51
|
Siracusano G, Tagliamonte M, Buonaguro L, Lopalco L. Cell Surface Proteins in Hepatocellular Carcinoma: From Bench to Bedside. Vaccines (Basel) 2020; 8:vaccines8010041. [PMID: 31991677 PMCID: PMC7157713 DOI: 10.3390/vaccines8010041] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/16/2020] [Accepted: 01/22/2020] [Indexed: 12/20/2022] Open
Abstract
Cell surface proteins act as the go-between in carrying the information from the extracellular environment to the intracellular signaling proteins. However, these proteins are often deregulated in neoplastic diseases, including hepatocellular carcinoma. This review discusses several recent studies that have investigated the role of cell surface proteins in the occurrence and progression of HCC, highlighting the possibility to use them as biomarkers of the disease and/or targets for vaccines and therapeutics.
Collapse
Affiliation(s)
- Gabriel Siracusano
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, 20132 Milan, Italy;
- Correspondence: ; Tel.: +39-022643-4957
| | - Maria Tagliamonte
- Cancer Immunoregulation Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori IRCCS, “Fondazione Pascale”, 80131 Naples, Italy; (M.T.); (L.B.)
| | - Luigi Buonaguro
- Cancer Immunoregulation Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori IRCCS, “Fondazione Pascale”, 80131 Naples, Italy; (M.T.); (L.B.)
| | - Lucia Lopalco
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, 20132 Milan, Italy;
| |
Collapse
|
52
|
Elgundi Z, Papanicolaou M, Major G, Cox TR, Melrose J, Whitelock JM, Farrugia BL. Cancer Metastasis: The Role of the Extracellular Matrix and the Heparan Sulfate Proteoglycan Perlecan. Front Oncol 2020; 9:1482. [PMID: 32010611 PMCID: PMC6978720 DOI: 10.3389/fonc.2019.01482] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 12/10/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer metastasis is the dissemination of tumor cells to new sites, resulting in the formation of secondary tumors. This process is complex and is spatially and temporally regulated by intrinsic and extrinsic factors. One important extrinsic factor is the extracellular matrix, the non-cellular component of tissues. Heparan sulfate proteoglycans (HSPGs) are constituents of the extracellular matrix, and through their heparan sulfate chains and protein core, modulate multiple events that occur during the metastatic cascade. This review will provide an overview of the role of the extracellular matrix in the events that occur during cancer metastasis, primarily focusing on perlecan. Perlecan, a basement membrane HSPG is a key component of the vascular extracellular matrix and is commonly associated with events that occur during the metastatic cascade. Its contradictory role in these events will be discussed and we will highlight the recent advances in cancer therapies that target HSPGs and their modifying enzymes.
Collapse
Affiliation(s)
- Zehra Elgundi
- Graduate School of Biomedical Engineering, UNSW Sydney, Sydney, NSW, Australia
| | - Michael Papanicolaou
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, UNSW Sydney, Darlinghurst, NSW, Australia.,School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - Gretel Major
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, UNSW Sydney, Darlinghurst, NSW, Australia
| | - Thomas R Cox
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, UNSW Sydney, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - James Melrose
- Graduate School of Biomedical Engineering, UNSW Sydney, Sydney, NSW, Australia.,Raymond Purves Bone and Joint Research Laboratories, Kolling Institute of Medical Research, Royal North Shore Hospital, University of Sydney, St Leonards, NSW, Australia
| | - John M Whitelock
- Graduate School of Biomedical Engineering, UNSW Sydney, Sydney, NSW, Australia
| | - Brooke L Farrugia
- Graduate School of Biomedical Engineering, UNSW Sydney, Sydney, NSW, Australia.,Department of Biomedical Engineering, Melbourne School of Engineering, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
53
|
Li N, Wei L, Liu X, Bai H, Ye Y, Li D, Li N, Baxa U, Wang Q, Lv L, Chen Y, Feng M, Lee B, Gao W, Ho M. A Frizzled-Like Cysteine-Rich Domain in Glypican-3 Mediates Wnt Binding and Regulates Hepatocellular Carcinoma Tumor Growth in Mice. Hepatology 2019; 70:1231-1245. [PMID: 30963603 PMCID: PMC6783318 DOI: 10.1002/hep.30646] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 04/02/2019] [Indexed: 12/12/2022]
Abstract
Wnt signaling is one of the key regulators of hepatocellular carcinoma (HCC) tumor progression. In addition to the classical receptor frizzled (FZD), various coreceptors including heparan sulfate proteoglycans (HSPGs) are involved in Wnt activation. Glypican-3 (GPC3) is an HSPG that is overexpressed in HCC and functions as a Wnt coreceptor that modulates HCC cell proliferation. These features make GPC3 an attractive target for liver cancer therapy. However, the precise interaction of GPC3 and Wnt and how GPC3, Wnt, and FZD cooperate with each other are poorly understood. In this study, we established a structural model of GPC3 containing a putative FZD-like cysteine-rich domain at its N-terminal lobe. We found that F41 and its surrounding residues in GPC3 formed a Wnt-binding groove that interacted with the middle region located between the lipid thumb domain and the index finger domain of Wnt3a. Mutating residues in this groove significantly inhibited Wnt3a binding, β-catenin activation, and the transcriptional activation of Wnt-dependent genes. In contrast with the heparan sulfate chains, the Wnt-binding groove that we identified in the protein core of GPC3 seemed to promote Wnt signaling in conditions when FZD was not abundant. Specifically, blocking this domain using an antibody inhibited Wnt activation. In HCC cells, mutating residue F41 on GPC3 inhibited activation of β-catenin in vitro and reduced xenograft tumor growth in nude mice compared with cells expressing wild-type GPC3. Conclusion: Our investigation demonstrates a detailed interaction of GPC3 and Wnt3a, reveals the precise mechanism of GPC3 acting as a Wnt coreceptor, and provides a potential target site on GPC3 for Wnt blocking and HCC therapy.
Collapse
Affiliation(s)
- Na Li
- Key Laboratory of Human Functional Genomics of Jiangsu Province, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Liwen Wei
- Key Laboratory of Human Functional Genomics of Jiangsu Province, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China.,Bio-medical Center, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, P.R. China
| | - Xiaoyu Liu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Hongjun Bai
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yvonne Ye
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dan Li
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.,School of Life Sciences, East China Normal University, Shanghai 200241, P.R. China
| | - Nan Li
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ulrich Baxa
- Electron Microscopy Laboratory, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Qun Wang
- School of Life Sciences, East China Normal University, Shanghai 200241, P.R. China
| | - Ling Lv
- Liver Transplantation Center of the First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province 210029, P.R. China
| | - Yun Chen
- Key Laboratory of Human Functional Genomics of Jiangsu Province, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Mingqian Feng
- Bio-medical Center, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, P.R. China
| | - Byungkook Lee
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wei Gao
- Key Laboratory of Human Functional Genomics of Jiangsu Province, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China.,Corresponding to: Dr. Wei Gao, School of Basic Medical Science, Nanjing Medical University, 101 Longmian Road, Xuehai Building, Room A110, Nanjing, Jiangsu, 211166, P.R. China. Tel: 86-25-86869471; Fax: 86-25-86869471, . Dr. Mitchell Ho, Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Room 5002, Bethesda, MD 20892-4264. Tel: (240)760-7848; Fax: (301)402-1344;
| | - Mitchell Ho
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.,Corresponding to: Dr. Wei Gao, School of Basic Medical Science, Nanjing Medical University, 101 Longmian Road, Xuehai Building, Room A110, Nanjing, Jiangsu, 211166, P.R. China. Tel: 86-25-86869471; Fax: 86-25-86869471, . Dr. Mitchell Ho, Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Room 5002, Bethesda, MD 20892-4264. Tel: (240)760-7848; Fax: (301)402-1344;
| |
Collapse
|
54
|
Tian Z, Hou X, Liu W, Han Z, Wei L. Macrophages and hepatocellular carcinoma. Cell Biosci 2019; 9:79. [PMID: 31572568 PMCID: PMC6761725 DOI: 10.1186/s13578-019-0342-7] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 09/16/2019] [Indexed: 02/08/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is among the most prevalent and lethal cancers in the human population. HCC is an inflammation-associated cancer caused by different etiological factors. The chronic inflammation leads to continuous cycles of hepatocytes destructive-regenerative process and contributes to HCC initiation and progression. Macrophages play a crucial role in chronic liver inflammation. The tumor microenvironment plays a key role in the progression of HCC. Tumor-associated macrophages are a well-known component of the tumor microenvironment and abundantly infiltrate HCC microenvironment. The roles of macrophages in the development and progression of HCC have been recognized. The deep understanding of macrophages in HCC will be critical for developing effective HCC therapy. Targeting of macrophages might provide novel therapeutic approaches for HCC patients and is an emerging field of interest. This review summarizes the knowledge on the contribution of macrophages in the development and progression of HCC, as well as potential immunotherapy being explored in targeting macrophages.
Collapse
Affiliation(s)
- Zhiqiang Tian
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, 225 Changhai Road, Shanghai, 200438 China
- Department of General Surgery, Wuxi People’s Hospital Affiliated Nanjing Medical University, 299 Qingyang Road, Wuxi, 214000 China
| | - Xiaojuan Hou
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, 225 Changhai Road, Shanghai, 200438 China
| | - Wenting Liu
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, 225 Changhai Road, Shanghai, 200438 China
| | - Zhipeng Han
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, 225 Changhai Road, Shanghai, 200438 China
| | - Lixin Wei
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, 225 Changhai Road, Shanghai, 200438 China
| |
Collapse
|
55
|
Ludwig AD, Labadie KP, Seo YD, Hamlin DK, Nguyen HM, Mahadev VM, Yeung RS, Wilbur DS, Park JO. Yttrium-90-Labeled Anti-Glypican 3 Radioimmunotherapy Halts Tumor Growth in an Orthotopic Xenograft Model of Hepatocellular Carcinoma. JOURNAL OF ONCOLOGY 2019; 2019:4564707. [PMID: 31636665 PMCID: PMC6766125 DOI: 10.1155/2019/4564707] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 08/21/2019] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is the second most lethal malignancy globally and is increasing in incidence in the United States. Unfortunately, there are few effective systemic treatment options, particularly for disseminated disease. Glypican-3 (GPC3) is a proteoglycan cell surface receptor overexpressed in most HCCs and provides a unique target for molecular therapies. We have previously demonstrated that PET imaging using a 89Zr-conjugated monoclonal anti-GPC3 antibody (αGPC3) can bind to minute tumors and allow imaging with high sensitivity and specificity in an orthotopic xenograft mouse model of HCC and that serum alpha-fetoprotein (AFP) levels are highly correlated with tumor size in this model. In the present study, we conjugated 90Y, a high-energy beta-particle-emitting radionuclide, to our αGPC3 antibody to develop a novel antibody-directed radiotherapeutic approach for HCC. Luciferase-expressing HepG2 human hepatoblastoma cells were orthotopically implanted in the livers of athymic nude mice, and tumor establishment was verified at 6 weeks after implantation by bioluminescent imaging and serum AFP concentration. Tumor burden by bioluminescence and serum AFP concentration was highly correlated in our model. Yttrium-90 was conjugated to αGPC3 using the chelating agent 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) and injected via the tail vein into the experimental mice at a dose of 200 μCi/mouse or 300 μCi/mouse. Control mice received DOTA-αGPC3 without radionuclide. At 30 days after a single dose of the radioimmunotherapy agent, mean serum AFP levels in control animals increased dramatically, while animals treated with 200 μCi only experienced a minor increase, indicating cessation of tumor growth, and animals treated with 300 μCi experienced a reduction in serum AFP concentration, indicating tumor shrinkage. Mean tumor-bearing liver weight in control animals was also significantly greater than that in animals that received either dose of 90Y-αGPC3. These results were achieved without significant toxicity as measured by body condition scoring and body weight. The results of this preclinical pilot demonstrate that GPC3 can be used as a target for radioimmunotherapy in an orthotopic mouse model of HCC and may be a target of clinical significance, particularly for disseminated HCC.
