1
|
Wang J, Wang Y, Zhou H, Yu G, Xu H, Gao D, Li M, Wang Y, Xu B. Identification of the specific characteristics of neuroendocrine prostate cancer: Immune status, hub genes and treatment. Transl Oncol 2025; 54:102320. [PMID: 39999729 PMCID: PMC11908612 DOI: 10.1016/j.tranon.2025.102320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 01/13/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Castration-resistant prostate cancer (CRPC) marks the advanced phase of prostate malignancy, manifested through two principal subtypes: castration-resistant adenocarcinoma (CRPC-adeno) and neuroendocrine prostate cancer (NEPC). This study aims to identify unique central regulatory genes, assess the immunological landscape, and explore potential therapeutic strategies specifically tailored to NEPC. We discovered 1444 differentially expressed genes (DEGs) distinguishing between the two cancer types and identified 12 critical hub genes. Notably, CHST1, MPPED2, and RIPPLY3 emerged as closely associated with the immune cell infiltration pattern, establishing them as top candidates. Prognostic analysis highlighted the potential critical roles of CHST1 and MPPED2 in prostate cancer development, findings corroborated through in vitro and in vivo assays. Moreover, we validated the functions and expression levels of CHST1, MPPED2, and RIPPLY3 in NEPC using cell lines, animal models and human tissues. In the final step, we found that imatinib might be the drug specific to NEPC, which was further confirmed by in vitro cell assay. Our results revealed the clinical characteristics, molecular features, immune cell infiltration pattern in CRPC-adeno and NEPC, and identified and confirmed CHST1, MPPED2, and RIPPLY3 as the critical genes in the development in prostate cancer and NEPC. We also predicted and validated imatinib as the potential specific drugs to NEPC.
Collapse
Affiliation(s)
- Jianqing Wang
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, China
| | - Yu Wang
- Vancouver Prostate Centre, Vancouver, BC, Canada; Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Huihui Zhou
- Department of Pathology, Affiliated Yuhuangding Hospital of Qingdao University, China
| | - Guopeng Yu
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, China
| | - Huan Xu
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, China
| | - Dajun Gao
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, China
| | - Minglun Li
- Urologic and Hematologic Oncology, Department of Radiation Oncology, LMU, University Hospital, Munich, Germany.
| | - Yuzhuo Wang
- Vancouver Prostate Centre, Vancouver, BC, Canada; Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.
| | - Bin Xu
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, China.
| |
Collapse
|
2
|
Kim H, Woo CG, Son SM, Lee YP, Kim HK, Yang Y, Kwon J, Lee KH, Lee HC, Lee OJ, Han HS. Targeted Suppression of CEACAM6 via pHLIP-Delivered RNAs in Pancreatic Ductal Adenocarcinoma. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:598. [PMID: 40282889 PMCID: PMC12028928 DOI: 10.3390/medicina61040598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025]
Abstract
Background and Objectives: Carcinoembryonic antigen-related cell adhesion molecule 6 (CEACAM6) is involved in pancreatic cancer progression and is an attractive therapeutic target for pancreatic cancer. In this study, we evaluated the therapeutic efficacy of small-interfering RNA (siRNA) targeting CEACAM6 (siCEACAM6) and the CEACAM6-suppressive microRNA-29a (miR-29a) in a pancreatic ductal adenocarcinoma xenograft mouse model using pH-low insertion peptide (pHLIP) technology, which targets the acidic tumor microenvironment. Materials and Methods: The delivery vectors for siRNA and miRNA were constructed by conjugating the peptide nucleic acid forms of siCEACAM6 and miR-29a to a peptide with a pHLIP, enabling the transport of siRNA and miRNA across the plasma membrane. The tumor-suppressive effects of pHLIP-siCEACAM6 and pHLIP-miR-29a were assessed in vivo using a BALB/c xenograft mouse model with the injection of the CFPAC-1 human pancreatic ductal adenocarcinoma cell line. Results: The treatment of CFPAC-1 cells with pHLIP-siCEACAM6 and pHLIP-miR-29a under acidic pH conditions suppressed CEACAM6 expression and decreased cell viability. In a xenograft mouse model, the intravenous injection of pHLIP-siCEACAM6 and pHLIP-miR-29a suppressed tumor growth by up to 25.1% (p < 0.01) and 21.2% (p < 0.01), respectively, compared to the control mice treated with pHLIP-scr. Conclusions: Our results demonstrated the efficacy of the pHLIP-mediated delivery of siCEACAM6 and miR-29a as a promising therapeutic strategy in a pancreatic ductal adenocarcinoma xenograft mouse model. The pHLIP technology, which targets the acidic tumor microenvironment, represents an innovative approach to the delivery of small RNAs to pancreatic ductal adenocarcinoma cells, providing new potential strategies for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Hongsik Kim
- Department of Internal Medicine, Chungbuk National University Hospital, College of Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea; (H.K.); (Y.-P.L.); (H.-K.K.); (Y.Y.); (J.K.); (K.-H.L.)
| | - Chang-Gok Woo
- Department of Pathology, Chungbuk National University Hospital, College of Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea; (C.-G.W.); (S.-M.S.); (H.-C.L.); (O.-J.L.)
| | - Seung-Myoung Son
- Department of Pathology, Chungbuk National University Hospital, College of Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea; (C.-G.W.); (S.-M.S.); (H.-C.L.); (O.-J.L.)
| | - Yong-Pyo Lee
- Department of Internal Medicine, Chungbuk National University Hospital, College of Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea; (H.K.); (Y.-P.L.); (H.-K.K.); (Y.Y.); (J.K.); (K.-H.L.)
| | - Hee-Kyung Kim
- Department of Internal Medicine, Chungbuk National University Hospital, College of Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea; (H.K.); (Y.-P.L.); (H.-K.K.); (Y.Y.); (J.K.); (K.-H.L.)
| | - Yaewon Yang
- Department of Internal Medicine, Chungbuk National University Hospital, College of Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea; (H.K.); (Y.-P.L.); (H.-K.K.); (Y.Y.); (J.K.); (K.-H.L.)
| | - Jihyun Kwon
- Department of Internal Medicine, Chungbuk National University Hospital, College of Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea; (H.K.); (Y.-P.L.); (H.-K.K.); (Y.Y.); (J.K.); (K.-H.L.)
| | - Ki-Hyeong Lee
- Department of Internal Medicine, Chungbuk National University Hospital, College of Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea; (H.K.); (Y.-P.L.); (H.-K.K.); (Y.Y.); (J.K.); (K.-H.L.)
| | - Ho-Chang Lee
- Department of Pathology, Chungbuk National University Hospital, College of Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea; (C.-G.W.); (S.-M.S.); (H.-C.L.); (O.-J.L.)
