. CA-125 concentrations may predict HF hospitalization/CV death in patients with HFrEF, but not those with HFpEF. CLINICAL TRIAL REGISTRATION EMPEROR-Reduced (NCT03057977), EMPEROR-Preserved (NCT03057951).
Collapse
Affiliation(s)
- João Pedro Ferreira
- Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Cardiovascular Research and Development Center (UnIC@RISE), Porto, Portugal
- Heart Failure Clinic, Internal Medicine Department, Centro Hospitalar de Vila Nova de Gaia/Espinho, Gaia, Portugal
- Centre d'Investigations Cliniques Plurithématique 14-33, Inserm U1116, CHRU, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Cardiovascular Research and Development Center, Université de Lorraine, Nancy, France
| | - Milton Packer
- Imperial College London, London, UK
- Baylor Heart and Vascular Institute, Baylor University Medical Center, Dallas, TX, USA
| | - Naveed Sattar
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Javed Butler
- Baylor Scott and White Research Institute, Dallas, TX, USA
- University of Mississippi Medical Center, Jackson, MS, USA
| | | | - Stefan D Anker
- Center for Regenerative Therapies, Berlin Institute of Health, Department of Cardiology, German Centre for Cardiovascular Research, partner site Berlin, Charité Universitätsmedizin Berlin, Berlin, Germany
| | | | | | - Mikhail Sumin
- Boehringer Ingelheim International GmbH, Ingelheim, Germany
| | - Serge Masson
- Roche Diagnostics International Ltd, Rotkreuz, Switzerland
| | - Faiez Zannad
- Centre d'Investigations Cliniques Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France
| | - James L Januzzi
- Massachusetts General Hospital and Baim Institute for Clinical Research, Boston, MA, USA
| |
Collapse
|
20
|
Mogere E, Mutebi M, Njau A, Mansour MH, Abuodha J, Okiro P. A rare case of breast carcinoma metastasis into a meningioma in a 64-year-old female patient. Radiol Case Rep 2024; 19:1519-1523. [PMID: 38304352 PMCID: PMC10832375 DOI: 10.1016/j.radcr.2023.11.035] [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: 09/12/2023] [Revised: 11/08/2023] [Accepted: 11/13/2023] [Indexed: 02/03/2024] Open
Abstract
This report discusses the occurrence of tumor-to-tumor metastasis-an atypical phenomenon in oncology where a secondary malignancy develops within an existing primary tumor. The case of a 64-year-old woman is presented, who, with a history of stage II invasive ductal carcinoma of the breast treated with mastectomy and chemoradiotherapy, developed neurological symptoms indicative of a secondary brain tumor. MRI and subsequent histopathological analysis post-craniotomy confirmed a meningioma with a metastatic breast carcinoma, demonstrating the clinical importance of considering tumor-to-tumor metastasis in similar patient histories.
Collapse
Affiliation(s)
- Edwin Mogere
- Section of Neurosurgery, Department of Surgery, Aga Khan University Hospital, Nairobi, Kenya
| | - Miriam Mutebi
- Section of Breast and Endocrine, Department of Surgery, Aga Khan University Hospital, Nairobi, Kenya
| | - Allan Njau
- Section of Anatomic Pathology, Department of Pathology, Aga Khan University Hospital, Nairobi, Kenya
| | - Manel Haj Mansour
- Section of Oncology, Department of Haemato-Oncology, Aga Khan University Hospital, Nairobi, Kenya
| | - Joseph Abuodha
- Section of Oncology, Department of Haemato-Oncology, Aga Khan University Hospital, Nairobi, Kenya
| | - Patricia Okiro
- Section of Anatomic Pathology, Department of Pathology, Aga Khan University Hospital, Nairobi, Kenya
| |
Collapse
|
21
|
Lumish MA, Kohn EC, Tew WP. Top advances of the year: Ovarian cancer. Cancer 2024; 130:837-845. [PMID: 38100616 DOI: 10.1002/cncr.35135] [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] [Indexed: 12/17/2023]
Abstract
Although cure rates remain low and effective screening strategies are elusive, the recent advances in systemic therapies over the past year highlighted in this review have prolonged survival for women with ovarian cancer. In 2022, the first antibody-drug conjugate for platinum-resistant ovarian cancer received accelerated US Food and Drug Administration (FDA) approval. Confirmatory studies examining the efficacy of mirvetuximab and other antibody-drug conjugates are underway. In the upfront setting, the first data establishing an overall survival benefit from poly(ADP-ribose) polymerase inhibitor maintenance was demonstrated after a 7-year follow-up period. In contrast, long-term updates from poly(ADP-ribose) polymerase inhibitor trials in the noncurative setting reported survival detriments, and the FDA withdrew the respective indications. Several trials attempted to improve upon the standard of care for platinum-sensitive ovarian carcinoma and those with rare ovarian cancer histologies (carcinosarcoma, clear cell carcinoma) but failed to demonstrate a clinically or statistically meaningful benefit. This leaves the open question of how to further optimize systemic therapy for advanced ovarian carcinoma to improve long-term survival and cure rates.
Collapse
Affiliation(s)
- Melissa A Lumish
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Elise C Kohn
- National Cancer Institute, Bethesda, Maryland, USA
| | - William P Tew
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
22
|
Pampeno C, Opp S, Hurtado A, Meruelo D. Sindbis Virus Vaccine Platform: A Promising Oncolytic Virus-Mediated Approach for Ovarian Cancer Treatment. Int J Mol Sci 2024; 25:2925. [PMID: 38474178 PMCID: PMC10932354 DOI: 10.3390/ijms25052925] [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: 12/13/2023] [Revised: 01/30/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
This review article provides a comprehensive overview of a novel Sindbis virus vaccine platform as potential immunotherapy for ovarian cancer patients. Ovarian cancer is the most lethal of all gynecological malignancies. The majority of high-grade serous ovarian cancer (HGSOC) patients are diagnosed with advanced disease. Current treatment options are very aggressive and limited, resulting in tumor recurrences and 50-60% patient mortality within 5 years. The unique properties of armed oncolytic Sindbis virus vectors (SV) in vivo have garnered significant interest in recent years to potently target and treat ovarian cancer. We discuss the molecular biology of Sindbis virus, its mechanisms of action against ovarian cancer cells, preclinical in vivo studies, and future perspectives. The potential of Sindbis virus-based therapies for ovarian cancer treatment holds great promise and warrants further investigation. Investigations using other oncolytic viruses in preclinical studies and clinical trials are also presented.
Collapse
Affiliation(s)
- Christine Pampeno
- Department of Pathology, NYU Grossman School of Medicine, New York University, New York, NY 10016, USA
| | | | - Alicia Hurtado
- Department of Pathology, NYU Grossman School of Medicine, New York University, New York, NY 10016, USA
| | - Daniel Meruelo
- Department of Pathology, NYU Grossman School of Medicine, New York University, New York, NY 10016, USA
| |
Collapse
|
23
|
Ghose A, McCann L, Makker S, Mukherjee U, Gullapalli SVN, Erekkath J, Shih S, Mahajan I, Sanchez E, Uccello M, Moschetta M, Adeleke S, Boussios S. Diagnostic biomarkers in ovarian cancer: advances beyond CA125 and HE4. Ther Adv Med Oncol 2024; 16:17588359241233225. [PMID: 38435431 PMCID: PMC10908239 DOI: 10.1177/17588359241233225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 01/26/2024] [Indexed: 03/05/2024] Open
Abstract
Ovarian cancer (OC) is the most lethal gynaecologic malignancy, attributed to its insidious growth, non-specific symptoms and late presentation. Unfortunately, current screening modalities are inadequate at detecting OC and many lack the appropriate specificity and sensitivity that is desired from a screening test. Nearly 70% of cases are diagnosed at stage III or IV with poor 5-year overall survival. Therefore, the development of a sensitive and specific biomarker for early diagnosis and screening for OC is of utmost importance. Currently, diagnosis is guided by CA125, the patient's menopausal status and imaging features on ultrasound scan. However, emerging evidence suggests that a combination of CA125 and HE4 (another serum biomarker) and patient characteristics in a multivariate index assay may provide a higher specificity and sensitivity than either CA125 and HE4 alone in the early detection of OC. Other attempts at combining various serum biomarkers into one multivariate index assay such as OVA1, ROMA and Overa have all shown promise. However, significant barriers exist before these biomarkers can be implemented in clinical practice. This article aims to provide an up-to-date review of potential biomarkers for screening and early diagnosis of OC which may have the potential to transform its diagnostic landscape.
