1
|
Gallery M, Zhang J, Bradley DP, Brauer P, Cvet D, Estevam J, Danaee H, Greenfield E, Li P, Manfredi M, Loke HK, Rabino C, Stringer B, Williamson M, Wyant T, Yang J, Zhu Q, Abu-Yousif A, Veiby OP. A monomethyl auristatin E-conjugated antibody to guanylyl cyclase C is cytotoxic to target-expressing cells in vitro and in vivo. PLoS One 2018; 13:e0191046. [PMID: 29370189 PMCID: PMC5784926 DOI: 10.1371/journal.pone.0191046] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 12/27/2017] [Indexed: 12/15/2022] Open
Abstract
Guanylyl cyclase C (GCC) is a cell-surface protein that is expressed by normal intestinal epithelial cells, more than 95% of metastatic colorectal cancers (mCRC), and the majority of gastric and pancreatic cancers. Due to strict apical localization, systemically delivered GCC-targeting agents should not reach GCC in normal intestinal tissue, while accessing antigen in tumor. We generated an investigational antibody-drug conjugate (TAK-264, formerly MLN0264) comprising a fully human anti-GCC monoclonal antibody conjugated to monomethyl auristatin E via a protease-cleavable peptide linker. TAK-264 specifically bound, was internalized by, and killed GCC-expressing cells in vitro in an antigen-density-dependent manner. In GCC-expressing xenograft models with similar GCC expression levels/patterns observed in human mCRC samples, TAK-264 induced cell death, leading to tumor regressions and long-term tumor growth inhibition. TAK-264 antitumor activity was generally antigen-density-dependent, although some GCC-expressing tumors were refractory to TAK-264-targeted high local concentrations of payload. These data support further evaluation of TAK-264 in the treatment of GCC-expressing tumors.
Collapse
Affiliation(s)
- Melissa Gallery
- Molecular & Cellular Oncology, Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, United States of America
| | - Julie Zhang
- Cancer Pharmacology, Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, United States of America
| | - Daniel P Bradley
- Biomedical Imaging, Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, United States of America
| | - Pamela Brauer
- Protein Sciences, Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, United States of America
| | - Donna Cvet
- Biomedical Imaging, Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, United States of America
| | - Jose Estevam
- Biomarker Assay & Exploratory Biology, Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, United States of America
| | - Hadi Danaee
- Biomarker Assay & Exploratory Biology, Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, United States of America
| | - Edward Greenfield
- Protein Sciences, Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, United States of America
| | - Ping Li
- Protein Sciences, Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, United States of America
| | - Mark Manfredi
- Cancer Pharmacology, Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, United States of America
| | - Huay-Keng Loke
- Oncology Biochemistry, Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, United States of America
| | - Claudia Rabino
- Molecular & Cellular Oncology, Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, United States of America
| | - Brad Stringer
- Molecular Pathology, Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, United States of America
| | - Mark Williamson
- US Medical Affairs, Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, United States of America
| | - Tim Wyant
- Translational Medicine, Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, United States of America
| | - Johnny Yang
- DMPK, Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, United States of America
| | - Qing Zhu
- DMPK, Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, United States of America
| | - Adnan Abu-Yousif
- Cancer Pharmacology, Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, United States of America
| | - O Petter Veiby
- Global Biotherapeutics, Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, United States of America
| |
Collapse
|
2
|
Almhanna K, Miron MLL, Wright D, Gracian AC, Hubner RA, Van Laethem JL, López CM, Alsina M, Muñoz FL, Bendell J, Firdaus I, Messersmith W, Ye Z, Fasanmade AA, Danaee H, Kalebic T. Phase II study of the antibody-drug conjugate TAK-264 (MLN0264) in patients with metastatic or recurrent adenocarcinoma of the stomach or gastroesophageal junction expressing guanylyl cyclase C. Invest New Drugs 2017; 35:235-241. [PMID: 28188407 DOI: 10.1007/s10637-017-0439-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 01/30/2017] [Indexed: 01/08/2023]
Abstract
Background The first-in-class antibody-drug conjugate TAK-264 (formerly MLN0264) consists of an antibody targeting guanylyl cyclase C (GCC) conjugated to monomethyl auristatin E (MMAE) via a peptide linker. This phase II study evaluated the efficacy and safety of TAK-264 in patients with adenocarcinoma of the stomach or gastroesophageal junction expressing GCC, who had progressed on ≥1 line of prior therapy. Methods This study used a two-stage design, with an interim analysis conducted after stage I to determine whether to continue to stage II or discontinue on the grounds of futility. Adult patients with gastric and gastroesophageal junction adenocarcinoma expressing low, intermediate, or high GCC levels received TAK-264 1.8 mg/kg as a 30-min intravenous infusion once every 21 days, for up to 1 year. The primary endpoint was objective response rate. Radiographic assessments of tumor burden were performed every 2 cycles (6 weeks). Results A total of 38 patients participated in the study. Patients received a median of 2 (range 1-14) cycles; 8 (21%) received at least 6 cycles. The most common adverse events were nausea (53%), fatigue (32%), and decreased appetite (29%). Grade ≥3 events including anemia, diarrhea, and neutropenia were seen in 14 (37%) patients. Systemic exposure to TAK-264 was maintained throughout each treatment cycle. Two patients (6%) with intermediate GCC expression had objective responses. Conclusions TAK-264 demonstrated a manageable safety profile in this patient population. The stage I interim analysis did not support continuation to stage II of the study.