Collapse
Affiliation(s)
- Andrew D. Ludwig
- Department of Surgery, University of Washington, Seattle, WA, USA
| | - Kevin P. Labadie
- Department of Surgery, University of Washington, Seattle, WA, USA
| | - Y. David Seo
- Department of Surgery, University of Washington, Seattle, WA, USA
| | - Donald K. Hamlin
- Department of Radiation Oncology, University of Washington, Seattle, WA, USA
| | - Holly M. Nguyen
- Department of Urology, University of Washington, Seattle, WA, USA
| | | | - Raymond S. Yeung
- Department of Surgery, University of Washington, Seattle, WA, USA
| | - D. S. Wilbur
- Department of Radiation Oncology, University of Washington, Seattle, WA, USA
| | - James O. Park
- Department of Surgery, University of Washington, Seattle, WA, USA
| |
Collapse
|
56
|
Nishida T, Kataoka H. Glypican 3-Targeted Therapy in Hepatocellular Carcinoma. Cancers (Basel) 2019; 11:E1339. [PMID: 31510063 PMCID: PMC6770328 DOI: 10.3390/cancers11091339] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/03/2019] [Accepted: 09/07/2019] [Indexed: 02/08/2023] Open
Abstract
Glypican-3 (GPC3) is an oncofetal glycoprotein attached to the cell membrane by a glycophosphatidylinositol anchor. GPC3 is overexpressed in some kinds of tumors, particularly hepatocellular carcinoma (HCC). The prognostic significance of serum GPC3 levels and GPC3 immunoreactivity in tumor cells has been defined in patients with HCC. In addition to its usefulness as a biomarker, GPC3 has attracted attention as a novel therapeutic target molecule, and clinical trials targeting GPC3 are in progress. The major mechanism of anti-GPC3 antibody (GPC3Ab) against cancer cells is antibody-dependent cellular cytotoxicity and/or complement-dependent cytotoxicity. Since GPC3Ab is associated with immune responses, a combination of protocols with immune checkpoint inhibitors has also been investigated. Moreover, some innovative approaches for GPC3-targeting therapy have emerged in recent years. This review introduces the results of recent clinical trials targeting GPC3 in HCC and summarizes the latest knowledge regarding the role of GPC3 in HCC progression and clinical application targeting GPC3.
Collapse
Affiliation(s)
- Takahiro Nishida
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan.
- Division of Gastrointestinal, Endocrine and Pediatric Surgery, Department of Surgery, University of Miyazaki Faculty of Medicine, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan.
| | - Hiroaki Kataoka
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan.
| |
Collapse
|
57
|
Wu JR, You RI, Hu CT, Cheng CC, Rudy R, Wu WS. Hydrogen peroxide inducible clone-5 sustains NADPH oxidase-dependent reactive oxygen species-c-jun N-terminal kinase signaling in hepatocellular carcinoma. Oncogenesis 2019; 8:40. [PMID: 31387985 PMCID: PMC6684519 DOI: 10.1038/s41389-019-0149-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 05/18/2019] [Accepted: 06/21/2019] [Indexed: 12/19/2022] Open
Abstract
Target therapy aiming at critical molecules within the metastatic signal pathways is essential for prevention of hepatocellular carcinoma (HCC) progression. Hic-5 (hydrogen peroxide inducible clone-5) which belongs to the paxillin superfamily, can be stimulated by a lot of metastatic factors, such as transforming growth factor (TGF-β), hepatocyte growth factor (HGF), and reactive oxygen species (ROS). Previous studies implicated Hic-5 cross-talks with the ROS-c-jun N-terminal kinase (JNK) signal cascade in a positive feedback manner. In this report, we addressed this issue in a comprehensive manner. By RNA interference and ectopic Hic-5 expression, we demonstrated Hic-5 was essential for activation of NADPH oxidase and ROS generation leading to activation of downstream JNK and c-jun transcription factor. This was initiated by interaction of Hic-5 with the regulator and adaptor of NADPH oxidase, Rac1 and Traf4, respectively, which may further phosphorylate the nonreceptor tyrosine kinase Pyk2 at Tyr881. On the other hand, promoter activity assay coupled with deletion mapping and site directed mutagenesis strategies demonstrated the distal c-jun and AP4 putative binding regions (943–1126 bp upstream of translational start site) were required for transcriptional activation of Hic-5. Thus Hic-5 was both downstream and upstream of NADPH oxidase-ROS-JNK-c-jun cascade. This signal circuit was essential for regulating the expression of epithelial mesenchymal transition (EMT) factors, such as Snail, Zeb1, E-cadherin, and matrix metalloproteinase 9, involved in HCC cell migration and metastasis. Due to the limited expression of Hic-5 in normal tissue, it can be a promising therapeutic target for preventing HCC metastasis.
Collapse
Affiliation(s)
- Jia-Ru Wu
- Department of Molecular Biology and Human Genetics, Hualien, Taiwan
| | - Ren-In You
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Chi-Tan Hu
- Division of Gastroenterology, Department of Medicine, Buddhist Tzu Chi General Hospital and Tzu Chi University, Hualien, Taiwan.,Research Centre for Hepatology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Chuan-Chu Cheng
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Rudy Rudy
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Wen-Sheng Wu
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien, Taiwan. .,Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan.
| |
Collapse
|
58
|
Immunotherapy in Hepatocellular Carcinoma: Is There a Light at the End of the Tunnel? Cancers (Basel) 2019; 11:cancers11081078. [PMID: 31366113 PMCID: PMC6721326 DOI: 10.3390/cancers11081078] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/10/2019] [Accepted: 07/23/2019] [Indexed: 12/16/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver cancer with dismal prognosis when diagnosed at advanced stages. Surgical resection of the primary tumor or orthotropic liver transplantation serves as a potential curative option. However, this approach is highly dependent on the hepatic reserve and baseline functional status of the patient. Liver directed therapies such as portal vein embolization (PVE), trans-arterial chemoembolization (TACE), and systemic chemotherapy are employed in non-surgical candidates. Sorafenib was the only approved systemic therapeutic agent for almost a decade until the recent approval of lenvatinib by the United States Food and Drug Administration (FDA) as an alternate first-line agent. Regorafenib, nivolumab, pembrolizumab and cabozantinib are approved by the FDA as second-line agents in patients who failed or could not tolerate sorafenib. Ramucirumab was recently FDA approved for the subset of patients that have high alfa-fetoprotein levels (>400 ng/mL). A better understanding of tumorigenesis and encouraging clinical trial results that evaluated immune-checkpoint inhibitors opened doors for immunotherapy in HCC. Immune checkpoint inhibitors have demonstrated a prolonged median overall and progression-free survival in a subset of patients with HCC. On-going translational and clinical research will hopefully provide us with a better understanding of tumor markers, genetic aberrations and other factors that determine the immunotherapy response in HCC. In this review, we sought to summarize the potential role and future directions of immunotherapy in the management of HCC.
Collapse
|
59
|
Xu W, Liu K, Chen M, Sun JY, McCaughan GW, Lu XJ, Ji J. Immunotherapy for hepatocellular carcinoma: recent advances and future perspectives. Ther Adv Med Oncol 2019; 11:1758835919862692. [PMID: 31384311 PMCID: PMC6651675 DOI: 10.1177/1758835919862692] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 06/17/2019] [Indexed: 12/13/2022] Open
Abstract
The introduction of immunotherapies has been a major development in the treatment of many advanced cancers, including hepatocellular carcinoma (HCC). We are entering a new era of systemic therapy for advanced HCC associated with an explosion of clinical trial activity. Data from phase I/II studies of checkpoint inhibitors in advanced HCC have been promising, with durable objective response rates of approximately 20% seen (in both first- and second-line settings) and acceptable safety profiles (including immune-mediated hepatitis). Phase III studies evaluating anti-programmed cell death protein 1 (anti-PD-1) and anti-programmed cell death ligand 1 (anti-PD-L1) antibodies compared with sorafenib are already underway. The potential synergistic effects of anti-PD-1/anti-PD-L1 when used in combination with agents against other checkpoint molecules, systemic therapies, as well as conventional surgical and locoregional therapies are also being explored in upcoming clinical trials. Aside from this, other strategies to harness the immune system, including chimeric antigen receptor-engineered T cells, natural killer cell therapies, and peptide vaccines directed against HCC antigens have entered phase I/II studies. Current limitations of immunotherapies and areas of future research include the accurate assessment and prediction of tumor response, overcoming the immunosuppressive effects of a hypoxic microenvironment, and the management of immune-related hepatitis in patients who already have limited liver reserve.
Collapse
Affiliation(s)
- Weiqi Xu
- Department of Hepatic Surgery and Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, China
| | - Ken Liu
- AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney, NSW, Australia, Sydney Medical School, The University of Sydney, Australia; and Liver Injury and Cancer Program, The Centenary Institute, Sydney, Australia
| | - Minjiang Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research and Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University; Affiliated Lishui Hospital of Zhejiang University; and The Central Hospital of Zhejiang Lishui, China
| | - Jin-Yu Sun
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, China, and Sparkfire Scientific Research Group, Nanjing Medical University, China
| | - Geoffrey W McCaughan
- AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney, NSW, Australia, Sydney Medical School, The University of Sydney, Australia; and Liver Injury and Cancer Program, The Centenary Institute, Sydney, Australia
| | - Xiao-Jie Lu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, 210029 China
| | - Jiansong Ji
- Department of Radiology and Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Fifth Affiliated Hospital of Wenzhou Medical University; Affiliated Lishui Hospital of Zhejiang University; and The Central Hospital of Zhejiang Lishui, China
| |
Collapse
|
60
|
Heczey A. Alliance of the Titans: An Effective Combination of a TKI with CAR T Cells. Mol Ther 2019; 27:1348-1349. [PMID: 31337604 DOI: 10.1016/j.ymthe.2019.07.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Affiliation(s)
- Andras Heczey
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
61
|
Role of cell surface proteoglycans in cancer immunotherapy. Semin Cancer Biol 2019; 62:48-67. [PMID: 31336150 DOI: 10.1016/j.semcancer.2019.07.012] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/05/2019] [Accepted: 07/17/2019] [Indexed: 12/23/2022]
Abstract
Over the past few decades, understanding how tumor cells evade the immune system and their communication with their tumor microenvironment, has been the subject of intense investigation, with the aim of developing new cancer immunotherapies. The current therapies against cancer such as monoclonal antibodies against checkpoint inhibitors, adoptive T-cell transfer, cytokines, vaccines, and oncolytic viruses have managed to improve the clinical outcome of the patients. However, in some tumor entities, the response is limited and could benefit from the identification of novel therapeutic targets. It is known that tumor-extracellular matrix interplay and matrix remodeling are necessary for anti-tumor and pro-tumoral immune responses. Proteoglycans are dominant components of the extracellular matrix and are a highly heterogeneous group of proteins characterized by the covalent attachment of a specific linear carbohydrate chain of the glycosaminoglycan type. At cell surfaces, these molecules modulate the expression and activity of cytokines, chemokines, growth factors, adhesion molecules, and function as signaling co-receptors. By these mechanisms, proteoglycans influence the behavior of cancer cells and their microenvironment during the progression of solid tumors and hematopoietic malignancies. In this review, we discuss why cell surface proteoglycans are attractive pharmacological targets in cancer, and we present current and recent developments in cancer immunology and immunotherapy utilizing proteoglycan-targeted strategies.
Collapse
|
62
|
Redirecting T cells to glypican-3 with 28.41BB.ζ and 28.ζ-41BBL CARs for hepatocellular carcinoma treatment. Protein Cell 2019; 9:664-669. [PMID: 29143211 PMCID: PMC6019662 DOI: 10.1007/s13238-017-0489-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
|
63
|
Liu Y, Di S, Shi B, Zhang H, Wang Y, Wu X, Luo H, Wang H, Li Z, Jiang H. Armored Inducible Expression of IL-12 Enhances Antitumor Activity of Glypican-3-Targeted Chimeric Antigen Receptor-Engineered T Cells in Hepatocellular Carcinoma. THE JOURNAL OF IMMUNOLOGY 2019; 203:198-207. [PMID: 31142602 DOI: 10.4049/jimmunol.1800033] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 04/28/2019] [Indexed: 12/23/2022]
Abstract
Adoptive immunotherapy based on chimeric antigen receptor-modified T (CAR-T) cells has been demonstrated as one of the most promising therapeutic strategies in the treatment of malignancies. However, CAR-T cell therapy has shown limited efficacy for the treatment of solid tumors. This is, in part, because of tumor heterogeneity and a hostile tumor microenvironment, which could suppress adoptively transferred T cell activity. In this study, we, respectively, engineered human- or murine-derived-armored glypican-3 (GPC3)-specific CAR-T cells capable of inducibly expressing IL-12 (GPC3-28Z-NFAT-IL-12) T cells. The results showed that GPC3-28Z-NFAT-IL-12 T cells could lyse GPC3+ tumor cells specifically and increase cytokine secretion compared with GPC3-28Z T cells in vitro. In vivo, GPC3-28Z-NFAT-IL-12 T cells augmented the antitumor effect when encountering GPC3+ large tumor burdens, which could be attributed to IL-12 increasing IFN-γ production, favoring T cells infiltration and persistence. Furthermore, in immunocompetent hosts, low doses of GPC3-m28Z-mNFAT-mIL-12 T cells exerted superior antitumor efficacy without prior conditioning in comparison with GPC3-m28Z T cells. Also, mIL-12 secretion decreased regulatory T cell infiltration in established tumors. In conclusion, these findings demonstrated that the inducible expression of IL-12 could boost CAR-T function with less potential side effects, both in immunodeficient and immunocompetent hosts. The inducibly expressed IL-12-armored GPC3-CAR-T cells could broaden the application of CAR-T-based immunotherapy to patients intolerant of lymphodepletion chemotherapy and might provide an alternative therapeutic strategy for patients with GPC3+ cancers.