Collapse
Affiliation(s)
- Aruni Ghose
- Department of Medical Oncology, Barts Cancer Centre, St. Bartholomew’s Hospital, Barts Health NHS Trust, London, UK
- Department of General Medicine, Newham University Hospital, Barts Health NHS Trust, London, UK
- Department of Medical Oncology, Medway NHS Foundation Trust, Gillingham, UK
- Department of Medical Oncology, Mount Vernon Cancer Centre, East and North Hertfordshire NHS Trust, London, UK
| | - Lucy McCann
- Department of General Medicine, Newham University Hospital, Barts Health NHS Trust, London, UK
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, UK
| | - Shania Makker
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- University College London Cancer Institute, London, UK
| | - Uma Mukherjee
- Department of Medical Oncology, Barts Cancer Centre, St. Bartholomew’s Hospital, Barts Health NHS Trust, London, UK
- University College London Cancer Institute, London, UK
| | | | - Jayaraj Erekkath
- Department of Medical Oncology, Northern Ireland Cancer Centre, Belfast City Hospital, Belfast Health and Social Care Trust, Belfast, UK
| | - Stephanie Shih
- Department of General Medicine, Newham University Hospital, Barts Health NHS Trust, London, UK
| | - Ishika Mahajan
- Department of Acute Medicine, Lincoln County Hospital, United Lincolnshire Hospitals NHS Trust, Lincoln, Lincolnshire, UK
- Department of Medical Oncology, Apollo Cancer Centre, Chennai, Tamil Nadu, India
| | - Elisabet Sanchez
- Department of Medical Oncology, Medway NHS Foundation Trust, Gillingham, UK
| | - Mario Uccello
- Department of Medical Oncology, Southampton General Hospital, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | | | - Sola Adeleke
- Department of Clinical Oncology, Cancer Centre at Guy’s, Guy’s and St. Thomas’ NHS Foundation Trust, London, UK
- Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King’s College London, Guy’s Campus, London, WC2R 2LS, UK
| | - Stergios Boussios
- Department of Medical Oncology, Medway NHS Foundation Trust, Gillingham, UK
- Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King’s College London, London, UK
- Kent and Medway Medical School, University of Kent, Canterbury, UK
- AELIA Organization, Thermi, Thessaloniki, Greece
| |
Collapse
|
24
|
Klotz DM, Link T, Wimberger P, Kuhlmann JD. A predictive and prognostic model for surgical outcome and prognosis in ovarian cancer computed by clinico-pathological and serological parameters (CA125, HE4, mesothelin). Clin Chem Lab Med 2024; 62:530-539. [PMID: 37816681 DOI: 10.1515/cclm-2023-0314] [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: 03/28/2023] [Accepted: 09/18/2023] [Indexed: 10/12/2023]
Abstract
OBJECTIVES Numerous prognostic models have been proposed for ovarian cancer, extending from single serological factors to complex gene-expression signatures. Nonetheless, these models have not been routinely translated into clinical practice. We constructed a robust and readily calculable model for predicting surgical outcome and prognosis of ovarian cancer patients by exploiting commonly available clinico-pathological factors and three selected serum parameters. METHODS Serum CA125, human epididymis protein 4 (HE4) and mesothelin (MSL) were quantified by Lumipulse® G chemiluminescent enzyme immunoassay (Fujirebio) in a total of 342 serum samples from 190 ovarian cancer patients, including 152 paired pre- and post-operative samples. RESULTS Detection of pre-operative HE4 and CA125 was the optimal marker combination for blood-based prediction of surgical outcome (AUC=0.86). We constructed a prognostic model, computed by serum levels of pre-operative CA125, post-operative HE4, post-operative MSL and surgical outcome. Prognostic performance of our model was superior to any of these parameters alone and was independent from BRCA1/2 mutational status. We subsequently transformed our model into a prognostic risk index, stratifying patients as "lower risk" or "higher risk". In "higher risk" patients, relapse or death was predicted with an AUC of 0.89 and they had a significantly shorter progression free survival (HR: 9.74; 95 % CI: 5.95-15.93; p<0.0001) and overall survival (HR: 5.62; 95 % CI: 3.16-9.99; p<0.0001) compared to "lower risk" patients. CONCLUSIONS We present a robust predictive/prognostic model for ovarian cancer, which could readily be implemented into routine diagnostics in order to identify ovarian cancer patients at high risk of recurrence.
Collapse
Affiliation(s)
- Daniel Martin Klotz
- Department of Gynecology and Obstetrics, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumour Diseases (NCT), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Theresa Link
- Department of Gynecology and Obstetrics, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumour Diseases (NCT), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Pauline Wimberger
- Department of Gynecology and Obstetrics, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumour Diseases (NCT), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jan Dominik Kuhlmann
- Department of Gynecology and Obstetrics, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumour Diseases (NCT), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
25
|
Lee K, Perry K, Xu M, Veillard I, Kumar R, Rao TD, Rueda BR, Spriggs DR, Yeku OO. Structural basis for antibody recognition of the proximal MUC16 ectodomain. J Ovarian Res 2024; 17:41. [PMID: 38374055 PMCID: PMC10875768 DOI: 10.1186/s13048-024-01373-9] [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: 08/09/2023] [Accepted: 02/13/2024] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND Mucin 16 (MUC16) overexpression is linked with cancer progression, metastasis, and therapy resistance in high grade serous ovarian cancer and other malignancies. The cleavage of MUC16 forms independent bimodular fragments, the shed tandem repeat sequence which circulates as a protein bearing the ovarian cancer biomarker (CA125) and a proximal membrane-bound component which is critical in MUC16 oncogenic behavior. A humanized, high affinity antibody targeting the proximal ectodomain represents a potential therapeutic agent against MUC16 with lower antigenic potential and restricted human tissue expression. RESULTS Here, we demonstrate the potential therapeutic versatility of the humanized antibody as a monoclonal antibody, antibody drug conjugate, and chimeric antigen receptor. We report the crystal structures of 4H11-scFv, derived from an antibody specifically targeting the MUC16 C-terminal region, alone and in complex with a 26-amino acid MUC16 segment resolved at 2.36 Å and 2.47 Å resolution, respectively. The scFv forms a robust interaction with an epitope consisting of two consecutive β-turns and a β-hairpin stabilized by 2 hydrogen bonds. The VH-VL interface within the 4H11-scFv is stabilized through an intricate network of 11 hydrogen bonds and a cation-π interaction. CONCLUSIONS Together, our studies offer insight into antibody-MUC16 ectodomain interaction and advance our ability to design agents with potentially improved therapeutic properties over anti-CA125 moiety antibodies.
Collapse
Affiliation(s)
- Kwangkook Lee
- Division of Hematology & Oncology, Department of Medicine, Massachusetts General Hospital-Harvard Medical School, Boston, MA, USA
- Division of Hematology and Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Kay Perry
- Department of Chemistry and Chemical Biology, Argonne National Laboratory, NE-CAT, Cornell University, Building 436E, 9700 S. Cass Avenue, Argonne, IL, 60439, USA
| | - Mengyao Xu
- Division of Hematology and Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Irva Veillard
- Division of Hematology and Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Raj Kumar
- Division of Hematology and Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Thapi Dharma Rao
- Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Bo R Rueda
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - David R Spriggs
- Division of Hematology & Oncology, Department of Medicine, Massachusetts General Hospital-Harvard Medical School, Boston, MA, USA
| | - Oladapo O Yeku
- Division of Hematology & Oncology, Department of Medicine, Massachusetts General Hospital-Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
26
|
Takeda-Uchimura Y, Ikezaki M, Akama TO, Ihara Y, Allain F, Nishitsuji K, Uchimura K. GlcNAc6ST2/CHST4 Is Essential for the Synthesis of R-10G-Reactive Keratan Sulfate/Sulfated N-Acetyllactosamine Oligosaccharides in Mouse Pleural Mesothelium. Molecules 2024; 29:764. [PMID: 38398516 PMCID: PMC10893525 DOI: 10.3390/molecules29040764] [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: 12/08/2023] [Revised: 01/25/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
We recently showed that 6-sulfo sialyl N-acetyllactosamine (LacNAc) in O-linked glycans recognized by the CL40 antibody is abundant in the pleural mesothelium under physiological conditions and that these glycans undergo complementary synthesis by GlcNAc6ST2 (encoded by Chst4) and GlcNAc6ST3 (encoded by Chst5) in mice. GlcNAc6ST3 is essential for the synthesis of R-10G-positive keratan sulfate (KS) in the brain. The predicted minimum epitope of the R-10G antibody is a dimeric asialo 6-sulfo LacNAc. Whether R-10G-reactive KS/sulfated LacNAc oligosaccharides are also present in the pleural mesothelium was unknown. The question of which GlcNAc6STs are responsible for R-10G-reactive glycans was an additional issue to be clarified. Here, we show that R-10G-reactive glycans are as abundant in the pulmonary pleura as CL40-reactive glycans and that GlcNAc6ST3 is only partially involved in the synthesis of these pleural R-10G glycans, unlike in the adult brain. Unexpectedly, GlcNAc6ST2 is essential for the synthesis of R-10G-positive KS/sulfated LacNAc oligosaccharides in the lung pleura. The type of GlcNAc6ST and the magnitude of its contribution to KS glycan synthesis varied among tissues in vivo. We show that GlcNAc6ST2 is required and sufficient for R-10G-reactive KS synthesis in the lung pleura. Interestingly, R-10G immunoreactivity in KSGal6ST (encoded by Chst1) and C6ST1 (encoded by Chst3) double-deficient mouse lungs was markedly increased. MUC16, a mucin molecule, was shown to be a candidate carrier protein for pleural R-10G-reactive glycans. These results suggest that R-10G-reactive KS/sulfated LacNAc oligosaccharides may play a role in mesothelial cell proliferation and differentiation. Further elucidation of the functions of sulfated glycans synthesized by GlcNAc6ST2 and GlcNAc6ST3, such as R-10G and CL40 glycans, in pathological conditions may lead to a better understanding of the underlying mechanisms of the physiopathology of the lung mesothelium.
Collapse
Affiliation(s)
- Yoshiko Takeda-Uchimura
- Univ. Lille, CNRS, UMR 8576—UGSF—Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France; (Y.T.-U.); (F.A.); or (K.N.)
| | - Midori Ikezaki
- Department of Biochemistry, School of Medicine, Wakayama Medical University, Wakayama 641-8509, Japan; (M.I.); (Y.I.)
| | - Tomoya O. Akama
- Department of Pharmacology, Kansai Medical University, Osaka 570-8506, Japan;
| | - Yoshito Ihara
- Department of Biochemistry, School of Medicine, Wakayama Medical University, Wakayama 641-8509, Japan; (M.I.); (Y.I.)
| | - Fabrice Allain
- Univ. Lille, CNRS, UMR 8576—UGSF—Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France; (Y.T.-U.); (F.A.); or (K.N.)
| | - Kazuchika Nishitsuji
- Univ. Lille, CNRS, UMR 8576—UGSF—Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France; (Y.T.-U.); (F.A.); or (K.N.)
