1
|
Qian J, Wang Q, Xiao L, Xiong W, Xian M, Su P, Yang M, Zhang C, Li Y, Zhong L, Ganguly S, Zu Y, Yi Q. Development of therapeutic monoclonal antibodies against DKK1 peptide-HLA-A2 complex to treat human cancers. J Immunother Cancer 2024; 12:e008145. [PMID: 38267222 PMCID: PMC10824003 DOI: 10.1136/jitc-2023-008145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2024] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND Targeted immunotherapy with monoclonal antibodies (mAbs) is an effective and safe method for the treatment of malignancies. Development of mAbs with improved cytotoxicity, targeting new and known tumor-associated antigens, therefore continues to be an active research area. We reported that Dickkopf-1 (DKK1) is a good target for immunotherapy of human cancers based on its wide expression in different cancers but not in normal tissues. As DKK1 is a secreted protein, mAbs binding directly to DKK1 have limited effects on cancer cells in vivo. METHODS The specificity and antibody-binding capacity of DKK1-A2 mAbs were determined using indirect ELISA, confocal imaging, QIFIKIT antibody-binding capacity and cell surface binding assays. The affinity of mAbs was determined using a surface plasmon resonance biosensor. A flow cytometry-based cell death was performed to detect tumor cell apoptosis. Antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) assays were used to evaluate the ability of DKK1-A2 mAbs to mediate ADCC and CDC activities against tumor cells in vitro. Flow cytometry data were collected with an FACSymphony A3 cell analyzer and analyzed with FlowJo V.10.1 software. Human cancer xenograft mouse models were used to determine the in vivo therapeutic efficacy and the potential safety and toxicity of DKK1-A2 mAbs. In situ TUNEL assay was performed to detect apoptosis in tumors and mouse organs. RESULTS We generated novel DKK1-A2 mAbs that recognize the DKK1 P20 peptide presented by human HLA-A*0201 (HLA-A2) molecules (DKK1-A2 complexes) that are naturally expressed by HLA-A2+DKK1+ cancer cells. These mAbs directly induced apoptosis in HLA-A2+DKK1+ hematologic and solid cancer cells by activating the caspase-9 cascade, effectively lysed the cancer cells in vitro by mediating CDC and ADCC and were therapeutic against established cancers in their xenograft mouse models. As DKK1 is not detected in most human tissues, DKK1-A2 mAbs neither bound to or killed HLA-A2+ blood cells in vitro nor caused tissue damage in tumor-free or tumor-bearing HLA-A2-transgenic mice. CONCLUSION Our study suggests that DKK1-A2 mAbs may be a promising therapeutic agent to treat human cancers.
Collapse
Affiliation(s)
- Jianfei Qian
- Center for Translational Research in Hematological Malignancies, Houston Methodist Neal Cancer Center/Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Qiang Wang
- Center for Translational Research in Hematological Malignancies, Houston Methodist Neal Cancer Center/Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Liuling Xiao
- Center for Translational Research in Hematological Malignancies, Houston Methodist Neal Cancer Center/Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Wei Xiong
- Center for Translational Research in Hematological Malignancies, Houston Methodist Neal Cancer Center/Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Miao Xian
- Center for Translational Research in Hematological Malignancies, Houston Methodist Neal Cancer Center/Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Pan Su
- Center for Translational Research in Hematological Malignancies, Houston Methodist Neal Cancer Center/Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Maojie Yang
- Center for Translational Research in Hematological Malignancies, Houston Methodist Neal Cancer Center/Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Chuanchao Zhang
- Center for Translational Research in Hematological Malignancies, Houston Methodist Neal Cancer Center/Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Yabo Li
- Center for Translational Research in Hematological Malignancies, Houston Methodist Neal Cancer Center/Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Ling Zhong
- Center for Translational Research in Hematological Malignancies, Houston Methodist Neal Cancer Center/Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Siddhartha Ganguly
- Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, USA
| | - Youli Zu
- Department of Pathology and Genomic Medicine, Institute for Academic Medicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Qing Yi
- Center for Translational Research in Hematological Malignancies, Houston Methodist Neal Cancer Center/Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA
| |
Collapse
|
2
|
Urata S, Yamaguchi S, Nambu A, Sudo K, Nakae S, Yasuda J. The roles of BST-2 in murine B cell development and on virus propagation. Microbiol Immunol 2023; 67:105-113. [PMID: 36604771 DOI: 10.1111/1348-0421.13049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 12/13/2022] [Accepted: 01/04/2023] [Indexed: 01/07/2023]
Abstract
The bone marrow (BM) stromal cell antigen-2 (BST-2), also known as tetherin, CD317, PDCA-1, or HM1.24, is a membrane protein overexpressed in several types of tumors and may act as a promising target for cancer treatment via antibody-dependent cellular cytotoxicity. BST-2 is also expressed in human BM stromal cells (BMSC), which support B cell development. While the activity of BST-2 as an antiviral factor has been demonstrated, the expression patterns and the role of BST-2 on B-cell development and activation have not been investigated, especially in vivo. In this study, Bst2 knockout (Bst2-/- ) mice were generated to assess the role of BST-2 on B cell development and activation. It was observed that BST-2 was not expressed in BMSC or all B cell progenitors even in wild-type mice and does not play a significant role in B cell development. In addition, the loss of BST-2 had no effect on B cell activation. Furthermore and in contrast to the well-known antiviral role of BST-2, infection of vesicular stomatitis Indiana virus to the BM cells collected from the Bst2-/- mice produced less infectious virus compared with that from the WT mice. These results suggest that murine BST-2 is different from human BST-2 in the expression pattern, physiological function, in vivo, and might possess positive role on VSV replication.
Collapse
Affiliation(s)
- Shuzo Urata
- Department of Emerging Infectious Diseases, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan.,National Research Center for the Control and Prevention of Infectious Diseases (CCPID), Nagasaki University, Nagasaki, Japan
| | - Sachiko Yamaguchi
- Laboratory of Systems Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Aya Nambu
- Laboratory of Systems Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Katsuko Sudo
- Pre-clinical Research Center, Tokyo Medical University, Tokyo, Japan
| | - Susumu Nakae
- Laboratory of Systems Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan.,Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency, Saitama, Japan
| | - Jiro Yasuda
- Department of Emerging Infectious Diseases, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan.,National Research Center for the Control and Prevention of Infectious Diseases (CCPID), Nagasaki University, Nagasaki, Japan.,Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
3
|
Shi Z, Gu J, Yao Y, Wu Z. Identification of a predictive gene signature related to pyroptosis for the prognosis of cutaneous melanoma. Medicine (Baltimore) 2022; 101:e30564. [PMID: 36086707 PMCID: PMC10980462 DOI: 10.1097/md.0000000000030564] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 08/11/2022] [Indexed: 12/24/2022] Open
Abstract
Pyroptosis is a form of inflammatory programmed cell death. However, because of no specific molecular biomarker, pyroptosis has not been considered as a novel therapeutic method to treat cutaneous melanoma (CM). Here, we identified pyroptosis genes that associate with the prognosis of CM patients and constructed an effective model for the prognostic prediction of CM patients. To identify genes related to pyroptosis that are differentially expressed in CM, we obtained gene expression data of CM patients and normal skin tissues from the Cancer Genome Atlas and the Genotype-Tissue Expression databases, and used another cohort obtained from Gene Expression Omnibus database for validation. Three genes (BST2, GBP5, and AIM2) that were associated with prognosis were found and incorporated into our prognostic model. Furthermore, we divided the patients into 2 groups: a high-risk group and a low-risk group. Functional analyses indicated that our model was correlated with patient survival and cancer growth. Multivariate and univariate Cox regressions revealed that the constructed model could serve as an independent prognostic factor for CM patients. Meanwhile, compared with other clinical characteristics, our model significantly improved the diagnostic accuracy. Gene function analysis revealed that pyroptosis genes BST2, GBP5, and AIM2 were differentially expressed in CM patients and positively associated with patient prognosis. Finally, a risk score was used to generate nomograms that displayed favorable discriminatory abilities for CM. In summary, our model could significantly predict the prognosis of CM patients and be used for the development of CM therapy.