Collapse
Affiliation(s)
- Ying Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China; and
| | - Shengmeng Di
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China; and
| | - Bizhi Shi
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China; and
| | | | - Yi Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China; and
| | - Xiuqi Wu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China; and
| | - Hong Luo
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China; and
| | - Huamao Wang
- CARsgen Therapeutics, Shanghai 200233, China
| | - Zonghai Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China; and .,CARsgen Therapeutics, Shanghai 200233, China
| | - Hua Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China; and
| |
Collapse
|
64
|
Busato D, Mossenta M, Baboci L, Di Cintio F, Toffoli G, Dal Bo M. Novel immunotherapeutic approaches for hepatocellular carcinoma treatment. Expert Rev Clin Pharmacol 2019; 12:453-470. [PMID: 30907177 DOI: 10.1080/17512433.2019.1598859] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION The introduction of immune checkpoint inhibitors has been lately proposed for the treatment of hepatocellular carcinoma (HCC) with respect to other cancer types. Several immunotherapeutic approaches are now under evaluation for HCC treatment including: i) antibodies acting as immune checkpoint inhibitors; ii) antibodies targeting specific tumor-associated antigens; iii) chimeric antigen receptor redirected T (CAR-T) cells targeting specific tumor-associated antigens; iv) vaccination strategies with tumor-specific epitopes. Areas covered: The review provides a wide description of the clinical trials investigating the efficacy of the main immunotherapeutic approaches proposed for the treatment of patients affected by HCC. Expert opinion: The balancing between immunostimulative and immunosuppressive factors in the context of HCC tumor microenvironment results in heterogeneous response rates to immunotherapeutic approaches such as checkpoint inhibitors, among HCC patients. In this context, it becomes crucial the identification of predictive factors determining the treatment response. A multiple approach using different biomarkers could be useful to identify the subgroup of HCC patients responsive to the treatment with a checkpoint inhibitor (as an example, nivolumab) as single agent, and to identify those patients in which other treatment regimens, such as the combination with sorafenib, or with locoregional therapies, could be more effective.
Collapse
Affiliation(s)
- Davide Busato
- a Experimental and Clinical Pharmacology Unit , Centro di Riferimento Oncologico di Aviano (CRO), IRCCS , Aviano (PN) , Italy.,b Department of Life Sciences , University of Trieste , Trieste , Italy
| | - Monica Mossenta
- a Experimental and Clinical Pharmacology Unit , Centro di Riferimento Oncologico di Aviano (CRO), IRCCS , Aviano (PN) , Italy.,b Department of Life Sciences , University of Trieste , Trieste , Italy
| | - Lorena Baboci
- a Experimental and Clinical Pharmacology Unit , Centro di Riferimento Oncologico di Aviano (CRO), IRCCS , Aviano (PN) , Italy
| | - Federica Di Cintio
- a Experimental and Clinical Pharmacology Unit , Centro di Riferimento Oncologico di Aviano (CRO), IRCCS , Aviano (PN) , Italy.,b Department of Life Sciences , University of Trieste , Trieste , Italy
| | - Giuseppe Toffoli
- a Experimental and Clinical Pharmacology Unit , Centro di Riferimento Oncologico di Aviano (CRO), IRCCS , Aviano (PN) , Italy
| | - Michele Dal Bo
- a Experimental and Clinical Pharmacology Unit , Centro di Riferimento Oncologico di Aviano (CRO), IRCCS , Aviano (PN) , Italy
| |
Collapse
|
65
|
Shimizu Y, Suzuki T, Yoshikawa T, Endo I, Nakatsura T. Next-Generation Cancer Immunotherapy Targeting Glypican-3. Front Oncol 2019; 9:248. [PMID: 31024850 PMCID: PMC6469401 DOI: 10.3389/fonc.2019.00248] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 03/18/2019] [Indexed: 12/16/2022] Open
Abstract
Glypican-3 (GPC3), a 65 kD protein consisting of 580 amino acids, is a heparan sulfate proteoglycan bound to the cell membrane by glycosylphosphatidylinositol. This protein is expressed in the liver and the kidney of healthy fetuses but is hardly expressed in adults, except in the placenta. Contrarily, GPC3 is specifically expressed in hepatocellular carcinoma (HCC), ovarian clear cell carcinoma, melanoma, squamous cell carcinoma of the lung, hepatoblastoma, nephroblastoma (Wilms tumor), yolk sac tumor, and some pediatric cancers. Although the precise function of GPC3 remains unclear, it has been strongly suggested that it is related to the malignant transformation of HCC. We identified GPC3 as a promising target for cancer immunotherapy and have been working on the development of cancer immunotherapeutic agents targeting it through clinical trials. In some trials, it was revealed that the GPC3 peptide vaccines we developed using human leukocyte antigen-A24- and A2-restricted GPC3-derived peptides could induce GPC3-specific cytotoxic T cells in most vaccinated patients and thereby improve their prognosis. To further improve the clinical efficacy of cancer immunotherapy targeting GPC3, we are also developing next-generation therapeutic strategies using T cells engineered to express antigen-specific T-cell receptor or chimeric antigen receptor. In addition, we have successfully monitored the levels of serum full-length GPC3 protein, which is somehow secreted in the blood. The utility of GPC3 as a biomarker for predicting tumor recurrence and treatment efficacy is now being considered. In this review article, we summarize the results of clinical trials carried out by our team and describe the novel agent targeting the cancer-specific shared antigen, GPC3.
Collapse
Affiliation(s)
- Yasuhiro Shimizu
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan.,Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Toshihiro Suzuki
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Toshiaki Yoshikawa
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tetsuya Nakatsura
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| |
Collapse
|
66
|
Abstract
Hepatocellular carcinoma (HCC) has an increasing incidence and dismal prognosis, with few systemic treatments approved, including several small molecule tyrosine kinase inhibitors. The application of immune checkpoint inhibitors (ICIs) to HCC has resulted in durable activity, and further evaluation is ongoing. In this review, we discuss the immunologic principles and the mechanism of action of the ICIs and present the relevant clinical data. Furthermore, we provide an overview of the current and emerging immunotherapeutic approaches for HCC, such as combination treatments, vaccines, and cellular therapies.
Collapse
Affiliation(s)
- Charalampos S Floudas
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Rm B2L312, 10 Center Drive, Bethesda, MD, 20892-1078, USA.
| | - Gagandeep Brar
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Rm B2L312, 10 Center Drive, Bethesda, MD, 20892-1078, USA
| | - Tim F Greten
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Rm B2L312, 10 Center Drive, Bethesda, MD, 20892-1078, USA
- Liver Cancer Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
67
|
Akazawa Y, Suzuki T, Yoshikawa T, Mizuno S, Nakamoto Y, Nakatsura T. Prospects for immunotherapy as a novel therapeutic strategy against hepatocellular carcinoma. World J Meta-Anal 2019; 7:80-95. [DOI: 10.13105/wjma.v7.i3.80] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 03/12/2019] [Accepted: 03/16/2019] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a highly aggressive malignant disease, with a poor clinical prognosis. Many standard therapies are often considered for HCC treatment today; however, these conventional therapies often fail to achieve sufficiently effective clinical results. Today, HCC therapy is set to undergo a major revolution, owing to rapid developments in cancer immunotherapy, particularly immune checkpoint inhibitor therapy. Cancer immunotherapy is a novel and promising treatment strategy that differs significantly from conventional therapies in its approach to achieve antitumor effects. In fact, many cancer immunotherapies have been tested worldwide and shown to be effective against various types of cancer; HCC is no exception to this trend. For example, we identified a specific cancer antigen called glypican-3 (GPC3) and performed clinical trials of GPC3-targeted peptide vaccine immunotherapy in patients with HCC. Here, we present an overview of the immune mechanisms for development and progression of HCC, our GPC3-based immunotherapy, and immune checkpoint inhibitor therapy against HCC. Finally, we discuss the future prospects of cancer immunotherapy against HCC. We believe that this review and discussion of cancer immunotherapy against HCC could stimulate more interest in this promising strategy for cancer therapy and help in its further development.
Collapse
Affiliation(s)
- Yu Akazawa
- Toshiaki Yoshioka, Shoichi Mizuno, Tetsuya Nakatsura, Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan
| | - Toshihiro Suzuki
- Toshiaki Yoshioka, Shoichi Mizuno, Tetsuya Nakatsura, Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
| | | | | | - Yasunari Nakamoto
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan
| | | |
Collapse
|
68
|
Pradier MF, Reis B, Jukofsky L, Milletti F, Ohtomo T, Perez-Cruz F, Puig O. Case-control Indian buffet process identifies biomarkers of response to Codrituzumab. BMC Cancer 2019; 19:278. [PMID: 30922327 PMCID: PMC6438135 DOI: 10.1186/s12885-019-5472-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 03/14/2019] [Indexed: 02/08/2023] Open
Abstract
Background Codrituzumab, a humanized monoclonal antibody against Glypican-3 (GPC3), which is expressed in hepatocellular carcinoma (HCC), was tested in a randomized phase II trial in advanced HCC patients who had failed prior systemic therapy. Biomarker analysis was performed to identify a responder population that benefits from treatment. Methods A novel statistical method based on the Indian buffet process (IBP) was used to identify biomarkers predictive of response to treatment with Codrituzumab. The IBP is a novel method that allows flexibility in analysis design, and which is sensitive to slight, but meaningful between-group differences in biomarkers in very complex datasets Results The IBP model identified several subpopulations of patients having defined biomarker values. Tumor necrosis and viable cell content in the tumor were identified as prognostic markers of disease progression, as were the well-known HCC prognostic markers of disease progression, alpha-fetoprotein and Glypican-3 expression. Predictive markers of treatment response included natural killer (NK) cell surface markers and parameters influencing NK cell activity, all related to the mechanism of action of this drug Conclusions The Indian buffet process can be effectively used to detect statistically significant signals with high sensitivity in complex and noisy biological data Trial registration NCT01507168, January 6, 2012 Electronic supplementary material The online version of this article (10.1186/s12885-019-5472-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Melanie F Pradier
- Harvard University, Cambridge, MA, USA. .,University Carlos III in Madrid, Leganés, Spain.
| | - Bernhard Reis
- Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Lori Jukofsky
- Translational Medicine Oncology, Roche Innovation Center New York, New York, USA
| | - Francesca Milletti
- pRED Informatics, Roche Innovation Center New York, New York, USA.,Kite, a Gilead Company, Santa Monica, CA, USA
| | | | - Fernando Perez-Cruz
- Swiss Data Science Center, ETH Zurich, Switzerland.,University Carlos III in Madrid, Leganés, Spain
| | - Oscar Puig
- Translational Medicine Oncology, Roche Innovation Center New York, New York, USA.,Translational Medicine, Bristol-Myers Squibb, Township, Lawrenceville, NJ, USA
| |
Collapse
|
69
|
Ortiz MV, Roberts SS, Glade Bender J, Shukla N, Wexler LH. Immunotherapeutic Targeting of GPC3 in Pediatric Solid Embryonal Tumors. Front Oncol 2019; 9:108. [PMID: 30873384 PMCID: PMC6401603 DOI: 10.3389/fonc.2019.00108] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 02/05/2019] [Indexed: 12/23/2022] Open
Abstract
Glypican 3 (GPC3) is a heparan sulfate proteoglycan and cell surface oncofetal protein which is highly expressed on a variety of pediatric solid embryonal tumors including the majority of hepatoblastomas, Wilms tumors, rhabdoid tumors, certain germ cell tumor subtypes, and a minority of rhabdomyosarcomas. Via both its core protein and heparan sulfate side chains, GPC3 activates the canonical Wnt/β-catenin pathway, which is frequently overexpressed in these malignancies. Loss of function mutations in GPC3 lead to Simpson-Golabi-Behmel Syndrome, an X-linked overgrowth condition with a predisposition to GPC3-expressing cancers including hepatoblastoma and Wilms tumor. There are several immunotherapeutic approaches to targeting GPC3, including vaccines, monoclonal antibodies, antibody-drug conjugates, bispecific antibodies, cytolytic T lymphocytes, and CAR T cells. These therapies offer a potentially novel means to target these pediatric solid embryonal tumors. A key pediatric-specific consideration of GPC3-targeted immunotherapeutics is that GPC3 can be physiologically expressed in normal tissues during the first year of life, particularly in the liver and kidney. In summary, this article reviews the current evidence for targeting childhood cancers with GPC3-directed immunotherapies.