- Department of Biochemistry, School of Medicine, Wakayama Medical University, Wakayama 641-8509, Japan; (M.I.); (Y.I.)
| | - Kenji Uchimura
- Univ. Lille, CNRS, UMR 8576—UGSF—Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France; (Y.T.-U.); (F.A.); or (K.N.)
| |
Collapse
|
27
|
Yousef A, Yousef M, Zeineddine MA, More A, Fanaeian M, Chowdhury S, Knafl M, Edelkamp P, Ito I, Gu Y, Pattalachinti V, Naini ZA, Zeineddine FA, Peterson J, Alfaro K, Foo WC, Jin J, Bhutiani N, Higbie V, Scally CP, Kee B, Kopetz S, Goldstein D, Strach M, Williamson A, Aziz O, Barriuso J, Uppal A, White MG, Helmink B, Fournier KF, Raghav KP, Taggart MW, Overman MJ, Shen JP. Serum Tumor Markers and Outcomes in Patients With Appendiceal Adenocarcinoma. JAMA Netw Open 2024; 7:e240260. [PMID: 38416491 PMCID: PMC10902735 DOI: 10.1001/jamanetworkopen.2024.0260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 01/02/2024] [Indexed: 02/29/2024] Open
Abstract
Importance Serum tumor markers carcinoembryonic antigen (CEA), carbohydrate antigen 19-9 (CA19-9), and cancer antigen 125 (CA125) have been useful in the management of gastrointestinal and gynecological cancers; however, there is limited information regarding their utility in patients with appendiceal adenocarcinoma. Objective To assess the association of serum tumor markers (CEA, CA19-9, and CA125) with clinical outcomes and pathologic and molecular features in patients with appendiceal adenocarcinoma. Design, Setting, and Participants This is a retrospective cohort study at a single tertiary care comprehensive cancer center. The median (IQR) follow-up time was 52 (21-101) months. Software was used to query the MD Anderson internal patient database to identify patients with a diagnosis of appendiceal adenocarcinoma and at least 1 tumor marker measured at MD Anderson between March 2016 and May 2023. Data were analyzed from January to December 2023. Main Outcomes and Measures Association of serum tumor markers with survival in patients with appendiceal adenocarcinoma. Cox proportional hazards regression analyses were also performed to assess associations between clinical factors (serum tumor marker levels, demographics, and patient and disease characteristics) and patient outcomes (overall survival). Results A total of 1338 patients with appendiceal adenocarcinoma were included, with a median (range) age at diagnosis of 56.5 (22.3-89.6) years. The majority of the patients had metastatic disease (1080 patients [80.7%]). CEA was elevated in 742 of the patients tested (56%), while CA19-9 and CA125 were elevated in 381 patients (34%) and 312 patients (27%), respectively. Individually, elevation of CEA, CA19-9, or CA125 were associated with worse 5-year survival; elevated vs normal was 81% vs 95% for CEA (hazard ratio [HR], 4.0; 95% CI, 2.9-5.6), 84% vs 92% for CA19-9 (HR, 2.2; 95% CI, 1.4-3.4), and 69% vs 93% for CA125 (HR, 4.6; 95% CI, 2.7-7.8) (P < .001 for all). Quantitative evaluation of tumor markers was associated with outcomes. Patients with highly elevated (top 10th percentile) CEA, CA19-9, or CA125 had markedly worse survival, with 5-year survival rates of 59% for CEA (HR, 9.8; 95% CI, 5.3-18.0), 64% for CA19-9 (HR, 6.0; 95% CI, 3.0-11.7), and 57% for CA125 (HR, 7.6; 95% CI, 3.5-16.5) (P < .001 for all). Although metastatic tumors had higher levels of all tumor markers, when restricting survival analysis to 1080 patients with metastatic disease, elevated CEA, CA19-9, or CA125 were all still associated worse survival (HR for CEA, 3.4; 95% CI, 2.5-4.8; P < .001; HR for CA19-9, 1.8; 95% CI, 1.2-2.7; P = .002; and HR for CA125, 3.9; 95% CI, 2.4-6.4; P < .001). Interestingly, tumor grade was not associated with CEA or CA19-9 level, while CA-125 was slightly higher in high-grade tumors relative to low-grade tumors (mean value, 18.3 vs 15.0; difference, 3.3; 95% CI, 0.9-3.7; P < .001). Multivariable analysis identified an incremental increase in the risk of death with an increase in the number of elevated tumor markers, with an 11-fold increased risk of death in patients with all 3 tumor markers elevated relative to those with none elevated. Somatic mutations in KRAS and GNAS were associated with significantly higher levels of CEA and CA19-9. Conclusions and Relevance In this retrospective study of serum tumor markers in patients with appendiceal adenocarcinoma, CEA, CA19-9, and CA125 were associated with overall survival in appendiceal adenocarcinoma. Given their value, all 3 biomarkers should be included in the initial workup of patients with a diagnosis of appendiceal adenocarcinoma.
Collapse
Affiliation(s)
- Abdelrahman Yousef
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Mahmoud Yousef
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Mohammad A. Zeineddine
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Aditya More
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Mohammad Fanaeian
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Saikat Chowdhury
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Mark Knafl
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston
| | - Paul Edelkamp
- Department of Data Engineering and Analytics, University of Texas MD Anderson Cancer Center, Houston
| | - Ichiaki Ito
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Yue Gu
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Vinay Pattalachinti
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Zahra Alavi Naini
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Fadl A. Zeineddine
- Department of Internal Medicine, Houston Methodist Hospital, Houston, Texas
| | - Jennifer Peterson
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Kristin Alfaro
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Wai Chin Foo
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston
| | - Jeff Jin
- Department of Enterprise Development and Integration, University of Texas MD Anderson Cancer Center, Houston
| | - Neal Bhutiani
- Department of Colon and Rectal Surgery, University of Texas MD Anderson Cancer Center, Houston
| | - Victoria Higbie
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Christopher P. Scally
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Bryan Kee
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | | | - Madeleine Strach
- Colorectal and Peritoneal Oncology Centre, The Christie NHS Foundation Trust, Manchester, United Kingdom
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
- Faculty of Medicine and Health, The University of Sydney, Darlington, Victoria, Australia
| | - Andrew Williamson
- Department of Medical Oncology, The Christie National Health Service Foundation Trust, Manchester, United Kingdom
| | - Omer Aziz
- Colorectal and Peritoneal Oncology Centre, The Christie NHS Foundation Trust, Manchester, United Kingdom
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Jorge Barriuso
- Colorectal and Peritoneal Oncology Centre, The Christie NHS Foundation Trust, Manchester, United Kingdom
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Abhineet Uppal
- Department of Colon and Rectal Surgery, University of Texas MD Anderson Cancer Center, Houston
| | - Michael G. White
- Department of Colon and Rectal Surgery, University of Texas MD Anderson Cancer Center, Houston
| | - Beth Helmink
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Keith F. Fournier
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Kanwal P. Raghav
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Melissa W. Taggart
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston
| | - Michael J. Overman
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - John Paul Shen
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
28
|
Deng M, Tang F, Chang X, Liu P, Ji X, Hao M, Wang Y, Yang R, Ma Q, Zhang Y, Miao J. Immunotherapy for Ovarian Cancer: Disappointing or Promising? Mol Pharm 2024; 21:454-466. [PMID: 38232985 DOI: 10.1021/acs.molpharmaceut.3c00986] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Ovarian cancer, one of the deadliest malignancies, lacks effective treatment, despite advancements in surgical techniques and chemotherapy. Thus, new therapeutic approaches are imperative to improving treatment outcomes. Immunotherapy, which has demonstrated considerable success in managing various cancers, has already found its place in clinical practice. This review aims to provide an overview of ovarian tumor immunotherapy, including its basics, key strategies, and clinical research data supporting its potential. In particular, this discussion highlights promising strategies such as checkpoint inhibitors, vaccines, and pericyte transfer, both individually and in combination. However, the advancement of new immunotherapies necessitates large controlled randomized trials, which will undoubtedly shape the future of ovarian cancer treatment.
Collapse
Affiliation(s)
- Mengqi Deng
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
| | - Fan Tang
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
| | - Xiangyu Chang
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
| | - Penglin Liu
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
| | - Xuechao Ji
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
| | - Menglin Hao
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
| | - Yixiao Wang
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
| | - Ruiye Yang
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
| | - Qingqing Ma
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
- Nanyuan Hospital of Fengtai District, Beijing 100006, China
| | - Yubo Zhang
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
- Qingdao Hospital, University of Health and Rehabilitation Sciences, Shandong 266011, China
| | - Jinwei Miao
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
| |
Collapse
|
29
|
Wang CW, Weaver SD, Boonpattrawong N, Schuster-Little N, Patankar M, Whelan RJ. A Revised Molecular Model of Ovarian Cancer Biomarker CA125 (MUC16) Enabled by Long-read Sequencing. CANCER RESEARCH COMMUNICATIONS 2024; 4:253-263. [PMID: 38197671 PMCID: PMC10829539 DOI: 10.1158/2767-9764.crc-23-0327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 12/01/2023] [Accepted: 01/04/2024] [Indexed: 01/11/2024]
Abstract
The biomarker CA125, a peptide epitope located in several tandem repeats of the mucin MUC16, is the gold standard for monitoring regression and recurrence of high-grade serous ovarian cancer in response to therapy. However, the CA125 epitope along with several structural features of the MUC16 molecule are ill defined. One central aspect still unresolved is the number of tandem repeats in MUC16 and how many of these repeats contain the CA125 epitope. Studies from the early 2000s assembled short DNA reads to estimate that MUC16 contained 63 repeats.Here, we conduct Nanopore long-read sequencing of MUC16 transcripts from three primary ovarian tumors and established cell lines (OVCAR3, OVCAR5, and Kuramochi) for a more exhaustive and accurate estimation and sequencing of the MUC16 tandem repeats.The consensus sequence derived from these six sources was confirmed by proteomics validation and agrees with recent additions to the NCBI database. We propose a model of MUC16 containing 19-not 63-tandem repeats. In addition, we predict the structure of the tandem repeat domain using the deep learning algorithm, AlphaFold.The predicted structure displays an SEA domain and unstructured linker region rich in proline, serine, and threonine residues in all 19 tandem repeats. These studies now pave the way for a detailed characterization of the CA125 epitope. Sequencing and modeling of the MUC16 tandem repeats along with their glycoproteomic characterization, currently underway in our laboratories, will help identify novel epitopes in the MUC16 molecule that improve on the sensitivity and clinical utility of the current CA125 assay. SIGNIFICANCE Despite its crucial role in clinical management of ovarian cancer, the exact molecular sequence and structure of the biomarker, CA125, are not defined. Here, we combine long-read sequencing, mass spectrometry, and in silico modeling to provide the foundational dataset for a more complete characterization of the CA125 epitope.