| | - Ok-Jun Lee
- Department of Pathology, Chungbuk National University Hospital, College of Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea; (C.-G.W.); (S.-M.S.); (H.-C.L.); (O.-J.L.)
| | - Hye-Sook Han
- Department of Internal Medicine, Chungbuk National University Hospital, College of Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea; (H.K.); (Y.-P.L.); (H.-K.K.); (Y.Y.); (J.K.); (K.-H.L.)
| |
Collapse
|
3
|
Shin S, Baek DS, Mellors JW, Dimitrov DS, Li W. Development of Fully Human Antibodies Targeting SIRPα and PLA2G7 for Cancer Therapy. Antibodies (Basel) 2025; 14:21. [PMID: 40136470 PMCID: PMC11939323 DOI: 10.3390/antib14010021] [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: 10/30/2024] [Revised: 01/12/2025] [Accepted: 02/25/2025] [Indexed: 03/27/2025] Open
Abstract
Background: Macrophages play an important role in eliminating diseased and damaged cells through programmed cell death. Signal regulatory protein alpha (SIRPα) is a crucial immune checkpoint primarily expressed on myeloid cells and macrophages. It initiates a 'do not eat me' signal when engaged with CD47, which is typically expressed at elevated levels on multiple solid tumors. The phospholipase A2 Group 7 (PLA2G7), which is mainly secreted by macrophages, interacts with oxidized low-density lipoprotein (oxLDL) and associates with several vascular diseases and cancers. Methods: To identify potent fully human monoclonal antibodies (mAbs) against human SIRPα and PLA2G7, we conducted bio-panning of phage antibody libraries. Results: We isolated one human Fab (1B3) and VH (1A3) for SIRPα, as well as one human Fab (1H8) and one VH (1A9) for PLA2G7; the 1B3 Fab and 1A3 VH are competitively bound to SIRPα, interfering with CD47 binding. The 1B3 IgG and 1A3 VH-Fc augmented macrophage-mediated phagocytic activity when combined with the anti-EGFR antibody, cetuximab. The anti-PLA2G7 antibodies exhibited high specificity for the PLA2G7 antigen and effectively blocked the PLA2G7 enzymatic activity with half-maximal inhibitory concentrations (IC50) in the single-digit nanomolar range. Additionally, 1H8 IgG and its derivative bispecific antibody exhibited the ability to block PLA2G7-mediated tumor cell migration. Conclusions: Our anti-SIRPα mAbs are expected to serve as potent and fully human immune checkpoint inhibitors of SIRPα, enhancing the antitumor responses of SIRPα-positive immune cells. Moreover, our anti-PLA2G7 mAbs represent promising fully human PLA2G7 enzymatic blockade antibodies with the potential to enhance both anti-tumor and anti-aging responses. Anti-SIRPα and PLA2G7 mAbs can modulate macrophage phagocytic activity and inflammatory responses against tumors.
Collapse
Affiliation(s)
- Seungmin Shin
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; (D.-S.B.); (J.W.M.); (D.S.D.)
| | - Du-San Baek
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; (D.-S.B.); (J.W.M.); (D.S.D.)
- GLPG US, 1401 Forbes Avenue, Pittsburgh, PA 15219, USA
| | - John W. Mellors
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; (D.-S.B.); (J.W.M.); (D.S.D.)
- GLPG US, 1401 Forbes Avenue, Pittsburgh, PA 15219, USA
| | - Dimiter S. Dimitrov
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; (D.-S.B.); (J.W.M.); (D.S.D.)
- GLPG US, 1401 Forbes Avenue, Pittsburgh, PA 15219, USA
| | - Wei Li
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; (D.-S.B.); (J.W.M.); (D.S.D.)
| |
Collapse
|
4
|
Wang F, Zhang R, Zhou Z, Shi R, Peng F, Xu Y, Yang S, Wang Z, Zhang P, Tu R, Zhang C, Liu X, Cai J. CAR-T therapy for endocrine neoplasms: novel targets and combination of therapies. Front Endocrinol (Lausanne) 2025; 16:1517525. [PMID: 40007813 PMCID: PMC11850254 DOI: 10.3389/fendo.2025.1517525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
Endocrine malignancies constitute a heterogeneous tumour group with diverse biological characteristics. While typically indolent, they encompass aggressive types and presence of any metastatic sign indicates a high probability of recurrence and a diminished response to conventional therapies. Chimeric antigen receptor (CAR)-T cell immunotherapy has constituted a revolutionary advance in cancer treatment and exhibited significant potential for application in endocrine cancer. However, limited effectiveness was displayed in clinical application, which necessitates the exploration of novel modalities. Identification of specific and safe targets for endocrine cancer is the initial stage towards establishing a successful CAR-T treatment. Various therapies under investigation offer potential enhancements to CAR T cell efficacy through diverse mechanisms. Herein, we summarize recent advances in identifying targets of endocrine cancer for CAR therapy and provide an overview of combinatorial approaches.
Collapse
Affiliation(s)
- Fang Wang
- Department of Otolaryngology-Head and Neck Surgery, Xinyang Central Hospital, Xinyang, Henan, China
| | - Ruiqi Zhang
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Henan Joint International Pediatric Urodynamic Laboratory, Zhengzhou, China
| | - Zhaokai Zhou
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Henan Joint International Pediatric Urodynamic Laboratory, Zhengzhou, China
| | - Run Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fu Peng
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yudi Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shuai Yang
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Henan Joint International Pediatric Urodynamic Laboratory, Zhengzhou, China
| | - Zhan Wang
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Henan Joint International Pediatric Urodynamic Laboratory, Zhengzhou, China
| | - Pengpeng Zhang
- Department of Lung Cancer, Tianjin Lung Cancer Center, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Rui Tu
- Department of Ultrasound, Xinyang Central Hospital, Xinyang, Henan, China
| | - Chun Zhang
- Department of Gastroenterology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xingchen Liu
- Department of Gynaecology, Xinyang Central Hospital, Xinyang, Henan, China
| | - Jun Cai
- Department of Gynaecology, Xinyang Central Hospital, Xinyang, Henan, China
| |
Collapse
|
5
|
Vicart A, Holland C, Fraser K, Gervais F, Aspinall-O'Dea M, Brown N, Siddals K, Greiner G, Carreira V, Galbreath E, Willer M, Kaliyaperumal S, Wood C, MacLachlan T, Clark E. Applications of Cell-Based Protein Array Technology to Preclinical Safety Assessment of Biological Products. Toxicol Pathol 2025; 53:31-44. [PMID: 39871795 DOI: 10.1177/01926233241311259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
Off-target evaluation is essential in preclinical safety assessments of novel biotherapeutics, supporting lead molecule selection, endpoint selection in toxicology studies, and regulatory requirements for first-in-human trials. Off-target interaction of a therapeutic antibody and antibody derivatives has been historically assessed via the Tissue Cross-Reactivity (TCR) study, in which the candidate molecule is used as a reagent in immunohistochemistry (IHC) to assess binding of the candidate molecule to a panel of human tissue sections. The TCR approach is limited by the performance of the therapeutic as an IHC reagent, which is often suboptimal to outright infeasible. Furthermore, binding of the therapeutic in IHC conditions typically has poor in vitro to in vivo translation and lacks qualitative data of the identity of putative off-targets limiting the decisional value of the data. More recently, cell-based protein arrays (CBPA) that allow for screening against a large portion of the human membrane proteome and secretome have emerged as a complement, and likely a higher value alternative, to IHC-based off-target assessment. These arrays identify specific protein interactions and may be useful for testing nontraditional antibody-based therapeutic formats that are unsuitable for TCR studies. This article presents an overview of CBPA technologies in the context of TCR and off-target assessment studies. Selected case examples and strategic considerations covering a range of different modalities are presented.