Collapse
Affiliation(s)
- Michael V Ortiz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Stephen S Roberts
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Julia Glade Bender
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Neerav Shukla
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Leonard H Wexler
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
70
|
Kawaida M, Yamazaki K, Tsujikawa H, Fukuma M, Abe Y, Kitago M, Shinoda M, Kitagawa Y, Sakamoto M. Diffuse and canalicular patterns of glypican-3 expression reflect malignancy of hepatocellular carcinoma. Pathol Int 2019; 69:125-134. [PMID: 30729617 DOI: 10.1111/pin.12767] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 12/20/2018] [Indexed: 01/08/2023]
Abstract
Glypican-3 (GPC3) is expressed in most hepatocellular carcinomas (HCCs). To investigate the significance of various GPC3 staining patterns in HCC, we classified 134 HCC patients into three groups: those with diffuse GPC3 staining, canalicular GPC3 staining, and others (including negative staining). HCCs with diffuse staining were correlated with poor differentiation, high Ki-67 indices, high serum α-fetoprotein (AFP) levels, and early recurrence. In contrast, HCCs with canalicular staining were well differentiated with lower AFP levels. Overall survival in this group was better than that of the other two groups. Comparative analysis of GPC3 staining patterns with markers for HCC subclassification showed that diffuse staining was correlated with the expression of biliary/stem cell markers, whereas canalicular staining was correlated with expression of the markers of WNT-activated HCCs. Induction of leucine-rich repeat-containing G-protein-coupled receptor 5 (LGR5), known as a target of the WNT signaling pathway, in HCC cells resulted in reduced GPC3 expression in vitro. The LGR5-induced cells formed tumors with canaliculus-like structures in mice and showed canalicular GPC3 staining. The current findings showed the significance of recognizing distinct GPC3 staining patterns, i.e., diffuse and canalicular, which may reflect different carcinogenetic mechanisms and indicate the level of malignancy of HCC.
Collapse
Affiliation(s)
- Miho Kawaida
- Department of Pathology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Ken Yamazaki
- Department of Pathology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hanako Tsujikawa
- Department of Pathology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Mariko Fukuma
- Department of Pathology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yuta Abe
- Department of Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Minoru Kitago
- Department of Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Masahiro Shinoda
- Department of Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Michiie Sakamoto
- Department of Pathology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| |
Collapse
|
71
|
Sung PS, Jang JW. Natural Killer Cell Dysfunction in Hepatocellular Carcinoma: Pathogenesis and Clinical Implications. Int J Mol Sci 2018; 19:ijms19113648. [PMID: 30463262 PMCID: PMC6274919 DOI: 10.3390/ijms19113648] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/16/2018] [Accepted: 11/16/2018] [Indexed: 12/14/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is currently the third leading cause of malignancy-related mortalities worldwide. Natural killer (NK) cells are involved in the critical role of first line immunological defense against cancer development. Defects in NK cell functions are recognized as important mechanisms for immune evasion of tumor cells. NK cell function appears to be attenuated in HCC, and many previous reports suggested that NK cells play a critical role in controlling HCC, suggesting that boosting the activity of dysfunctional NK cells can enhance tumor cell killing. However, the detailed mechanisms of NK cell dysfunction in tumor microenvironment of HCC remain largely unknown. A better understanding of the mechanisms of NK cell dysfunction in HCC will help in the NK cell-mediated eradication of cancer cells and prolong patient survival. In this review, we describe the various mechanisms underlying human NK cell dysfunction in HCC. Further, we summarize current advances in the approaches to enhance endogenous NK cell function and in adoptive NK cell therapies, to cure this difficult-to-treat cancer.
Collapse
Affiliation(s)
- Pil Soo Sung
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
- The Catholic Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
| | - Jeong Won Jang
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
- The Catholic Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
| |
Collapse
|
72
|
Li N, Gao W, Zhang YF, Ho M. Glypicans as Cancer Therapeutic Targets. Trends Cancer 2018; 4:741-754. [PMID: 30352677 PMCID: PMC6209326 DOI: 10.1016/j.trecan.2018.09.004] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 08/29/2018] [Accepted: 09/06/2018] [Indexed: 12/14/2022]
Abstract
Glypicans are a group of cell-surface glycoproteins in which heparan sulfate (HS) glycosaminoglycan chains are covalently linked to a protein core. The glypican gene family is broadly conserved across animal species and plays important roles in biological processes. Glypicans can function as coreceptors for multiple signaling molecules known for regulating cell growth, motility, and differentiation. Some members of the glypican family, including glypican 2 (GPC2) and glypican 3 (GPC3), are expressed in childhood cancers and liver cancers, respectively. Antibody-based therapies targeting glypicans are being investigated in preclinical and clinical studies, with the goal of treating solid tumors that do not respond to standard therapies. These studies may establish glypicans as a new class of therapeutic targets for treating cancer.
Collapse
Affiliation(s)
- Nan Li
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wei Gao
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China
| | - Yi-Fan Zhang
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA
| | - Mitchell Ho
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
73
|
De Stefano F, Chacon E, Turcios L, Marti F, Gedaly R. Novel biomarkers in hepatocellular carcinoma. Dig Liver Dis 2018; 50:1115-1123. [PMID: 30217732 DOI: 10.1016/j.dld.2018.08.019] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/09/2018] [Accepted: 08/13/2018] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related deaths and the fifth most common cancer worldwide. Most of these patients are seen with advanced disease at the time of presentation. In spite of its high prevalence, there are not many therapeutic options available for patients with advanced-stage HCC. There is an urgent need for improving early detection and prognostication of patients with HCC. In addition, the development of new therapies targeting specific pathways involved in the pathogenesis of HCC should be a major goal for future research, with the objective of improving outcomes of patients with HCC. Biomarkers represent a relatively easy and noninvasive way to detect and estimate disease prognosis. In spite of the numerous efforts to find molecules as possible biomarkers, there is not a single ideal marker in HCC. Many new findings have shown promising results both in diagnosing and treating HCC. In this review, we summarized the most recent and relevant biomarkers in HCC.
Collapse
Affiliation(s)
- Felice De Stefano
- Transplant and Hepatobiliary Center, Department of Surgery, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Eduardo Chacon
- Transplant and Hepatobiliary Center, Department of Surgery, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Lilia Turcios
- Transplant and Hepatobiliary Center, Department of Surgery, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Francesc Marti
- Transplant and Hepatobiliary Center, Department of Surgery, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Roberto Gedaly
- Transplant and Hepatobiliary Center, Department of Surgery, University of Kentucky College of Medicine, Lexington, KY, United States.
| |
Collapse
|
74
|
Shojaei S, Hashemi SM, Ghanbarian H, Salehi M, Mohammadi-Yeganeh S. Effect of mesenchymal stem cells-derived exosomes on tumor microenvironment: Tumor progression versus tumor suppression. J Cell Physiol 2018; 234:3394-3409. [PMID: 30362503 DOI: 10.1002/jcp.27326] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 08/08/2018] [Indexed: 12/19/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells with the potential to differentiate into different cell types. Owing to their immunosuppressive and anti-inflammatory properties, they are widely used in regenerative medicine, but they have a dual effect on cancer progression and exert both growth-stimulatory or -inhibitory effects on different cancer types. It has been proposed that these controversial effects of MSC in tumor microenvironment (TME) are mediated by their polarization to proinflammatory or anti-inflammatory phenotype. In addition, they can polarize the immune system cells that in turn influence tumor progression. One of the mechanisms involved in the TME communications is extracellular vesicles (EVs). MSCs, as one of cell populations in TME, produce a large amount of EVs that can influence tumor development. Similar to MSC, MSC-EVs can exert both anti- or protumorigenic effects. In the current study, we will investigate the current knowledge related to MSC role in cancer progression with a focus on the MSC-EV content in limiting tumor growth, angiogenesis, and metastasis. We suppose MSC-EVs can be used as safe vehicles for delivering antitumor agents to TME.
Collapse
Affiliation(s)
- Samaneh Shojaei
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Ghanbarian
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Salehi
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samira Mohammadi-Yeganeh
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
75
|
Brar G, Greten TF, Brown ZJ. Current frontline approaches in the management of hepatocellular carcinoma: the evolving role of immunotherapy. Therap Adv Gastroenterol 2018; 11:1756284818808086. [PMID: 30377451 PMCID: PMC6202741 DOI: 10.1177/1756284818808086] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/24/2018] [Indexed: 02/04/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a major cause of cancer-associated mortality worldwide and is expected to rise. Patients with early-stage disease may have a good prognosis with a 5-year survival rate of greater than 70%. However, the majority of patients are diagnosed with late-stage disease with a dismal overall survival rate of less than 16%. Therefore, there is a great need for advances in the treatment of advanced HCC, which for approximately the past decade, has been sorafenib. Immunotherapy is an evolving cancer treatment and has shown promise in treating patients with advanced HCC. In this review, we discuss the current standard of care for advanced HCC and then discuss the evolving role of immunotherapies.
Collapse
Affiliation(s)
- Gagandeep Brar
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tim F. Greten
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Institutes of Health, Building 10, Room 3B43, Bethesda, MD 20892, USA
| | - Zachary J. Brown
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
76
|
Preclinical and Clinical Therapeutic Strategies Affecting Tumor-Associated Macrophages in Hepatocellular Carcinoma. J Immunol Res 2018; 2018:7819520. [PMID: 30410942 PMCID: PMC6206557 DOI: 10.1155/2018/7819520] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 09/07/2018] [Accepted: 09/27/2018] [Indexed: 12/23/2022] Open
Abstract
Hepatocellular carcinoma (HCC) most often develops in patients with underlying liver disease characterized by chronic nonresolving inflammation. Tumor-associated macrophages (TAMs) are one of the most abundant immune cell populations within the tumoral microenvironment. As key actors of cancer-related inflammation, they promote tumor growth by suppression of effective anticancer immunity, stimulation of angiogenesis, and tissue remodeling. Therefore, they have become an attractive and promising target for immunotherapy. The heterogeneity of TAM subtypes and their origin and dynamic phenotype during the initiation and progression of HCC has been partially unraveled and forms the base for the development of therapeutic agents. Current approaches are aimed at decreasing the population of TAMs by depleting macrophages present in the tumor, blocking the recruitment of bone marrow-derived monocytes, and/or functionally reprogramming TAMs to antitumoral behavior. In this review, the preclinical evolution and hitherto clinical trials for TAM-targeted therapy in HCC will be highlighted.
Collapse
|
77
|
Tanaka Y, Tateishi R, Koike K. Proteoglycans Are Attractive Biomarkers and Therapeutic Targets in Hepatocellular Carcinoma. Int J Mol Sci 2018; 19:ijms19103070. [PMID: 30297672 PMCID: PMC6213444 DOI: 10.3390/ijms19103070] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/03/2018] [Accepted: 10/04/2018] [Indexed: 12/11/2022] Open
Abstract
Proteoglycans, which consist of a protein core and glycosaminoglycan chains, are major components of the extracellular matrix and play physiological roles in maintaining tissue homeostasis. In the carcinogenic tissue microenvironment, proteoglycan expression changes dramatically. Altered proteoglycan expression on tumor and stromal cells affects cancer cell signaling pathways, which alters growth, migration, and angiogenesis and could facilitate tumorigenesis. This dysregulation of proteoglycans has been implicated in the pathogenesis of diseases such as hepatocellular carcinoma (HCC) and the underlying mechanism has been studied extensively. This review summarizes the current knowledge of the roles of proteoglycans in the genesis and progression of HCC. It focuses on well-investigated proteoglycans such as serglycin, syndecan-1, glypican 3, agrin, collagen XVIII/endostatin, versican, and decorin, with particular emphasis on the potential of these factors as biomarkers and therapeutic targets in HCC regarding the future perspective of precision medicine toward the "cure of HCC".