Collapse
Affiliation(s)
- Chien-Wei Wang
- Department of Chemistry, University of Kansas, Lawrence, Kansas
| | - Simon D. Weaver
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana
- Integrated Biomedical Sciences Graduate Program, University of Notre Dame, Notre Dame, Indiana
| | - Nicha Boonpattrawong
- Department of Obstetrics and Gynecology, University of Wisconsin–Madison, Madison, Wisconsin
| | | | - Manish Patankar
- Department of Obstetrics and Gynecology, University of Wisconsin–Madison, Madison, Wisconsin
| | | |
Collapse
|
30
|
Ubhenin AE, Adefolalu AA, Oriakhi K, Adamude FA, Dingwoke EJ, Ikebuiro JO, Chiwendu BC, Muhammad ML, Omage K. Caesalpinia pulcherrima lowered serum carcinoembryonic antigen and antigen 125 in 7,12-Dimethylbenz[ a]anthracene-induced Mammary Carcinogenesis in Female Albino Rats. Heliyon 2024; 10:e23401. [PMID: 38187255 PMCID: PMC10770447 DOI: 10.1016/j.heliyon.2023.e23401] [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: 02/07/2023] [Revised: 08/07/2023] [Accepted: 12/03/2023] [Indexed: 01/09/2024] Open
Abstract
Aim This study is aimed at evaluating the anticancer effect of the aqueous extract of Caesalpinia pulcherrima (L.) Sw in 7,12-Dimethlbenz[a]anthracene (DMBA) - induced mammary cancer. Methods Tumors were induced via a single intraperitoneal injection of DMBA (dissolved in olive oil) at a dose of 80 mg/kg body weight to the test rats and allowed to develop for about four months. They were treated with cyclophosphamide and an aqueous extract of Caesalpinia pulcherrima at doses of 10 and 250 mg/kg body weight, respectively, for 28 days. Serum levels of cancer antigen 125 (CA125), carcinoembryonic antigen (CEA) activity, cyclooxygenase-2 (COX-2), and cytochrome p450 oxidase (cytp450) activity, as well as other diagnostic enzymes, were estimated. Results The result revealed that DMBA is associated with a significant (p < 0.05) increase in the serum levels of CA125, CEA, COX-2, cytp450, lactate dehydrogenase (LDH), alkaline phosphatase (ALP), aspartate aminotransferase (AST), and alanine aminotransferase (ALT) of the rats, thus suggesting tumor-promoting and hepatotoxic effects of DMBA. There was also a significant (p < 0.05) reduction of serum levels of these cancer and liver biomarker enzymes in the groups treated with cyclophosphamide and Caesalpinia pulcherrima compared to the untreated group, thus suggesting anticancer activity of Caesalpinia pulcherrima. The anticancer effect of Caesalpinia pulcherrima was further confirmed by the disappearance of infiltrative fibrous cells and the absence of inflammatory cells from the photomicrographs of the rats treated with Caesalpinia pulcherrima. Conclusion Our findings show that Caesalpinia pulcherrima possesses anticancer activity, and could protect against mammary cancer.
Collapse
Affiliation(s)
- Abraham Ehinomhen Ubhenin
- Department of Biochemistry, Faculty of Medical Sciences, Federal University Lafia, Nasarawa State, Nigeria
| | - Adedotun A. Adefolalu
- Department of Biochemistry, Faculty of Medical Sciences, Federal University Lafia, Nasarawa State, Nigeria
| | - Kelly Oriakhi
- Department of Medical Biochemistry, Faculty of Medical Sciences, University of Benin, Benin City, Nigeria
| | - Fatima Adis Adamude
- Department of Biochemistry, Faculty of Medical Sciences, Federal University Lafia, Nasarawa State, Nigeria
| | - Emeka John Dingwoke
- Department of Tropical Diseases, UNESCO-International Center for Biotechnology, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Joshua Onyeka Ikebuiro
- Department of Biochemistry, Faculty of Medical Sciences, Federal University Lafia, Nasarawa State, Nigeria
| | - Benjamin Comfort Chiwendu
- Department of Biochemistry, Faculty of Medical Sciences, Federal University Lafia, Nasarawa State, Nigeria
| | - Mabruqah Liman Muhammad
- Department of Biochemistry, Faculty of Medical Sciences, Federal University Lafia, Nasarawa State, Nigeria
| | - Kingsley Omage
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, United States
| |
Collapse
|
31
|
Nguyen MQ, Kim DH, Shim HJ, Ta HKK, Vu TL, Nguyen TKO, Lim JC, Choe H. Novel Anti-Mesothelin Nanobodies and Recombinant Immunotoxins with Pseudomonas Exotoxin Catalytic Domain for Cancer Therapeutics. Mol Cells 2023; 46:764-777. [PMID: 38052492 PMCID: PMC10701305 DOI: 10.14348/molcells.2023.0155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/10/2023] [Accepted: 10/17/2023] [Indexed: 12/07/2023] Open
Abstract
Recombinant immunotoxins (RITs) are fusion proteins consisting of a targeting domain linked to a toxin, offering a highly specific therapeutic strategy for cancer treatment. In this study, we engineered and characterized RITs aimed at mesothelin, a cell surface glycoprotein overexpressed in various malignancies. Through an extensive screening of a large nanobody library, four mesothelin-specific nanobodies were selected and genetically fused to a truncated Pseudomonas exotoxin (PE24B). Various optimizations, including the incorporation of furin cleavage sites, maltose-binding protein tags, and tobacco etch virus protease cleavage sites, were implemented to improve protein expression, solubility, and purification. The RITs were successfully overexpressed in Escherichia coli, achieving high solubility and purity post-purification. In vitro cytotoxicity assays on gastric carcinoma cell lines NCI-N87 and AGS revealed that Meso(Nb2)-PE24B demonstrated the highest cytotoxic efficacy, warranting further characterization. This RIT also displayed selective binding to human and monkey mesothelins but not to mouse mesothelin. The competitive binding assays between different RIT constructs revealed significant alterations in IC50 values, emphasizing the importance of nanobody specificity. Finally, a modification in the endoplasmic reticulum retention signal at the C-terminus further augmented its cytotoxic activity. Our findings offer valuable insights into the design and optimization of RITs, showcasing the potential of Meso(Nb2)-PE24B as a promising therapeutic candidate for targeted cancer treatment.
Collapse
Affiliation(s)
- Minh Quan Nguyen
- Department of Physiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | | | | | - Huynh Kim Khanh Ta
- Department of Physiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Thi Luong Vu
- Department of Physiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Thi Kieu Oanh Nguyen
- Department of Physiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | | | - Han Choe
- Department of Physiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| |
Collapse
|
32
|
Thapa BV, Banerjee M, Glimm T, Saini DK, Bhat R. The senescent mesothelial matrix accentuates colonization by ovarian cancer cells. Cell Mol Life Sci 2023; 81:2. [PMID: 38043093 PMCID: PMC10694112 DOI: 10.1007/s00018-023-05017-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 10/04/2023] [Accepted: 10/24/2023] [Indexed: 12/05/2023]
Abstract
Ovarian cancer is amongst the most morbid of gynecological malignancies due to its diagnosis at an advanced stage, a transcoelomic mode of metastasis, and rapid transition to chemotherapeutic resistance. Like all other malignancies, the progression of ovarian cancer may be interpreted as an emergent outcome of the conflict between metastasizing cancer cells and the natural defense mounted by microenvironmental barriers to such migration. Here, we asked whether senescence in coelom-lining mesothelia, brought about by drug exposure, affects their interaction with disseminated ovarian cancer cells. We observed that cancer cells adhered faster on senescent human and murine mesothelial monolayers than on non-senescent controls. Time-lapse epifluorescence microscopy showed that mesothelial cells were cleared by a host of cancer cells that surrounded the former, even under sub-confluent conditions. A multiscale computational model predicted that such colocalized mesothelial clearance under sub-confluence requires greater adhesion between cancer cells and senescent mesothelia. Consistent with the prediction, we observed that senescent mesothelia expressed an extracellular matrix with higher levels of fibronectin, laminins and hyaluronan than non-senescent controls. On senescent matrix, cancer cells adhered more efficiently, spread better, and moved faster and persistently, aiding the spread of cancer. Inhibition assays using RGD cyclopeptides suggested the adhesion was predominantly contributed by fibronectin and laminin. These findings led us to propose that the senescence-associated matrisomal phenotype of peritoneal barriers enhances the colonization of invading ovarian cancer cells contributing to the metastatic burden associated with the disease.
Collapse
Affiliation(s)
- Bharat Vivan Thapa
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore, 560012, India
- Undergraduate Program, Indian Institute of Science, Bangalore, 560012, India
| | - Mallar Banerjee
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore, 560012, India
| | - Tilmann Glimm
- Department of Mathematics, Western Washington University, Bellingham, WA, 98229, USA
| | - Deepak K Saini
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore, 560012, India.
- Department of Bioengineering, Indian Institute of Science, Bangalore, 560012, India.
| | - Ramray Bhat
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore, 560012, India.
- Department of Bioengineering, Indian Institute of Science, Bangalore, 560012, India.
| |
Collapse
|
33
|
Gao H, Ji K, Bao L, Chen H, Lin C, Feng M, Tao L, Wang M. Establishment and verification of prediction model of occult peritoneal metastasis in advanced gastric cancer. World J Surg Oncol 2023; 21:320. [PMID: 37833730 PMCID: PMC10571475 DOI: 10.1186/s12957-023-03188-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/17/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND To investigate the risk factors associated with the development of occult peritoneal metastasis in advanced gastric cancer, and establish and externally validate a nomogram for predicting the occurrence of occult peritoneal metastasis in patients with advanced gastric cancer. METHODS A total of 111 patients with advanced gastric cancer who underwent laparoscopic exploration or peritoneal lavage cytology examination at the Affiliated Drum Tower Hospital of Nanjing University Medical School from August 2014 to December 2021 were retrospectively analyzed. The patients diagnosed between 2019 and 2021 were assigned to the training set (n = 64), while those diagnosed between 2014 and 2016 constituted the external validation set (n = 47). In the training set, patients were classified into two groups based on preoperative imaging and postoperative pathological data: the occult peritoneal metastasis group (OPMG) and the peritoneal metastasis negative group (PMNG). In the validation set, patients were classified into the occult peritoneal metastasis group (CY1P0, OPMG) and the peritoneal metastasis negative group (CY0P0, PMNG) based on peritoneal lavage cytology results. A nomogram was constructed using univariate and multivariate analyses. The performance of the nomogram was evaluated using Harrell's C-index, the area under the receiver operating characteristic curve (AUC), decision curve analysis (DCA), and calibration plots. RESULTS This study analyzed 22 potential variables of OPM in 111 gastric cancer patients who underwent laparoscopic exploration or peritoneal lavage cytology examination. Logistic regression analysis results showed that Lauren classification, CLDN18.2 score and CA125 were independent risk factors for OPM in patients with gastric cancer. We developed a simple and easy-to-use prediction nomogram of occult peritoneal metastasis in advanced gastric cancer. This nomogram had an excellent diagnostic performance. The AUC of the bootstrap model in the training set was 0.771 and in the validation set was 0.711. This model showed a good fitting and calibration and positive net benefits in decision curve analysis. CONCLUSION We have developed a prediction nomogram of OPM for gastric cancer. This novel nomogram has the potential to enhance diagnostic accuracy for occult peritoneal metastasis in gastric cancer patients.