Collapse
Affiliation(s)
- Khaldoun Almhanna
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, USA.
| | | | | | | | | | | | | | - Maria Alsina
- Vall d'Hebron University Hospital and VHIO-Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | | | - Johanna Bendell
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN, USA
| | | | | | - Zhan Ye
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, USA
| | - Adedigbo A Fasanmade
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, USA
| | - Hadi Danaee
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, USA
| | - Thea Kalebic
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, USA
| |
Collapse
|
3
|
Almhanna K, Prithviraj GK, Veiby P, Kalebic T. Antibody-drug conjugate directed against the guanylyl cyclase antigen for the treatment of gastrointestinal malignancies. Pharmacol Ther 2016; 170:8-13. [PMID: 27765652 DOI: 10.1016/j.pharmthera.2016.10.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Antibody-directed cancer chemotherapy in the form of antibody-drug conjugates (ADCs) may improve the therapeutic index with the potential to enhance efficacy and decrease systemic toxicity. ADCs consist of three key components including an antibody that specifically binds to the target, a toxic agent and a linker which releases the toxic agent inside tumor cells. A novel ADC, MLN0264 (TAK-264) was recently investigated in patients with gastrointestinal (GI) malignancies. TAK-264 is an anti- guanylyl cyclase C (GCC) antibody conjugated via a protease-cleavable linker to the potent anti-microtubule agent monomethyl auristatin E (MMAE) (linker and toxin licensed from Seattle Genetics). Following binding to GCC, the ADC is internalized and transported to lysosomes where MMAE is released to bind to tubulin, leading to cell cycle arrest and apoptosis. This GCC targeting ADC has been evaluated in clinical studies in patients with advanced gastrointestinal malignancies. The early findings from Phase 1 study have shown preliminary activity signals in gastric, gastroesophageal, and pancreatic cancer. Results from two phase II studies in pancreatic and gastoesophageal adenocarcinoma showed only limited activity. Antibody-drug-conjugates offer a promising therapeutic modality aimed at providing target-directed cancer chemotherapy. Herein we discuss the GCC target and gastrointestinal malignancies where GCC based targeted therapies could further evolve and offer a significant clinical benefit.
Collapse
Affiliation(s)
- Khaldoun Almhanna
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, FOB-2, Tampa, FL 334612, USA.
| | - Gopi K Prithviraj
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, FOB-2, Tampa, FL 334612, USA
| | - Petter Veiby
- Biotherapeutics Discovery, Takeda Pharmaceuticals International, Co 35 Landsdowne Street, Cambridge, MA 02139, USA
| | - Thea Kalebic
- Clinical Research, Takeda Pharmaceuticals International, Oncology Co 35 Landsdowne Street, Cambridge, MA 02139, USA
| |
Collapse
|
4
|
Zhang J, Kuai X, Song M, Chen X, Yu Z, Zhang H, Mao Z. microRNA-32 inhibits the proliferation and invasion of the SGC-7901 gastric cancer cell line in vitro.. Oncol Lett 2013; 7:270-274. [PMID: 24348862 PMCID: PMC3861597 DOI: 10.3892/ol.2013.1667] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2013] [Accepted: 10/24/2013] [Indexed: 01/07/2023] Open
Abstract
microRNAs (miRNAs) are a class of endogenously expressed, small non-coding RNAs, which suppress their target mRNAs at the post-transcriptional level. miRNAs play key roles in tumor metastasis. The aim of the present study was to investigate the expression of miRNA-32 (miR-32) on the biological behavior of the human gastric cancer cell line, SGC-7901. SGC-7901 cells were transfected with miR-32-mimic, miR-32-inhibitor and empty plasmid vectors using Lipofectamine™ 2000. The expression of GFP was observed by fluorescent microscopy and miR-32 gene expression was detected by quantitative polymerase chain reaction. The cell counting kit-8 assay was performed to evaluate the effect of miR-32 expression on cell proliferation in vitro. Alterations in the migration and metastatic potential of SGC-7901 cells, prior to and following miR-32 gene transfection, were assayed by cell chemotactic migration and invasion tests. The results of the current study showed that the proliferation rate of the transfected SGC-7901 cells overexpressing miR-32 is reduced and cell chemotactic migration and invasion potentials is markedly reduced following miR-32-mimic transfection (P<0.05). In addition, the results demonstrated that overexpression of miR-32 greatly inhibits the proliferation and decreases the migration and invasion capabilities of SGC-7901 cells in vitro.