Collapse
Affiliation(s)
- Axel Vicart
- Novartis Institute for Biomedical Research, Basel, Switzerland
| | - Cam Holland
- Johnson & Johnson, Spring House, Pennsylvania, USA
| | | | | | | | - Nick Brown
- Charles River Laboratories, High Peak, UK
| | | | | | | | | | | | | | - Charles Wood
- Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut, USA
| | - Tim MacLachlan
- Novartis Institute for Biomedical Research, Cambridge, Massachusetts, USA
| | - Elizabeth Clark
- Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut, USA
| |
Collapse
|
6
|
Fayyaz A, Haqqi A, Khan R, Irfan M, Khan K, Reiner Ž, Sharifi-Rad J, Calina D. Revolutionizing cancer treatment: the rise of personalized immunotherapies. Discov Oncol 2024; 15:756. [PMID: 39692978 DOI: 10.1007/s12672-024-01638-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024] Open
Abstract
Interest in biological therapy for cancer has surged due to its precise targeting of cancer cells and minimized impact on surrounding healthy tissues. This review discusses various biological cancer therapies, highlighting advanced alternatives over conventional chemotherapy alone. It explores DNA and RNA-based vaccines, T-cell modifications, adoptive cell transfer, CAR T cell therapy, angiogenesis inhibitors, and the combination of immunotherapy with chemotherapy, offering a holistic view of the potential in cancer treatment. Additionally, it discusses the role of nanotechnology in increasing the efficacy of cancer-targeting drugs, as well as cytokine and immunoconjugate therapies for bolstering immune system effectiveness against neoplastic cells. The potential of gene potential for precise targeting of cancer-linked genes and the application of oncolytic viruses against virus-associated cancers are also discussed. The review identifies significant advancements in the targeted treatment of cancer by biological methods. It acknowledges the challenges, including drug resistance and the need for high specificity in certain therapies, while also highlighting the effectiveness of cancer vaccines, modified T-cells, and oncolytic viruses. Biological therapies are a promising frontier in cancer treatment, offering the potential for more personalized and effective therapeutic strategies. Despite existing challenges, ongoing research and clinical trials are fundamental for overcoming current limitations and enhancing the efficacy of biological therapies in cancer care.
Collapse
Affiliation(s)
- Amna Fayyaz
- Atta-Ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Aleena Haqqi
- School of Medical Laboratory Technology, Faculty of Allied Health Sciences, Minhaj University Lahore (MUL), Lahore, 54000, Pakistan
| | - Rashid Khan
- Department of Pharmacy, Punjab University College of Pharmacy University of Punjab Lahore, Lahore, 54000, Pakistan
| | - Muhammad Irfan
- Atta-Ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Khushbukhat Khan
- Cancer Clinical Research Unit, Trials360, Lahore, 54000, Pakistan.
| | - Željko Reiner
- Department for Metabolic Diseases, University Hospital Center Zagreb, Zagreb, Croatia
- Polish Mother's Memorial Hospital Research Institute, Lodz, Poland
| | - Javad Sharifi-Rad
- Universidad Espíritu Santo, Samborondón, 092301, Ecuador.
- Centro de Estudios Tecnológicos, Universitarios del Golfo, Veracruz, Mexico.
- Department of Medicine, College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| |
Collapse
|
7
|
Kumar A, Duffieux F, Gagnaire M, Rapisarda C, Bertrand T, Rak A. Structural insights into epitope-paratope interactions of a monoclonal antibody targeting CEACAM5-expressing tumors. Nat Commun 2024; 15:9377. [PMID: 39477960 PMCID: PMC11525548 DOI: 10.1038/s41467-024-53746-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/18/2024] [Indexed: 11/02/2024] Open
Abstract
Carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) are overexpressed in some tumor types. The antibody-drug conjugate tusamitamab ravtansine specifically recognizes the A3-B3 domains of human CEACAM5 (hCEACAM5). To understand this specificity, here we map the epitope-paratope interface between the A3-B3 domains of hCEACAM5 (hCEACAM5A3-B3) and the antigen-binding fragment of tusamitamab (tusa Fab). We use hydrogen/deuterium exchange mass spectrometry to identify the tusa Fab paratope, which involves heavy chain (HC) residues 101-109 and light chain residues 48-54 and 88-104. Using surface plasmon resonance, we demonstrate that alanine variants of HC residues 96-108 abolish binding to hCEACAM5, suggesting that these residues are critical for tusa-Fab-antigen complex formation. The cryogenic electron microscopy structure of the hCEACAM5A3-B3- tusa Fab complex (3.11 Å overall resolution) reveals a discontinuous epitope involving residues in the A3-B3 domains and an N-linked mannose at residue Asn612. Conformational constraints on the epitope-paratope interface enable tusamitamab to target hCEACAM5A3-B3 and distinguish CEACAM5 from other CEACAMs.
