1
|
Dadgar N, Arunachalam AK, Hong H, Phoon YP, Arpi-Palacios JE, Uysal M, Wehrle CJ, Aucejo F, Ma WW, Melenhorst JJ. Advancing Cholangiocarcinoma Care: Insights and Innovations in T Cell Therapy. Cancers (Basel) 2024; 16:3232. [PMID: 39335203 PMCID: PMC11429565 DOI: 10.3390/cancers16183232] [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: 08/21/2024] [Revised: 09/16/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
Cholangiocarcinoma (CCA) is a rare and aggressive malignancy originating from the bile ducts, with poor prognosis and limited treatment options. Traditional therapies, such as surgery, chemotherapy, and radiation, have shown limited efficacy, especially in advanced cases. Recent advancements in immunotherapy, particularly T cell-based therapies like chimeric antigen receptor T (CAR T) cells, tumor-infiltrating lymphocytes (TILs), and T cell receptor (TCR)-based therapies, have opened new avenues for improving outcomes in CCA. This review provides a comprehensive overview of the current state of T cell therapies for CCA, focusing on CAR T cell therapy. It highlights key challenges, including the complex tumor microenvironment and immune evasion mechanisms, and the progress made in preclinical and clinical trials. The review also discusses ongoing clinical trials targeting specific CCA antigens, such as MUC1, EGFR, and CD133, and the evolving role of precision immunotherapy in enhancing treatment outcomes. Despite significant progress, further research is needed to optimize these therapies for solid tumors like CCA. By summarizing the most recent clinical results and future directions, this review underscores the promising potential of T cell therapies in revolutionizing CCA treatment.
Collapse
Affiliation(s)
- Neda Dadgar
- Cleveland Clinic Foundation, Enterprise Cancer Institute, Translational Hematology & Oncology Research, Cleveland, OH 44114, USA;
| | - Arun K. Arunachalam
- Cleveland Clinic Foundation, Lerner Research Institute, Center for Immunotherapy and Precision Immuno-Oncology, Cleveland, OH 44195, USA; (A.K.A.); (H.H.); (Y.P.P.); (J.E.A.-P.); (M.U.)
| | - Hanna Hong
- Cleveland Clinic Foundation, Lerner Research Institute, Center for Immunotherapy and Precision Immuno-Oncology, Cleveland, OH 44195, USA; (A.K.A.); (H.H.); (Y.P.P.); (J.E.A.-P.); (M.U.)
| | - Yee Peng Phoon
- Cleveland Clinic Foundation, Lerner Research Institute, Center for Immunotherapy and Precision Immuno-Oncology, Cleveland, OH 44195, USA; (A.K.A.); (H.H.); (Y.P.P.); (J.E.A.-P.); (M.U.)
| | - Jorge E. Arpi-Palacios
- Cleveland Clinic Foundation, Lerner Research Institute, Center for Immunotherapy and Precision Immuno-Oncology, Cleveland, OH 44195, USA; (A.K.A.); (H.H.); (Y.P.P.); (J.E.A.-P.); (M.U.)
| | - Melis Uysal
- Cleveland Clinic Foundation, Lerner Research Institute, Center for Immunotherapy and Precision Immuno-Oncology, Cleveland, OH 44195, USA; (A.K.A.); (H.H.); (Y.P.P.); (J.E.A.-P.); (M.U.)
| | - Chase J. Wehrle
- Cleveland Clinic Foundation, Digestive Diseases & Surgery Institute, Cleveland, OH 44195, USA; (C.J.W.); (F.A.)
| | - Federico Aucejo
- Cleveland Clinic Foundation, Digestive Diseases & Surgery Institute, Cleveland, OH 44195, USA; (C.J.W.); (F.A.)
| | - Wen Wee Ma
- Cleveland Clinic Foundation, Taussig Cancer Institute, Cleveland, OH 44106, USA;
| | - Jan Joseph Melenhorst
- Cleveland Clinic Foundation, Lerner Research Institute, Center for Immunotherapy and Precision Immuno-Oncology, Cleveland, OH 44195, USA; (A.K.A.); (H.H.); (Y.P.P.); (J.E.A.-P.); (M.U.)