Collapse
Affiliation(s)
- Zhaoyang Shi
- Department of Hand Plastic Surgery, The First People’s Hospital of Linping District, Hangzhou, China
| | - Jiaying Gu
- Department of Laboratory, Integrated Traditional Chinese and Western Medicine Hospital of Linping District, Hangzhou, China
| | - Yi Yao
- Department of Hand Plastic Surgery, The First People’s Hospital of Linping District, Hangzhou, China
| | - Zhengyuan Wu
- Department of Hand Plastic Surgery, The First People’s Hospital of Linping District, Hangzhou, China
| |
Collapse
|
4
|
Disease Biomarkers in Gastrointestinal Malignancies. DISEASE MARKERS 2016; 2016:4714910. [PMID: 27445424 PMCID: PMC4947494 DOI: 10.1155/2016/4714910] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 04/28/2016] [Indexed: 01/21/2023]
|
5
|
Abstract
Multiple myeloma (MM) is a B-cell malignancy characterized by the clonal proliferation of malignant plasma cells in the bone marrow and the development of osteolytic bone lesions. MM has emerged as a paradigm within the cancers for the success of drug discovery and translational medicine. This article discusses immunotherapy as an encouraging option for the goal of inducing effective and long-lasting therapeutic outcome. Divided into two distinct approaches, passive or active, immunotherapy, which targets tumor-associated antigens has shown promising results in multiple preclinical and clinical studies.
Collapse
Affiliation(s)
- Jooeun Bae
- Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA.
| | - Nikhil C Munshi
- Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Kenneth C Anderson
- Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA
| |
Collapse
|
6
|
Zhang M, He J, Liu Z, Lu Y, Zheng Y, Li H, Xu J, Liu H, Qian J, Orlowski RZ, Kwak LW, Yi Q, Yang J. Anti-β₂-microglobulin monoclonal antibodies overcome bortezomib resistance in multiple myeloma by inhibiting autophagy. Oncotarget 2015; 6:8567-78. [PMID: 25895124 PMCID: PMC4496167 DOI: 10.18632/oncotarget.3251] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 01/29/2015] [Indexed: 01/19/2023] Open
Abstract
Our previous studies showed that anti-β2M monoclonal antibodies (mAbs) have strong and direct apoptotic effects on multiple myeloma (MM) cells, suggesting that anti-β2M mAbs might be developed as a novel therapeutic agent. In this study, we investigated the anti-MM effects of combination treatment with anti-β2M mAbs and bortezomib (BTZ). Our results showed that anti-β2M mAbs enhanced BTZ-induced apoptosis of MM cell lines and primary MM cells. Combination treatment could also induce apoptosis of BTZ-resistant MM cells, and the enhanced effect depended on the surface expression of β2M on MM cells. BTZ up-regulated the expression of autophagy proteins, whereas combination with anti-β2M mAbs inhibited autophagy. Sequence analysis of the promoter region of beclin 1 identified 3 putative NF-κB-binding sites from -615 to -789 bp. BTZ treatment increased, whereas combination with anti-β2M mAbs reduced, NF-κB transcription activities in MM cells, and combination treatment inhibited NF-κB p65 binding to the beclin 1 promoter. Furthermore, anti-β2M mAbs and BTZ combination treatment had anti-MM activities in an established MM mouse model. Thus, our studies provide new insight and support for the clinical development of an anti-β2M mAb and BTZ combination treatment to overcome BTZ drug resistance and improve MM patient survival.
Collapse
MESH Headings
- Active Transport, Cell Nucleus/drug effects
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Apoptosis/drug effects
- Apoptosis Regulatory Proteins/biosynthesis
- Apoptosis Regulatory Proteins/genetics
- Autophagy/drug effects
- Beclin-1
- Bortezomib/pharmacology
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/physiology
- Drug Screening Assays, Antitumor
- Drug Synergism
- Humans
- Lysosomal Membrane Proteins/biosynthesis
- Lysosomal Membrane Proteins/genetics
- Male
- Membrane Proteins/biosynthesis
- Membrane Proteins/genetics
- Mice
- Mice, SCID
- Microtubule-Associated Proteins/biosynthesis
- Microtubule-Associated Proteins/genetics
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Neoplasm Proteins/immunology
- RNA, Bacterial
- RNA, Small Interfering/genetics
- Signal Transduction/drug effects
- Transcription Factor RelA/antagonists & inhibitors
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
- beta 2-Microglobulin/antagonists & inhibitors
- beta 2-Microglobulin/biosynthesis
- beta 2-Microglobulin/genetics
- beta 2-Microglobulin/immunology
Collapse
Affiliation(s)
- Mingjun Zhang
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jin He
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Zhiqiang Liu
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yong Lu
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Yuhuan Zheng
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Haiyan Li
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jingda Xu
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Huan Liu
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jianfei Qian
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Robert Z. Orlowski
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Larry W. Kwak
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Qing Yi
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jing Yang
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Cancer Research Institute and Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
7
|
Mahauad-Fernandez WD, DeMali KA, Olivier AK, Okeoma CM. Bone marrow stromal antigen 2 expressed in cancer cells promotes mammary tumor growth and metastasis. Breast Cancer Res 2014; 16:493. [PMID: 25499888 PMCID: PMC4308845 DOI: 10.1186/s13058-014-0493-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 12/02/2014] [Indexed: 01/01/2023] Open
Abstract
Introduction Several innate immunity genes are overexpressed in human cancers and their roles remain controversial. Bone marrow stromal antigen 2 (BST-2) is one such gene whose role in cancer is not clear. BST-2 is a unique innate immunity gene with both antiviral and pro-tumor functions and therefore can serve as a paradigm for understanding the roles of other innate immunity genes in cancers. Methods Meta-analysis of tumors from breast cancer patients obtained from the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) datasets were evaluated for levels of BST-2 expression and for tumor aggressiveness. In vivo, we examined the effect of knockdown of BST-2 in two different murine carcinoma cells on tumor growth, metastasis, and survival. In vitro, we assessed the effect of carcinoma cell BST-2 knockdown and/or overexpression on adhesion, anchorage-independent growth, migration, and invasion. Results BST-2 in breast tumors and mammary cancer cells is a strong predictor of tumor size, tumor aggressiveness, and host survival. In humans, BST-2 mRNA is elevated in metastatic and invasive breast tumors. In mice, orthotopic implantation of mammary tumor cells lacking BST-2 increased tumor latency, decreased primary tumor growth, reduced metastases to distal organs, and prolonged host survival. Furthermore, we found that the cellular basis for the role of BST-2 in promoting tumorigenesis include BST-2-directed enhancement in cancer cell adhesion, anchorage-independency, migration, and invasion. Conclusions BST-2 contributes to the emergence of neoplasia and malignant progression of breast cancer. Thus, BST-2 may (1) serve as a biomarker for aggressive breast cancers, and (2) be a novel target for breast cancer therapeutics. Electronic supplementary material The online version of this article (doi:10.1186/s13058-014-0493-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wadie D Mahauad-Fernandez
- Department of Microbiology, Carver College of Medicine, University of Iowa, 51 Newton Road, Iowa City, IA, 52242-1109, USA. .,Interdisciplinary Graduate Program in Molecular and Cellular Biology (MCB), University of Iowa, 500 Newton Road, Iowa City, IA, 52242-1109, USA.
| | - Kris A DeMali
- Interdisciplinary Graduate Program in Molecular and Cellular Biology (MCB), University of Iowa, 500 Newton Road, Iowa City, IA, 52242-1109, USA. .,Department of Biochemistry, Carver College of Medicine, University of Iowa, 51 Newton Road, Iowa City, IA, 52242-1109, USA.