Collapse
Affiliation(s)
- Anand Kumar
- Integrated Drug Discovery, Sanofi R&D, Paris, France
| | | | | | | | | | - Alexey Rak
- Integrated Drug Discovery, Sanofi R&D, Paris, France.
| |
Collapse
|
8
|
Porter LH, Harrison SG, Risbridger GP, Lister N, Taylor RA. Left out in the cold: Moving beyond hormonal therapy for the treatment of immunologically cold prostate cancer with CAR T cell immunotherapies. J Steroid Biochem Mol Biol 2024; 243:106571. [PMID: 38909866 DOI: 10.1016/j.jsbmb.2024.106571] [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: 05/13/2024] [Revised: 06/11/2024] [Accepted: 06/17/2024] [Indexed: 06/25/2024]
Abstract
Prostate cancer is primarily hormone-dependent, and medical treatments have focused on inhibiting androgen biosynthesis or signaling through various approaches. Despite significant advances with the introduction of androgen receptor signalling inhibitors (ARSIs), patients continue to progress to castration-resistant prostate cancer (CRPC), highlighting the need for targeted therapies that extend beyond hormonal blockade. Chimeric Antigen Receptor (CAR) T cells and other engineered immune cells represent a new generation of adoptive cellular therapies. While these therapies have significantly enhanced outcomes for patients with hematological malignancies, ongoing research is exploring the broader use of CAR T therapy in solid tumors, including advanced prostate cancer. In general, CAR T cell therapies are less effective against solid cancers with the immunosuppressive tumor microenvironment hindering T cell infiltration, activation and cytotoxicity following antigen recognition. In addition, inherent tumor heterogeneity exists in patients with advanced prostate cancer that may prevent durable therapeutic responses using single-target agents. These barriers must be overcome to inform clinical trial design and improve treatment efficacy. In this review, we discuss the innovative and rationally designed strategies under investigation to improve the clinical translation of cellular immunotherapy in prostate cancer and maximise therapeutic outcomes for these patients.
Collapse
Affiliation(s)
- L H Porter
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
| | - S G Harrison
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
| | - G P Risbridger
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Cancer Immunology Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Cabrini Institute, Cabrini Health, Malvern, VIC 3144, Australia
| | - Natalie Lister
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
| | - R A Taylor
- Cancer Immunology Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Cabrini Institute, Cabrini Health, Malvern, VIC 3144, Australia; Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Physiology, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
9
|
Hernandez-Gamarra M, Salgado-Roo A, Dominguez E, Goiricelaya Seco EM, Veiga-Rúa S, Pedrera-Garbayo LF, Carracedo Á, Allegue C. CARTAR: a comprehensive web tool for identifying potential targets in chimeric antigen receptor therapies using TCGA and GTEx data. Brief Bioinform 2024; 25:bbae326. [PMID: 38975894 PMCID: PMC11229032 DOI: 10.1093/bib/bbae326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/24/2024] [Accepted: 06/26/2024] [Indexed: 07/09/2024] Open
Abstract
Chimeric antigen receptor (CAR) therapy has emerged as a ground-breaking advancement in cancer treatment, harnessing the power of engineered human immune cells to target and eliminate cancer cells. The escalating interest and investment in CAR therapy in recent years emphasize its profound significance in clinical research, positioning it as a rapidly expanding frontier in the field of personalized cancer therapies. A crucial step in CAR therapy design is choosing the right target as it determines the therapy's effectiveness, safety and specificity against cancer cells, while sparing healthy tissues. Herein, we propose a suite of tools for the identification and analysis of potential CAR targets leveraging expression data from The Cancer Genome Atlas and Genotype-Tissue Expression Project, which are implemented in CARTAR website. These tools focus on pinpointing tumor-associated antigens, ensuring target selectivity and assessing specificity to avoid off-tumor toxicities and can be used to rationally designing dual CARs. In addition, candidate target expression can be explored in cancer cell lines using the expression data for the Cancer Cell Line Encyclopedia. To our best knowledge, CARTAR is the first website dedicated to the systematic search of suitable candidate targets for CAR therapy. CARTAR is publicly accessible at https://gmxenomica.github.io/CARTAR/.
Collapse
Affiliation(s)
- Miguel Hernandez-Gamarra
- Genomic Medicine Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Av. Barcelona, 15706 A Coruña, Spain
- C005, Instituto de Investigación Sanitaria de Santiago (IDIS), Travesía da Choupana, 15706 A Coruña, Spain
| | - Alba Salgado-Roo
- Genomic Medicine Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Av. Barcelona, 15706 A Coruña, Spain
- C005, Instituto de Investigación Sanitaria de Santiago (IDIS), Travesía da Choupana, 15706 A Coruña, Spain
| | - Eduardo Dominguez
- C005, Instituto de Investigación Sanitaria de Santiago (IDIS), Travesía da Choupana, 15706 A Coruña, Spain
| | | | - Sara Veiga-Rúa
- Genomic Medicine Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Av. Barcelona, 15706 A Coruña, Spain
- C005, Instituto de Investigación Sanitaria de Santiago (IDIS), Travesía da Choupana, 15706 A Coruña, Spain
| | - Lucía F Pedrera-Garbayo
- Genomic Medicine Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Av. Barcelona, 15706 A Coruña, Spain
- C005, Instituto de Investigación Sanitaria de Santiago (IDIS), Travesía da Choupana, 15706 A Coruña, Spain
| | - Ángel Carracedo
- Genomic Medicine Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Av. Barcelona, 15706 A Coruña, Spain
- C005, Instituto de Investigación Sanitaria de Santiago (IDIS), Travesía da Choupana, 15706 A Coruña, Spain
- Fundación Pública Galega de Medicina Xenómica (FPGMX), Travesía da Choupana, 15706 A Coruña, Spain
- CB06/07/0088, Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Av. Monforte de Lemos, 28029 Madrid, Spain
| | - Catarina Allegue
- Genomic Medicine Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Av. Barcelona, 15706 A Coruña, Spain
- C005, Instituto de Investigación Sanitaria de Santiago (IDIS), Travesía da Choupana, 15706 A Coruña, Spain
- CB06/07/0088, Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Av. Monforte de Lemos, 28029 Madrid, Spain
| |
Collapse
|
10
|
Zhu XY, Li QX, Kong Y, Huang KK, Wang G, Wang YJ, Lu J, Hua GQ, Wu YL, Ying TL. A novel human single-domain antibody-drug conjugate targeting CEACAM5 exhibits potent in vitro and in vivo antitumor activity. Acta Pharmacol Sin 2024; 45:609-618. [PMID: 38030799 PMCID: PMC10834580 DOI: 10.1038/s41401-023-01200-9] [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: 05/09/2023] [Accepted: 11/13/2023] [Indexed: 12/01/2023]
Abstract
Leveraging the specificity of antibody to deliver cytotoxic agent into tumor, antibody-drug conjugates (ADCs) have become one of the hotspots in the development of anticancer therapies. Although significant progress has been achieved, there remain challenges to overcome, including limited penetration into solid tumors and potential immunogenicity. Fully human single-domain antibodies (UdAbs), with their small size and human nature, represent a promising approach for addressing these challenges. Carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5) is a glycosylated cell surface protein that rarely expressed in normal adult tissues but overexpressed in diverse cancers, taking part in tumorigenesis, progression, and metastasis. In this study, we investigated the therapeutic potential of UdADC targeting CEACAM5. We performed biopanning in our library and obtained an antibody candidate B9, which bound potently and specifically to CEACAM5 protein (KD = 4.84 nM) and possessed excellent biophysical properties (low aggregation tendency, high homogeneity, and thermal stability). The conjugation of B9 with a potent cytotoxic agent, monomethyl auristatin E (MMAE), exhibited superior antitumor efficacy against CEACAM5-expressing human gastric cancer cell line MKN-45, human pancreatic carcinoma cell line BxPC-3 and human colorectal cancer cell line LS174T with IC50 values of 38.14, 25.60, and 101.4 nM, respectively. In BxPC-3 and MKN-45 xenograft mice, administration of UdADC B9-MMAE (5 mg/kg, i.v.) every 2 days for 4 times markedly inhibited the tumor growth without significant change in body weight. This study may have significant implications for the design of next-generation ADCs.