Collapse
Affiliation(s)
- Hengfei Gao
- Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- Medical School of Nanjing University, Nanjing, China
| | - Kangkang Ji
- Department of Gastrointestinal, Fuyang People's Hospital, Fuyang, China
| | - Linsen Bao
- Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- Medical School of Nanjing University, Nanjing, China
| | - Hao Chen
- Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- Medical School of Nanjing University, Nanjing, China
| | - Chen Lin
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Min Feng
- Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
- Medical School of Nanjing University, Nanjing, China.
| | - Liang Tao
- Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
- Medical School of Nanjing University, Nanjing, China.
| | - Meng Wang
- Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
- Medical School of Nanjing University, Nanjing, China.
| |
Collapse
|
34
|
Schlam I, Moges R, Morganti S, Tolaney SM, Tarantino P. Next-generation antibody-drug conjugates for breast cancer: Moving beyond HER2 and TROP2. Crit Rev Oncol Hematol 2023; 190:104090. [PMID: 37562695 DOI: 10.1016/j.critrevonc.2023.104090] [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: 04/03/2023] [Revised: 08/02/2023] [Accepted: 08/07/2023] [Indexed: 08/12/2023] Open
Abstract
Antibody-drug conjugates (ADCs) have reshaped the treatment of several malignancies, including breast cancer. Two ADCs are currently approved for the treatment of each breast cancer subtype, including the HER2 targeted ADCs trastuzumab emtansine (T-DM1) and trastuzumab deruxtecan (T-DXd), and the TROP2-targeted ADC sacituzumab govitecan. Each of the ADC components (antibody, linker, and payload) plays a key role in determining the efficacy and toxicity profile of an individual ADC, and their modification can lead to major changes in the clinical profile of these agents. Leveraging the knowledge from three decades of development in the field, several novel ADCs are currently being investigated. Some approaches include targeting different antigens beyond the established HER2/TROP2, or evaluating innovative constructs, such as bispecific ADCs, ADCs with dual payload, immune-modulating ADCs, radionuclide drug conjugates, and masked ADCs, among others. In this review article we discuss the evolving landscape of novel ADCs, highlighting opportunities and challenges emerging in the field.
Collapse
Affiliation(s)
- Ilana Schlam
- Department of Hematology and Oncology, Tufts Medical Center, Boston, MA, USA
| | - Ruth Moges
- Department of Internal Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Stefania Morganti
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Division of New Drugs and Early Drug Development, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Onco-Hematology, University of Milan, Milan, Italy; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sara M Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Paolo Tarantino
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Division of New Drugs and Early Drug Development, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Onco-Hematology, University of Milan, Milan, Italy.
| |
Collapse
|
35
|
Hagerty BL, Takabe K. Biology of Mesothelin and Clinical Implications: A Review of Existing Literature. World J Oncol 2023; 14:340-349. [PMID: 37869242 PMCID: PMC10588497 DOI: 10.14740/wjon1655] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 08/30/2023] [Indexed: 10/24/2023] Open
Abstract
Since its discovery in 1992, mesothelin (MSLN) has generated significant interest as a therapeutic target. A number of characteristics make it ideal for this purpose. First, it is not expressed on the parenchyma of any vital organs. Second, it is differentially expressed on a number of cancer types that have relatively poor prognosis and lack effective systemic options. Third, it is expressed on the cell membrane making it accessible to large molecule targeted therapies. However, unlike other drug targets that have been exploited for therapeutic benefit, the precise function of MSLN, why it is expressed in certain cancers, and its biological role have not been clearly elucidated. Here the existing literature on the cellular function and expression patterns of MSLN across tumor types is reviewed in order to gain further understanding of this intriguing molecule. In doing so, we conclude that there remains significant ambiguity surrounding its function and role in cellular and tumor biology. Furthermore, the expression of MSLN and its relation of prognosis seems to depend on the type of tumor. Finally, the unified mechanism by which MSLN acts as a protein that conveys tumor aggressiveness remains elusive. What is clear is that there is much yet to be discovered in this realm and doing so may have large implications for treatment of otherwise lethal malignancies.
Collapse
Affiliation(s)
- Brendan L Hagerty
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Kazuaki Takabe
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Gastroenterological Surgery, Yokohama City University School of Medicine, Yokohama, Kanagawa, Japan
- Department of Surgery, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY, USA
- Department of Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo, Japan
- Department of Breast Surgery, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
36
|
Thrash HL, Pendergast AM. Multi-Functional Regulation by YAP/TAZ Signaling Networks in Tumor Progression and Metastasis. Cancers (Basel) 2023; 15:4701. [PMID: 37835395 PMCID: PMC10572014 DOI: 10.3390/cancers15194701] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/14/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
The Hippo pathway transcriptional co-activators, YES-associated protein (YAP) and Transcriptional Co-Activator with PDZ Binding Motif (TAZ), have both been linked to tumor progression and metastasis. These two proteins possess overlapping and distinct functions, and their activities lead to the expression of genes involved in multiple cellular processes, including cell proliferation, survival, and migration. The dysregulation of YAP/TAZ-dependent cellular processes can result in altered tumor growth and metastasis. In addition to their well-documented roles in the regulation of cancer cell growth, survival, migration, and invasion, the YAP/TAZ-dependent signaling pathways have been more recently implicated in cellular processes that promote metastasis and therapy resistance in several solid tumor types. This review highlights the role of YAP/TAZ signaling networks in the regulation of tumor cell plasticity mediated by hybrid and reversible epithelial-mesenchymal transition (EMT) states, and the promotion of cancer stem cell/progenitor phenotypes. Mechanistically, YAP and TAZ regulate these cellular processes by targeting transcriptional networks. In this review, we detail recently uncovered mechanisms whereby YAP and TAZ mediate tumor growth, metastasis, and therapy resistance, and discuss new therapeutic strategies to target YAP/TAZ function in various solid tumor types. Understanding the distinct and overlapping roles of YAP and TAZ in multiple cellular processes that promote tumor progression to metastasis is expected to enable the identification of effective therapies to treat solid tumors through the hyper-activation of YAP and TAZ.
Collapse
Affiliation(s)
| | - Ann Marie Pendergast
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
37
|
Yousef A, Yousef M, Zeineddine M, More A, Chowdhury S, Knafl M, Edelkamp P, Ito I, Gu Y, Pattalachinti V, Naini ZA, Zeineddine F, Peterson J, Alfaro K, Foo WC, Jin J, Bhutiani N, Higbie V, Scally C, Kee B, Kopetz S, Goldstein D, Uppal A, White MG, Helmink B, Fournier K, Raghav K, Taggart M, Overman MJ, Shen JP. The Clinical Significance of CEA, CA19-9, and CA125 in Management of Appendiceal Adenocarcinoma. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.10.23295319. [PMID: 37745596 PMCID: PMC10516068 DOI: 10.1101/2023.09.10.23295319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Importance Serum tumor markers CEA, CA19-9, & CA125 have been useful in the management of gastrointestinal and gynecological cancers, however there is limited information regarding their utility in patients with appendiceal adenocarcinoma. Objective Assessing the association of serum tumor markers (CEA, CA19-9, and CA125) with clinical outcomes, pathologic, and molecular features in patients with appendiceal adenocarcinoma. Design This is a retrospective study with results reported in 2023. The median follow-up time was 43 months. Setting Single tertiary care comprehensive cancer center. Participants Under an approved Institutional Review Board protocol, the Palantir Foundry software system was used to query the MD Anderson internal patient database to identify patients with a diagnosis of appendiceal adenocarcinoma and at least one tumor marker measured at MD Anderson between 2016 and 2023. Results A total of 1,338 patients with appendiceal adenocarcinoma were included, with a median age of 56.5 years. The majority of the patients had metastatic disease (80.7%). CEA was elevated in more than half of the patients tested (56%), while CA19-9 and CA125 were elevated in 34% and 27%, respectively. Individually, elevation of CEA, CA19-9, or CA125 were associated with worse 5-year survival; 82% vs 95%, 84% vs 92%, and 69% vs 93% elevated vs normal for CEA, CA19-9, and CA125 respectively (all p<0.0001). Quantitative evaluation of tumor markers increased prognostic ability. Patients with highly elevated (top 10th percentile) CEA, CA19-9 or CA125 had markedly worse survival with 5-year survival rates of 59%, 64%, and 57%, respectively (HR vs. normal : 9.8, 6.0, 7.6, all p<0.0001). Although metastatic tumors had higher levels of all tumor markers, when restricting survival analysis to 1080 patients with metastatic disease elevated CEA, CA19-9 or CA125 were all still associated worse survival (HR vs. normal : 3.4, 1.8, 3.9, p<0.0001 for CEA and CA125, p=0.0019 for CA19-9). Interestingly tumor grade was not associated with CEA or CA19-9 level, while CA-125 was slightly higher in high relative to low-grade tumors (18.3 vs. 15.0, p=0.0009). Multivariable analysis identified an incremental increase in the risk of death with an increase in the number of elevated tumor markers, with a 11-fold increased risk of death in patients with all three tumor markers elevated relative to those with none elevated. Mutation in KRAS and GNAS were associated with significantly higher levels of CEA and CA19-9. Conclusions These findings demonstrate the utility of measuring CEA, CA19-9, and CA125 in the management of appendiceal adenocarcinoma. Given their prognostic value, all three biomarkers should be included in the initial workup of patients diagnosed with appendiceal adenocarcinoma.
