1
|
Mathiyazhagan J, Rajesh C, Sagar S, Caffrey TC, Huang Y, Mohs AM, Swanson BJ, Hollingsworth MA, Brooks CL, Radhakrishnan P. Humanized Anti-MUC16 Antibody-Conjugated Contrast Agents for Magnetic Resonance Imaging of Pancreatic Cancer. Cancers (Basel) 2025; 17:957. [PMID: 40149293 PMCID: PMC11940418 DOI: 10.3390/cancers17060957] [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: 02/06/2025] [Revised: 03/05/2025] [Accepted: 03/08/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Pancreatic ductal adenocarcinoma (PDAC) is diagnosed at a late stage with distant metastasis in an overwhelming 50% of cases, and the prognosis is poor. Treating this extremely aggressive disease with standard-of-care therapies has led to modest benefits in overall survival, mainly due to a lack of targeted early treatment modalities, as early detection has not yet been possible. Mucin-16 (MUC16) is a glycoprotein overexpressed in more than 60% of patients with PDAC and is a tumor-specific biomarker. Methods: In this study, a magnetic resonance imaging (MRI) probe to facilitate the detection of early and late lesions of PDAC is developed by conjugating a MUC16-targeted humanized antibody (huAR9.6) with gadolinium. Results: In preclinical mouse models, this MUC16-targeted MRI probe demonstrates effective contrast enhancement in early lesions of PDAC in the subcutaneous setting and allows for the detection of late-stage pancreatic cancer tumors in an orthotopic model. The probe did not induce any toxicity in vital organs at the administered doses. Conclusions: This study establishes that synthesizing a MUC16-targeted MRI probe is feasible and allows for the better high-resolution contrast enhancement of MUC16+ PDAC lesions to facilitate detection and possibly better treatment strategies.
Collapse
Affiliation(s)
- Jayasindu Mathiyazhagan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Christabelle Rajesh
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Satish Sagar
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Thomas C. Caffrey
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ying Huang
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Aaron M. Mohs
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Benjamin J. Swanson
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Michael A. Hollingsworth
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Cory L. Brooks
- Department of Chemistry and Biochemistry, California State University Fresno, Fresno, CA 93740, USA
| | - Prakash Radhakrishnan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
2
|
Huynh AS, Cohen AS, Doligalski M, Casagni TJ, Moberg VE, Huang X, Morse J, Abrahams D, Lloyd MC, Centeno BA, Baldwin MK, McLaughlin ML, Vagner J, Morse DL. Intraoperative Guidance of Pancreatic Cancer Resection Using a Toll-like Receptor 2-Targeted Fluorescence Molecular Imaging Agent. CANCER RESEARCH COMMUNICATIONS 2024; 4:2877-2887. [PMID: 39320054 PMCID: PMC11536076 DOI: 10.1158/2767-9764.crc-24-0244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/01/2024] [Accepted: 09/23/2024] [Indexed: 09/26/2024]
Abstract
To increase the achievement of negative R0 surgical margins and increase the low survival rates of pancreatic cancer, improvements in assessing tumor margins during surgical resections are needed. This can be accomplished by using pancreatic cancer-targeted fluorescence molecular imaging agents to intraoperatively detect tumor margins in real time. Because Toll-like receptor 2 (TLR2) is broadly expressed among many cancer types including pancreatic adenocarcinomas, a high-affinity TLR2-targeted fluorescence molecular imaging agent (TLR2L-800) was developed. We investigate the potential for increased survival by employing real-time intraoperative tumor detection in a preclinical orthotopic human pancreatic xenograft tumor model using TLR2L-800. Three cohorts of nude mice bearing orthotopic human pancreatic xenograft tumors were intravenously injected with TLR2L-800. At 24 hours postinjection, one cohort underwent in vivo fluorescence-guided surgical removal of tumors using a real-time fluorescence imaging platform, a second cohort underwent visible light surgery (VLS), and a third cohort did not undergo surgery. A fourth, nontumor-bearing cohort was administered TLR2L-800 with no surgery. At 41 days postsurgery, the survival rates were 53% for the fluorescence-guided surgery (FGS) group and 0% for both the VLS and the tumor-bearing no-surgery group. The overall 200-day survival rate of 35% for the FGS group was significant compared with 0% for the VLS group (P value = 0.0018). This study demonstrates the potential of increasing disease-free survival for patients with pancreatic cancer by increasing the attainment of R0 margins using a novel tumor-targeted lipopeptide ligand-based fluorescence molecular imaging agent, TLR2L-800, during real-time FGS. SIGNIFICANCE Human TLR2 is broadly expressed among pancreatic adenocarcinomas, and the highly specific TLR2L-800 fluorescence molecular imaging agent has potential for use in fluorescence-guided surgery to increase R0 margins and improve patient survival.