Collapse
Affiliation(s)
- Yasuo Tanaka
- Graduate School of Medicine, Department of Gastroenterology, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Ryosuke Tateishi
- Graduate School of Medicine, Department of Gastroenterology, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Kazuhiko Koike
- Graduate School of Medicine, Department of Gastroenterology, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| |
Collapse
|
78
|
Song X, Shang W, Peng L, Jiang H, Wang K, Fang C, Tian J. Novel GPC3-binding WS 2-Ga 3+-PEG-peptide nanosheets for in vivo bimodal imaging-guided photothermal therapy. Nanomedicine (Lond) 2018; 13:1681-1693. [PMID: 30091395 DOI: 10.2217/nnm-2017-0367] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Aim: The diagnosis and treatment of hepatocarcinoma (HCC) is believed to be improved due to the development of specific targeting probes by molecular imaging methods. GPC3, which is a hepatocellular carcinoma (HCC)-specific tumor marker, anchors at most HCC cells. To target this cell membrane protein, we developed a novel nanoparticle by conjugating WS2-Ga3+-PEG and a short peptide with favorable specificity and affinity to the GPC3 protein. Materials & methods: In in vitro assay, several physical properties of the novel probe were evaluated. In in vivo assay, MRI, photoacoustic imaging and photothermal therapy were performed in the subcutaneous HepG2-bearing mice with the novel probe. Results & conclusion: The effect of imaging and photothermal therapy was significant. We revealed that the novel nanosheet WS2-Ga3+-PEG-peptide is promising to detect and treat HCC-expressing GPC3.
Collapse
Affiliation(s)
- Xiaodong Song
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China.,CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, PR China.,Beijing Key Laboratory of Molecular Imaging, Beijing 100190, PR China.,Guangdong Provincial Clinical & Engineering Center of Digital Medicine, Guangzhou, PR China
| | - Wenting Shang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, PR China.,Beijing Key Laboratory of Molecular Imaging, Beijing 100190, PR China
| | - Li Peng
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, PR China.,Beijing Key Laboratory of Molecular Imaging, Beijing 100190, PR China
| | - Hongmei Jiang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, PR China.,Beijing Key Laboratory of Molecular Imaging, Beijing 100190, PR China
| | - Kun Wang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, PR China.,Beijing Key Laboratory of Molecular Imaging, Beijing 100190, PR China
| | - Chihua Fang
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China.,Guangdong Provincial Clinical & Engineering Center of Digital Medicine, Guangzhou, PR China
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, PR China.,Beijing Key Laboratory of Molecular Imaging, Beijing 100190, PR China
| |
Collapse
|
79
|
Moek KL, Fehrmann RSN, van der Vegt B, de Vries EGE, de Groot DJA. Glypican 3 Overexpression across a Broad Spectrum of Tumor Types Discovered with Functional Genomic mRNA Profiling of a Large Cancer Database. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:1973-1981. [PMID: 29935166 DOI: 10.1016/j.ajpath.2018.05.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 05/15/2018] [Accepted: 05/24/2018] [Indexed: 12/27/2022]
Abstract
Glypican 3 (GPC3), a membrane-bound heparan sulfate proteoglycan, is overexpressed in approximately 70% to 80% of hepatocellular carcinomas, but is not expressed commonly in healthy tissues. This raised interest in GPC3 as a drug target and several GPC3-targeting drugs are in clinical development. We therefore predicted GPC3 protein overexpression across tumors and validated these predictions. Functional genomic mRNA profiling was applied to the expression profiles of 18,055 patient-derived tumor samples to predict GPC3 overexpression at the protein level in 60 tumor types and subtypes using healthy tissues as reference. For validation, predictions were compared with immunohistochemical (IHC) staining of a breast cancer tissue microarray and literature data reporting IHC GPC3 overexpression in various solid, hematologic, and pediatric tumors. The percentage of samples with predicted GPC3 overexpression was 77% for hepatocellular carcinomas (n = 364), 45% for squamous cell lung cancers (n = 405), and 19% for head and neck squamous cell cancers (n = 344). Breast cancer tissue microarray analysis showed GPC3 expression ranged from 12% to 17% in subgroups based on estrogen receptor and human epidermal growth factor receptor 2 status. In 28 of 34 tumor types for which functional genomic mRNA data could be compared with IHC there was a relative difference of ≤10%. This study provides a data-driven prioritization of tumor types and subtypes for future research with GPC3-targeting therapies.
Collapse
Affiliation(s)
- Kirsten L Moek
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Rudolf S N Fehrmann
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Bert van der Vegt
- Department of Pathology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Derk J A de Groot
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
80
|
Lipovšek D, Carvajal I, Allentoff AJ, Barros A, Brailsford J, Cong Q, Cotter P, Gangwar S, Hollander C, Lafont V, Lau WL, Li W, Moreta M, O'Neil S, Pinckney J, Smith MJ, Su J, Terragni C, Wallace MA, Wang L, Wright M, Marsh HN, Bryson JW. Adnectin-drug conjugates for Glypican-3-specific delivery of a cytotoxic payload to tumors. Protein Eng Des Sel 2018; 31:159-171. [PMID: 30247737 PMCID: PMC6158766 DOI: 10.1093/protein/gzy013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 05/21/2018] [Accepted: 05/30/2018] [Indexed: 12/12/2022] Open
Abstract
Tumor-specific delivery of cytotoxic agents remains a challenge in cancer therapy. Antibody-drug conjugates (ADC) deliver their payloads to tumor cells that overexpress specific tumor-associated antigens-but the multi-day half-life of ADC leads to high exposure even of normal, antigen-free, tissues and thus contributes to dose-limiting toxicity. Here, we present Adnectin-drug conjugates, an alternative platform for tumor-specific delivery of cytotoxic payloads. Due to their small size (10 kDa), renal filtration eliminates Adnectins from the bloodstream within minutes to hours, ensuring low exposure to normal tissues. We used an engineered cysteine to conjugate an Adnectin that binds Glypican-3, a membrane protein overexpressed in hepatocellular carcinoma, to a cytotoxic derivative of tubulysin, with the drug-to-Adnectin ratio of 1. We demonstrate specific, nanomolar binding of this Adnectin-drug conjugate to human and murine Glypican-3; its high thermostability; its localization to target-expressing tumor cells in vitro and in vivo, its fast clearance from normal tissues and its efficacy against Glypican-3-positive mouse xenograft models.
Collapse
Affiliation(s)
- Daša Lipovšek
- Molecular Discovery Technologies, Bristol-Myers Squibb, Waltham, MA, USA
| | - Irvith Carvajal
- Molecular Discovery Technologies, Bristol-Myers Squibb, Waltham, MA, USA
| | | | - Anthony Barros
- Preclinical Candidate Optimization, Bristol-Myers Squibb, Lawrenceville, NJ, USA
| | - John Brailsford
- Radiochemistry, Bristol-Myers Squibb, Lawrenceville, NJ, USA
| | - Qiang Cong
- Discovery Chemistry Oncology, Bristol-Myers Squibb, Redwood City, CA, USA
| | - Pete Cotter
- Molecular Discovery Technologies, Bristol-Myers Squibb, Waltham, MA, USA
| | - Sanjeev Gangwar
- Discovery Chemistry Oncology, Bristol-Myers Squibb, Redwood City, CA, USA
| | - Cris Hollander
- Molecular Discovery Technologies, Bristol-Myers Squibb, Waltham, MA, USA
| | - Virginie Lafont
- Molecular Discovery Technologies, Bristol-Myers Squibb, Lawrenceville, NJ, USA
| | - Wai Leung Lau
- Molecular Discovery Technologies, Bristol-Myers Squibb, Waltham, MA, USA
| | - Wenying Li
- Preclinical Candidate Optimization, Bristol-Myers Squibb, Lawrenceville, NJ, USA
| | - Miguel Moreta
- Molecular Discovery Technologies, Bristol-Myers Squibb, Waltham, MA, USA
| | - Steven O'Neil
- Molecular Discovery Technologies, Bristol-Myers Squibb, Waltham, MA, USA
| | - Jason Pinckney
- Molecular Discovery Technologies, Bristol-Myers Squibb, Waltham, MA, USA
| | - Michael J Smith
- Chemical and Synthetic Development, Bristol-Myers Squibb, New Brunswick, NJ, USA
| | - Julie Su
- Molecular Discovery Technologies, Bristol-Myers Squibb, Waltham, MA, USA
| | - Christina Terragni
- Molecular Discovery Technologies, Bristol-Myers Squibb, Waltham, MA, USA
| | | | - Lifei Wang
- Preclinical Candidate Optimization, Bristol-Myers Squibb, Lawrenceville, NJ, USA
| | - Martin Wright
- Molecular Discovery Technologies, Bristol-Myers Squibb, Waltham, MA, USA
| | - H Nicholas Marsh
- Molecular Discovery Technologies, Bristol-Myers Squibb, Waltham, MA, USA
| | - James W Bryson
- Molecular Discovery Technologies, Bristol-Myers Squibb, Waltham, MA, USA
| |
Collapse
|
81
|
Kumar N, Khakoo SI. Hepatocellular carcinoma: Prospects for natural killer cell immunotherapy. HLA 2018; 92:3-11. [PMID: 29667374 DOI: 10.1111/tan.13275] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 04/11/2018] [Accepted: 04/12/2018] [Indexed: 12/12/2022]
Abstract
Liver disease is a growing cause of death in the United Kingdom and the incidence of hepatocellular carcinoma (HCC) is rising (http://www.cancerresearchuk.org/). The combination of an immunosuppressive environment within the liver and suboptimal host anti-tumour immune responses may account for the poor survival outcome of HCC. Understanding how tumours evade immune recognition coupled with new insights into the unique immunological environment within the liver will be critical to developing liver-specific immunotherapies.
Collapse
Affiliation(s)
- N Kumar
- Section of Hepatology, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - S I Khakoo
- Department of Clinical and Experimental Sciences, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, UK
| |
Collapse
|
82
|
Dhungel B, Andrzejewski S, Jayachandran A, Shrestha R, Ramlogan-Steel CA, Layton CJ, Steel JC. Evaluation of the Glypican 3 promoter for transcriptional targeting of hepatocellular carcinoma. Gene Ther 2018; 25:115-128. [DOI: 10.1038/s41434-018-0002-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 12/01/2017] [Accepted: 12/27/2017] [Indexed: 12/19/2022]
|
83
|
Nakamura M, Xu C, Diack C, Ohishi N, Lee RM, Iida S, Kawanishi T, Ohtomo T, Abou-Alfa GK, Chen YC. Time-to-event modelling of effect of codrituzumab on overall survival in patients with hepatocellular carcinoma. Br J Clin Pharmacol 2018; 84:944-951. [PMID: 29381229 DOI: 10.1111/bcp.13530] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 01/18/2018] [Accepted: 01/19/2018] [Indexed: 11/30/2022] Open
Abstract
AIMS Codrituzumab (GC33) is a recombinant, humanized mAb that binds to glypican-3 (GPC3), an oncofetal protein highly expressed in hepatocellular carcinoma (HCC). This investigation aimed to identify clinically relevant factors that may affect the overall survival (OS) in HCC patients treated with codrituzumab and to quantitatively annotate their effects. METHODS Codrituzumab exposure was estimated by a population pharmacokinetics model with a nonlinear elimination pathway. Analysis of OS was performed using a time-to-event model in 181 patients with advanced HCC. The model was tested with the addition of various covariates, including levels of immune biomarkers, such as CD16 (measured in terms of molecules of equivalent soluble fluorophore; CD16MESF ) and CD4, codrituzumab exposure and potential prognostic biomarkers of HCC such as baseline tumour size and soluble GPC3. RESULTS The time-to-event model estimated a prolonged OS (>3 months) in patients with codrituzumab exposure of ≥230 μg ml-1 and high CD16MESF level (>5.26 × 105 MESF at least). The Weibull model was selected as the base hazard model. The baseline tumour size was included in the hazard model as a parameter independent of the drug effect. A logistic model was applied to explain the effects of drug exposure and CD16MESF level. CONCLUSIONS The final model indicates that adequate drug exposure plus a favourable immune environment are associated with prolonged OS. This quantitative model should be further validated with emerging data so as to guide study design in future clinical trials.