Collapse
Affiliation(s)
- Abdelrahman Yousef
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mahmoud Yousef
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mohammad Zeineddine
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Aditya More
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Saikat Chowdhury
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mark Knafl
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paul Edelkamp
- Department of Data Engineering & Analytics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ichiaki Ito
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yue Gu
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vinay Pattalachinti
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zahra Alavi Naini
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fadl Zeineddine
- Department of Internal Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Jennifer Peterson
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kristin Alfaro
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wai Chin Foo
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeff Jin
- Department of Enterprise Dev & Integration, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Neal Bhutiani
- Department of Colon and Rectal Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Victoria Higbie
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christopher Scally
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bryan Kee
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Abhineet Uppal
- Department of Colon and Rectal Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael G. White
- Department of Colon and Rectal Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Beth Helmink
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Keith Fournier
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kanwal Raghav
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Melissa Taggart
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael J. Overman
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John Paul Shen
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
38
|
Zhao H, Wu L, Dai J, Sun K, Zi Z, Guan J, Zhang L. Ligand-based adoptive T cell targeting CA125 in ovarian cancer. J Transl Med 2023; 21:596. [PMID: 37670338 PMCID: PMC10481596 DOI: 10.1186/s12967-023-04271-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/13/2023] [Indexed: 09/07/2023] Open
Abstract
BACKGROUND Ovarian cancer (OC) is a highly aggressive gynecological malignancy prevalent worldwide. Most OC cases are typically diagnosed at advanced stages, which has led to a 5-year overall survival rate of less than 35% following conventional treatment. Furthermore, immune checkpoint inhibitor therapy has shown limited efficacy in the treatment of patients with OC, and CAR-T therapy has also demonstrated modest results owing to inadequate T cell infiltration. Therefore, novel strategies must be developed to enhance T cell persistence and trafficking within the OC tumor microenvironment. METHODS In this study, we developed a novel adoptive T-cell therapy for ovarian cancer based on a chimeric antigen receptor structure. We used a ligand-receptor binding motif to enhance the therapeutic effect of targeting CA125. Since mesothelin can naturally bind to CA125 with high affinity, we concatenated the core-binding fragment of mesothelin with the 4-1BB and CD3ζ signal fragments to assemble a novel CA125-targeting chimeric receptor (CR). The CAR structure targeting CA125 derived from the 4H11 antibody was also constructed. CR- and CAR-encoding RNA were electroporated into T cells to evaluate their antitumor activity both in vitro and in vivo. RESULTS While CR-T or CAR-T cells exhibited moderate activity against two ovarian cancer cell lines, T cells co-expressing CR and CAR exhibited a superior killing effect compared to T cells expressing either CR or CAR alone. Furthermore, upon interaction with ovarian tumors, the ability of CR and CAR T cells to release activation markers and functional cytokines increased significantly. Similarly, CR and CAR co-expressing T cells persistently controlled the growth of transplanted ovarian cancer tumors in NSG mice and significantly prolonged the overall survival of tumor-challenged mice. Transcriptome sequencing revealed that the survival and cytotoxicity of T cells co-expressing CR and CAR were significantly altered compared with those of T cells expressing either CR or CAR. CONCLUSION Our findings demonstrate that CA125 targeting CR and CAR can synergistically kill ovarian cancer cells, indicating that CA125 targeting by the two binding motifs simultaneously in tumors may improve the therapeutic outcomes of ovarian cancer treatment.
Collapse
Affiliation(s)
- Haihong Zhao
- Department of Obstetrics and Gynecology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, 200240, China
| | - Lina Wu
- Department of Obstetrics and Gynecology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, 200240, China
| | - Jiemin Dai
- Department of Obstetrics and Gynecology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, 200240, China
| | - Ke Sun
- Department of Obstetrics and Gynecology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, 200240, China
| | - Zhenguo Zi
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Junhua Guan
- Department of Obstetrics and Gynecology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, 200240, China.
| | - Liwen Zhang
- Department of Obstetrics and Gynecology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, 200240, China.
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
39
|
Shen Y, Li Y, Wang Z, Xu W, Wang W, Chen X. The prognostic value of FAR and a novel FAR-CA125 score in resectable gastric signet ring cell carcinoma patients. J Cancer Res Clin Oncol 2023; 149:9597-9608. [PMID: 37222811 DOI: 10.1007/s00432-023-04870-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 05/19/2023] [Indexed: 05/25/2023]
Abstract
BACKGROUND The fibrinogen to albumin ratio (FAR) is increasingly regarded as a potential biomarker for predicting prognosis in variety of malignant tumors, but not in gastric signet ring cell carcinoma (GSRC). This study seeks to examine the prognostic value of the FAR and explore a novel FAR-CA125 score (FCS) in resectable GSRC patients. METHODS A retrospective cohort was conducted including 330 GSRC patients who underwent curative resection. Kaplan-Meier (K-M) and Cox regression were used to analysis the prognostic value of FAR and FCS. And a predictive nomogram model was developed. RESULTS The optimal cut-off values for CA125 and FAR were 9.88 and 0.0697, respectively, according to the receiver operating characteristic curve (ROC). Th area under the ROC curve of FCS is higher than CA125 and FAR. 330 patients were grouped into three groups according to the FCS. High FCS was related to males, anemia, tumor size, TNM stage, lymph node metastasis, tumor invasion depth, SII, and pathological subtypes. K-M analysis showed that high FCS and FAR were associated with poor survival. In the multivariate analysis, FCS, TNM stage, and SII were independent prognostic factors for poor OS in resectable GSRC patients. And the predictive accuracy of clinical nomogram contained FCS was better than TNM stage. CONCLUSION This study indicated that the FCS is a prognostic, and effective biomarker for patients with surgically resectable GSRC. Such developed FCS-based nomogram could be effective tools to assist the clinicians to determine the treatment strategy.
Collapse
Affiliation(s)
- Yimin Shen
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Yuanyuan Li
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Zhou Wang
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Wei Xu
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Wenjie Wang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Xiao Chen
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China.
| |
Collapse
|
40
|
McNamara B, Chang Y, Goreshnik A, Santin AD. Value of Antibody Drug Conjugates for Gynecological Cancers: A Modern Appraisal Following Recent FDA Approvals. Int J Womens Health 2023; 15:1353-1365. [PMID: 37663226 PMCID: PMC10474218 DOI: 10.2147/ijwh.s400537] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 08/09/2023] [Indexed: 09/05/2023] Open
Abstract
Antibody drug conjugates (ADCs) are a new class of targeted anti-cancer therapies that combine a monoclonal tumor surface receptor-targeting antibody with a highly cytotoxic molecule payload. They enable delivery of cytotoxic therapy more directly to tumor cells and minimize delivery to healthy tissues. This review summarizes the existing literature about ADC therapies approved for use in gynecologic malignancies, relevant preclinical studies, as well as ongoing clinical trials.
Collapse
Affiliation(s)
- Blair McNamara
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Yifan Chang
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Ashley Goreshnik
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Alessandro D Santin
- Department of Obstetrics, Gynecology, and Reproductive Sciences Yale University School of Medicine, New Haven, CT, 06520, USA
| |
Collapse
|
41
|
Wang Y, Liu L, Yu Y. Mucins and mucinous ovarian carcinoma: Development, differential diagnosis, and treatment. Heliyon 2023; 9:e19221. [PMID: 37664708 PMCID: PMC10468386 DOI: 10.1016/j.heliyon.2023.e19221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 09/05/2023] Open
Abstract
Mucinous ovarian carcinoma (MOC) is a rare histological type of epithelial ovarian cancer. It has poor response to conventional platinum-based chemotherapy regimens and PARPi-based maintenance treatment, resulting in short survival and poor prognosis in advanced-disease patients. MOC is characterized by mucus that is mainly composed of mucin in the cystic cavity. Our review discusses in detail the role of mucins in MOC. Mucins are correlated with MOC development. Furthermore, they are valuable in the differential diagnosis of primary and secondary ovarian mucinous tumors. Some types of mucins have been studied in the context of chemoresistance and targeted therapy for ovarian cancer. This review may provide a new direction for the diagnosis and treatment of advanced MOC.
Collapse
Affiliation(s)
- Yicong Wang
- Department of Obstetrics and Gynecology, Dalian Municipal Central Hospital, Dalian, China
| | - Lifeng Liu
- Department of Obstetrics and Gynecology, Dalian Municipal Central Hospital, Dalian, China
| | - Yongai Yu
- Department of Obstetrics and Gynecology, Dalian Municipal Central Hospital, Dalian, China
| |
Collapse
|
42
|
Grasso L, Jiang Q, Hassan R, Nicolaides NC, Kline JB. NAV-003, a bispecific antibody targeting a unique mesothelin epitope and CD3ε with improved cytotoxicity against humoral immunosuppressed tumors. Eur J Immunol 2023; 53:e2250309. [PMID: 37146241 PMCID: PMC10524251 DOI: 10.1002/eji.202250309] [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: 12/05/2022] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 05/07/2023]
Abstract
Mesothelin (MSLN) is a cell surface protein overexpressed in a number of cancer types. Several antibody- and cellular-based MSLN targeting agents have been tested in clinical trials where their therapeutic efficacy has been moderate at best. Previous studies using antibody and Chimeric Antigen Receptor-T cells (CAR-T) strategies have shown the importance of particular MSLN epitopes for optimal therapeutic response, while other studies have found that certain MSLN-positive tumors can produce proteins that can bind to subsets of IgG1-type antibodies and suppress their immune effector activities. In an attempt to develop an improved anti-MSLN targeting agent, we engineered a humanized divalent anti-MSLN/anti-CD3ε bispecific antibody that avoids suppressive factors, can target a MSLN epitope proximal to the tumor cell surface, and is capable of effectively binding, activating, and redirecting T cells to the surface of MSLN-positive tumor cells. NAV-003 has shown significantly improved tumor cell killing against lines producing immunosuppressive proteins in vitro and in vivo. Moreover, NAV-003 demonstrated good tolerability in mice and efficacy against patient-derived mesothelioma xenografts co-engrafted with human peripheral blood mononuclear cells. Together these data support the potential for NAV-003 clinical development and human proof-of-concept studies in patients with MSLN-expressing cancers.