Collapse
Affiliation(s)
- Jianfeng Zhang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Xiaoling Kuai
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Mengjiao Song
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Xiaoqi Chen
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Zhihua Yu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Hong Zhang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Zhenbiao Mao
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
5
|
Zhang JF, Zhang JG, Kuai XL, Zhang H, Jiang W, Ding WF, Li ZL, Zhu HJ, Mao ZB. Reactivation of the homeotic tumor suppressor gene CDX2 by 5-aza-2'-deoxycytidine-induced demethylation inhibits cell proliferation and induces caspase-independent apoptosis in gastric cancer cells. Exp Ther Med 2013; 5:735-741. [PMID: 23408490 PMCID: PMC3570199 DOI: 10.3892/etm.2013.901] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Accepted: 01/10/2013] [Indexed: 12/17/2022] Open
Abstract
The DNA methylation inhibitor 5-aza-2′-deoxycytidine (5-aza-CdR) is widely used as an anticancer drug for the treatment of leukemia and solid tumors. Gastric cancer (GC) patients who were positive for caudal type homeobox transcription factor 2 (CDX2) expression showed a higher survival rate compared with those who were CDX2 negative, which suggests that CDX2 performs a tumor suppressor role. However, the molecular mechanisms leading to the inactivation of CDX2 remain unclear. In the present study we demonstrated that the expression levels of CDX2 and DNA methyltransferase enzyme 1 (DNMT1) mRNA were significantly higher in GC compared with distal non-cancerous tissue. The expression of CDX2 mRNA was significantly correlated with Lauren classification, TNM stage and lymph node metastasis. DNMT1 mRNA expression was significantly correlated with TNM stage, pathological differentiation and lymph node metastasis. The expression of CDX2 mRNA was inversely correlated with that of DNMT1 mRNA in GC. Hypermethylation of the CDX2 gene promoter region, extremely low expression levels of CDX2 mRNA and no expression of CDX2 protein were the characteristics observed in MKN-45 and SGC-7901 GC cell lines. Following the treatment of MKN-45 cells with 5-aza-CdR, the hypermethylated CDX2 gene promoter region was demethylated and expression of CDX2 was upregulated, while DNMT1 expression was downregulated. Furthermore, a concentration- and time-dependent growth inhibition as well as increased apoptosis were observed. Caspase-3, −8 and −9 activities increased in a concentration-dependent manner following exposure to different concentrations of 5-aza-CdR. Therefore, our data show that the overexpression of DNMT1 and methylation of the CDX2 gene promoter region is likely to be responsible for CDX2 silencing in GC. 5-Aza-CdR may effectively induce re-expression of the CDX2 gene, inhibit cell proliferation and enhance the caspase-independent apoptosis of MKN-45 cells in vitro.
Collapse
Affiliation(s)
- Jian-Feng Zhang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Meng H, Zhang JF, Mao ZB, Xue SM, Wu XH, Ju SQ. GC-C gene silencing inhibits the growth of subcutaneously implanted gastric tumors in nude mice. Shijie Huaren Xiaohua Zazhi 2012; 20:1919-1926. [DOI: 10.11569/wcjd.v20.i21.1919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To study the effect of GC-C gene silencing on the biological behaviors of subcutaneously implanted gastric tumors and to explore the possible mechanisms involved.
METHODS: Non-transfected SGC-7901 cells (SGC-7901 group), SGC-7901 cells transfected cell with an empty plasmid (PRNA group), and those transfected with a shRNA targeting the GC-C gene (GC-C-shRNA group) were subcutaneously inoculated into nude mice. Then, the histologically intact xenograft tissues were subcutaneously transplanted to nude mice to observe the condition of nude mice and determine the rate of tumor survival and growth velocity. The levels of GC-C mRNA and protein were determined by RFQ-PCR and Western blot, respectively. The heteromorphism and necrosis of tumor cells were observed after HE staining. The expression of CXCR4 protein was detected by Western blot and immunohistochemistry.