Collapse
Affiliation(s)
- Mikiko Nakamura
- Translational Clinical Research Science & Strategy Dept., Chugai Pharmaceutical Co., Ltd., Tokyo, Japan
| | - Chao Xu
- Translational and Clinical Research Center, Hoffmann-La Roche Inc., New York City, USA
| | - Cheikh Diack
- Clinical Pharmacology, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Norihisa Ohishi
- Translational Clinical Research Science & Strategy Dept., Chugai Pharmaceutical Co., Ltd., Tokyo, Japan
| | - Ruey-Min Lee
- Translational and Clinical Research Center, Hoffmann-La Roche Inc., New York City, USA
| | - Satofumi Iida
- Translational Clinical Research Science & Strategy Dept., Chugai Pharmaceutical Co., Ltd., Tokyo, Japan
| | - Takehiko Kawanishi
- Translational Clinical Research Science & Strategy Dept., Chugai Pharmaceutical Co., Ltd., Tokyo, Japan
| | - Toshihiko Ohtomo
- Project Planning & Coordination Dept., Chugai Pharmaceutical Co., Ltd., Tokyo, Japan
| | | | - Ya-Chi Chen
- Translational and Clinical Research Center, Hoffmann-La Roche Inc., New York City, USA
| |
Collapse
|
84
|
Carrasquillo JA, O'Donoghue JA, Beylergil V, Ruan S, Pandit-Taskar N, Larson SM, Smith-Jones PM, Lyashchenko SK, Ohishi N, Ohtomo T, Abou-Alfa GK. I-124 codrituzumab imaging and biodistribution in patients with hepatocellular carcinoma. EJNMMI Res 2018; 8:20. [PMID: 29508107 PMCID: PMC5838028 DOI: 10.1186/s13550-018-0374-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 02/22/2018] [Indexed: 01/03/2023] Open
Abstract
Background I-124 codrituzumab (aka GC33), an antibody directed at Glypican 3, was evaluated in patients with hepatocellular carcinoma (HCC). Fourteen patients with HCC underwent baseline imaging with I-124 codrituzumab (~ 185 MBq, 10 mg). Seven of these patients undergoing sorafenib/immunotherapy with 2.5 or 5 mg/kg of cold codrituzumab had repeat imaging, with co-infusion of I-124 codrituzumab, as part of their immunotherapy treatment. Three patients who progressed while on sorafenib/immunotherapy were re-imaged after a 4-week washout period to assess for the presence of antigen. Serial positron emission tomography (PET) imaging and pharmacokinetics were performed following I-124 codrituzumab. An ELISA assay was used to determine “cold” codrituzumab serum pharmacokinetics and compare it to that of I-124 codrituzumab. Correlation of imaging results was performed with IHC. Short-term safety assessment was also evaluated. Results Thirteen patients had tumor localization on baseline I-124 codrituzumab; heterogeneity in tumor uptake was noted. In three patients undergoing repeat imaging while on immunotherapy/sorafenib, evidence of decreased I-124 codrituzumab uptake was noted. All three patients who underwent imaging after progression while on immunotherapy continued to have I-124 codrituzumab tumor uptake. Pharmacokinetics of I-124 codrituzumab was similar to that of other intact IgG. No significant adverse events were observed related to the I-124 codrituzumab. Conclusions I-124 codrituzumab detected tumor localization in most patients with HCC. Pharmacokinetics was similar to that of other intact iodinated humanized IgG. No visible cross-reactivity with normal organs was observed. Electronic supplementary material The online version of this article (10.1186/s13550-018-0374-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jorge A Carrasquillo
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
| | - Joseph A O'Donoghue
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Volkan Beylergil
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Shutian Ruan
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Neeta Pandit-Taskar
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Steven M Larson
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Peter M Smith-Jones
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.,Department of Psychiatry and Behavioral Science, Stony Brook University Hospital, 101 Nicolls Road, Stony Brook, NY, 11794, USA.,Department of Radiology, Stony Brook University Hospital, 101 Nicolls Road, Stony Brook, NY, 11794, USA
| | - Serge K Lyashchenko
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Norihisa Ohishi
- Chugai Pharmaceutical Co., Ltd., 1-1 Nihonbashi-Muromachi 2-Chome Chuo-ku, Tokyo, 103-8324, Japan
| | - Toshihiko Ohtomo
- Chugai Pharmaceutical Co., Ltd., 1-1 Nihonbashi-Muromachi 2-Chome Chuo-ku, Tokyo, 103-8324, Japan
| | - Ghassan K Abou-Alfa
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| |
Collapse
|
85
|
Shimizu Y, Suzuki T, Yoshikawa T, Tsuchiya N, Sawada Y, Endo I, Nakatsura T. Cancer immunotherapy-targeted glypican-3 or neoantigens. Cancer Sci 2018; 109:531-541. [PMID: 29285841 PMCID: PMC5834776 DOI: 10.1111/cas.13485] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 12/22/2017] [Accepted: 12/25/2017] [Indexed: 12/17/2022] Open
Abstract
Immune checkpoint inhibitors have ushered in a new era in cancer therapy, although other therapies or combinations thereof are still needed for many patients for whom these drugs are ineffective. In this light, we have identified glypican‐3 an HLA‐24, HLA‐A2 restriction peptide with extreme cancer specificity. In this paper, we summarize results from a number of related clinical trials showing that glypican‐3 peptide vaccines induce specific CTLs in most patients (UMIN Clinical Trials Registry: UMIN000001395, UMIN000005093, UMIN000002614, UMN000003696, and UMIN000006357). We also describe the current state of personalized cancer immunotherapy based on neoantigens, and assess, based on our own research and experience, the potential of such therapy to elicit cancer regression. Finally, we discuss the future direction of cancer immunotherapy.
Collapse
Affiliation(s)
- Yasuhiro Shimizu
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan.,Department of Gastroenterological Surgery, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Toshihiro Suzuki
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Toshiaki Yoshikawa
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Nobuhiro Tsuchiya
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan.,Department of Gastroenterological Surgery, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Yu Sawada
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan.,Department of Gastroenterological Surgery, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Itaru Endo
- Department of Gastroenterological Surgery, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Tetsuya Nakatsura
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| |
Collapse
|
86
|
Yu M, Luo H, Fan M, Wu X, Shi B, Di S, Liu Y, Pan Z, Jiang H, Li Z. Development of GPC3-Specific Chimeric Antigen Receptor-Engineered Natural Killer Cells for the Treatment of Hepatocellular Carcinoma. Mol Ther 2018; 26:366-378. [PMID: 29339014 PMCID: PMC5835122 DOI: 10.1016/j.ymthe.2017.12.012] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 12/07/2017] [Accepted: 12/14/2017] [Indexed: 02/06/2023] Open
Abstract
Chimeric antigen receptor (CAR)-modified natural killer (NK) cells represent a promising immunotherapeutic modality for cancer treatment. However, their potential utilities have not been explored in hepatocellular carcinoma (HCC). Glypian-3 (GPC3) is a rational immunotherapeutic target for HCC. In this study, we developed GPC3-specific NK cells and explored their potential in the treatment of HCC. The NK-92/9.28.z cell line was established by engineering NK-92, a highly cytotoxic NK cell line with second-generation GPC3-specific CAR. Exposure of GPC3+ HCC cells to this engineered cell line resulted in significant in vitro cytotoxicity and cytokine production. In addition, soluble GPC3 and TGF-β did not significantly inhibit the cytotoxicity of NK-92/9.28.z cells in vitro, and no significant difference in anti-tumor activities was observed in hypoxic (1%) conditions. Potent anti-tumor activities of NK-92/9.28.z cells were observed in multiple HCC xenografts with both high and low GPC3 expression, but not in those without GPC3 expression. Obvious infiltration of NK-92/9.28.z cells, decreased tumor proliferation, and increased tumor apoptosis were observed in the GPC3+ HCC xenografts. Similarly, efficient retargeting on primary NK cells was achieved. These results justified clinical translation of this GPC3-specific, NK cell-based therapeutic as a novel treatment option for patients with GPC3+ HCC.
Collapse
Affiliation(s)
- Min Yu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Luo
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingliang Fan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiuqi Wu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bizhi Shi
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shengmeng Di
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zeyan Pan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hua Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zonghai Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; CARsgen Therapeutics, Shanghai, China.
| |
Collapse
|
87
|
In vitro modeling of hepatocellular carcinoma molecular subtypes for anti-cancer drug assessment. Exp Mol Med 2018; 50:e419. [PMID: 29303513 PMCID: PMC5992986 DOI: 10.1038/emm.2017.164] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 05/11/2017] [Indexed: 12/21/2022] Open
Abstract
Tractable experimental model that accounts for inter-tumor molecular heterogeneity is a key element of anti-cancer drug development. Hepatocellular carcinoma is known to exhibit highly heterogeneous molecular aberrations across the tumors, including somatic genetic and epigenetic alterations. Previous studies showed that molecular tumor subtypes determined by transcriptome, as a comprehensive functional readout, are reproducibly observed across global patient populations irrespective of geographic and etiological variations. Here we demonstrate that transcriptomic hepatocellular carcinoma subtypes, S1 and S2, determined by our previous transcriptome meta-analysis of multiple clinical hepatocellular carcinoma cohorts, are presented in a panel of hepatoma cell lines widely used by the research community. Interestingly, cell line that resembles gene expression pattern of S3 subtype, representing less aggressive tumors, was not identified in the panel. MYC pathway-activated S2-like cell lines showed higher sensitivity to a small molecule BET bromodomain inhibitor, (+)-JQ1, which has anti-MYC activity. These results support the use of hepatoma cell lines as models to evaluate molecular subtype-specific drug response, which is expected to lead to development of tailored, precision care of the patients with hepatocellular carcinoma.
Collapse
|
88
|
Pillai SG, Li S, Siddappa CM, Ellis MJ, Watson MA, Aft R. Identifying biomarkers of breast cancer micrometastatic disease in bone marrow using a patient-derived xenograft mouse model. Breast Cancer Res 2018; 20:2. [PMID: 29291741 PMCID: PMC5748947 DOI: 10.1186/s13058-017-0927-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 12/12/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Disseminated tumor cells (DTCs) found in the bone marrow (BM) of patients with breast cancer portend a poor prognosis and are thought to be intermediaries in the metastatic process. To assess the clinical relevance of a mouse model for identifying possible prognostic and predictive biomarkers of these cells, we have employed patient-derived xenografts (PDX) for propagating and molecularly profiling human DTCs. METHODS Previously developed mouse xenografts from five breast cancer patients were further passaged by implantation into NOD/SCID mouse mammary fat pads. BM was collected from long bones at early, serial passages and analyzed for human-specific gene expression by qRT-PCR as a surrogate biomarker for the detection of DTCs. Microarray-based gene expression analyses were performed to compare expression profiles between primary xenografts, solid metastasis, and populations of BM DTCs. Differential patterns of gene expression were then compared to previously generated microarray data from primary human BM aspirates from patients with breast cancer and healthy volunteers. RESULTS Human-specific gene expression of SNAI1, GSC, FOXC2, KRT19, and STAM2, presumably originating from DTCs, was detected in the BM of all xenograft mice that also developed metastatic tumors. Human-specific gene expression was undetectable in the BM of those xenograft lines with no evidence of distant metastases and in non-transplanted control mice. Comparative gene expression analysis of BM DTCs versus the primary tumor of one mouse line identified multiple gene transcripts associated with epithelial-mesenchymal transition, aggressive clinical phenotype, and metastatic disease development. Sixteen of the PDX BM associated genes also demonstrated a statistically significant difference in expression in the BM of healthy volunteers versus the BM of breast cancer patients with distant metastatic disease. CONCLUSION Unique and reproducible patterns of differential gene expression can be identified that presumably originate from BM DTCs in mouse PDX lines. Several of these identified genes are also detected in the BM of patients with breast cancer who develop early metastases, which suggests that they may be clinically relevant biomarkers. The PDX model may also provide a clinically relevant system for analyzing and targeting these intermediaries of metastases.