Collapse
Affiliation(s)
- Luigi Grasso
- Navrogen Inc., 1837 University Circle, Cheyney, PA 19319
| | - Qun Jiang
- Thoracic and GI Malignancies Branch, CCR, NCI, NIH, Bethesda, Maryland
| | - Raffit Hassan
- Thoracic and GI Malignancies Branch, CCR, NCI, NIH, Bethesda, Maryland
| | | | | |
Collapse
|
43
|
Tominaga S, Ojima T, Miyazawa M, Iwamoto H, Kitadani J, Maruoka S, Hayata K, Yamaue H. Induced pluripotent stem cell-derived dendritic cell vaccine therapy genetically modified on the ubiquitin-proteasome system. Gene Ther 2023; 30:552-559. [PMID: 36959396 DOI: 10.1038/s41434-023-00388-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 01/16/2023] [Accepted: 01/27/2023] [Indexed: 03/25/2023]
Abstract
We previously reported that dendritic cells (DCs) transduced with the full-length tumor-associated antigen (TAA) gene induced TAA-specific cytotoxic T lymphocytes (CTLs) to elicit antitumor responses. To overcome the issue of quantity and quality of DCs required for DC vaccine therapy, we focused on induced pluripotent stem cells (iPSCs) as a new tool for obtaining DCs and reported efficacy of iPSCs-derived DCs (iPSDCs). However, in clinical application of iPSDC vaccine therapy, further enhancement of the antitumor effect is necessary. In this study, we targeted mesothelin (MSLN) as a potentially useful TAA, and focused on the ubiquitin-proteasome system to enhance antigen-presenting ability of iPSDCs. The CTLs induced by iPSDCs transduced with MSLN gene (iPSDCs-MSLN) from healthy donors showed cytotoxic activity against autologous lymphoblastoid cells (LCLs) expressing MSLN (LCLs-MSLN). The CTLs induced by iPSDCs transduced ubiquitin-MSLN fusion gene exhibited higher cytotoxic activity against LCLs-MSLN than the CTLs induced by iPSDCs-MSLN. The current study was designed that peripheral T-cell tolerance to MSLN could be overcome by the immunization of genetically modified iPSDCs simultaneously expressing ubiquitin and MSLN, leading to a strong cytotoxicity against tumors endogenously expressing MSLN. Therefore, this strategy may be promising for clinical application as an effective cancer vaccine therapy.
Collapse
Affiliation(s)
- Shinta Tominaga
- Second Department of Surgery, Wakayama Medical University, Wakayama, Japan
| | - Toshiyasu Ojima
- Second Department of Surgery, Wakayama Medical University, Wakayama, Japan.
| | - Motoki Miyazawa
- Second Department of Surgery, Wakayama Medical University, Wakayama, Japan
| | - Hiromitsu Iwamoto
- Second Department of Surgery, Wakayama Medical University, Wakayama, Japan
| | - Junya Kitadani
- Second Department of Surgery, Wakayama Medical University, Wakayama, Japan
| | - Shimpei Maruoka
- Second Department of Surgery, Wakayama Medical University, Wakayama, Japan
| | - Keiji Hayata
- Second Department of Surgery, Wakayama Medical University, Wakayama, Japan
| | - Hiroki Yamaue
- Second Department of Surgery, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
44
|
Fu X, Wang Q, Du H, Hao H. CXCL8 and the peritoneal metastasis of ovarian and gastric cancer. Front Immunol 2023; 14:1159061. [PMID: 37377954 PMCID: PMC10291199 DOI: 10.3389/fimmu.2023.1159061] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
CXCL8 is the most representative chemokine produced autocrine or paracrine by tumor cells, endothelial cells and lymphocytes. It can play a key role in normal tissues and tumors by activating PI3K-Akt, PLC, JAK-STAT, and other signaling pathways after combining with CXCR1/2. The incidence of peritoneal metastasis in ovarian and gastric cancer is extremely high. The structure of the peritoneum and various peritoneal-related cells supports the peritoneal metastasis of cancers, which readily produces a poor prognosis, low 5-year survival rate, and the death of patients. Studies show that CXCL8 is excessively secreted in a variety of cancers. Thus, this paper will further elaborate on the mechanism of CXCL8 and the peritoneal metastasis of ovarian and gastric cancer to provide a theoretical basis for the proposal of new methods for the prevention, diagnosis, and treatment of cancer peritoneal metastasis.
Collapse
|
45
|
Bukkuri A, Adler FR. Biomarkers or biotargets? Using competition to lure cancer cells into evolutionary traps. Evol Med Public Health 2023; 11:264-276. [PMID: 37599857 PMCID: PMC10439788 DOI: 10.1093/emph/eoad017] [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: 12/23/2022] [Revised: 03/23/2023] [Indexed: 08/22/2023] Open
Abstract
Background and Objectives Cancer biomarkers provide information on the characteristics and extent of cancer progression and help inform clinical decision-making. However, they can also play functional roles in oncogenesis, from enabling metastases and inducing angiogenesis to promoting resistance to chemotherapy. The resulting evolution could bias estimates of cancer progression and lead to suboptimal treatment decisions. Methodology We create an evolutionary game theoretic model of cell-cell competition among cancer cells with different levels of biomarker production. We design and simulate therapies on top of this pre-existing game and examine population and biomarker dynamics. Results Using total biomarker as a proxy for population size generally underestimates chemotherapy efficacy and overestimates targeted therapy efficacy. If biomarker production promotes resistance and a targeted therapy against the biomarker exists, this dynamic can be used to set an evolutionary trap. After chemotherapy selects for a high biomarker-producing cancer cell population, targeted therapy could be highly effective for cancer extinction. Rather than using the most effective therapy given the cancer's current biomarker level and population size, it is more effective to 'overshoot' and utilize an evolutionary trap when the aim is extinction. Increasing cell-cell competition, as influenced by biomarker levels, can help prime and set these traps. Conclusion and Implications Evolution of functional biomarkers amplify the limitations of using total biomarker levels as a measure of tumor size when designing therapeutic protocols. Evolutionarily enlightened therapeutic strategies may be highly effective, assuming a targeted therapy against the biomarker is available.
Collapse
Affiliation(s)
- Anuraag Bukkuri
- Tissue Development and Evolution Research Group, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Cancer Biology and Evolution Program and Department of Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Frederick R Adler
- Department of Mathematics, University of Utah, Salt Lake City, UT, USA
- School of Biological Sciences, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
46
|
Chaudhary S, Appadurai MI, Maurya SK, Nallasamy P, Marimuthu S, Shah A, Atri P, Ramakanth CV, Lele SM, Seshacharyulu P, Ponnusamy MP, Nasser MW, Ganti AK, Batra SK, Lakshmanan I. MUC16 promotes triple-negative breast cancer lung metastasis by modulating RNA-binding protein ELAVL1/HUR. Breast Cancer Res 2023; 25:25. [PMID: 36918912 PMCID: PMC10012760 DOI: 10.1186/s13058-023-01630-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 03/01/2023] [Indexed: 03/16/2023] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is highly aggressive with an increased metastatic incidence compared to other breast cancer subtypes. However, due to the absence of clinically reliable biomarkers and targeted therapy in TNBC, outcomes are suboptimal. Hence, there is an urgent need to understand biological mechanisms that lead to identifying novel therapeutic targets for managing metastatic TNBC. METHODS The clinical significance of MUC16 and ELAVL1 or Hu antigen R (HuR) was examined using breast cancer TCGA data. Microarray was performed on MUC16 knockdown and scramble TNBC cells and MUC16-associated genes were identified using RNA immunoprecipitation and metastatic cDNA array. Metastatic properties of MUC16 were evaluated using tail vein experiment. MUC16 and HuR downstream pathways were confirmed by ectopic overexpression of MUC16-carboxyl-terminal (MUC16-Cter), HuR and cMyc as well as HuR inhibitors (MS-444 and CMLD-2) in TNBC cells. RESULTS MUC16 was highly expressed in TNBC and correlated with its target HuR. Depletion of MUC16 showed decreased invasion, migration, and colony formation abilities of human and mouse TNBC cells. Mice injected with MUC16 depleted cells were less likely to develop lung metastasis (P = 0.001). Notably, MUC16 and HuR were highly expressed in the lung tropic TNBC cells and lung metastases. Mechanistically, we identified cMyc as a HuR target in TNBC using RNA immunoprecipitation and metastatic cDNA array. Furthermore, MUC16 knockdown and pharmacological inhibition of HuR (MS-444 and CMLD-2) in TNBC cells showed a reduction in cMyc expression. MUC16-Cter or HuR overexpression models indicated MUC16/HuR/cMyc axis in TNBC cell migration. CONCLUSIONS Our study identified MUC16 as a TNBC lung metastasis promoter that acts through HuR/cMyc axis. This study will form the basis of future studies to evaluate the targeting of both MUC16 and HuR in TNBC patients.
Collapse
Affiliation(s)
- Sanjib Chaudhary
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Muthamil Iniyan Appadurai
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Shailendra Kumar Maurya
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Palanisamy Nallasamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Saravanakumar Marimuthu
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Ashu Shah
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Pranita Atri
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Chirravuri Venkata Ramakanth
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Subodh M Lele
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198-5900, USA
| | - Parthasarathy Seshacharyulu
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198-5900, USA
| | - Mohd W Nasser
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Apar Kishor Ganti
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
- Division of Oncology-Hematology, Department of Internal Medicine, VA Nebraska Western Iowa Health Care System, University of Nebraska Medical Center, Omaha, NE, 68105-1850, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA.
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198-5900, USA.