| | - Alicia K Olivier
- Department of Pathology, Carver College of Medicine, University of Iowa, 51 Newton Road, Iowa City, IA, 52242-1109, USA. .,Department of Pathobiology and Population Medicine, College of Veterinary Medicine, Mississippi State University, 240 Wise Center Drive, Starkville, MS, 39762-6100, USA.
| | - Chioma M Okeoma
- Department of Microbiology, Carver College of Medicine, University of Iowa, 51 Newton Road, Iowa City, IA, 52242-1109, USA. .,Interdisciplinary Graduate Program in Molecular and Cellular Biology (MCB), University of Iowa, 500 Newton Road, Iowa City, IA, 52242-1109, USA.
| |
Collapse
|
8
|
Wang L, Jin N, Schmitt A, Greiner J, Malcherek G, Hundemer M, Mani J, Hose D, Raab MS, Ho AD, Chen BA, Goldschmidt H, Schmitt M. T cell-based targeted immunotherapies for patients with multiple myeloma. Int J Cancer 2014; 136:1751-68. [PMID: 25195787 DOI: 10.1002/ijc.29190] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 08/28/2014] [Accepted: 09/03/2014] [Indexed: 12/17/2022]
Abstract
Despite high-dose chemotherapy followed by autologs stem-cell transplantation as well as novel therapeutic agents, multiple myeloma (MM) remains incurable. Following the general trend towards personalized therapy, targeted immunotherapy as a new approach in the therapy of MM has emerged. Better progression-free survival and overall survival after tandem autologs/allogeneic stem cell transplantation suggest a graft versus myeloma effect strongly supporting the usefulness of immunological therapies for MM patients. How to induce a powerful antimyeloma effect is the key issue in this field. Pivotal is the definition of appropriate tumor antigen targets and effective methods for expansion of T cells with clinical activity. Besides a comprehensive list of tumor antigens for T cell-based approaches, eight promising antigens, CS1, Dickkopf-1, HM1.24, Human telomerase reverse transcriptase, MAGE-A3, New York Esophageal-1, Receptor of hyaluronic acid mediated motility and Wilms' tumor gene 1, are described in detail to provide a background for potential clinical use. Results from both closed and on-going clinical trials are summarized in this review. On the basis of the preclinical and clinical data, we elaborate on three encouraging therapeutic options, vaccine-enhanced donor lymphocyte infusion, chimeric antigen receptors-transfected T cells as well as vaccines with multiple antigen peptides, to pave the way towards clinically significant immune responses against MM.
Collapse
Affiliation(s)
- Lei Wang
- Department of Internal Medicine V, University Clinic Heidelberg, University of Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Targeted therapy for HM1.24 (CD317) on multiple myeloma cells. BIOMED RESEARCH INTERNATIONAL 2014; 2014:965384. [PMID: 25143955 PMCID: PMC4124849 DOI: 10.1155/2014/965384] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Accepted: 07/02/2014] [Indexed: 11/18/2022]
Abstract
Multiple myeloma (MM) still remains an incurable disease, at least because of the existence of cell-adhesion mediated drug-resistant MM cells and/or continuous recruitment of presumed MM cancer stem cell-like cells (CSCs). As a new alternative treatment modality, immunological approaches using monoclonal antibodies (mAbs) and/or cytotoxic T lymphocytes (CTLs) are now attracting much attention as a novel strategy attacking MM cells. We have identified that HM1.24 [also known as bone marrow stromal cell antigen 2 (BST2) or CD317] is overexpressed on not only mature MM cells but also MM CSCs. We then have developed a humanized mAb to HM1.24 and defucosylated version of the mAb to adapt to clinical practice. Moreover, we have successfully induced HM1.24-specific CTLs against MM cells. The combination of these innovative therapeutic modalities may likely exert an anti-MM activity by evading the drug resistance mechanism and eliminating presumed CSCs in MM.
Collapse
|
10
|
Zhang M, Qian J, Lan Y, Lu Y, Li H, Hong B, Zheng Y, He J, Yang J, Yi Q. Anti-β₂M monoclonal antibodies kill myeloma cells via cell- and complement-mediated cytotoxicity. Int J Cancer 2014; 135:1132-41. [PMID: 24474467 DOI: 10.1002/ijc.28745] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Accepted: 01/15/2014] [Indexed: 12/26/2022]
Abstract
Our previous studies showed that anti-β2M monoclonal antibodies (mAbs) at high doses have direct apoptotic effects on myeloma cells, suggesting that anti-β2M mAbs might be developed as a novel therapeutic agent. In this study, we investigated the ability of the mAbs at much lower concentrations to indirectly kill myeloma cells by utilizing immune effector cells or molecules. Our results showed that anti-β2M mAbs effectively lysed MM cells via antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC), which were correlated with and dependent on the surface expression of β2M on MM cells. The presence of MM bone marrow stromal cells or addition of IL-6 did not attenuate anti-β2M mAb-induced ADCC and CDC activities against MM cells. Furthermore, anti-β2M mAbs only showed limited cytotoxicity toward normal B cells and nontumorous mesenchymal stem cells, indicating that the ADCC and CDC activities of the anti-β2M mAbs were more prone to the tumor cells. Lenalidomide potentiated in vitro ADCC activity against MM cells and in vivo tumor inhibition capacity induced by the anti-β2M mAbs by enhancing the activity of NK cells. These results support clinical development of anti-β2M mAbs, both as a monotherapy and in combination with lenalidomide, to improve MM patient outcome.
Collapse
Affiliation(s)
- Mingjun Zhang
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Harada T, Ozaki S, Oda A, Tsuji D, Ikegame A, Iwasa M, Udaka K, Fujii S, Nakamura S, Miki H, Kagawa K, Kuroda Y, Kawai S, Itoh K, Yamada-Okabe H, Matsumoto T, Abe M. Combination with a defucosylated anti-HM1.24 monoclonal antibody plus lenalidomide induces marked ADCC against myeloma cells and their progenitors. PLoS One 2013; 8:e83905. [PMID: 24386306 PMCID: PMC3873421 DOI: 10.1371/journal.pone.0083905] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 11/08/2013] [Indexed: 12/03/2022] Open
Abstract
The immunomodulatory drug lenalidomide (Len) has drawn attention to potentiate antibody-dependent cellular cytotoxicity (ADCC)-mediated immunotherapies. We developed the defucosylated version (YB-AHM) of humanized monoclonal antibody against HM1.24 (CD317) overexpressed in multiple myeloma (MM) cells. In this study, we evaluated ADCC by YB-AHM and Len in combination against MM cells and their progenitors. YB-AHM was able to selectively kill via ADCC MM cells in bone marrow samples from patients with MM with low effector/target ratios, which was further enhanced by treatment with Len. Interestingly, Len also up-regulated HM1.24 expression on MM cells in an effector-dependent manner. HM1.24 was found to be highly expressed in a drug-resistant clonogenic “side population” in MM cells; and this combinatory treatment successfully reduced SP fractions in RPMI 8226 and KMS-11 cells in the presence of effector cells, and suppressed a clonogenic potential of MM cells in colony-forming assays. Collectively, the present study suggests that YB-AHM and Len in combination may become an effective therapeutic strategy in MM, warranting further study to target drug-resistant MM clonogenic cells.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Antibodies, Monoclonal, Humanized/immunology
- Antibodies, Monoclonal, Humanized/metabolism
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibody-Dependent Cell Cytotoxicity/drug effects
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Antineoplastic Combined Chemotherapy Protocols
- Cell Line, Tumor
- Drug Synergism
- Female
- GPI-Linked Proteins/immunology
- GPI-Linked Proteins/metabolism
- Glycosylation
- Humans
- Immunotherapy
- Lenalidomide
- Male
- Middle Aged
- Multiple Myeloma/immunology
- Multiple Myeloma/pathology
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/immunology