Collapse
Affiliation(s)
- Sreeraj G. Pillai
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110 USA
| | - Shunqiang Li
- Department of Internal Medicine, Division of Medical Oncology, Washington University School of Medicine, St. Louis, MO USA
| | - Chidananda M. Siddappa
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110 USA
| | - Matthew J Ellis
- Baylor College of Medicine, Lester and Sue Smith Breast Center, Houston, TX USA
| | - Mark A. Watson
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO USA
- Siteman Cancer Center at the Washington University School of Medicine, St. Louis, MO USA
| | - Rebecca Aft
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110 USA
- Siteman Cancer Center at the Washington University School of Medicine, St. Louis, MO USA
- John Cochran Veterans Administration Hospital, St. Louis, MO USA
| |
Collapse
|
89
|
Nikitovic D, Berdiaki A, Spyridaki I, Krasanakis T, Tsatsakis A, Tzanakakis GN. Proteoglycans-Biomarkers and Targets in Cancer Therapy. Front Endocrinol (Lausanne) 2018; 9:69. [PMID: 29559954 PMCID: PMC5845539 DOI: 10.3389/fendo.2018.00069] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 02/16/2018] [Indexed: 12/18/2022] Open
Abstract
Proteoglycans (PGs), important constituents of the extracellular matrix, have been associated with cancer pathogenesis. Their unique structure consisting of a protein core and glycosaminoglycan chains endowed with fine modifications constitutes these molecules as capable cellular effectors important for homeostasis and contributing to disease progression. Indeed, differential expression of PGs and their interacting proteins has been characterized as specific for disease evolvement in various cancer types. Importantly, PGs to a large extent regulate the bioavailability of hormones, growth factors, and cytokines as well as the activation of their respective receptors which regulate phenotypic diversibility, gene expression and rates of recurrence in specific tumor types. Defining and targeting these effectors on an individual patient basis offers ground for the development of newer therapeutic approaches which may act as either supportive or a substitute treatment to the standard therapy protocols. This review discusses the roles of PGs in cancer progression, developing technologies utilized for the defining of the PG "signature" in disease, and how this may facilitate the generation of tailor-made cancer strategies.
Collapse
Affiliation(s)
- Dragana Nikitovic
- Laboratory of Anatomy-Histology-Embryology, Medical School, University of Crete, Heraklion, Greece
| | - Aikaterini Berdiaki
- Laboratory of Anatomy-Histology-Embryology, Medical School, University of Crete, Heraklion, Greece
| | - Ioanna Spyridaki
- Laboratory of Anatomy-Histology-Embryology, Medical School, University of Crete, Heraklion, Greece
| | - Theodoros Krasanakis
- Laboratory of Anatomy-Histology-Embryology, Medical School, University of Crete, Heraklion, Greece
| | - Aristidis Tsatsakis
- Laboratory of Toxicology, Medical School, University of Crete, Heraklion, Greece
| | - George N Tzanakakis
- Laboratory of Anatomy-Histology-Embryology, Medical School, University of Crete, Heraklion, Greece
| |
Collapse
|
90
|
van Sprundel RG, van den Ingh TS, Schotanus BA, van Wolferen ME, Penning LC, Rothuizen J, Spee B. Cellular characteristics of keratin 19-positive canine hepatocellular tumours explain its aggressive behaviour. Vet Rec Open 2017; 4:e000212. [PMID: 29118993 PMCID: PMC5663258 DOI: 10.1136/vetreco-2016-000212] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 08/09/2017] [Accepted: 08/14/2017] [Indexed: 01/03/2023] Open
Abstract
The expression of the hepatic progenitor cell marker keratin 19 (K19) in canine hepatocellular carcinomas is linked with a poor prognosis. To better understand this aggressive behaviour, K19-positive hepatocellular carcinomas (n=5) and K19-negative hepatocellular adenomas (n=6) were immunohistochemically stained for proteins involved in malignant tumour development. The K19-positive carcinomas showed marked positivity for platelet-derived growth factor receptor alpha polypeptide (PDGFRα), laminin, integrin beta-1/CD29, B-cell-specific Moloney murine leukaemia virus Integration site 1, glypican-3 (GPC-3) and prominin-1/CD133, in contrast with K19-negative hepatocellular adenomas. Conversely, neurofibromatosis type 2 was highly expressed in the hepatocellular adenomas in contrast with the hepatocellular carcinomas. This expression pattern is clearly in line with the observed aggressive behaviour. The presence of the malignancy markers PDGFRα and GPC-3 might make it possible to develop specific strategies to intervene in tumour growth and to devise novel serological tests and personalised treatment methods for canine hepatocellular carcinomas.
Collapse
Affiliation(s)
- Renee G van Sprundel
- Clinical Sciences of Companion Animals, Utrecht University, Utrecht, The Netherlands
| | | | - Baukje A Schotanus
- Clinical Sciences of Companion Animals, Utrecht University, Utrecht, The Netherlands
| | | | - Louis C Penning
- Clinical Sciences of Companion Animals, Utrecht University, Utrecht, The Netherlands
| | - Jan Rothuizen
- Clinical Sciences of Companion Animals, Utrecht University, Utrecht, The Netherlands
| | - Bart Spee
- Clinical Sciences of Companion Animals, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
91
|
Wu Q, Pi L, Le Trinh T, Zuo C, Xia M, Jiao Y, Hou Z, Jo S, Puszyk W, Pham K, Nelson DR, Robertson K, Ostrov D, Rameshwar P, Xia CQ, Liu C. A Novel Vaccine Targeting Glypican-3 as a Treatment for Hepatocellular Carcinoma. Mol Ther 2017; 25:2299-2308. [PMID: 28865999 DOI: 10.1016/j.ymthe.2017.08.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Revised: 08/07/2017] [Accepted: 08/07/2017] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) has a high morbidity and mortality rate worldwide, with limited treatment options. Glypican-3 (GPC3) is a glycosylphosphatidylinositol-anchored glycoprotein that is overexpressed in most HCC tissues but not in normal tissues. GPC3-targeting antibody therapy shows limited response in a clinical trial due to the lack of a tumor-specific cytotoxic T lymphocyte (CTL) response. Here, in C57/B6 mice, we demonstrated that intravenous infusion of GPC3-coupled lymphocytes (LC/GPC3+) elicited robust GPC3-specific antibody and CTL responses, which effectively restricted proliferation and lysed cultured-HCC cells. Treatment with LC/GPC3+ induced durable tumor regression in HCC-bearing C57/B6 mice. Administration of LC/GPC3+ induced elevated levels of the cytotoxic T cell bioactive factors tumor necrosis factor alpha (TNF-α), interferon-γ (IFN-γ), granzyme B, and perforin, and substantially increased the number of infiltrating CD8+ T cells in tumor tissues. Moreover, immune responses elicited by LC/GPC3+ selectively suppressed GPC3+ tumors, but didn't affect the GPC3- tumors in BALB/c mice. Our findings provide the first preclinical evidence that intravenous infusion of the LC/GPC3+ complex can induce a strong anti-HCC effect through regulating systemic and local immune responses. These results indicate that the LC/GPC3+ complex could be developed as precision therapeutics for HCC patients in the future.
Collapse
Affiliation(s)
- Qunfeng Wu
- Department of Pathology and Laboratory Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA; Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Liya Pi
- Department of Pediatrics, University of Florida, Gainesville, FL 32611, USA
| | - Thu Le Trinh
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA; Department of Immunology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Chaohui Zuo
- Department of Pathology and Laboratory Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA; Department of Gastroduodenal and Pancreatic Surgery, Translational Medicine Research Center of Liver Cancer, Hunan Province Cancer Hospital and Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan Province 410013, P.R. China
| | - Man Xia
- Department of Pathology and Laboratory Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA; Laboratory of Digestive Oncology, Hunan Province Cancer Institute, Changsha, Hunan Province 410013, P.R. China
| | - Yu Jiao
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32611, USA
| | - Zhouhua Hou
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, P.R. China
| | - Sung Jo
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - William Puszyk
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Kien Pham
- Department of Pathology and Laboratory Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA; Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - David R Nelson
- Department of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Keith Robertson
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Rochester, MN 85259, USA
| | - David Ostrov
- Department of Pathology and Laboratory Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Pranela Rameshwar
- Department of Medicine, Hematology/Oncology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Chang Qing Xia
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA.
| | - Chen Liu
- Department of Pathology and Laboratory Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA; Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
92
|
Natural killer cells in hepatocellular carcinoma: current status and perspectives for future immunotherapeutic approaches. Front Med 2017; 11:509-521. [DOI: 10.1007/s11684-017-0546-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 04/26/2017] [Indexed: 12/21/2022]
|
93
|
T Cell-Activating Mesenchymal Stem Cells as a Biotherapeutic for HCC. MOLECULAR THERAPY-ONCOLYTICS 2017; 6:69-79. [PMID: 28856237 PMCID: PMC5562179 DOI: 10.1016/j.omto.2017.07.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 07/25/2017] [Indexed: 01/10/2023]
Abstract
The outcome for advanced stage hepatocellular carcinoma (HCC) remains poor, highlighting the need for novel therapies. Genetically modified mesenchymal stem cells (MSCs) are actively being explored as cancer therapeutics due to their inherent ability to migrate to tumor sites. We reasoned that MSCs can be genetically modified to redirect T cells to Glypican-3 (GPC3)+ HCC, and genetically modified these with viral vectors encoding a GPC3/CD3 bispecific T cell engager (GPC3-ENG), a bispecifc T cell engager specific for an irrelevant antigen (EGFRvIII), and/or costimulatory molecules (CD80 and 41BBL). Coculture of GPC3+ cells, GPC3-ENG MSCs, and T cells resulted in T cell activation, as judged by interferon γ (IFNγ) production and killing of tumor cells by T cells. Modification of GPC3-ENG MSCs with CD80 and 41BBL was required for antigen-dependent interleukin-2 (IL-2) production by T cells and resulted in faster tumor cell killing by redirected T cells. In vivo, GPC3-ENG MSCs ± costimulatory molecules had antitumor activity in the HUH7 HCC xenograft model, resulting in a survival advantage. In conclusion, MSCs genetically modified to express GPC3-ENG ± costimulatory molecules redirect T cells to GPC3+ tumor cells and have potent antitumor activity. Thus, further preclinical exploration of our modified approach to GPC3-targeted immunotherapy for HCC is warranted.
Collapse
|
94
|
Zhou F, Shang W, Yu X, Tian J. Glypican-3: A promising biomarker for hepatocellular carcinoma diagnosis and treatment. Med Res Rev 2017. [PMID: 28621802 DOI: 10.1002/med.21455] [Citation(s) in RCA: 221] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Liver cancer is the second leading cause of cancer-related deaths, and hepatocellular carcinoma (HCC) is the most common type. Therefore, molecular targets are urgently required for the early detection of HCC and the development of novel therapeutic approaches. Glypican-3 (GPC3), an oncofetal proteoglycan anchored to the cell membrane, is normally detected in the fetal liver but not in the healthy adult liver. However, in HCC patients, GPC3 is overexpressed at both the gene and protein levels, and its expression predicts a poor prognosis. Mechanistic studies have revealed that GPC3 functions in HCC progression by binding to molecules such as Wnt signaling proteins and growth factors. Moreover, GPC3 has been used as a target for molecular imaging and therapeutic intervention in HCC. To date, GPC3-targeted magnetic resonance imaging, positron emission tomography, and near-infrared imaging have been investigated for early HCC detection, and various immunotherapeutic protocols targeting GPC3 have been developed, including the use of humanized anti-GPC3 cytotoxic antibodies, treatment with peptide/DNA vaccines, immunotoxin therapies, and genetic therapies. In this review, we summarize the current knowledge regarding the structure, function, and biology of GPC3 with a focus on its clinical potential as a diagnostic molecule and a therapeutic target in HCC immunotherapy.
Collapse
Affiliation(s)
- Fubo Zhou
- Department of Interventional Ultrasound, Chinese PLA General Hospital, Beijing, 100853, China
| | - Wenting Shang
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Xiaoling Yu
- Department of Interventional Ultrasound, Chinese PLA General Hospital, Beijing, 100853, China
| | - Jie Tian
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| |
Collapse
|
95
|
Vongchan P, Linhardt RJ. Characterization of a new monoclonal anti-glypican-3 antibody specific to the hepatocellular carcinoma cell line, HepG2. World J Hepatol 2017; 9:368-384. [PMID: 28321273 PMCID: PMC5340992 DOI: 10.4254/wjh.v9.i7.368] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 12/19/2016] [Accepted: 01/14/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To characterize the antigen on HepG2 cell that is specifically recognized by a new monoclonal antibody raised against human liver heparan sulfate proteoglycan (HSPG), clone 1E4-1D9.