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA.
| | - Imayavaramban Lakshmanan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA.
| |
Collapse
|
47
|
Chelariu-Raicu A, Mahner S, Moore KN, Lorusso D, Coleman RL. Integrating antibody drug conjugates in the management of gynecologic cancers. Int J Gynecol Cancer 2023; 33:420-429. [PMID: 36878560 DOI: 10.1136/ijgc-2022-003701] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023] Open
Abstract
The clinical development of antibody drug conjugates (ADCs) in ovarian cancer began in 2008 with farletuzumab, a humanized monoclonal antibody, and vintafolide, an antigen drug conjugate, both targeting alpha folate receptor. Over the years, this novel class of drugs expanded to agents with a more sophisticated design and structure, targeting tissue factor (TF) in cervical cancer or human epidermal growth factor receptor 2 (HER2) in endometrial cancer. Despite the impressive number of patients included in clinical trials investigating different ADCs across gynecological cancers, it was only recently that the Food and Drug Administration (FDA) granted accelerated approvals to the first ADCs in gynecologic cancer. In September 2021, the FDA approved tisotumab vedotin (TV) in recurrent or metastatic cervical cancer with disease progression on or after chemotherapy. This was followed in November 2022, by the approval of mirvetuximab soravtansine (MIRV) for adult patients with folate receptor alpha (FRα) positive, platinum-resistant epithelial ovarian, fallopian tube, or primary peritoneal cancer, who have received one to three prior systemic treatment regimens. Currently, the field of ADCs is rapidly expanding and more than 20 ADC formulations are in clinical trials for the treatment of ovarian, cervical and endometrial tumors. This review summarizes key evidence supporting their use and therapeutic indications, including results from late-stage development trials investigating MIRV in ovarian cancer and TV in cervical cancer. We also outline new concepts in the field of ADCs, including promising targets such as NaPi2 and novel drug delivery platforms such as dolaflexin with a scaffold-linker. Finally, we briefly present challenges in the clinical management of ADC toxicities and the emerging role of ADC combination therapies, including chemotherapy, anti-angiogenic and immunotherapeutic agents.
Collapse
Affiliation(s)
- Anca Chelariu-Raicu
- Department of Obstetrics and Gynecology, Breast Center, Gynecologic Cancer Center and CCC Munich, LMU University Hospital, Munich, Germany
- German Cancer Research Center, German Cancer Consortium (DKTK), Munich, Germany
| | - Sven Mahner
- Department of Obstetrics and Gynecology, Breast Center, Gynecologic Cancer Center and CCC Munich, LMU University Hospital, Munich, Germany
- German Cancer Research Center, German Cancer Consortium (DKTK), Munich, Germany
| | - Kathleen Nadine Moore
- Division of Gynecologic Oncology, Stephenson Cancer Center, University of Oklahoma, Oklahoma city, Oklahoma, USA
| | - Domenica Lorusso
- Gynecologic Oncology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Catholic Univeristy of Sacred Heart, Rome, Italy
| | | |
Collapse
|
48
|
Lin HJ, Hu RM, Chen HC, Lin CC, Lee CY, Chou CY. CA125 for the Diagnosis of Advanced Urothelial Carcinoma of the Bladder: A Systematic Review and Meta-Analysis. Cancers (Basel) 2023; 15:cancers15030813. [PMID: 36765770 PMCID: PMC9913454 DOI: 10.3390/cancers15030813] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/23/2023] [Accepted: 01/26/2023] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Urothelial carcinoma of the bladder (UCB) is the second most common genitourinary cancer. This study aims to assess the diagnostic accuracy of CA125 in advanced UCB. METHODS We searched prevalent studies in PubMed, the Cochrane Library, Scopus, Embase, the Web of Science China National Knowledge Infrastructure database, and Wanfang data before October 2022. Pooled sensitivity, specificity, and summary receiver operating characteristics were used to assess the diagnostic value of CA125. RESULTS One thousand six hundred forty-one patients from 14 studies were analyzed. UCB stage T3-4N1 was defined as advanced UCB in ten studies; T2-4 was used in three studies; and N1M1 in one study. Patients' age was between 21 to 92, and 21% to 48.6% of patients were female. The pooled sensitivity was 0.695 (95% confidence interval (CI): 0.426-0.875). The pooled specificity was 0.846 (95% CI: 0.713-0.924). The diagnostic odds ratio was 8.138 (95% CI: 4.559-14.526). The AUC was 0.797. CONCLUSION CA125 may provide significant diagnostic accuracy in identifying muscle-invasive, lymph node-involved, and distant metastatic tumors in patients with urothelial carcinoma of the bladder. Limited studies have been conducted on the prognostic role of CA125. More studies are needed for a meta-analysis on the prognostic role of CA125 in UCB.
Collapse
Affiliation(s)
- Hsuan-Jen Lin
- Division of Nephrology, Asia University Hospital, Taichung 41354, Taiwan
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung 41354, Taiwan
| | - Rouh-Mei Hu
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung 41354, Taiwan
| | - Hung-Chih Chen
- Division of Nephrology, Asia University Hospital, Taichung 41354, Taiwan
| | - Chung-Chih Lin
- Division of Nephrology, Asia University Hospital, Taichung 41354, Taiwan
| | - Chi-Yu Lee
- Division of Nephrology, Asia University Hospital, Taichung 41354, Taiwan
| | - Che-Yi Chou
- Division of Nephrology, Asia University Hospital, Taichung 41354, Taiwan
- College of Medicine, China Medical University, Taichung 404332, Taiwan
- Division of Nephrology and Kidney Institute, China Medical University Hospital, Taichung 404332, Taiwan
- Department of Post-baccalaureate Veterinary Medicine, Asia University, Taichung 41354, Taiwan
- Correspondence: ; Tel.: +886-4-3706-1668-1878; Fax: +886-4-3706-1668-1338
| |
Collapse
|
49
|
Bakun OV, Koval HD, Dudka YA, Oshchepkova IA, Makoviichuk KY. INFLUENCE OF PROBIOTICS ON THE MESOTHELIN LEVEL IN WOMEN WITH ENDOMETRIOSIS ASSOCIATED WITH INFERTILITY IN COMPLEX PREPARATION FOR ASSISTED REPRODUCTIVE TECHNOLOGIES. WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2023; 76:2455-2459. [PMID: 38112364 DOI: 10.36740/wlek202311118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
OBJECTIVE The aim: To study the determination of Mesothelin level in women with endometriosis associated with infertility and estimate influence of probiotic on endometriosis according of Mesothelin level in complex preparation before assisted reproductive technologies. PATIENTS AND METHODS Materials and methods: In this study, we conducted a retrospective analysis of the medical records of 40 infertile women who underwent assisted reproductive technologies while also using the probiotic "Femina Probiz." We divided the participants into two groups. The control group comprised 11 women who had tubal infertility due to a previous inflammatory condition but were otherwise found to be in good health through comprehensive clinical and laboratory assessments. These women, aged between 21 and 42 with an average age of 29.75 years, did not use the probiotic "Femina Probiz." The main group consisted of 29 women diagnosed with external genital endometriosis who were undergoing assisted reproductive technologies. Women in the main group received the probiotic "Femina Probiz" from Unic Biotech Ltd, India. They took one tablet twice a day for one month as part of their overall treatment before undergoing assisted reproductive technologies. We measured the Mesothelin levels before and after this preparation phase. This study was conducted at Bukovinian State Medical University and Centre of Reproductive Medicine. It's worth noting that the primary infertility incidence was significantly higher in the main group of patients. RESULTS Results: In the main group, we observed that the Mesothelin level was 0.73±0.01, which was significantly higher than the post-preparation level (0.59±0.01). In contrast, the control group had a Mesothelin level of 0.49±0.01. Interestingly, we noted that the Mesothelin level in patients increased approximately twofold before preparation compared to those who had undergone preparation. This suggests that the use of the probiotic led to a sharp reduction in the elevated Mesothelin levels. Consequently, the significant decrease in Mesothelin levels after using the probiotic indicates its effectiveness and potential utility in the preparation phase of assisted reproductive technologies programs. CONCLUSION Conclusions: The elevated Mesothelin levels indicate a strong association between the pathogenesis of endometriosis and inflammation, as well as damage to the peritoneum. The incorporation of a probiotic as part of a comprehensive preparation regimen prior to assisted reproductive technologies notably enhances the overall health of patients and leads to a reduction in Mesothelin levels. Based on our findings, we highly recommend the inclusion of this probiotic preparation in clinical practice.
Collapse
Affiliation(s)
- Oksana V Bakun
- BUKOVINIAN STATE MEDICAL UNIVERSITY, CHERNIVTSI, UKRAINE
| | - Halyna D Koval
- BUKOVINIAN STATE MEDICAL UNIVERSITY, CHERNIVTSI, UKRAINE
| | | | | | | |
Collapse
|
50
|
Evaluation of serum CA125-Tn glycoform in peritoneal dissemination and surgical completeness of high-grade serous ovarian cancer. J Ovarian Res 2022; 15:134. [PMID: 36564848 PMCID: PMC9784250 DOI: 10.1186/s13048-022-01066-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/22/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Peritoneal dissemination is the predominant feature of malignant progression in ovarian cancer and is a major cause of poor surgical outcomes and clinical prognoses. Abnormal glycosylation of carbohydrate antigen 125 (CA125) may be involved in peritoneal implantation and metastasis. Here, we evaluated the clinical relevance of CA125-Tn glycoform in the assessment of high-grade serous ovarian cancer (HGSOC). METHODS A total of 72 patients diagnosed with HGSOC were included. Pre-treatment serum CA125-Tn levels were measured using an antibody-lectin enzyme-linked immunosorbent assay. The association of CA125-Tn with clinical factors was analyzed in all cases, whereas its association with peritoneal dissemination, residual disease, and progression-free survival was analyzed in stage III-IV cases. RESULTS Pre-treatment serum CA125-Tn levels were significantly higher in advanced-stage HGSOC patients than in early-stage patients (P = 0.029). In advanced-stage patients, the pre-treatment CA125-Tn level increased with an increase in Fagotti's score (P = 0.004) and with the extension of peritoneal dissemination (P = 0.011). The pre-treatment CA125-Tn level increased with the volume of residual disease (P = 0.005). The association between CA125-Tn level and suboptimal surgery remained significant even after adjustment for treatment type and stage. Pre-treatment CA125-Tn levels were also related to disease recurrence. CONCLUSION Serum CA125-Tn level could be a novel biomarker for peritoneal dissemination and a promising predictor of surgical completeness in ovarian cancer. Patients with lower CA125-Tn levels were more likely to have no residual disease. CA125-Tn could help surgeons to adopt optimized treatment strategies for patients with advanced ovarian cancer as a pre-treatment evaluator.
Collapse
|