- Neoplastic Stem Cells/pathology
- Side-Population Cells/drug effects
- Side-Population Cells/pathology
- Thalidomide/analogs & derivatives
- Thalidomide/pharmacology
- Up-Regulation/drug effects
Collapse
Affiliation(s)
- Takeshi Harada
- Department of Medicine and Bioregulatory Sciences, Graduate School of Medical Sciences, University of Tokushima, Tokushima, Japan
| | - Shuji Ozaki
- Department of Hematology, Tokushima Prefectural Central Hospital, Tokushima, Japan
- * E-mail:
| | - Asuka Oda
- Department of Medicine and Bioregulatory Sciences, Graduate School of Medical Sciences, University of Tokushima, Tokushima, Japan
| | - Daisuke Tsuji
- Department of Medicinal Biotechnology, Graduate School of Pharmaceutical Sciences, University of Tokushima, Tokushima, Japan
| | - Akishige Ikegame
- Division of Medical Technology, Tokushima University Hospital, Tokushima, Japan
| | - Masami Iwasa
- Department of Medicine and Bioregulatory Sciences, Graduate School of Medical Sciences, University of Tokushima, Tokushima, Japan
| | - Kengo Udaka
- Department of Medicine and Bioregulatory Sciences, Graduate School of Medical Sciences, University of Tokushima, Tokushima, Japan
| | - Shiro Fujii
- Department of Medicine and Bioregulatory Sciences, Graduate School of Medical Sciences, University of Tokushima, Tokushima, Japan
| | - Shingen Nakamura
- Department of Medicine and Bioregulatory Sciences, Graduate School of Medical Sciences, University of Tokushima, Tokushima, Japan
| | - Hirokazu Miki
- Department of Medicine and Bioregulatory Sciences, Graduate School of Medical Sciences, University of Tokushima, Tokushima, Japan
| | - Kumiko Kagawa
- Department of Medicine and Bioregulatory Sciences, Graduate School of Medical Sciences, University of Tokushima, Tokushima, Japan
| | - Yoshiaki Kuroda
- Department of Hematology and Oncology, RIRBM, Hiroshima University, Hiroshima, Japan
| | - Shigeto Kawai
- Research Division, Forerunner Pharma Research Co. Ltd., Tokyo, Japan
| | - Kohji Itoh
- Department of Medicinal Biotechnology, Graduate School of Pharmaceutical Sciences, University of Tokushima, Tokushima, Japan
| | | | - Toshio Matsumoto
- Department of Medicine and Bioregulatory Sciences, Graduate School of Medical Sciences, University of Tokushima, Tokushima, Japan
| | - Masahiro Abe
- Department of Medicine and Bioregulatory Sciences, Graduate School of Medical Sciences, University of Tokushima, Tokushima, Japan
| |
Collapse
|
12
|
Allegra A, Penna G, Alonci A, Russo S, Greve B, Innao V, Minardi V, Musolino C. Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma. Eur J Haematol 2013; 90:441-68. [DOI: 10.1111/ejh.12107] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2013] [Indexed: 12/12/2022]
Affiliation(s)
| | - Giuseppa Penna
- Division of Haematology; University of Messina; Messina; Italy
| | - Andrea Alonci
- Division of Haematology; University of Messina; Messina; Italy
| | - Sabina Russo
- Division of Haematology; University of Messina; Messina; Italy
| | - Bruna Greve
- Division of Haematology; University of Messina; Messina; Italy
| | - Vanessa Innao
- Division of Haematology; University of Messina; Messina; Italy
| | - Viviana Minardi
- Division of Haematology; University of Messina; Messina; Italy
| | | |
Collapse
|
13
|
Immunogenic targets for specific immunotherapy in multiple myeloma. Clin Dev Immunol 2012; 2012:820394. [PMID: 22611422 PMCID: PMC3352660 DOI: 10.1155/2012/820394] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Accepted: 02/05/2012] [Indexed: 12/22/2022]
Abstract
Multiple myeloma remains an incurable disease although the prognosis has been improved by novel therapeutics and agents recently. Relapse occurs in the majority of patients and becomes fatal finally. Immunotherapy might be a powerful intervention to maintain a long-lasting control of minimal residual disease or to even eradicate disseminated tumor cells. Several tumor-associated antigens have been identified in patients with multiple myeloma. These antigens are expressed in a tumor-specific or tumor-restricted pattern, are able to elicit immune response, and thus could serve as targets for immunotherapy. This review discusses immunogenic antigens with therapeutic potential for multiple myeloma.
Collapse
|
14
|
Potent in vitro and in vivo activity of an Fc-engineered humanized anti-HM1.24 antibody against multiple myeloma via augmented effector function. Blood 2012; 119:2074-82. [PMID: 22246035 DOI: 10.1182/blood-2011-06-364521] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
HM1.24, an immunologic target for multiple myeloma (MM) cells, has not been effectively targeted with therapeutic monoclonal antibodies (mAbs). In this study, we investigated in vitro and in vivo anti-MM activities of XmAb5592, a humanized anti-HM1.24 mAb with Fc-domain engineered to significantly enhance FcγR binding and associated immune effector functions. XmAb5592 increased antibody-dependent cellular cytotoxicity (ADCC) several fold relative to the anti-HM1.24 IgG1 analog against both MM cell lines and primary patient myeloma cells. XmAb5592 also augmented antibody dependent cellular phagocytosis (ADCP) by macrophages. Natural killer (NK) cells became more activated by XmAb5592 than the IgG1 analog, evidenced by increased cell surface expression of granzyme B-dependent CD107a and MM cell lysis, even in the presence of bone marrow stromal cells. XmAb5592 potently inhibited tumor growth in mice bearing human MM xenografts via FcγR-dependent mechanisms, and was significantly more effective than the IgG1 analog. Lenalidomide synergistically enhanced in vitro ADCC against MM cells and in vivo tumor inhibition induced by XmAb5592. A single dose of 20 mg/kg XmAb5592 effectively depleted both blood and bone marrow plasma cells in cynomolgus monkeys. These results support clinical development of XmAb5592, both as a monotherapy and in combination with lenalidomide, to improve patient outcome of MM.
Collapse
|
15
|
In vivo expression profile of the antiviral restriction factor and tumor-targeting antigen CD317/BST-2/HM1.24/tetherin in humans. Proc Natl Acad Sci U S A 2011; 108:13688-93. [PMID: 21808013 DOI: 10.1073/pnas.1101684108] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Human CD317 is an intrinsic immunity factor that restricts the release of enveloped viruses, including the major pathogens HIV and Lassa virus, from infected cells in culture. Its importance for infection control in humans is unclear, due in part to its incompletely defined in vivo expression pattern. CD317 also has been proposed as a selective target for immunotherapy of multiple myeloma. To provide a framework for studies of the biological functions, regulation, and therapeutic potential of CD317, we performed microarray-based expression profiling in 468 tissue samples from 25 healthy organs from more than 210 patients. We found that CD317 protein was expressed to varying degrees in all organs tested and detected in a number of specialized cell types, including hepatocytes, pneumocytes, ducts of major salivary glands, pancreas and kidney, Paneth cells, epithelia, Leydig cells, plasma cells, bone marrow stromal cells, monocytes, and vascular endothelium. Although many of these cell types are in vivo targets for pathogenic viruses, restriction by CD317 or virus-encoded antagonists has been documented in only some of them. Limited cell type-dependent coexpression of CD317 with the IFN biomarker MxA in vivo and lack of responsive stimulation in organ explants suggest that interferons may only partially regulate CD317. This in vivo expression profiling sheds light on the biology and species-specificity of CD317, identifies multiple thus far unknown interaction sites of viruses with this restriction factor, and refutes the concept of its restricted constitutive expression and primary IFN inducibility. CD317's widespread expression calls into question its suitability as a target for immunotherapy.