Collapse
Affiliation(s)
- Xiao-Yi Zhu
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, 200032, China
| | - Quan-Xiao Li
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, 200032, China
| | - Yu Kong
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, 200032, China
| | - Ke-Ke Huang
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, 200032, China
| | - Gang Wang
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yun-Ji Wang
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jun Lu
- Auckland Bioengineering Institute, University of Auckland, Auckland, 1010, New Zealand
| | - Guo-Qiang Hua
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Yan-Ling Wu
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, 200032, China.
| | - Tian-Lei Ying
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, 200032, China.
| |
Collapse
|
11
|
Saini S, Sreekumar A, Nathani S, Asante DM, Simmons MN. A novel exosome based therapeutic intervention against neuroendocrine prostate cancer. Sci Rep 2024; 14:2816. [PMID: 38307935 PMCID: PMC10837194 DOI: 10.1038/s41598-024-53269-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 01/30/2024] [Indexed: 02/04/2024] Open
Abstract
Neuroendocrine prostate cancer (NEPC) is a highly lethal variant of castration-resistant prostate cancer (CRPC) with poor survival rates. Current treatment options for NEPC are limited to highly toxic platinum drugs highlighting the urgent need for new therapies. This study aimed to develop a novel therapeutic approach using engineered exosomes against NEPC. Exosomes were modified to target CEACAM5, an NEPC surface antigen, by attaching CEACAM5 antibodies to HEK293T exosomes. These exosomes were loaded with drugs inhibiting EZH2 and the androgen receptor (AR) as recent research shows a persistent role of AR in NEPC wherein it plays a concerted role with EZH2 in driving neuronal gene programs. In vitro experiments with NEPC cell lines demonstrated that CEACAM5-targeted exosomes were specifically taken up by NEPC cells, leading to reduced cellular viability and decreased expression of neuronal markers. Further in vivo tests using a NEPC patient-derived xenograft model (LuCaP145.1) showed significant tumor regression in mice treated with engineered exosomes compared to control mice receiving IgG-labeled exosomes. These results suggest that CEACAM5-engineered exosomes hold promise as a targeted therapy for NEPC. Importantly, our exosome engineering strategy is versatile and can be adapted to target various surface antigens in prostate cancer and other diseases.
Collapse
Affiliation(s)
- Sharanjot Saini
- Department of Biochemistry and Molecular Biology, Augusta University, 1410 Laney Walker Boulevard, Augusta, GA, 30912, USA.
- Department of Urology, Augusta University, Augusta, GA, USA.
| | - Amritha Sreekumar
- Department of Biochemistry and Molecular Biology, Augusta University, 1410 Laney Walker Boulevard, Augusta, GA, 30912, USA
| | - Sandip Nathani
- Department of Biochemistry and Molecular Biology, Augusta University, 1410 Laney Walker Boulevard, Augusta, GA, 30912, USA
| | - Diana M Asante
- Department of Biochemistry and Molecular Biology, Augusta University, 1410 Laney Walker Boulevard, Augusta, GA, 30912, USA
| | | |
Collapse
|
12
|
Jancewicz I, Śmiech M, Winiarska M, Zagozdzon R, Wisniewski P. New CEACAM-targeting 2A3 single-domain antibody-based chimeric antigen receptor T-cells produce anticancer effects in vitro and in vivo. Cancer Immunol Immunother 2024; 73:30. [PMID: 38279989 PMCID: PMC10821984 DOI: 10.1007/s00262-023-03602-4] [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: 08/29/2023] [Accepted: 11/20/2023] [Indexed: 01/29/2024]
Abstract
Recently, a breakthrough immunotherapeutic strategy of chimeric antigen receptor (CAR) T-cells has been introduced to hematooncology. However, to apply this novel treatment in solid cancers, one must identify suitable molecular targets in the tumors of choice. CEACAM family proteins are involved in the progression of a range of malignancies, including pancreatic and breast cancers, and pose attractive targets for anticancer therapies. In this work, we used a new CEACAM-targeted 2A3 single-domain antibody-based chimeric antigen receptor T-cells to evaluate their antitumor properties in vitro and in animal models. Originally, 2A3 antibody was reported to target CEACAM6 molecule; however, our in vitro co-incubation experiments showed activation and high cytotoxicity of 2A3-CAR T-cells against CEACAM5 and/or CEACAM6 high human cell lines, suggesting cross-reactivity of this antibody. Moreover, 2A3-CAR T-cells tested in vivo in the BxPC-3 xenograft model demonstrated high efficacy against pancreatic cancer xenografts in both early and late intervention treatment regimens. Our results for the first time show an enhanced targeting toward CEACAM5 and CEACAM6 molecules by the new 2A3 sdAb-based CAR T-cells. The results strongly support the further development of 2A3-CAR T-cells as a potential treatment strategy against CEACAM5/6-overexpressing cancers.
Collapse
Affiliation(s)
- Iga Jancewicz
- 4Cell Therapies S.A., 59C Bojkowska Street, 44-100, Gliwice, Poland
| | - Magdalena Śmiech
- 4Cell Therapies S.A., 59C Bojkowska Street, 44-100, Gliwice, Poland
| | - Magdalena Winiarska
- 4Cell Therapies S.A., 59C Bojkowska Street, 44-100, Gliwice, Poland
- Department of Immunology, Medical University of Warsaw, 5 Nielubowicza St., Building F, 02-097, Warsaw, Poland
| | - Radoslaw Zagozdzon
- 4Cell Therapies S.A., 59C Bojkowska Street, 44-100, Gliwice, Poland.
- Laboratory of Cellular and Genetic Therapies, Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland.