Collapse
Affiliation(s)
- Amanda S. Huynh
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Allison S. Cohen
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Michael Doligalski
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Todd J. Casagni
- Department of Comparative Medicine, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Valerie E. Moberg
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Xuan Huang
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Jennifer Morse
- Department of Comparative Medicine, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Dominique Abrahams
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Mark C. Lloyd
- Analytic Microscopy Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Barbara A. Centeno
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Margaret K. Baldwin
- Department of Comparative Medicine, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Mark L. McLaughlin
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, West Virginia
| | - Josef Vagner
- BIO5 Institute, University of Arizona, Tucson, Arizona
| | - David L. Morse
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
- Small Animal Imaging Laboratory, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
- Department of Oncologic Sciences, University of South Florida, Tampa, Florida
- Department of Physics, University of South Florida, Tampa, Florida
- Department of Medical Engineering, University of South Florida, Tampa, Florida
| |
Collapse
|
3
|
Muilenburg KM, Ehrhorn EG, Olson MT, Isder CC, Klute KA, Talmon GA, Carlson MA, Ly QP, Mohs AM. MUC16 Retention after Neoadjuvant Chemotherapy in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2024; 16:3439. [PMID: 39456534 PMCID: PMC11506185 DOI: 10.3390/cancers16203439] [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: 08/20/2024] [Revised: 09/27/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis. Currently, surgical resection is the only potentially curative treatment. Unfortunately, less than 20% of PDAC patients are eligible for surgical resection at diagnosis. In the past few decades, neoadjuvant chemotherapy treatment (NCT) has been investigated as a way to downstage PDAC tumors for surgical resection. Fluorescence-guided surgery (FGS) is a technique that can aid in increasing complete resection rates by enhancing the tumor through passive or active targeting of a contrast agent. In active targeting, a probe (e.g., antibody) binds a protein differentially upregulated in the tumor compared to normal tissue. Mucin 16 (MUC16), a transmembrane glycoprotein, has recently been explored as an FGS target in preclinical tumor models. However, the impact of chemotherapy on MUC16 expression is unknown. Methods: To investigate this issue, immunohistochemistry was performed on PDAC patient samples. Results: We found that MUC16 expression was retained after NCT in patient samples (mean expression = 5.7) with minimal change in expression between the matched diagnostic (mean expression = 3.66) and PDAC NCT patient samples (mean expression = 4.5). Conclusions: This study suggests that MUC16 is a promising target for FGS and other targeted therapies in PDAC patients treated with NCT.
Collapse
Affiliation(s)
- Kathryn M. Muilenburg
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA; (K.M.M.); (C.C.I.)
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA; (E.G.E.); (M.T.O.); (K.A.K.); (G.A.T.); (M.A.C.); (Q.P.L.)
| | - Evie G. Ehrhorn
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA; (E.G.E.); (M.T.O.); (K.A.K.); (G.A.T.); (M.A.C.); (Q.P.L.)
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA
| | - Madeline T. Olson
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA; (E.G.E.); (M.T.O.); (K.A.K.); (G.A.T.); (M.A.C.); (Q.P.L.)
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA
| | - Carly C. Isder
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA; (K.M.M.); (C.C.I.)
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA; (E.G.E.); (M.T.O.); (K.A.K.); (G.A.T.); (M.A.C.); (Q.P.L.)
| | - Kelsey A. Klute
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA; (E.G.E.); (M.T.O.); (K.A.K.); (G.A.T.); (M.A.C.); (Q.P.L.)