Collapse
|
16
|
Tai YT, Anderson KC. Antibody-based therapies in multiple myeloma. BONE MARROW RESEARCH 2011; 2011:924058. [PMID: 22046572 PMCID: PMC3200112 DOI: 10.1155/2011/924058] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Accepted: 01/04/2011] [Indexed: 01/06/2023]
Abstract
The unmet need for improved multiple myeloma (MM) therapy has stimulated clinical development of monoclonal antibodies (mAbs) targeting either MM cells or cells of the bone marrow (BM) microenvironment. In contrast to small-molecule inhibitors, therapeutic mAbs present the potential to specifically target tumor cells and directly induce an immune response to lyse tumor cells. Unique immune-effector mechanisms are only triggered by therapeutic mAbs but not by small molecule targeting agents. Although therapeutic murine mAbs or chimeric mAbs can cause immunogenicity, the advancement of genetic recombination for humanizing rodent mAbs has allowed large-scale production and designation of mAbs with better affinities, efficient selection, decreasing immunogenicity, and improved effector functions. These advancements of antibody engineering technologies have largely overcome the critical obstacle of antibody immunogenicity and enabled the development and subsequent Food and Drug Administration (FDA) approval of therapeutic Abs for cancer and other diseases.
Collapse
Affiliation(s)
- Yu-Tzu Tai
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, 44 Binney Street, Boston, MA 02115, USA
| | - Kenneth C. Anderson
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, 44 Binney Street, Boston, MA 02115, USA
| |
Collapse
|
17
|
Ishiguro T, Kawai S, Habu K, Sugimoto M, Shiraiwa H, Iijima S, Ozaki S, Matsumoto T, Yamada-Okabe H. A defucosylated anti-CD317 antibody exhibited enhanced antibody-dependent cellular cytotoxicity against primary myeloma cells in the presence of effectors from patients. Cancer Sci 2010; 101:2227-33. [PMID: 20701608 PMCID: PMC11158282 DOI: 10.1111/j.1349-7006.2010.01663.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The humanized monoclonal antibody (mAb) against CD317 antigen (anti-HM1.24 antibody; AHM), which is highly expressed on multiple myeloma (MM), induces antibody-dependent cellular cytotoxicity (ADCC). However, the antitumor activity of AHM in the clinical setting has not been clearly demonstrated. In this study, we produced defucosylated AHM and evaluated its potency for clinical application by performing autologous ADCC assays against primary MM cells from patients. Defucosylated AHM that was produced in rat myeloma YB2/0 cells expressing a low level of fucosyltransferase (FUT8) showed significant ADCC activity against three out of six primary MM cells in the presence of autologous PBMC, whereas conventional AHM did not. The results indicate that the potency of AHM to induce ADCC against primary MM cells was insufficient, but was significantly augmented by defucosylation. To generate more homogenous defucosylated monoclonal antibodies (mAb) for fermentation, we disrupted the GFT gene that encodes a GDP-fucose transporter in a CHO/DXB11 cell line by sequential homologous recombination. Analysis of the N-linked oligosaccharide in the defucosylated AHM produced by the established GFT(-/-)CHO cell line showed that a majority (93.4%) of the oligosaccharide was fucose free. The GFT(-/-) cells stably produced defucosylated mAb over passages. These results demonstrate that GTF(-/-)CHO-produced defucosylated AHM (GFTKO-AHM) will be a promising new therapeutic antibody against MM in the clinical setting.
Collapse
Affiliation(s)
- Takahiro Ishiguro
- Pharmaceutical Research Department 3, Chugai Pharmaceutical Co. Ltd, Kanagawa, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Amano J, Masuyama N, Hirota Y, Tanaka Y, Igawa Y, Shiokawa R, Okutani T, Miyayama T, Nanami M, Ishigai M. Antigen-dependent internalization is related to rapid elimination from plasma of humanized anti-HM1.24 monoclonal antibody. Drug Metab Dispos 2010; 38:2339-46. [PMID: 20823293 DOI: 10.1124/dmd.110.035709] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Anti-HM1.24 monoclonal antibody (AHM) is a humanized anti-HM1.24 monoclonal antibody that binds to the HM1.24 antigen, a protein that is highly expressed in multiple myeloma cells. The pharmacokinetics of AHM was determined in experiments in which AHM was administered intravenously to cynomolgus monkeys. The area under the plasma concentration-time curve increased by more than the dose ratio between 2 and 20 mg/kg, and nonlinear pharmacokinetics was observed. The elimination half-life of AHM from the plasma was 7.56 h at 2 mg/kg and 28.6 h at 20 mg/kg, which was shorter than that observed for other therapeutic humanized monoclonal antibodies, such as trastuzumab and bevacizumab. Although antibodies to AHM were detected in all monkeys on or after 10 days of administration, there was a temporal disassociation between the rapid elimination of AHM and the appearance of anti-AHM antibodies. HM1.24 antigen-dependent internalization and intracellular metabolism of AHM were investigated in peripheral blood mononuclear, KPMM2, and U937 cells. In all cases, AHM was rapidly internalized from the cell surface; this internalization was significantly prevented by phenylarsine oxide in KPMM2 cells, an inhibitor of receptor-mediated endocytosis, and the internalized AHM was subsequently degraded within the cells. Furthermore, immunofluorescence microscopy revealed that the internalized AHM is delivered to and degraded in late endosomes/lysosomes. Taken together, our results suggest that the rapid elimination of AHM from plasma in monkey is due to HM1.24 antigen-dependent internalization followed by delivery to the lysosomes.
Collapse
Affiliation(s)
- Jun Amano
- Chugai Pharmaceutical Co., Ltd., Fuji-Gotenba Research Laboratories, 1-135 Komakado, Gotenba-shi, Shizuoka 412-8513, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Changes in the quality of antibodies produced by Chinese hamster ovary cells during the death phase of cell culture. J Biosci Bioeng 2010; 109:281-7. [DOI: 10.1016/j.jbiosc.2009.09.043] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Revised: 09/16/2009] [Accepted: 09/16/2009] [Indexed: 11/18/2022]
|
20
|
Evaluation of Chinese hamster ovary cell stability during repeated batch culture for large-scale antibody production. J Biosci Bioeng 2010; 109:274-80. [DOI: 10.1016/j.jbiosc.2009.09.044] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2009] [Revised: 09/15/2009] [Accepted: 09/15/2009] [Indexed: 11/18/2022]
|
21
|
Miyakawa K, Ryo A, Murakami T, Ohba K, Yamaoka S, Fukuda M, Guatelli J, Yamamoto N. BCA2/Rabring7 promotes tetherin-dependent HIV-1 restriction. PLoS Pathog 2009; 5:e1000700. [PMID: 20019814 PMCID: PMC2788703 DOI: 10.1371/journal.ppat.1000700] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Accepted: 11/18/2009] [Indexed: 01/28/2023] Open
Abstract
Host cell factors can either positively or negatively regulate the assembly and egress of HIV-1 particles from infected cells. Recent reports have identified a previously uncharacterized transmembrane protein, tetherin/CD317/BST-2, as a crucial host restriction factor that acts during a late budding step in HIV-1 replication by inhibiting viral particle release. Although tetherin has been shown to promote the retention of nascent viral particles on the host cell surface, the precise molecular mechanisms that occur during and after these tethering events remain largely unknown. We here report that a RING-type E3 ubiquitin ligase, BCA2 (Breast cancer-associated gene 2; also called Rabring7, ZNF364 or RNF115), is a novel tetherin-interacting host protein that facilitates the restriction of HIV-1 particle production in tetherin-positive cells. The expression of human BCA2 in “tetherin-positive” HeLa, but not in “tetherin-negative” HOS cells, resulted in a strong restriction of HIV-1 particle production. Upon the expression of tetherin in HOS cells, BCA2 was capable of inhibiting viral particle production as in HeLa cells. The targeted depletion of endogenous BCA2 by RNA interference (RNAi) in HeLa cells reduced the intracellular accumulation of viral particles, which were nevertheless retained on the plasma membrane. BCA2 was also found to facilitate the internalization of HIV-1 virions into CD63+ intracellular vesicles leading to their lysosomal degradation. These results indicate that BCA2 accelerates the internalization and degradation of viral particles following their tethering to the cell surface and is a co-factor or enhancer for the tetherin-dependent restriction of HIV-1 release from infected cells. Human cells possess multiple systems that render them resistant to viral infection. Recently, a transmembrane protein, tetherin, has been identified as an antiviral host factor in HIV-1-infected cells. Tetherin retains newly assembled virions at the plasma membrane and prevents viral release from the infected cells. However, the precise molecular mechanisms following the virion tethering remain largely unknown. In our current study, we have identified a RING-type E3 ubiquitin ligase, BCA2, which co-localizes and interacts with tetherin in human cells. BCA2 was found to facilitate the internalization of HIV-1 particles captured by tetherin on the plasma membrane and to enhance the targeting of viral particles to the lysosomes. Conversely, the targeted depletion of endogenous BCA2 reduces the intracellular accumulation of viral particles. Additionally, the expression of a small viral protein Vpu, an antagonist of tetherin, counteracts the antiviral effects of BCA2. These results suggest that BCA2 is a potential antiviral factor that collaborates with tetherin to facilitate the degradation of nascent HIV-1 particles during “post-tethering” processes.