| |
Collapse
|
2
|
Theocharopoulos C, Ziogas IA, Douligeris CC, Efstathiou A, Kolorizos E, Ziogas DC, Kontis E. Antibody-drug conjugates for hepato-pancreato-biliary malignancies: "Magic bullets" to the rescue? Cancer Treat Rev 2024; 129:102806. [PMID: 39094332 DOI: 10.1016/j.ctrv.2024.102806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 07/17/2024] [Accepted: 07/28/2024] [Indexed: 08/04/2024]
Abstract
Hepato-Pancreato-Biliary (HPB) malignancies constitute a highly aggressive group of cancers that have a dismal prognosis. Patients not amenable to curative intent surgical resection are managed with systemic chemotherapy which, however, confers little survival benefit. Antibody-Drug Conjugates (ADCs) are tripartite compounds that merge the intricate selectivity and specificity of monoclonal antibodies with the cytodestructive potency of attached supertoxic payloads. In view of the unmet need for drugs that will enhance the survival rates of HPB cancer patients, the assessment of ADCs for treating HPB malignancies has become the focus of extensive clinical and preclinical investigation, showing encouraging preliminary results. In the current review, we offer a comprehensive overview of the growing body of evidence on ADC approaches tested for HPB malignancies. Starting from a concise discussion of the functional principles of ADCs, we summarize here all available data from preclinical and clinical studies evaluating ADCs in HPB cancers.
Collapse
Affiliation(s)
| | - Ioannis A Ziogas
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| | | | | | | | - Dimitrios C Ziogas
- First Department of Internal Medicine, Laikon General Hospital, School of Medicine, National Kapodistrian University of Athens, Athens 11527, Greece
| | - Elissaios Kontis
- Department of Surgery, Metaxa Cancer Hospital, Piraeus 18537, Greece
| |
Collapse
|
3
|
Uchida S, Serada S, Suzuki Y, Funajima E, Kitakami K, Dobashi K, Tamatani S, Sato Y, Beppu T, Ogasawara K, Naka T. Glypican-1-targeted antibody-drug conjugate inhibits the growth of glypican-1-positive glioblastoma. Neoplasia 2024; 50:100982. [PMID: 38417223 PMCID: PMC10915784 DOI: 10.1016/j.neo.2024.100982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 01/21/2024] [Accepted: 02/21/2024] [Indexed: 03/01/2024]
Abstract
Glioblastoma is the deadliest form of brain tumor. The presence of the blood-brain barrier (BBB) significantly hinders chemotherapy, necessitating the development of innovative treatment options for this tumor. This report presents the in vitro and in vivo efficacy of an antibody-drug conjugate (ADC) that targets glypican-1 (GPC1) in glioblastoma. The GPC1-ADC was created by conjugating a humanized anti-GPC1 antibody (clone T2) with monomethyl auristatin E (MMAE) via maleimidocaproyl-valine-citrulline-p-aminobenzyloxycarbonyl linkers. Immunohistochemical staining analysis of a glioblastoma tissue microarray revealed that GPC1 expression was elevated in more than half of the cases. GPC1-ADC, when bound to GPC1, was efficiently and rapidly internalized in glioblastoma cell lines. It inhibited the growth of GPC1-positive glioma cell lines by inducing cell cycle arrest in the G2/M phase and triggering apoptosis in vitro. We established a heterotopic xenograft model by subcutaneously implanting KALS-1 and administered GPC1-ADC intravenously. GPC1-ADC significantly inhibited tumor growth and increased the number of mitotic cells. We also established an orthotopic xenograft model by intracranially implanting luciferase-transfected KS-1-Luc#19. After injecting Evans blue and resecting brain tissues, dye leakage was observed in the implantation area, confirming BBB disruption. We administered GPC1-ADC intravenously and measured the luciferase activity using an in vivo imaging system. GPC1-ADC significantly inhibited tumor growth and extended survival. In conclusion, GPC1-ADC demonstrated potent intracranial activity against GPC1-positive glioblastoma in an orthotopic xenograft model. These results indicate that GPC1-ADC could represent a groundbreaking new therapy for treating glioblastoma beyond the BBB.