- Department of Internal Medicine, University of Nebraska Medical Center, 42nd and Emile, Omaha, NE 68198, USA
| | - Geoffrey A. Talmon
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA; (E.G.E.); (M.T.O.); (K.A.K.); (G.A.T.); (M.A.C.); (Q.P.L.)
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE 68198, USA
| | - Mark A. Carlson
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA; (E.G.E.); (M.T.O.); (K.A.K.); (G.A.T.); (M.A.C.); (Q.P.L.)
- Department of Surgery, University of Nebraska Medical Center, 983280 Nebraska Medical Center, Omaha, NE 68198, USA
| | - Quan P. Ly
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA; (E.G.E.); (M.T.O.); (K.A.K.); (G.A.T.); (M.A.C.); (Q.P.L.)
- Department of Surgery, University of Nebraska Medical Center, 983280 Nebraska Medical Center, Omaha, NE 68198, USA
| | - Aaron M. Mohs
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA; (K.M.M.); (C.C.I.)
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA; (E.G.E.); (M.T.O.); (K.A.K.); (G.A.T.); (M.A.C.); (Q.P.L.)
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, S 45th St, Omaha, NE 68198, USA
| |
Collapse
|
4
|
Aguilar EN, Sagar S, Murray BR, Rajesh C, Lei EK, Michaud SA, Goodlett DR, Caffrey TC, Grandgenett PM, Swanson B, Brooks TM, Black AR, van Faassen H, Hussack G, Henry KA, Hollingsworth MA, Brooks CL, Radhakrishnan P. Structural Basis for Multivalent MUC16 Recognition and Robust Anti-Pancreatic Cancer Activity of Humanized Antibody AR9.6. Mol Cancer Ther 2024; 23:836-853. [PMID: 38394685 PMCID: PMC11660185 DOI: 10.1158/1535-7163.mct-23-0868] [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: 12/07/2023] [Revised: 01/25/2024] [Accepted: 02/21/2024] [Indexed: 02/25/2024]
Abstract
Mucin-16 (MUC16) is a target for antibody-mediated immunotherapy in pancreatic ductal adenocarcinoma (PDAC) among other malignancies. The MUC16-specific monoclonal antibody AR9.6 has shown promise for PDAC immunotherapy and imaging. Here, we report the structural and biological characterization of the humanized AR9.6 antibody (huAR9.6). The structure of huAR9.6 was determined in complex with a MUC16 SEA (Sea urchin sperm, Enterokinase, Agrin) domain. Binding of huAR9.6 to recombinant, shed, and cell-surface MUC16 was characterized, and anti-PDAC activity was evaluated in vitro and in vivo. HuAR9.6 bound a discontinuous, SEA domain epitope with an overall affinity of 88 nmol/L. Binding affinity depended on the specific SEA domain(s) present, and glycosylation modestly enhanced affinity driven by favorable entropy and enthalpy and via distinct transition state thermodynamic pathways. Treatment with huAR9.6 reduced the in vitro growth, migration, invasion, and clonogenicity of MUC16-positive PDAC cells and patient-derived organoids (PDO). HuAR9.6 blocked MUC16-mediated ErbB and AKT activation in PDAC cells, PDOs, and patient-derived xenografts and induced antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity. More importantly, huAR9.6 treatment caused substantial PDAC regression in subcutaneous and orthotopic tumor models. The mechanism of action of huAR9.6 may depend on dense avid binding to homologous SEA domains on MUC16. The results of this study validate the translational therapeutic potential of huAR9.6 against MUC16-positive PDACs.