- Department of Regenerative Medicine, The Maria Sklodowska-Curie National Research Institute of Oncology, 5 Roentgena Street, 02-781, Warsaw, Poland.
| | - Pawel Wisniewski
- 4Cell Therapies S.A., 59C Bojkowska Street, 44-100, Gliwice, Poland.
| |
Collapse
|
13
|
Chu X, Shin S, Baek DS, Zhang L, Conard A, Shi M, Kim YJ, Adams C, Hines M, Liu X, Chen C, Sun Z, Jelev DV, Mellors JW, Dimitrov DS, Li W. Discovery of a novel highly specific, fully human PSCA antibody and its application as an antibody-drug conjugate in prostate cancer. MAbs 2024; 16:2387240. [PMID: 39113562 PMCID: PMC11312989 DOI: 10.1080/19420862.2024.2387240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 07/25/2024] [Accepted: 07/29/2024] [Indexed: 08/11/2024] Open
Abstract
Prostate stem cell antigen (PSCA) is expressed in all stages of prostate cancer, including in advanced androgen-independent tumors and bone metastasis. PSCA may associate with prostate carcinogenesis and lineage plasticity in prostate cancer. PSCA is also a promising theranostic marker for a variety of other solid tumors, including pancreatic adenocarcinoma and renal cell carcinoma. Here, we identified a novel fully human PSCA antibody using phage display methodology. The structure-based affinity maturation yielded a high-affinity binder, F12, which is highly specific and does not bind to 6,000 human membrane proteins based on a membrane proteome array assay. F12 targets PSCA amino acids 63-69 as tested by the peptide scanning microarray, and it cross-reacts with the murine PSCA. IgG1 F12 efficiently internalizes into PSCA-expressing tumor cells. The antimitotic reagent monomethyl auristatin E (MMAE)-conjugated IgG1 F12 (ADC, F12-MMAE) exhibits dose-dependent efficacy and specificity in a human prostate cancer PC-3-PSCA xenograft NSG mouse model. This is a first reported ADC based on a fully human PSCA antibody and MMAE that is characterized in a xenograft murine model, which warrants further optimizations and investigations in additional preclinical tumor models, including prostate and other solid tumors.
Collapse
Affiliation(s)
- Xiaojie Chu
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh Medical School, Pittsburgh, PA, USA
| | - Seungmin Shin
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh Medical School, Pittsburgh, PA, USA
| | | | - Liyong Zhang
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Megan Shi
- Computational and System Biology, School of Medicine, University of Pittsburgh, Pittsburgh, USA
| | | | | | - Maggie Hines
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh Medical School, Pittsburgh, PA, USA
| | - Xianglei Liu
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh Medical School, Pittsburgh, PA, USA
| | - Chuan Chen
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh Medical School, Pittsburgh, PA, USA
| | | | - Dontcho V. Jelev
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh Medical School, Pittsburgh, PA, USA
| | - John W. Mellors
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh Medical School, Pittsburgh, PA, USA
- GLPG, Pittsburgh, PA, USA
| | - Dimiter S. Dimitrov
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh Medical School, Pittsburgh, PA, USA
- GLPG, Pittsburgh, PA, USA
| | - Wei Li
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh Medical School, Pittsburgh, PA, USA
| |
Collapse
|
14
|
Wang R, Li Q, Chu X, Li N, Liang H, He F. Sequencing and Bioinformatics analysis of lncRNA/circRNA-miRNA-mRNA in Glioblastoma multiforme. Metab Brain Dis 2023; 38:2289-2300. [PMID: 37389689 DOI: 10.1007/s11011-023-01256-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/20/2023] [Indexed: 07/01/2023]
Abstract
Evidence suggests that non-coding RNAs have a role in glioblastoma multiforme (GBM), although the regulatory mechanisms controlled by competing endogenous RNAs (ceRNAs) in GBM are still poorly understood and infrequently described. This research extensively analyzed circRNA, lncRNA, miRNA, and mRNA expression changes in GBM patients. RNA-sequencing analyses were conducted to investigate differentially expressed genes (DEGs), lncRNAs (DELs), miRNAs (DEMs), and circRNAs (DECs) in the GBM. In this study, researchers found that GBM patients and healthy controls differed in the presence of 1224 DECs, 1406 DELs, 229 DEMs, and 2740 DEGs. PPI network analysis demonstrated that CEACAM5, CXCL17, FAM83A, TMPRSS4, and GGPRC5A were hub genes and enriched in modules. Then a ceRNA network was constructed with 8 circRNA, 7 lncRNAs, 16 miRNAs, and 17 mRNAs. Overall, the ceRNA interaction axes that were found may prove to be pivotal therapeutic targets for treating GBM.
Collapse
Affiliation(s)
- Renjie Wang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
- Department of Neurosurgery, Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin, 300162, China
| | - Qi Li
- Tianjin Hospital, Tianjin University, Tianjin, 300050, China
| | - Xiaolei Chu
- Tianjin Hospital, Tianjin University, Tianjin, 300050, China
| | - Nan Li
- Department of Neurosurgery, Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin, 300162, China
| | - Haiqian Liang
- Department of Neurosurgery, Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin, 300162, China.
| | - Feng He
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China.
| |
Collapse
|
15
|
Saha N, Baek DS, Mendoza RP, Robev D, Xu Y, Goldgur Y, De La Cruz MJ, de Stanchina E, Janes PW, Xu K, Dimitrov DS, Nikolov DB. Fully human monoclonal antibody targeting activated ADAM10 on colorectal cancer cells. Biomed Pharmacother 2023; 161:114494. [PMID: 36917886 PMCID: PMC10499537 DOI: 10.1016/j.biopha.2023.114494] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/27/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023] Open
Abstract
Metastasis and chemoresistance in colorectal cancer are mediated by certain poorly differentiated cancer cells, known as cancer stem cells, that are maintained by Notch downstream signaling initiated upon Notch cleavage by the metalloprotease ADAM10. It has been shown that ADAM10 overexpression correlates with aberrant signaling from Notch, erbBs, and other receptors, as well as a more aggressive metastatic phenotype, in a range of cancers including colon, gastric, prostate, breast, ovarian, uterine, and leukemia. ADAM10 inhibition, therefore, stands out as an important and new approach to deter the progression of advanced CRC. For targeting the ADAM10 substrate-binding region, which is located outside of the catalytic domain of the protease, we generated a human anti-ADAM10 monoclonal antibody named 1H5. Structural and functional characterization of 1H5 reveals that it binds to the substrate-binding cysteine-rich domain and recognizes an activated ADAM10 conformation present on tumor cells. The mAb inhibits Notch cleavage and proliferation of colon cancer cell lines in vitro and in mouse models. Consistent with its binding to activated ADAM10, the mAb augments the catalytic activity of ADAM10 towards small peptide substrates in vitro. Most importantly, in a mouse model of colon cancer, when administered in combination with the therapeutic agent Irinotecan, 1H5 causes highly effective tumor growth inhibition without any discernible toxicity effects. Our singular approach to target the ADAM10 substrate-binding region with therapeutic antibodies could overcome the shortcomings of previous intervention strategies of targeting the protease active site with small molecule inhibitors that exhibit musculoskeletal toxicity.