Collapse
Affiliation(s)
- Shun Uchida
- Department of Neurosurgery, School of Medicine Iwate Medical University, Yahaba, Japan; Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Yahaba, Japan
| | - Satoshi Serada
- Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Yahaba, Japan.
| | - Yuji Suzuki
- Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Yahaba, Japan; Division of Allergy and Rheumatology, Department of Internal Medicine, School of Medicine Iwate Medical University, Yahaba, Japan
| | - Eiji Funajima
- Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Yahaba, Japan
| | - Kei Kitakami
- Department of Neurosurgery, School of Medicine Iwate Medical University, Yahaba, Japan; Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Yahaba, Japan
| | - Kazumasa Dobashi
- Department of Neurosurgery, School of Medicine Iwate Medical University, Yahaba, Japan; Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Yahaba, Japan
| | | | - Yuichi Sato
- Department of Neurosurgery, School of Medicine Iwate Medical University, Yahaba, Japan
| | - Takaaki Beppu
- Department of Neurosurgery, School of Medicine Iwate Medical University, Yahaba, Japan
| | - Kuniaki Ogasawara
- Department of Neurosurgery, School of Medicine Iwate Medical University, Yahaba, Japan
| | - Testuji Naka
- Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Yahaba, Japan; Division of Allergy and Rheumatology, Department of Internal Medicine, School of Medicine Iwate Medical University, Yahaba, Japan.
| |
Collapse
|
4
|
Zhang Y, Yan HJ, Wu J. The Tumor Immune Microenvironment plays a Key Role in Driving the Progression of Cholangiocarcinoma. Curr Cancer Drug Targets 2024; 24:681-700. [PMID: 38213139 DOI: 10.2174/0115680096267791231115101107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 10/09/2023] [Accepted: 10/13/2023] [Indexed: 01/13/2024]
Abstract
Cholangiocarcinoma (CCA) is an epithelial cancer distinguished by bile duct cell differentiation and is also a fibroproliferative tumor. It is characterized by a dense mesenchyme and a complex tumor immune microenvironment (TME). The TME comprises both cellular and non-cellular components. The celluar component includes CCA cells, immune cells and mesenchymal cells represented by the cancer-associated fibroblasts (CAFs), while the non-cellular component is represented by mesenchymal elements such as the extracellular matrix (ECM). Recent studies have demonstrated the important role of the TME in the development, progression, and treatment resistance of CCA. These cell-associated prognostic markers as well as intercellular connections, may serve as potential therapeutic targets and could inspire new treatment approaches for CCA in the future. This paper aims to summarize the current understanding of CCA's immune microenvironment, focusing on immune cells, mesenchymal cells, ECM, intercellular interactions, and metabolism within the microenvironment.
Collapse
Affiliation(s)
- Ye Zhang
- Department of Oncology, The Third Affiliated Hospital of Soochow University, 185 Juqian St, Changzhou, 213003, China
| | - Hai-Jiao Yan
- Department of Oncology, The Third Affiliated Hospital of Soochow University, 185 Juqian St, Changzhou, 213003, China
| | - Jun Wu
- Department of Oncology, The Third Affiliated Hospital of Soochow University, 185 Juqian St, Changzhou, 213003, China
| |
Collapse
|
5
|
Hadfield MJ, DeCarli K, Bash K, Sun G, Almhanna K. Current and Emerging Therapeutic Targets for the Treatment of Cholangiocarcinoma: An Updated Review. Int J Mol Sci 2023; 25:543. [PMID: 38203714 PMCID: PMC10779232 DOI: 10.3390/ijms25010543] [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: 10/02/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Cholangiocarcinoma is a malignancy of the bile ducts that is often associated with late diagnosis, poor overall survival, and limited treatment options. The standard of care therapy for cholangiocarcinoma has been cytotoxic chemotherapy with modest improvements in overall survival with the addition of immune checkpoint inhibitors. The discovery of actionable mutations has led to the advent of targeted therapies against FGFR and IDH-1, which has expanded the treatment landscape for this patient population. Significant efforts have been made in the pre-clinical space to explore novel immunotherapeutic approaches, as well as antibody-drug conjugates. This review provides an overview of the current landscape of treatment options, as well as promising future therapeutic targets.