Collapse
Affiliation(s)
- Eric N. Aguilar
- Department of Chemistry and Biochemistry, California State University Fresno, Fresno, CA, USA
| | - Satish Sagar
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Brandy R. Murray
- Department of Chemistry and Biochemistry, California State University Fresno, Fresno, CA, USA
| | - Christabelle Rajesh
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Eric K. Lei
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Sarah A. Michaud
- University of Victoria-Genome BC Proteomics Centre, Victoria, BC, Canada
| | - David R. Goodlett
- University of Victoria-Genome BC Proteomics Centre, Victoria, BC, Canada
| | - Thomas C. Caffrey
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Paul M. Grandgenett
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Benjamin Swanson
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Teresa M. Brooks
- Department of Chemistry and Biochemistry, California State University Fresno, Fresno, CA, USA
| | - Adrian R. Black
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Henk van Faassen
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Greg Hussack
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Kevin A. Henry
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Michael A. Hollingsworth
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Cory L. Brooks
- Department of Chemistry and Biochemistry, California State University Fresno, Fresno, CA, USA
| | - Prakash Radhakrishnan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
5
|
Liu Q, Zhang X, Song Y, Si J, Li Z, Dong Q. Construction and analysis of a reliable five-gene prognostic signature for colon adenocarcinoma associated with the wild-type allelic state of the COL6A6 gene. Transl Cancer Res 2024; 13:2475-2496. [PMID: 38881933 PMCID: PMC11170513 DOI: 10.21037/tcr-23-463] [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: 03/18/2023] [Accepted: 11/29/2023] [Indexed: 06/18/2024]
Abstract
BACKGROUND Tumors emerge by acquiring a number of mutations over time. The first mutation provides a selective growth advantage compared to adjacent epithelial cells, allowing the cell to create a clone that can outgrow the cells that surround it. Subsequent mutations determine the risk of the tumor progressing to metastatic cancer. Some secondary mutations may inhibit the aggressiveness of the tumor while still increasing the survival of the clone. Meaningful mutations in genes may provide a strong molecular foundation for developing novel therapeutic strategies for cancer. METHODS The somatic mutation and prognosis in colon adenocarcinoma (COAD) were analyzed. The copy number variation (CNV) and differentially expressed genes (DEGs) between the collagen type VI alpha 6 chain (COL6A6) mutation (COL6A6-MUT) and the COL6A6 wild-type (COL6A6-WT) subgroups were evaluated. The independent prognostic signatures based on COL6A6-allelic state were determined to construct a Cox model. The biological characteristics and the immune microenvironment between the two risk groups were compared. RESULTS COL6A6 was found to be highly mutated in COAD at a frequency of 9%. Patients with COL6A6-MUT had a good overall survival (OS) compared to those with COL6A6-WT, who had a different CNV pattern. Significant differences in gene expression were established for 593 genes between the COL6A6-MUT and COL6A6-WT samples. Among them, MUC16, ASNSP1, PRR18, PEG10, and RPL26P8 were determined to be independent prognostic factors. The internally validated prognostic risk model, constructed using these five genes, demonstrated its value by revealing a significant difference in patient prognosis between the high-risk and low-risk groups. Specifically, patients in the high-risk group exhibited a considerably worse prognosis than did those in the low-risk group. The high-risk group had a significantly higher proportion of patients over 60 years of age and patients in stage III. Moreover, the tumor immune dysfunction and exclusion (TIDE) score and the expression of human leukocyte antigen (HLA) family genes were all higher in the high-risk group than that in the low-risk group. CONCLUSIONS The allelic state of COL6A6 and the five associated DEGs were identified as novel biomarkers for the diagnosis and prognosis of COAD and may be therapeutic targets in COAD.