Collapse
Affiliation(s)
- Nayanendu Saha
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States.
| | - Du-San Baek
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, United States
| | - Rachelle P Mendoza
- Department of Pathology, University of Chicago, Chicago, IL 60637, United States
| | - Dorothea Robev
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Yan Xu
- Department of Veterinary Biosciences, Ohio State University, Columbus, OH 43210, United States
| | - Yehuda Goldgur
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - M Jason De La Cruz
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Elisa de Stanchina
- Antitumor Assessment Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Peter W Janes
- Tumour Targeting Program, Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Victoria 3084, Australia
| | - Kai Xu
- Department of Veterinary Biosciences, Ohio State University, Columbus, OH 43210, United States; Department of Microbial Infection and Immunity, Ohio State University, Columbus, OH 43210, United States
| | - Dimiter S Dimitrov
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, United States
| | - Dimitar B Nikolov
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States.
| |
Collapse
|
16
|
Chu X, Baek DS, Li W, Shyp T, Mooney B, Hines MG, Morin GB, Sorensen PH, Dimitrov DS. Human antibodies targeting ENPP1 as candidate therapeutics for cancers. Front Immunol 2023; 14:1070492. [PMID: 36761762 PMCID: PMC9905232 DOI: 10.3389/fimmu.2023.1070492] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 01/09/2023] [Indexed: 01/27/2023] Open
Abstract
Ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1) is a type II transmembrane glycoprotein expressed in many tissues. High expression levels of ENPP1 have been observed in many cancer types such as lung cancer, ovarian cancer, and breast cancer. Such overexpression has been associated with poor prognosis in these diseases. Hence, ENPP1 is a potential target for immunotherapy across multiple cancers. Here, we isolated and characterized two high-affinity and specific anti-ENPP1 Fab antibody candidates, 17 and 3G12, from large phage-displayed human Fab libraries. After conversion to IgG1, the binding of both antibodies increased significantly due to avidity effects. Based on these antibodies, we generated antibody-drug conjugates (ADCs), IgG-based bispecific T-cell engagers (IbTEs), and CAR T-cells which all exhibited potent killing of ENPP1-expressing cells. Thus, these various antibody-derived modalities are promising therapeutic candidates for cancers expressing human ENPP1.
Collapse
Affiliation(s)
- Xiaojie Chu
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh Medical School, Pittsburgh, PA, United States
| | - Du-San Baek
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh Medical School, Pittsburgh, PA, United States
| | - Wei Li
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh Medical School, Pittsburgh, PA, United States
| | - Taras Shyp
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Brian Mooney
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Margaret G Hines
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh Medical School, Pittsburgh, PA, United States
| | - Gregg B Morin
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, University of British Columbia, Vancouver, BC, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Poul H Sorensen
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Dimiter S Dimitrov
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh Medical School, Pittsburgh, PA, United States.,Abound Bio, Pittsburgh, PA, United States
| |
Collapse
|
17
|
Kim YJ, Li W, Zhelev DV, Mellors JW, Dimitrov DS, Baek DS. Chimeric antigen receptor-T cells are effective against CEACAM5 expressing non-small cell lung cancer cells resistant to antibody-drug conjugates. Front Oncol 2023; 13:1124039. [PMID: 36923424 PMCID: PMC10010383 DOI: 10.3389/fonc.2023.1124039] [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: 12/14/2022] [Accepted: 02/08/2023] [Indexed: 03/02/2023] Open
Abstract
Chimeric antigen receptor-T (CAR-T) cells and antibody-drug conjugates (ADCs) are promising therapeutic strategies in oncology. The carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5) is overexpressed in tumors including non-small cell lung cancer (NSCLC) and pancreatic ductal adenocarcinoma (PDAC), and is an attractive target for therapies based on CAR-T cell or/and ADCs. We previously developed a highly specific antibody-based CAR-T cells targeting CEACAM5 and the tumoricidal effect of CAR-T cells was proved against neuro-endocrine prostate cancer (NEPC) cells expressing CEACAM5. Here, we compare the anti-tumor efficacy of our CAR-T cells with that of an anti-CEACAM5 ADC being clinically evaluated against NSCLC. Our anti-CEACAM5 CAR-T cells showed cytotoxicity in a CEACAM5 surface concentration dependent manner and reduced tumor growth in both ADC-responsive and -non-responsive CEACAM5-expressing NSCLC cells in vitro and in vivo. In contrast, the ADC exhibited cytotoxicity independent on the CEACAM5 cell surface concentration. Even though clinical translation of CEACAM5 targeting CAR-T cell therapies is still in preclinical stage, our CAR-T cell approach could provide a potential therapeutic strategy for CEACAM5-positive cancer patients with resistance to ADCs.
Collapse
Affiliation(s)
- Ye-Jin Kim
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Wei Li
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Doncho V Zhelev
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - John W Mellors
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Abound Bio, Pittsburgh, PA, United States
| | - Dimiter S Dimitrov
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Abound Bio, Pittsburgh, PA, United States
| | - Du-San Baek
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
18
|
Wang X, Li Y, Pan M, Lu T, Wang M, Wang Z, Liu C, Hu G. CEACAM5 inhibits the lymphatic metastasis of head and neck squamous cell carcinoma by regulating epithelial-mesenchymal transition via inhibiting MDM2. Clin Sci (Lond) 2022; 136:1691-1710. [PMID: 36377775 PMCID: PMC9702577 DOI: 10.1042/cs20220581] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/14/2022] [Accepted: 11/14/2022] [Indexed: 10/30/2024]
Abstract
Lymph node (LN) metastasis affects both the management and prognosis of head and neck squamous cell carcinoma (HNSCC). Here, we explored the relationship between lymphatic metastasis and CEA family member 5 (CEACAM5), including its possible regulatory role in HNSCC. The levels of CEACAM5 in tissues from patients with HNSCC, with and without LN metastases, were assessed by transcriptome sequencing. The associations between CEACAM5 and the N stage of LN metastasis in HNSCC were predicted through The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases and a pan-cancer analysis of CEACAM5 expression in 33 common human tumors was conducted. CEACAM5 levels were analyzed in tumor and normal tissue specimens from HNSCC patients and the correlation between CEACAM5 levels and prognosis was evaluated. The influence of CEACAM5 on cell proliferation, invasion, migration, and apoptosis was investigated in HNSCC cell lines, as were the downstream regulatory mechanisms. A mouse model of LN metastasis was constructed. CEACAM5 levels were significantly higher in HNSCC tissue without LN metastasis than in that with LN metastasis. Similar findings were obtained for the clinical specimens. CEACAM5 levels were associated with better clinical prognosis. CEACAM5 was found to inhibit the proliferation and migration and promote the apoptosis of HNSCC cells. A mouse xenograft model showed that CEACAM5 inhibited LN metastasis. In conclusions, CEACAM5 inhibited epithelial-mesenchymal transition (EMT) in HNSCC by reducing murine double minute 2 (MDM2) expression and thereby suppressing LN metastasis. CEACAM5 has potential as both a prognostic marker and a therapeutic target in HNSCC.