Collapse
Affiliation(s)
- Matthew J. Hadfield
- Division of Hematology/Oncology, Department of Medicine, The Warren Alpert School of Medicine of Brown University, Providence, RI 02806, USA; (M.J.H.); (G.S.)
| | - Kathryn DeCarli
- Division of Hematology/Oncology, Department of Medicine, The Warren Alpert School of Medicine of Brown University, Providence, RI 02806, USA; (M.J.H.); (G.S.)
| | - Kinan Bash
- Department of Graduate Studies, University of New England, Biddeford, ME 04005, USA;
| | - Grace Sun
- Division of Hematology/Oncology, Department of Medicine, The Warren Alpert School of Medicine of Brown University, Providence, RI 02806, USA; (M.J.H.); (G.S.)
| | - Khaldoun Almhanna
- Division of Hematology/Oncology, Department of Medicine, The Warren Alpert School of Medicine of Brown University, Providence, RI 02806, USA; (M.J.H.); (G.S.)
| |
Collapse
|
6
|
Watabe T, Kabayama K, Naka S, Yamamoto R, Kaneda K, Serada S, Ooe K, Toyoshima A, Wang Y, Haba H, Kurimoto K, Kobayashi T, Shimosegawa E, Tomiyama N, Fukase K, Naka T. Immuno-PET and Targeted α-Therapy Using Anti-Glypican-1 Antibody Labeled with 89Zr or 211At: A Theranostic Approach for Pancreatic Ductal Adenocarcinoma. J Nucl Med 2023; 64:1949-1955. [PMID: 37827841 PMCID: PMC10690121 DOI: 10.2967/jnumed.123.266313] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/06/2023] [Indexed: 10/14/2023] Open
Abstract
Glypican-1 (GPC1) is overexpressed in several solid cancers and is associated with tumor progression, whereas its expression is low in normal tissues. This study aimed to evaluate the potential of an anti-GPC1 monoclonal antibody (GPC1 mAb) labeled with 89Zr or 211At as a theranostic target in pancreatic ductal adenocarcinoma. Methods: GPC1 mAb clone 01a033 was labeled with 89Zr or 211At with a deferoxamine or decaborane linker, respectively. The internalization ability of GPC1 mAb was evaluated by fluorescence conjugation using a confocal microscope. PANC-1 xenograft mice (n = 6) were intravenously administered [89Zr]GPC1 mAb (0.91 ± 0.10 MBq), and PET/CT scanning was performed for 7 d. Uptake specificity was confirmed through a comparative study using GPC1-positive (BxPC-3) and GPC1-negative (BxPC-3 GPC1-knockout) xenografts (each n = 3) and a blocking study. DNA double-strand breaks were evaluated using the γH2AX antibody. The antitumor effect was evaluated by administering [211At]GPC1 mAb (∼100 kBq) to PANC-1 xenograft mice (n = 10). Results: GPC1 mAb clone 01a033 showed increased internalization ratios over time. One day after administration, a high accumulation of [89Zr]GPC1 mAb was observed in the PANC-1 xenograft (SUVmax, 3.85 ± 0.10), which gradually decreased until day 7 (SUVmax, 2.16 ± 0.30). The uptake in the BxPC-3 xenograft was significantly higher than in the BxPC-3 GPC1-knockout xenograft (SUVmax, 4.66 ± 0.40 and 2.36 ± 0.36, respectively; P = 0.05). The uptake was significantly inhibited in the blocking group compared with the nonblocking group (percentage injected dose per gram, 7.3 ± 1.3 and 12.4 ± 3.0, respectively; P = 0.05). DNA double-strand breaks were observed by adding 150 kBq of [211At]GPC1 and were significantly suppressed by the internalization inhibitor (dynasore), suggesting a substantial contribution of the internalization ability to the antitumor effect. Tumor growth suppression was observed in PANC-1 mice after the administration of [211At]GPC1 mAb. Internalization inhibitors (prochlorperazine) significantly inhibited the therapeutic effect of [211At]GPC1 mAb, suggesting an essential role in targeted α-therapy. Conclusion: [89Zr]GPC1 mAb PET showed high tumoral uptake in the early phase after administration, and targeted α-therapy using [211At]GPC1 mAb showed tumor growth suppression. GPC1 is a promising target for future applications for the precise diagnosis of pancreatic ductal adenocarcinoma and GPC1-targeted theranostics.