Collapse
Affiliation(s)
- Qun Liu
- Second Department of Gastroenterology, Qingdao Municipal Hospital, Dalian Medical University, Qingdao, China
| | - Xiaohua Zhang
- Gastroenterology Center, Qingdao Traditional Chinese Medicine Hospital (Qingdao Hiser Hospital), Qingdao Hiser Hospital Affiliated of Qingdao University, Qingdao, China
| | - Yan Song
- Outpatient Department, Qingdao Traditional Chinese Medicine Hospital (Qingdao Hiser Hospital), Qingdao Hiser Hospital Affiliated of Qingdao University, Qingdao, China
| | - Junli Si
- Second Department of Gastroenterology, Qingdao Municipal Hospital, Dalian Medical University, Qingdao, China
| | - Zhaoshui Li
- Qingdao University, Qingdao Medical College, Qingdao, China
| | - Quanjiang Dong
- Central Laboratories, Department of Gastroenterology, Qingdao Municipal Hospital, Dalian Medical University, Qingdao, China
| |
Collapse
|
6
|
Tu HF, Wong M, Tseng SH, Ingavat N, Olczak P, Notarte KI, Hung CF, Roden RBS. Virus-like particle vaccine displaying an external, membrane adjacent MUC16 epitope elicits ovarian cancer-reactive antibodies. J Ovarian Res 2024; 17:19. [PMID: 38225646 PMCID: PMC10790439 DOI: 10.1186/s13048-023-01325-9] [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: 05/30/2023] [Accepted: 12/12/2023] [Indexed: 01/17/2024] Open
Abstract
BACKGROUND MUC16 is a heavily glycosylated cell surface mucin cleaved in the tumor microenvironment to shed CA125. CA125 is a serum biomarker expressed by > 95% of non-mucinous advanced stage epithelial ovarian cancers. MUC16/CA125 contributes to the evasion of anti-tumor immunity, peritoneal spread and promotes carcinogenesis; consequently, it has been targeted with antibody-based passive and active immunotherapy. However, vaccination against this self-antigen likely requires breaking B cell tolerance and may trigger autoimmune disease. Display of self-antigens on virus-like particles (VLPs), including those produced with human papillomavirus (HPV) L1, can efficiently break B cell tolerance. RESULTS A 20 aa juxta-membrane peptide of the murine MUC16 (mMUC16) or human MUC16 (hMUC16) ectodomain was displayed either via genetic insertion into an immunodominant loop of HPV16 L1-VLPs between residues 136/137, or by chemical coupling using malemide to cysteine sulfhydryl groups on their surface. Female mice were vaccinated intramuscularly three times with either DNA expressing L1-MUC16 fusions via electroporation, or with alum-formulated VLP chemically-coupled to MUC16 peptides. Both regimens were well tolerated, and elicited MUC16-specific serum IgG, although titers were higher in mice vaccinated with MUC16-coupled VLP on alum as compared to L1-MUC16 DNA vaccination. Antibody responses to mMUC16-targeted vaccination cross-reacted with hMUC16 peptide, and vice versa; both were reactive with the surface of CA125+ OVCAR3 cells, but not SKOV3 that lack detectable CA125 expression. Interestingly, vaccination of mice with mMUC16 peptide mixed with VLP and alum elicited mMUC16-specific IgG, implying VLPs provide robust T help and that coupling may not be required to break tolerance to this epitope. CONCLUSION Vaccination with VLP displaying the 20 aa juxta-membrane MUC16 ectodomain, which includes the membrane proximal cleavage site, is likely to be well tolerated and induce IgG targeting ovarian cancer cells, even after CA125 is shed.
Collapse
Affiliation(s)
- Hsin-Fang Tu
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Margaret Wong
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Ssu-Hsueh Tseng
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Nattha Ingavat
- Downstream Processing (DSP), Bioprocessing Technology Institute (BTI), Agency for Science, Technology, and Research (A*STAR), Singapore, 138632, Singapore
| | - Pola Olczak
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Kin Israel Notarte
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21287, USA
- Department of Oncology, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Richard B S Roden
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21287, USA.
- Department of Oncology, Johns Hopkins University, Baltimore, MD, 21287, USA.
- Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, MD, 21287, USA.
| |
Collapse
|
7
|
Song Y, Yuan M, Wang G. Update value and clinical application of MUC16 (cancer antigen 125). Expert Opin Ther Targets 2023; 27:745-756. [PMID: 37584221 DOI: 10.1080/14728222.2023.2248376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/25/2023] [Accepted: 08/10/2023] [Indexed: 08/17/2023]
Abstract
INTRODUCTION The largest transmembrane mucin, mucin 16 (MUC16), contains abundant glycosylation sites on the molecular surface, allowing it to participate in various molecular pathways. When cells lose polarity and become cancerous, MUC16 is overexpressed, and more of the extracellular region (cancer antigen [CA]125) is released into serum and possibly, promote the development of diseases. Thus, MUC16 plays an indispensable role in clinical research and application. AREAS COVERED This review summarizes the update proposed role of MUC16 in carcinogenesis and metastasis. Most importantly, we prospect its potential value in targeted therapy after screening 1226 articles published within the last 10 years from PubMed. Two reviewers screened each record and each report retrieved independently. We have summarized the progress of MUC16/CA125 in basic research and clinical application, and predicted its possible future development directions. EXPERT OPINION As an important noninvasive co-factor in the diagnosis of gynecological diseases, MUC16 has been used for a long time, especially in the diagnosis and treatment of ovarian cancer. The overexpression of MUC16 plays a very obvious role in regulating inflammatory response, supporting immune suppression, and promoting the proliferation, division, and metastasis of cancer cells. In the next 20 years, there will be a luxuriant clinical application of MUC16 as a target for immune monitoring and immunotherapy.