Collapse
Affiliation(s)
- Kei Miyakawa
- AIDS Research Center, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
- Department of Molecular Virology, Graduate School of Medicine, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Akihide Ryo
- AIDS Research Center, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
- * E-mail: (AR); (NY)
| | - Tsutomu Murakami
- AIDS Research Center, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Kenji Ohba
- AIDS Research Center, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Shoji Yamaoka
- Department of Molecular Virology, Graduate School of Medicine, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Mitsunori Fukuda
- Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - John Guatelli
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Naoki Yamamoto
- AIDS Research Center, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
- * E-mail: (AR); (NY)
| |
Collapse
|
22
|
van Rhee F, Szmania SM, Dillon M, van Abbema AM, Li X, Stone MK, Garg TK, Shi J, Moreno-Bost AM, Yun R, Balasa B, Ganguly B, Chao D, Rice AG, Zhan F, Shaughnessy JD, Barlogie B, Yaccoby S, Afar DEH. Combinatorial efficacy of anti-CS1 monoclonal antibody elotuzumab (HuLuc63) and bortezomib against multiple myeloma. Mol Cancer Ther 2009; 8:2616-24. [PMID: 19723891 PMCID: PMC2748787 DOI: 10.1158/1535-7163.mct-09-0483] [Citation(s) in RCA: 140] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Monoclonal antibody (mAb) therapy for multiple myeloma, a malignancy of plasma cells, has not been clinically efficacious in part due to a lack of appropriate targets. We recently reported that the cell surface glycoprotein CS1 (CD2 subset 1, CRACC, SLAMF7, CD319) was highly and universally expressed on myeloma cells while having restricted expression in normal tissues. Elotuzumab (formerly known as HuLuc63), a humanized mAb targeting CS1, is currently in a phase I clinical trial in relapsed/refractory myeloma. In this report we investigated whether the activity of elotuzumab could be enhanced by bortezomib, a reversible proteasome inhibitor with significant activity in myeloma. We first showed that elotuzumab could induce patient-derived myeloma cell killing within the bone marrow microenvironment using a SCID-hu mouse model. We next showed that CS1 gene and cell surface protein expression persisted on myeloma patient-derived plasma cells collected after bortezomib administration. In vitro bortezomib pretreatment of myeloma targets significantly enhanced elotuzumab-mediated antibody-dependent cell-mediated cytotoxicity, both for OPM2 myeloma cells using natural killer or peripheral blood mononuclear cells from healthy donors and for primary myeloma cells using autologous natural killer effector cells. In an OPM2 myeloma xenograft model, elotuzumab in combination with bortezomib exhibited significantly enhanced in vivo antitumor activity. These findings provide the rationale for a clinical trial combining elotuzumab and bortezomib, which will test the hypothesis that combining both drugs would result in enhanced immune lysis of myeloma by elotuzumab and direct targeting of myeloma by bortezomib.
Collapse
Affiliation(s)
- Frits van Rhee
- University of Arkansas for Medical Sciences, Myeloma Institute for Research and Therapy, Little Rock AR 72205, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Kawai S, Azuma Y, Fujii E, Furugaki K, Ozaki S, Matsumoto T, Kosaka M, Yamada-Okabe H. Interferon-alpha enhances CD317 expression and the antitumor activity of anti-CD317 monoclonal antibody in renal cell carcinoma xenograft models. Cancer Sci 2008; 99:2461-6. [PMID: 19032371 PMCID: PMC11160063 DOI: 10.1111/j.1349-7006.2008.00968.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2008] [Revised: 08/03/2008] [Accepted: 08/08/2008] [Indexed: 12/11/2022] Open
Abstract
A murine (mAHM) and a humanized (AHM) monoclonal antibody against CD317 (also called tetherin, BST2, or HM1.24 antigen), expressed preferentially in neoplastic B cells such as multiple myeloma, exhibited antitumor effects as a result of antibody-dependent cellular cytotoxicity (ADCC). The putative interferon (IFN) response elements IRF-1/2 and ISGF3 are present in the promoter of the CD317 gene, and IFN has been used for the treatment of not only myeloproliferative diseases but also solid tumors such as renal cell carcinoma (RCC) and melanoma. Therefore, we examined the effects of IFN on the expression of CD317 and on the antitumor activity of AHM and mAHM in RCC and melanoma. Flow cytometry and in vitro ADCC assays with human or mouse effector cells demonstrated that IFN-alpha markedly increased the amount of cell surface CD317 and augmented the ADCC activity of mAHM and AHM in RCC cells and to a lesser extent in melanoma cells. Administration of IFN-alpha to mice bearing RCC xenografts also increased the expression of CD317 in tumor cells. When coadministered with IFN-alpha, mAHM exhibited more profound antitumor activity in both IFN-alpha-sensitive and -insensitive RCC xenograft models. Thus, AHM in combination with IFN-alpha may be an effective therapy for the treatment of RCC.
Collapse
MESH Headings
- Animals
- Antibodies, Anti-Idiotypic/immunology
- Antibodies, Anti-Idiotypic/pharmacology
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Antigens, CD/genetics
- Antigens, CD/immunology
- Carcinoma, Renal Cell/immunology
- Carcinoma, Renal Cell/therapy
- Cell Line, Tumor
- Fluorescein-5-isothiocyanate/metabolism
- Fluorescent Dyes/metabolism
- GPI-Linked Proteins
- Humans
- Immunohistochemistry
- Interferon-alpha/immunology
- Interferon-alpha/pharmacology
- Kidney Neoplasms/immunology
- Kidney Neoplasms/therapy
- Male
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/immunology
- Mice
- Mice, Nude
- Mice, SCID
- Tumor Burden
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Shigeto Kawai
- Pharmaceutical Research Department, Chugai Pharmaceutical, 200 Kajiwara, Kamakura, Kanagawa 247-8530, Japan
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Tai YT, Dillon M, Song W, Leiba M, Li XF, Burger P, Lee AI, Podar K, Hideshima T, Rice AG, van Abbema A, Jesaitis L, Caras I, Law D, Weller E, Xie W, Richardson P, Munshi NC, Mathiot C, Avet-Loiseau H, Afar DEH, Anderson KC. Anti-CS1 humanized monoclonal antibody HuLuc63 inhibits myeloma cell adhesion and induces antibody-dependent cellular cytotoxicity in the bone marrow milieu. Blood 2008; 112:1329-37. [PMID: 17906076 PMCID: PMC2515112 DOI: 10.1182/blood-2007-08-107292] [Citation(s) in RCA: 384] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2007] [Accepted: 09/23/2007] [Indexed: 12/16/2022] Open
Abstract
Currently, no approved monoclonal antibody (mAb) therapies exist for human multiple myeloma (MM). Here we characterized cell surface CS1 as a novel MM antigen and further investigated the potential therapeutic utility of HuLuc63, a humanized anti-CS1 mAb, for treating human MM. CS1 mRNA and protein was highly expressed in CD138-purified primary tumor cells from the majority of MM patients (more than 97%) with low levels of circulating CS1 detectable in MM patient sera, but not in healthy donors. CS1 was expressed at adhesion-promoting uropod membranes of polarized MM cells, and short interfering RNA (siRNA) targeted to CS1 inhibited MM cell adhesion to bone marrow stromal cells (BMSCs). HuLuc63 inhibited MM cell binding to BMSCs and induced antibody-dependent cellular cytotoxicity (ADCC) against MM cells in dose-dependent and CS1-specific manners. HuLuc63 triggered autologous ADCC against primary MM cells resistant to conventional or novel therapies, including bortezomib and HSP90 inhibitor; and pretreatment with conventional or novel anti-MM drugs markedly enhanced HuLuc63-induced MM cell lysis. Administration of HuLuc63 significantly induces tumor regression in multiple xenograft models of human MM. These results thus define the functional significance of CS1 in MM and provide the preclinical rationale for testing HuLuc63 in clinical trials, either alone or in combination.