Collapse
Affiliation(s)
- Tadashi Watabe
- Department of Nuclear Medicine and Tracer Kinetics, Graduate School of Medicine, Osaka University, Suita, Japan;
- Institute for Radiation Sciences, Osaka University, Suita, Japan
| | - Kazuya Kabayama
- Institute for Radiation Sciences, Osaka University, Suita, Japan
- Department of Chemistry, Graduate School of Science, Osaka University, Toyonaka, Japan
- Forefront Research Center, Graduate School of Science, Osaka University, Toyonaka, Japan
| | - Sadahiro Naka
- Department of Pharmacy, Osaka University Hospital, Suita, Japan
| | - Ryuku Yamamoto
- Department of Chemistry, Graduate School of Science, Osaka University, Toyonaka, Japan
| | - Kazuko Kaneda
- Institute for Radiation Sciences, Osaka University, Suita, Japan
- Forefront Research Center, Graduate School of Science, Osaka University, Toyonaka, Japan
| | - Satoshi Serada
- Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Yahaba, Japan
| | - Kazuhiro Ooe
- Institute for Radiation Sciences, Osaka University, Suita, Japan
| | | | - Yang Wang
- Nishina Center for Accelerator-Based Science, RIKEN, Saitama, Japan
| | - Hiromitsu Haba
- Nishina Center for Accelerator-Based Science, RIKEN, Saitama, Japan
| | - Kenta Kurimoto
- Department of Pharmacy, Osaka University Hospital, Suita, Japan
| | - Takanori Kobayashi
- Department of Nuclear Medicine and Tracer Kinetics, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Eku Shimosegawa
- Department of Molecular Imaging in Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Noriyuki Tomiyama
- Institute for Radiation Sciences, Osaka University, Suita, Japan
- Department of Radiology, Graduate School of Medicine, Osaka University, Suita, Japan; and
| | - Koichi Fukase
- Institute for Radiation Sciences, Osaka University, Suita, Japan
- Department of Chemistry, Graduate School of Science, Osaka University, Toyonaka, Japan
- Forefront Research Center, Graduate School of Science, Osaka University, Toyonaka, Japan
| | - Tetsuji Naka
- Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Yahaba, Japan
- Division of Allergy and Rheumatology, Department of Internal Medicine, School of Medicine, Iwate Medical University, Yahaba, Japan
| |
Collapse
|
7
|
Pratap A, Qualman A, Garrett H, Westbrook L, The E, Mitra S, Cordero M, Monge KM, Idrovo JP, Chauhan A, Cheng L, Cohen MJ, Mungo B, Wani S, Meguid RA, McCarter MD, Meng X. Silencing Glypican-1 enhances the antitumor effects of Pictilisib via downregulating PI3K/Akt/ERK signaling in chemo-resistant esophageal adenocarcinoma. Mol Cell Oncol 2023; 10:2238873. [PMID: 37649964 PMCID: PMC10464651 DOI: 10.1080/23723556.2023.2238873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/17/2023] [Accepted: 07/17/2023] [Indexed: 09/01/2023]
Abstract
Poorly differentiated esophageal adenocarcinoma (PDEAC) has a dismal prognosis. Glypican-1(GPC-1) is known to be upregulated in several cancer types in contrast to healthy tissues, rendering it as a biomarker. Nevertheless, the potential therapeutic targeting of GPC-1 has not been explored in PDEAC. There is accumulating evidence that GPC-1, via upregulation of PI3K/Akt/ERK signaling, plays a crucial role in the progression and chemoresistance in cancer. Pictilisib, a class I pan PI3K inhibitor, has shown promising antitumor results in clinical trials, however, has not gained widespread success due to acquired drug resistance. This study investigated the role of GPC-1 in chemo-resistant PDEAC and appraises the impact of targeted silencing of GPC-1 on the antitumor effects of Pictilisib in PDEAC cell lines. Immunohistochemistry assays in PDEAC tissue specimens demonstrated a pronounced intensity of staining with GPC-1. Upregulation of GPC-1 was found to be correlated with advanced stage and poor prognosis. In-vitro studies examined the influence of GPC-1 knockdown and Pictilisib, both as individual agents and in combination, on cytotoxicity, cell cycle distribution, apoptosis, and gene expression profiles. Silencing GPC-1 alone showed significantly reduced cell viability, migration, colony formation, epithelial-mesenchymal transition, and stemness in PDEAC cells. Significantly, knockdown of GPC-1 combined with low-dose Pictilisib led to enhancement of cytotoxicity, cell cycle arrest, and apoptosis in ESO-26 and OE-33 cells. In the xenograft mouse model, the combination of Pictilisib and GPC-1 knockdown exhibited synergy. These findings suggest that GPC-1 represents a promising target to augment chemosensitivity in esophageal adenocarcinoma.