Collapse
Affiliation(s)
- Yaan Song
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Medical Integration and Practice Center, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Gynecology Laboratory, Shandong Provincial Hospital, Jinan, Shandong, China
| | - Ming Yuan
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Gynecology Laboratory, Shandong Provincial Hospital, Jinan, Shandong, China
| | - Guoyun Wang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Gynecology Laboratory, Shandong Provincial Hospital, Jinan, Shandong, China
| |
Collapse
|
8
|
Muilenburg KM, Isder CC, Radhakrishnan P, Batra SK, Ly QP, Carlson MA, Bouvet M, Hollingsworth MA, Mohs AM. Mucins as contrast agent targets for fluorescence-guided surgery of pancreatic cancer. Cancer Lett 2023; 561:216150. [PMID: 36997106 PMCID: PMC10150776 DOI: 10.1016/j.canlet.2023.216150] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/16/2023] [Accepted: 03/26/2023] [Indexed: 03/31/2023]
Abstract
Pancreatic cancer is difficult to resect due to its unique challenges, often leading to incomplete tumor resections. Fluorescence-guided surgery (FGS), also known as intraoperative molecular imaging and optical surgical navigation, is an intraoperative tool that can aid surgeons in complete tumor resection through an increased ability to detect the tumor. To target the tumor, FGS contrast agents rely on biomarkers aberrantly expressed in malignant tissue compared to normal tissue. These biomarkers allow clinicians to identify the tumor and its stage before surgical resection and provide a contrast agent target for intraoperative imaging. Mucins, a family of glycoproteins, are upregulated in malignant tissue compared to normal tissue. Therefore, these proteins may serve as biomarkers for surgical resection. Intraoperative imaging of mucin expression in pancreatic cancer can potentially increase the number of complete resections. While some mucins have been studied for FGS, the potential ability to function as a biomarker target extends to the entire mucin family. Therefore, mucins are attractive proteins to investigate more broadly as FGS biomarkers. This review summarizes the biomarker traits of mucins and their potential use in FGS for pancreatic cancer.
Collapse
Affiliation(s)
- Kathryn M Muilenburg
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 505 S 45th St, Omaha, NE, 68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45th St, Omaha, NE, 68198, USA.
| | - Carly C Isder
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 505 S 45th St, Omaha, NE, 68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45th St, Omaha, NE, 68198, USA.
| | - Prakash Radhakrishnan
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45th St, Omaha, NE, 68198, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 505 S 45th St, Omaha, NE, 68198, USA.
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, S 45th St, Omaha, NE, 68198, USA.
| | - Quan P Ly
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45th St, Omaha, NE, 68198, USA; Department of Surgery, University of Nebraska Medical Center, 983280 Nebraska Medical Center, Omaha, NE, 68198-3280, USA.
| | - Mark A Carlson
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45th St, Omaha, NE, 68198, USA; Department of Surgery, University of Nebraska Medical Center, 983280 Nebraska Medical Center, Omaha, NE, 68198-3280, USA.
| | - Michael Bouvet
- Department of Surgery, University of California San Diego, 9500 Gilman Dr, La Jolla, CA, 92093, USA; VA San Diego Healthcare System, 3350 La Jolla Village Dr, San Diego, CA, 92161, USA.
| | - Michael A Hollingsworth
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45th St, Omaha, NE, 68198, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 505 S 45th St, Omaha, NE, 68198, USA.
| | - Aaron M Mohs
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 505 S 45th St, Omaha, NE, 68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45th St, Omaha, NE, 68198, USA; Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, S 45th St, Omaha, NE, 68198, USA.
| |
Collapse
|