Collapse
Affiliation(s)
- Yu-Tzu Tai
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Wang W, Nishioka Y, Ozaki S, Jalili A, Verma VK, Hanibuchi M, Abe S, Minakuchi K, Matsumoto T, Sone S. Chimeric and humanized anti-HM1.24 antibodies mediate antibody-dependent cellular cytotoxicity against lung cancer cells. Lung Cancer 2008; 63:23-31. [PMID: 18524412 DOI: 10.1016/j.lungcan.2008.04.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2007] [Revised: 04/02/2008] [Accepted: 04/21/2008] [Indexed: 11/20/2022]
Abstract
HM1.24 antigen (CD317) was originally identified as a cell surface protein that is preferentially overexpressed on multiple myeloma cells. Immunotherapy using anti-HM1.24 antibody has been performed in patients with multiple myeloma as a phase I study. The aim of this study was to evaluate the anti-tumor activity of mouse-human chimeric and humanized anti-HM1.24 monoclonal antibodies (mAbs) against lung cancer cells in vitro. Human peripheral blood lymphocytes and monocytes separated from mononuclear cells (PBMCs) were used as effector cells. Antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) of chimeric and humanized anti-HM1.24 mAbs against lung cancer cells were determined by chromium-release assay. In some experiments, target or effector cells were pretreated with various cytokines. Chimeric and humanized anti-HM1.24 mAbs effectively induced ADCC against lung cancer cells mediated more efficiently by lymphocytes than monocytes. The cytotoxic activity correlated with the level of HM1.24 expression on lung cancer cells. Natural killer cells were identified as the major effector cells in ADCC mediated by the anti-HM1.24 mAb. The treatment of lymphocytes or monocytes with IL-2, IL-12, IL-15, M-CSF, or IFN-gamma significantly increased the ADCC activity. Moreover, the culture of lung cancer cells with IFN-beta or IFN-gamma augmented their susceptibility to ADCC and CDC. PBMCs from patients with lung cancer induced a level of ADCC comparable to that induced by PBMCs from healthy donors. Chimeric or humanized anti-HM1.24 mAbs have potential as a new therapeutic tool in lung cancer, and in combination with interleukins and interferons, could be useful for enhancing ADCC.
Collapse
Affiliation(s)
- Wei Wang
- Department of Internal Medicine and Molecular Therapeutics, Institute of Health Biosciences, University of Tokushima Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Sekimoto E, Ozaki S, Ohshima T, Shibata H, Hashimoto T, Abe M, Kimura N, Hattori K, Kawai S, Kinoshita Y, Yamada-Okabe H, Tsuchiya M, Matsumoto T. A single-chain Fv diabody against human leukocyte antigen-A molecules specifically induces myeloma cell death in the bone marrow environment. Cancer Res 2007; 67:1184-92. [PMID: 17283154 DOI: 10.1158/0008-5472.can-06-2236] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cross-linked human leukocyte antigen (HLA) class I molecules have been shown to mediate cell death in neoplastic lymphoid cells. However, clinical application of an anti-HLA class I antibody is limited by possible side effects due to widespread expression of HLA class I molecules in normal tissues. To reduce the unwanted Fc-mediated functions of the therapeutic antibody, we have developed a recombinant single-chain Fv diabody (2D7-DB) specific to the alpha2 domain of HLA-A. Here, we show that 2D7-DB specifically induces multiple myeloma cell death in the bone marrow environment. Both multiple myeloma cell lines and primary multiple myeloma cells expressed HLA-A at higher levels than normal myeloid cells, lymphocytes, or hematopoietic stem cells. 2D7-DB rapidly induced Rho activation and robust actin aggregation that led to caspase-independent death in multiple myeloma cells. This cell death was completely blocked by Rho GTPase inhibitors, suggesting that Rho-induced actin aggregation is crucial for mediating multiple myeloma cell death. Conversely, 2D7-DB neither triggered Rho-mediated actin aggregation nor induced cell death in normal bone marrow cells despite the expression of HLA-A. Treatment with IFNs, melphalan, or bortezomib enhanced multiple myeloma cell death induced by 2D7-DB. Furthermore, administration of 2D7-DB resulted in significant tumor regression in a xenograft model of human multiple myeloma. These results indicate that 2D7-DB acts on multiple myeloma cells differently from other bone marrow cells and thus provide the basis for a novel HLA class I-targeting therapy against multiple myeloma.
Collapse
Affiliation(s)
- Etsuko Sekimoto
- Department of Medicine and Bioregulatory Sciences, The University of Tokushima Graduate School of Health Biosciences, 3-18-15 Kuramoto, Tokushima 770-8503, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Kawai S, Koishihara Y, Iida SI, Ozaki S, Matsumoto T, Kosaka M, Yamada-Okabe H. Construction of a conventional non-radioisotope method to quantify HM1.24 antigens: correlation of HM1.24 levels and ADCC activity of the humanized antibody against HM1.24. Leuk Res 2006; 30:949-56. [PMID: 16473407 DOI: 10.1016/j.leukres.2005.11.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2005] [Accepted: 11/30/2005] [Indexed: 11/19/2022]
Abstract
A humanized monoclonal antibody (mAb) against HM1.24 (AHM) caused antibody-dependent cellular cytotoxicity (ADCC) against multiple myeloma (MM) cells. Here, we constructed a conventional non-radioisotope method that quantifies the amount of HM1.24 using fluorescein-labeled AHM. More than 10(4) molecules/cell of HM1.24 were detected in 12 out of 14 patients' MM cells, and a linear correlation was found between ADCC by AHM and the amounts of HM1.24. Thus, AHM is likely to be more efficacious against MM cells with high levels of HM1.24. This conventional non-RI method to quantify HM1.24 will be useful to select patients most likely to respond to AHM.