Collapse
Affiliation(s)
- Akshay Pratap
- Division of Gastrointestinal Tumor and Endocrine Surgery, University of Colorado, Aurora, CO, USA
| | - Andrea Qualman
- Division of Gastrointestinal Tumor and Endocrine Surgery, University of Colorado, Aurora, CO, USA
| | - Hedlund Garrett
- Department of Allergy and Clinical Immunology Flow Core, University of Colorado, Aurora, CO, USA
| | | | - Erlinda The
- Division of Cardiothoracic Surgery, University of Colorado, Aurora, CO, USA
| | - Sanchayita Mitra
- Division of Gastrointestinal Tumor and Endocrine Surgery, University of Colorado, Aurora, CO, USA
| | - Mila Cordero
- School of Science, Engineering, & Technology, St. Mary’s University, San Antonio, TX, USA
| | - Kenneth Meza Monge
- Division of Gastrointestinal Tumor and Endocrine Surgery, University of Colorado, Aurora, CO, USA
| | - Juan- Pablo Idrovo
- Division of Gastrointestinal Tumor and Endocrine Surgery, University of Colorado, Aurora, CO, USA
| | - Argudit Chauhan
- Department of Biomedical Engineering, University of Colorado, Boulder, USA
| | - Linling Cheng
- Division of Cardiothoracic Surgery, University of Colorado, Aurora, CO, USA
| | - Mitchell Jay Cohen
- Division of Gastrointestinal Tumor and Endocrine Surgery, University of Colorado, Aurora, CO, USA
| | - Benedetto Mungo
- Division of Surgical Oncology, University of Colorado, Aurora, CO, USA
| | - Sachin Wani
- Division of Gastroenterology and Hepatology, University of Colorado, Aurora, CO, USA
| | | | - Martin D McCarter
- Division of Surgical Oncology, University of Colorado, Aurora, CO, USA
| | - Xianzhong Meng
- Division of Cardiothoracic Surgery, University of Colorado, Aurora, CO, USA
| |
Collapse
|
8
|
Kuwatani M, Sakamoto N. Promising Highly Targeted Therapies for Cholangiocarcinoma: A Review and Future Perspectives. Cancers (Basel) 2023; 15:3686. [PMID: 37509347 PMCID: PMC10378186 DOI: 10.3390/cancers15143686] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/16/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
To overcome the poor prognosis of cholangiocarcinoma (CCA), highly targeted therapies, such as antibody-drug conjugates (ADCs), photodynamic therapy (PDT) with/without systemic chemotherapy, and experimental photoimmunotherapy (PIT), have been developed. Three preclinical trials have investigated the use of ADCs targeting specific antigens, namely HER2, MUC1, and glypican-1 (GPC1), for CCA. Trastuzumab emtansine demonstrated higher antiproliferative activity in CCA cells expressing higher levels of HER2. Similarly, "staphylococcal enterotoxin A-MUC1 antibody" and "anti-GPC1 antibody-monomethyl auristatin F" conjugates showed anticancer activity. PDT is effective in areas where appropriate photosensitizers and light coexist. Its mechanism involves photosensitizer excitation and subsequent reactive oxygen species production in cancer cells upon irradiation. Hematoporphyrin derivatives, temoporfin, phthalocyanine-4, talaporfin, and chlorine e6 derivatives have mainly been used clinically and preclinically in bile duct cancer. Currently, new forms of photosensitizers with nanotechnology and novel irradiation catheters are being developed. PIT is the most novel anti-cancer therapy developed in 2011 that selectively kills targeted cancer cells using a unique photosensitizer called "IR700" conjugated with an antibody specific for cancer cells. PIT is currently in the early stages of development for identifying appropriate CCA cell targets and irradiation devices. Future human and artificial intelligence collaboration has potential for overcoming challenges related to identifying universal CCA cell targets. This could pave the way for highly targeted therapies for CCA, such as ADC, PDT, and PIT.