METHODS The antigen recognized by mAb 1E4-1D9 was immunoprecipitated and its amino acid sequence was analyzed LC/MS. The transmembrane domain, number of cysteine residues, and glycosylation sites were predicted from these entire sequences. Data from amino acid analysis was aligned with glypican-3 (https://www.ebi.ac.uk/Tools/msa/clustalo/). The competitive reaction of mAb 1E4-1D9 and anti-glypican-3 on HepG2 cells was demonstrated by indirect immunofluorescence and analyzed by flow cytometry. Moreover, co-immunoprecipitation of mAb 1E4-1D9 and anti-glypican-3 was performed in HepG2 cells by Western immunoblotting. The recognition by mAb 1E4-1D9 of a specific epitope on solid tumor and hematopoietic cell lines was studied using indirect immunofluorescence and analyzed by flow cytometry.
RESULTS Monoclonal antibody 1E4-1D9 reacted with an HSPG isolated from human liver and a band of 67 kD was detected under both reducing and non-reducing conditions. The specific antigen pulled down by mAb 1E4-1D9, having a MW of 135 kD, was analyzed. The results showed two sequences of interest, gi30722350 (1478 amino acid) and gi60219551 (1378 amino acid). In both sequences no transmembrane regions were observed. Sequence number gi30722350 was 99.7% showed a match to FYCO1, a molecule involved in induction of autophagy. Sequence number gi60219551 contained 15 cysteines and 11 putative glycosylation sites with 6 predicted N-glycosylation sites. It was also matched with all PDZ domain proteins. Moreover, it showed an 85.7% match to glypican-3. Glypican-3 on HepG2 cells competitively reacted with both phycoerythrin-conjugated anti-glypican-3 and mAb 1E4-1C2 and resulted in an increase of double-stained cell population when higher concentration of mAb 1E4-1D9 was used. Moreover, antigens precipitated from HepG2 cell by anti-glypican-3 could be detected by mAb 1E4-1D9 and vice versa. The recognition of antigens, on other solid tumor cell lines, by mAb 1E4-1D9 was studied. The results demonstrated that mAb 1E4-1D9 reacted with Huh7, HepG2, HT29, MCF7, SW620, Caco2, B16F1, U937, K562 and Molt4 cells. It was also found to be weakly positive to SW1353 and HL60 and negative to H460 and Hela cell lines.
CONCLUSION All findings show that mAb 1E4-1D9 specifically recognizes glypican-3. Moreover, a new partner molecule of glypican-3, FYCO1 is proposed based on the results from co-precipitation studies.
Collapse
|
96
|
Abou-Alfa GK, Yen CJ, Hsu CH, O'Donoghue J, Beylergil V, Ruan S, Pandit-Taskar N, Gansukh B, Lyashchenko SK, Ma J, Wan P, Shao YY, Lin ZZ, Frenette C, O'Neil B, Schwartz L, Smith-Jones PM, Ohtomo T, Tanaka T, Morikawa H, Maki Y, Ohishi N, Chen YC, Agajanov T, Boisserie F, Di Laurenzio L, Lee R, Larson SM, Cheng AL, Carrasquilo JA. Phase Ib study of codrituzumab in combination with sorafenib in patients with non-curable advanced hepatocellular carcinoma (HCC). Cancer Chemother Pharmacol 2017; 79:421-429. [PMID: 28120036 PMCID: PMC5548107 DOI: 10.1007/s00280-017-3241-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 01/07/2017] [Indexed: 11/26/2022]
Abstract
PURPOSE Codrituzumab, a humanized antibody against glypican-3, is highly expressed in HCC. A phase I study evaluated the combination with sorafenib in HCC. PATIENTS AND METHODS In a 3 + 3 design, codrituzumab was given intravenously in various doses with sorafenib 400 mg twice daily to patients with advanced HCC, age ≥18, ECOG 0-1, Child-Pugh A and B7, adequate organ functions, and no prior systemic therapy, with tumor assessment by RECIST 1.0 and safety by CTCAE 3.0. PK and pre, during, and post-therapy 124I radiolabeled codrituzumab PET scan imaging were performed. RESULTS 41 patients were enrolled: 2.5 mg/kg weekly (qw) (12), 5 mg/kg qw (12), 10 mg/kg qw (3), 1600 mg every 2 weeks (q2w) (6), and 1600 mg qw (7). Two drug limiting toxicities occurred: grade 3 hyponatremia at 5 mg/kg and grade 3 hyponatremia and hyperglycemia at 1600 mg q2w. Adverse events occurred in 80% of patients, including at least one ≥grade 3: ten (25%) increased AST, three (7.5%) increased ALT, and ten (25%) increased lipase. There were no responses and nine (25.7%) had stable disease. PK C max and AUCt of codrituzumab and sorafenib were comparable to single-agent data. Thirteen out of 14 patients showed 124I radiolabeled codrituzumab uptake in tumor. In all three patients who underwent a post-progression PET, glypican-3 remained expressed. CONCLUSION Codrituzumab plus sorafenib were tolerated at 1600 mg q2w and 400 mg bid, respectively, with no responses. Codrituzumab exerts selective distribution to HCC cells, and GPC3 does not show any down-regulation post-progression (NCT00976170).
Collapse
Affiliation(s)
- Ghassan K Abou-Alfa
- Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
- Weill Cornell Medical College, New York, NY, USA.
| | - Chia-Jui Yen
- National Cheng-Kung University Hospital, Tainan, Taiwan, Republic of China
| | - Chih-Hung Hsu
- National Taiwan University Hospital, Taipei, Taiwan, Republic of China
| | | | - Volkan Beylergil
- Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Shutian Ruan
- Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | | | | | | | - Jennifer Ma
- Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Peter Wan
- Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Yu-Yun Shao
- National Taiwan University Hospital, Taipei, Taiwan, Republic of China
| | - Zhong-Zhe Lin
- National Taiwan University Hospital, Taipei, Taiwan, Republic of China
| | | | - Bert O'Neil
- Indiana University Simon Cancer Center, Indianapolis, IN, USA
| | | | | | | | | | | | - Yuko Maki
- Chugai Pharmaceutical Co. Ltd., Tokyo, Japan
| | | | - Ya-Chi Chen
- Translational and Clinical Research Center, Hoffmann-La Roche Inc., New York, USA
| | - Tamara Agajanov
- Translational and Clinical Research Center, Hoffmann-La Roche Inc., New York, USA
| | - Frederic Boisserie
- Translational and Clinical Research Center, Hoffmann-La Roche Inc., New York, USA
| | - Laura Di Laurenzio
- Translational and Clinical Research Center, Hoffmann-La Roche Inc., New York, USA
| | - Ray Lee
- Translational and Clinical Research Center, Hoffmann-La Roche Inc., New York, USA
| | - Steven M Larson
- Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Ann-Lii Cheng
- National Taiwan University Hospital, Taipei, Taiwan, Republic of China
| | - Jorge A Carrasquilo
- Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
97
|
Montalbano M, Georgiadis J, Masterson AL, McGuire JT, Prajapati J, Shirafkan A, Rastellini C, Cicalese L. Biology and function of glypican-3 as a candidate for early cancerous transformation of hepatocytes in hepatocellular carcinoma (Review). Oncol Rep 2017; 37:1291-1300. [PMID: 28098909 DOI: 10.3892/or.2017.5387] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 01/12/2017] [Indexed: 12/17/2022] Open
Abstract
Glypican-3 (GPC-3), a transmembrane heparan sulfate proteoglycan (HSPG), has recently been investigated as a player in tissue-dependent cellular signaling, specifically as a regulator of growth. Noteworthy, the regulatory protein has been implicated in both stimulatory and inhibitory pathways involving cell growth. Initially, GPC-3 was thought to act as a cell cycle regulator, as a loss-of-function mutation in the gene caused a hyper-proliferative state known as Simpson-Golabi-Behmel (SGB) overgrowth syndrome. Additionally, certain cancer types have displayed a downregulation of GPC-3 expression. More recently, the protein has been evaluated as a useful marker for hepatocellular carcinoma (HCC) due to its increased expression in the liver during times of growth. In contrast, the GPC-3 marker is not detectable in normal adult liver. Immunotherapy that targets GPC-3 and its affiliated proteins is under investigation as these new biomarkers may hold potential for the detection and treatment of HCC and other diseases in which GPC-3 may be overexpressed. Studies have reported that an overexpression of GPC-3 in HCC predicts a poorer prognosis. This prognostic value further pushes the question regarding GPC-3's role in the regulation and progression of HCC. This review will summarize the current knowledge regarding the clinical aspects of GPC-3, while also synthesizing the current literature with the aim to better understand this molecule's biological interactions at a molecular level, not only in the liver, but in the rest of the body as well. Due to the existing gap in the literature surrounding GPC-3, we believe further investigation of function, structure and domains, cellular localization, and other subfields is warranted to evaluate the protein as a whole, as well as its part in the study of HCC.
Collapse
Affiliation(s)
- Mauro Montalbano
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jeremias Georgiadis
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Ashlyn L Masterson
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Joshua T McGuire
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Janika Prajapati
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Ali Shirafkan
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Cristiana Rastellini
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Luca Cicalese
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
98
|
Suh HY, Peck CC, Yu KS, Lee H. Determination of the starting dose in the first-in-human clinical trials with monoclonal antibodies: a systematic review of papers published between 1990 and 2013. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:4005-4016. [PMID: 27994442 PMCID: PMC5153257 DOI: 10.2147/dddt.s121520] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A systematic review was performed to evaluate how the maximum recommended starting dose (MRSD) was determined in first-in-human (FIH) studies with monoclonal antibodies (mAbs). Factors associated with the choice of each MRSD determination method were also identified. PubMed was searched for FIH studies with mAbs published in English between January 1, 1990 and December 31, 2013, and the following information was extracted: MRSD determination method, publication year, therapeutic area, antibody type, safety factor, safety assessment results after the first dose, and number of dose escalation steps. Seventy-nine FIH studies with mAbs were identified, 49 of which clearly reported the MRSD determination method. The no observed adverse effects level (NOAEL)-based approach was the most frequently used method, whereas the model-based approach was the least commonly used method (34.7% vs 16.3%). The minimal anticipated biological effect level (MABEL)- or minimum effective dose (MED)-based approach was used more frequently in 2011–2013 than in 1990–2007 (31.6% vs 6.3%, P=0.036), reflecting a slow, but steady acceptance of the European Medicines Agency’s guidance on mitigating risks for FIH clinical trials (2007). The median safety factor was much lower for the MABEL- or MED-based approach than for the other MRSD determination methods (10 vs 32.2–53). The number of dose escalation steps was not significantly different among the different MRSD determination methods. The MABEL-based approach appears to be safer and as efficient as the other MRSD determination methods for achieving the objectives of FIH studies with mAbs faster.
Collapse
Affiliation(s)
- Hoon Young Suh
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Carl C Peck
- Department of Bioengineering and Therapeutic Sciences, School of Pharmacy, University of California, San Francisco, CA, USA
| | - Kyung-Sang Yu
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Howard Lee
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Seoul National University Hospital, Seoul, Korea; Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| |
Collapse
|
99
|
Goossens N, Hoshida Y. Tratamiento personalizado del carcinoma hepatocelular basado en información molecular: perspectivas futuras. Clin Liver Dis (Hoboken) 2016; 8:S43-S48. [PMID: 31041096 PMCID: PMC6490229 DOI: 10.1002/cld.601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Affiliation(s)
- Nicolas Goossens
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNY, EE. UU,Division of Gastroenterology and HepatologyGeneva University HospitalGinebraSuiza
| | - Yujin Hoshida
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNY, EE. UU
| |
Collapse
|
100
|
Abstract
Glypican-3 (GPC3), a member of heparan sulfate proteoglycans, attaches to the cell membrane and is frequently observed to be elevated in hepatocellular carcinoma (HCC). However, GPC3 is not detected in normal liver tissues and benign liver lesions. Consequently, GPC3 is currently being used as a diagnostic biomarker and HCC-specific positron emission computed tomography probe to identify HCCs in normal liver tissues and benign liver lesions. The overexpression of GPC-3 in serum or liver tissue also predicts poor prognosis for HCC patients. In addition, GPC3 promotes HCC growth and metastasis by activating the canonical Wnt and other signaling pathways. Targeting of GPC3, including GC33, HN3 and YP7, might offer new immunotherapeutic tools for HCC treatment.
Collapse
Affiliation(s)
- Yongle Wu
- Department of Gastroenterology and Hepatology
| | - Hui Liu
- Department of Pathology, Beijing You'an Hospital, Affiliated with Capital Medical University, Beijing, People's Republic of China
| | - Huiguo Ding
- Department of Gastroenterology and Hepatology
| |
Collapse
|