RESULTS: The rate of tumor survival and the condition of nude mice did not differ significantly among the three groups. Compared to the SGC-7901 group and PRNA group, the tumors grew more slowly and became smaller in the GC-C-shRNA group (all P < 0.05). The reduced rate of tumor growth velocity and volume in the GC-C-shRNA group were 33.7% and 33.2%, respectively, and GC-C gene silencing efficiency was 66.8% and 64.4%, respectively (P < 0.05). The expression of GC-C protein decreased by 50.0% and 49.6%, respectively (P < 0.05), and the degree of tumor heteromorphism and necrosis and expression of CXCR4 protein in the GC-C-shRNA group decreased obviously (all P < 0.05).
CONCLUSION: GC-C gene silencing slows the growth velocity of gastric tumors in vivo possibly in a CXCR4-dependent manner.
Collapse
|
7
|
Zhang JF, Mao ZB, Li ZL, Xue SM, Zhu HJ, Zhang H, Ni RZ. Ectopic expression of guanylyl cyclase C and endogenous ligand guanylin correlates significantly with Helicobacter pylori infection in gastric carcinogenesis. Med Oncol 2011; 29:1748-57. [PMID: 21972003 DOI: 10.1007/s12032-011-0067-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2011] [Accepted: 09/08/2011] [Indexed: 12/23/2022]
Abstract
The molecular mechanisms leading to gastric carcinogenesis still remain unclear. Recently, several studies demonstrated that over-expression of guanylyl cyclase C (GCC) has been detected in intestinal-type gastric cancer (GC) and precursor lesions. Our objective was to explore the expression levels of GCC and endogenous ligands guanylin (GN) and uroguanylin (UGN) and the correlation between Helicobacter pylori (H. pylori) and GCC, GN, and UGN expressions in patients at different stages from normal mucosa to superficial gastritis, atrophic gastritis, intestinal metaplasia (IM), dysplasia, and finally adenocarcinoma. The expression of GCC and GN was absent in the distal normal gastric tissues and superficial gastritis in all cases, whereas they were measured in IM, dysplasia, and GC. The expression of GCC and GN was closely related to intestinal-type GC. From superficial gastritis to gastric carcinomas, the H. pylori positive rate was 19.7, 33.3, 69.6, 80.0, and 82.1%, respectively. The positive correlation was found between GCC and GN in IM, dysplasia, and GC. Also, the positive correlation was found between GCC, GN, and H. pylori infection in them. These results demonstrate that the detection of GCC and GN will be beneficial to diagnosis human gastric carcinoma and precancerous lesions. Ectopic expression of GCC and GN in human gastric mucosa and H. pylori infection may play an important role in the carcinogenesis of the intestinal-type GC.
Collapse
Affiliation(s)
- Jian-Feng Zhang
- Department of Gastroenterology, Affiliated Hospital, Nantong University, 226001 Nantong, People's Republic of China.
| | | | | | | | | | | | | |
Collapse
|
8
|
Praharaj S, Overbey D, Giblin MF. Radiometallated peptides targeting guanylate cyclase C and the urokinase-type plasminogen activator receptor. Future Oncol 2010; 6:1325-37. [DOI: 10.2217/fon.10.91] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Research is currently underway worldwide into the development of receptor-specific radiopharmaceuticals for the imaging and treatment of cancer. The successful clinical development of radiolabeled somatostatin analogs for imaging and treatment of cancers overexpressing somatostatin receptors has catalyzed further preclinical investigation of other radiolabeled peptides for molecular imaging and peptide-receptor radiotherapy, including such well-studied peptide vectors as cholecystokinin, neurotensin, bombesin and RGD peptides. Within this larger context, this article will focus on the current status of two more recent additions to the list of molecular imaging targets – guanylate cyclase C, a specific marker for colorectal cancer, and the urokinase plasminogen activator receptor, a cell-surface receptor overexpressed in diverse cancer types.
Collapse
Affiliation(s)
- Snigdha Praharaj
- Harry S Truman Memorial Veterans’ Administration Hospital, Research Service, A004, 800 Hospital Drive, Columbia, MO 6520, USA
- Radiopharmaceutical Sciences Institute, Department of Radiology, University of Missouri-Columbia, MO, USA
| | - Douglas Overbey
- Harry S Truman Memorial Veterans’ Administration Hospital, Research Service, A004, 800 Hospital Drive, Columbia, MO 6520, USA
- University of Missouri-Columbia, MO, USA
| | | |
Collapse
|