Collapse
Affiliation(s)
- Masaki Kuwatani
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, North 14, West 5, Kita-ku, Sapporo 060-8648, Japan
| | - Naoya Sakamoto
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, North 14, West 5, Kita-ku, Sapporo 060-8648, Japan
| |
Collapse
|
9
|
Jeong MH, Son T, Tae YK, Park CH, Lee HS, Chung MJ, Park JY, Castro CM, Weissleder R, Jo JH, Bang S, Im H. Plasmon-Enhanced Single Extracellular Vesicle Analysis for Cholangiocarcinoma Diagnosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205148. [PMID: 36698298 PMCID: PMC10015870 DOI: 10.1002/advs.202205148] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 01/01/2023] [Indexed: 05/20/2023]
Abstract
Cholangiocarcinoma (CCA) is a fatal disease often detected late in unresectable stages. Currently, there are no effective diagnostic methods or biomarkers to detect CCA early with high confidence. Analysis of tumor-derived extracellular vesicles (tEVs) harvested from liquid biopsies can provide a new opportunity to achieve this goal. Here, an advanced nanoplasmonic sensing technology is reported, termed FLEX (fluorescence-amplified extracellular vesicle sensing technology), for sensitive and robust single EV analysis. In the FLEX assay, EVs are captured on a plasmonic gold nanowell surface and immunolabeled for cancer-associated biomarkers to identify tEVs. The underlying plasmonic gold nanowell structures then amplify EVs' fluorescence signals, an effective amplification process at the single EV level. The FLEX EV analysis revealed a wide heterogeneity of tEVs and their marker levels. FLEX also detected small tEVs not detected by conventional EV fluorescence imaging due to weak signals. Tumor markers (MUC1, EGFR, and EPCAM) are identified in CCA, and this marker combination is applied to detect tEVs in clinical bile samples. The FLEX assay detected CCA with an area under the curve of 0.93, significantly better than current clinical markers. The sensitive and accurate nanoplasmonic EV sensing technology can aid in early CCA diagnosis.
Collapse
Affiliation(s)
- Mi Ho Jeong
- Center for Systems BiologyMassachusetts General HospitalBostonMA02114USA
| | - Taehwang Son
- Center for Systems BiologyMassachusetts General HospitalBostonMA02114USA
| | - Yoo Keung Tae
- Division of GastroenterologyDepartment of Internal MedicineSeverance HospitalYonsei University College of MedicineSeoul03722Republic of Korea
| | - Chan Hee Park
- Division of GastroenterologyDepartment of Internal MedicineSeverance HospitalYonsei University College of MedicineSeoul03722Republic of Korea
| | - Hee Seung Lee
- Division of GastroenterologyDepartment of Internal MedicineSeverance HospitalYonsei University College of MedicineSeoul03722Republic of Korea
| | - Moon Jae Chung
- Division of GastroenterologyDepartment of Internal MedicineSeverance HospitalYonsei University College of MedicineSeoul03722Republic of Korea
| | - Jeong Youp Park
- Division of GastroenterologyDepartment of Internal MedicineSeverance HospitalYonsei University College of MedicineSeoul03722Republic of Korea
| | - Cesar M. Castro
- Center for Systems BiologyMassachusetts General HospitalBostonMA02114USA
- Cancer Center, Massachusetts General HospitalHarvard Medical SchoolBostonMA02114USA
| | - Ralph Weissleder
- Center for Systems BiologyMassachusetts General HospitalBostonMA02114USA
- Cancer Center, Massachusetts General HospitalHarvard Medical SchoolBostonMA02114USA
- Department of RadiologyMassachusetts General HospitalBostonMA02114USA
- Department of Systems BiologyHarvard Medical SchoolBostonMA02115USA
| | - Jung Hyun Jo
- Division of GastroenterologyDepartment of Internal MedicineSeverance HospitalYonsei University College of MedicineSeoul03722Republic of Korea
| | - Seungmin Bang
- Division of GastroenterologyDepartment of Internal MedicineSeverance HospitalYonsei University College of MedicineSeoul03722Republic of Korea