Collapse
Affiliation(s)
- Xudong Wang
- Department of Otorhinolaryngology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Department of Otorhinolaryngology, The People’s Hospital of Jianyang City, Jianyang, Sichuan 641400, China
| | - Yanshi Li
- Department of Otorhinolaryngology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Min Pan
- Department of Otorhinolaryngology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Tao Lu
- Department of Otorhinolaryngology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Min Wang
- Department of Otorhinolaryngology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Zhihai Wang
- Department of Otorhinolaryngology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Chuan Liu
- Department of Otorhinolaryngology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Guohua Hu
- Department of Otorhinolaryngology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
19
|
Qu C, Zhang H, Cao H, Tang L, Mo H, Liu F, Zhang L, Yi Z, Long L, Yan L, Wang Z, Zhang N, Luo P, Zhang J, Liu Z, Ye W, Liu Z, Cheng Q. Tumor buster - where will the CAR-T cell therapy 'missile' go? Mol Cancer 2022; 21:201. [PMID: 36261831 PMCID: PMC9580202 DOI: 10.1186/s12943-022-01669-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/26/2022] [Indexed: 11/10/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell (CAR-T cell) therapy based on gene editing technology represents a significant breakthrough in personalized immunotherapy for human cancer. This strategy uses genetic modification to enable T cells to target tumor-specific antigens, attack specific cancer cells, and bypass tumor cell apoptosis avoidance mechanisms to some extent. This method has been extensively used to treat hematologic diseases, but the therapeutic effect in solid tumors is not ideal. Tumor antigen escape, treatment-related toxicity, and the immunosuppressive tumor microenvironment (TME) limit their use of it. Target selection is the most critical aspect in determining the prognosis of patients receiving this treatment. This review provides a comprehensive summary of all therapeutic targets used in the clinic or shown promising potential. We summarize CAR-T cell therapies' clinical trials, applications, research frontiers, and limitations in treating different cancers. We also explore coping strategies when encountering sub-optimal tumor-associated antigens (TAA) or TAA loss. Moreover, the importance of CAR-T cell therapy in cancer immunotherapy is emphasized.
Collapse
Affiliation(s)
- Chunrun Qu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- XiangYa School of Medicine, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hao Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hui Cao
- Department of Psychiatry, The Second People's Hospital of Hunan Province, The Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Lanhua Tang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haoyang Mo
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- XiangYa School of Medicine, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fangkun Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Liyang Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhenjie Yi
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- XiangYa School of Medicine, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lifu Long
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- XiangYa School of Medicine, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Luzhe Yan
- XiangYa School of Medicine, Central South University, Changsha, Hunan, China
| | - Zeyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Nan Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- One-third Lab, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou, Zhengzhou, Henan, China
| | - Weijie Ye
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
20
|
Fanciulli G, Modica R, La Salvia A, Campolo F, Florio T, Mikovic N, Plebani A, Di Vito V, Colao A, Faggiano A. Immunotherapy of Neuroendocrine Neoplasms: Any Role for the Chimeric Antigen Receptor T Cells? Cancers (Basel) 2022; 14:cancers14163991. [PMID: 36010987 PMCID: PMC9406675 DOI: 10.3390/cancers14163991] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/14/2022] [Accepted: 08/17/2022] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Neuroendocrine neoplasms (NENs) comprise a heterogeneous group of tumors arising in different organs whose clinical course is variable according to histological differentiation and metastatic spread. Therapeutic options have recently expanded, but there is a need for new effective therapies, especially in less differentiated forms. Chimeric antigen receptor T cells (CAR-T) have shown efficacy in several cancers, mainly hematological, but data on NENs are scattered. We aimed to analyze the available preclinical and clinical data about CAR-T in NENs, to highlight their potential role in clinical practice. A significant therapeutic effect of CAR-T cells in NENs emerges from preclinical studies. Results from clinical trials are expected in order to define their effective role in these cancers. Abstract Neuroendocrine neoplasms (NENs) are a heterogeneous group of tumors with variable clinical presentation and prognosis. Surgery, when feasible, is the most effective and often curative treatment. However, NENs are frequently locally advanced or already metastatic at diagnosis. Consequently, additional local or systemic therapeutic approaches are required. Immunotherapy, based on chimeric antigen receptor T cells (CAR-T), is showing impressive results in several cancer treatments. The aim of this narrative review is to analyze the available data about the use of CAR-T in NENs, including studies in both preclinical and clinical settings. We performed an extensive search for relevant data sources, comprising full-published articles, abstracts from international meetings, and worldwide registered clinical trials. Preclinical studies performed on both cell lines and animal models indicate a significant therapeutic effect of CAR-T cells in NENs. Ongoing and future clinical trials will clarify the possible role of these drugs in patients with highly aggressive NENs.
Collapse
Affiliation(s)
- Giuseppe Fanciulli
- Neuroendocrine Tumour Unit, Department of Medicine, Surgery and Pharmacy, University of Sassari—Endocrine Unit, AOU Sassari, 07100 Sassari, Italy
- Correspondence:
| | - Roberta Modica
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, Federico II University of Naples, 80131 Naples, Italy
| | - Anna La Salvia
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Federica Campolo
- Department of Experimental Medicine, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Tullio Florio
- Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
- Scientific Institute for Research, Hospitalisation and Healthcare Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Nevena Mikovic
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, ENETS Center of Excellence, Sapienza University of Rome, 00189 Rome, Italy
| | - Alice Plebani
- Laboratory of Geriatric and Oncologic Neuroendocrinology Research, Istituto Auxologico Italiano IRCCS, Cusano Milanino, 20095 Milan, Italy
| | - Valentina Di Vito
- Department of Experimental Medicine, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Annamaria Colao
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, Federico II University of Naples, 80131 Naples, Italy
- UNESCO Chair, Education for Health and Sustainable Development, Federico II University, 80131 Naples, Italy
| | - Antongiulio Faggiano
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, ENETS Center of Excellence, Sapienza University of Rome, 00189 Rome, Italy
| |
Collapse
|