| | - Hyungsoon Im
- Center for Systems BiologyMassachusetts General HospitalBostonMA02114USA
- Department of RadiologyMassachusetts General HospitalBostonMA02114USA
| |
Collapse
|
10
|
Lipolysis-stimulated lipoprotein receptor-targeted antibody-drug conjugate demonstrates potent antitumor activity against epithelial ovarian cancer. Neoplasia 2022; 35:100853. [PMID: 36413881 PMCID: PMC9679668 DOI: 10.1016/j.neo.2022.100853] [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: 06/08/2022] [Revised: 11/05/2022] [Accepted: 11/08/2022] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Epithelial ovarian cancer (EOC) is a lethal malignant tumor, for which new treatment options are urgently required. Lipolysis-stimulated lipoprotein receptor (LSR) is widely expressed in EOC, and it is associated with poor prognosis. In this study, we developed an antibody-drug conjugate (ADC) targeting LSR as a new therapeutic approach to EOC. METHODS We, herein, developed novel anti-LSR monoclonal antibodies (mAbs) and an LSR-ADC by conjugating monomethyl auristatin E as a payload. We subsequently evaluated the in vitro and in vivo (on xenograft models) antitumor effect of the LSR-ADC. RESULTS An overexpression of LSR was observed not only in the primary EOC tumor but also in its lymph node and omental metastases. The EOC cell lines NOVC7-C and OVCAR3 strongly expressed LSR (as compared to ES2 cells). Both the anti-LSR mAb and the LSR-ADC were able to specifically bind to LSR-positive cells and were rapidly internalized and trafficked to the lysosomes. The LSR-ADC demonstrated a potent antitumor effect against NOVC-7C and OVCAR3, but little activity against ES2 cells. In vitro, the LSR-ADC exhibited a potent antitumor effect against NOVC-7C and OVCAR3. Moreover, in the OVCAR3 xenograft models as well as in the patient-derived xenograft models of LSR-positive EOC, the LSR-ADC significantly inhibited tumor growth. The LSR-ADC also suppressed the omental/bowel metastases in OVCAR3-Luc xenografts and improved the median survival. CONCLUSION The developed LSR-ADC demonstrated a significant antitumor activity against LSR-positive EOC cell lines and tumors. Our preclinical data support the use of the LSR-ADC as a novel therapy for patients with LSR-positive ovarian cancer.
Collapse
|
11
|
Ghosh S, Huda P, Fletcher N, Campbell D, Thurecht KJ, Walsh B. Clinical development of an anti-GPC-1 antibody for the treatment of cancer. Expert Opin Biol Ther 2022; 22:603-613. [DOI: 10.1080/14712598.2022.2033204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Saikat Ghosh
- Centre for Advanced Imaging (CAI)-Australian Institute for Bioengineering and Nanotechnology (AIBN), ARC Training Centre for Innovation in Biomedical Imaging Technologies, The University of Queensland, Brisbane, QLD, Australia
| | - Pie Huda
- Centre for Advanced Imaging (CAI)-Australian Institute for Bioengineering and Nanotechnology (AIBN), ARC Training Centre for Innovation in Biomedical Imaging Technologies, The University of Queensland, Brisbane, QLD, Australia
| | - Nicholas Fletcher
- Centre for Advanced Imaging (CAI)-Australian Institute for Bioengineering and Nanotechnology (AIBN), ARC Training Centre for Innovation in Biomedical Imaging Technologies, The University of Queensland, Brisbane, QLD, Australia
| | | | - Kristofer J. Thurecht
- Centre for Advanced Imaging (CAI)-Australian Institute for Bioengineering and Nanotechnology (AIBN), ARC Training Centre for Innovation in Biomedical Imaging Technologies, The University of Queensland, Brisbane, QLD, Australia
| | | |
Collapse
|
12
|
Li D, Lin S, Hong J, Ho M. Immunotherapy for hepatobiliary cancers: Emerging targets and translational advances. Adv Cancer Res 2022; 156:415-449. [DOI: 10.1016/bs.acr.2022.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|