Collapse
Affiliation(s)
- Shigeto Kawai
- Pharmaceutical Research Department III, Chugai Pharmaceutical Co. Ltd., 200 Kajiwara, Kamakura, Kanagawa 247-8530, Japan
| | | | | | | | | | | | | |
Collapse
|
28
|
Vidal-Laliena M, Romero X, March S, Requena V, Petriz J, Engel P. Characterization of antibodies submitted to the B cell section of the 8th Human Leukocyte Differentiation Antigens Workshop by flow cytometry and immunohistochemistry. Cell Immunol 2005; 236:6-16. [PMID: 16157322 DOI: 10.1016/j.cellimm.2005.08.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2005] [Accepted: 05/11/2005] [Indexed: 10/25/2022]
Abstract
The aim of this study was to characterize the reactivity of monoclonal antibodies (mAbs) that had been submitted to the HLDA8 Workshop. The lineage specificity of target molecules was tested by analyzing their expression patterns on blood cells, leukocytes, and lymphocyte subsets. The expression of target molecules during B cell development, ranging from early precursors to plasma cells, was analyzed using a large panel of B cell lines. Our results have permitted us to characterize the expression of 10 new CD molecules: CD316 (HM1.24, BST2), CD268 (BAFF-R, TNFRSF13C), CD269 (BCMA, TNFRF17), CD267 (TACI, TNFRSF13B), CD275 (ICOSL, B7H2), CD254 (TRANCE, TNFSF11), CD252 (OX40L TNFSF4), CD315 (CD9-P), CD316 (EWI-2, PGRL), and CD307 (IRTA-2 or FcRH5). Three of these new CDs, CD267, CD269, and CD307 presented a B cell-restricted expression pattern. MAbs against these novel cell-surface molecules may offer new tools for research, diagnosis, and therapy.
Collapse
Affiliation(s)
- Miriam Vidal-Laliena
- Immunology Unit, Department of Cellular Biology and Pathology, Medical School, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Casanova 143, Barcelona E-08036, Spain
| | | | | | | | | | | |
Collapse
|
29
|
Jalili A, Ozaki S, Hara T, Shibata H, Hashimoto T, Abe M, Nishioka Y, Matsumoto T. Induction of HM1.24 peptide-specific cytotoxic T lymphocytes by using peripheral-blood stem-cell harvests in patients with multiple myeloma. Blood 2005; 106:3538-45. [PMID: 16037388 DOI: 10.1182/blood-2005-04-1438] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
HM1.24 antigen is preferentially overexpressed in multiple myeloma (MM) cells but not in normal cells. To explore the potential of HM1.24 as a target for cellular immunotherapy, we selected 4 HM1.24-derived peptides that possess binding motifs for HLA-A2 or HLA-A24 by using 2 computer-based algorithms. The ability of these peptides to generate cytotoxic T lymphocytes (CTLs) was examined in 20 healthy donors and 6 patients with MM by a reverse immunologic approach. Dendritic cells (DCs) were induced from peripheral-blood mononuclear cells of healthy donors or peripheral-blood stem-cell (PBSC) harvests from patients with MM, and autologous CD8(+) T cells were stimulated with HM1.24 peptide-pulsed DCs. Both interferon-gamma-producing and cytotoxic responses were observed after stimulation with either HM1.24-126 or HM1.24-165 peptides in HLA-A2 or HLA-A24 individuals. The peptide-specific recognition of these CTLs was further confirmed by tetramer assay and cold target inhibition assay. Importantly, HM1.24-specific CTLs were also induced from PBSC harvests from patients with MM and these CTLs were able to kill MM cells in an HLA-restricted manner. These results indicate the existence of functional DCs and HM1.24-specific CTL precursors within PBSC harvests and provide the basis for cellular immunotherapy in combination with autologous PBSC transplantation in MM.
Collapse
Affiliation(s)
- Ali Jalili
- Department of Medicine and Bioregulatory Sciences, Institute of Health Biosciences, University of Tokushima Graduate School, Japan
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Ishida T, Iida S, Akatsuka Y, Ishii T, Miyazaki M, Komatsu H, Inagaki H, Okada N, Fujita T, Shitara K, Akinaga S, Takahashi T, Utsunomiya A, Ueda R. The CC Chemokine Receptor 4 as a Novel Specific Molecular Target for Immunotherapy in Adult T-Cell Leukemia/Lymphoma. Clin Cancer Res 2004; 10:7529-39. [PMID: 15569983 DOI: 10.1158/1078-0432.ccr-04-0983] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Adult T-cell leukemia/lymphoma (ATLL) is a peripheral T-cell neoplasm with dismal prognosis, and no optimal therapy has been developed. We tested the defucosylated chimeric anti-CC chemokine receptor 4 (CCR4) monoclonal antibody, KM2760, to develop a novel immunotherapy for this refractory tumor. In the presence of peripheral blood mononuclear cells (PBMCs) from healthy adult donors, KM2760 induced CCR4-specific antibody-dependent cellular cytotoxicity (ADCC) against CCR4-positive ATLL cell lines and primary tumor cells obtained from ATLL patients. We next examined the KM2760-induced ADCC against primary ATLL cells in an autologous setting. Antibody-dependent cellular cytotoxicity mediated by autologous effector cells was generally lower than that mediated by allogeneic control effector cells. However, a robust ADCC activity was induced in some cases, which was comparable with that mediated by allogeneic effector cells. It suggests that the ATLL patients' PBMCs retain substantial ADCC-effector function, although the optimal conditions for maximal effect have not yet been determined. In addition, we also found a high expression of FoxP3 mRNA and protein, a hallmark of regulatory T cells, in ATLL cells, indicating the possibility that ATLL cells originated from regulatory T cells. KM2760 reduced FoxP3 mRNA expression in normal PBMCs along with CCR4 mRNA by lysis of CCR4+ T cells in vitro. Our data suggest not only that the CCR4 molecule could be a suitable target for the novel antibody-based therapy for patients with ATLL but also that KM2760 may induce effective tumor immunity by reducing the number of regulatory T cells.
Collapse
MESH Headings
- Antibodies, Monoclonal/chemistry
- Blotting, Western
- CD3 Complex/biosynthesis
- Cell Line, Tumor
- Cell Proliferation
- DNA-Binding Proteins/biosynthesis
- Dose-Response Relationship, Drug
- Flow Cytometry
- Forkhead Transcription Factors
- Genotype
- Humans
- Immunotherapy/methods
- Leukemia-Lymphoma, Adult T-Cell/metabolism
- Leukocytes, Mononuclear/cytology
- RNA, Messenger/metabolism
- Receptors, CCR4
- Receptors, Chemokine/metabolism
- Receptors, IgG/genetics
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Takashi Ishida
- Department of Internal Medicine & Molecular Science, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abe M, Hiura K, Wilde J, Shioyasono A, Moriyama K, Hashimoto T, Kido S, Oshima T, Shibata H, Ozaki S, Inoue D, Matsumoto T. Osteoclasts enhance myeloma cell growth and survival via cell-cell contact: a vicious cycle between bone destruction and myeloma expansion. Blood 2004; 104:2484-91. [PMID: 15187021 DOI: 10.1182/blood-2003-11-3839] [Citation(s) in RCA: 231] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
AbstractMultiple myeloma (MM) expands in the bone marrow and causes devastating bone destruction by enhancing osteoclastic bone resorption in its vicinity, suggesting a close interaction between MM cells and osteoclasts (OCs). Here, we show that peripheral blood mononuclear cell-derived OCs enhanced growth and survival of primary MM cells as well as MM cell lines more potently than stromal cells, and that OCs protected MM cells from apoptosis induced by serum depletion or doxorubicin. OCs produced osteopontin (OPN) and interleukin 6 (IL-6), and adhesion of MM cells to OCs increased IL-6 production from OCs. In addition, IL-6 and OPN in combination enhanced MM cell growth and survival. However, the effects of OCs on MM cell growth and survival were only partially suppressed by a simultaneous addition of anti–IL-6 and anti-OPN antibodies and were completely abrogated by inhibition of cellular contact between MM cells and OCs. These results demonstrate that OCs enhance MM cell growth and survival through a cell-cell contact-mediated mechanism that is partially dependent on IL-6 and OPN. It is suggested that interactions of MM cells with OCs augment MM growth and survival and, thereby, form a vicious cycle, leading to extensive bone destruction and MM cell expansion.
Collapse
Affiliation(s)
- Masahiro Abe
- Department of Medicine and Bioregulatory Sciences, University of Tokushima Graduate School of Medicine, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Hönemann D, Kufer P, Rimpler MM, Chatterjee M, Friedl S, Riecher F, Bommert K, Dörken B, Bargou RC. A novel recombinant bispecific single-chain antibody, bscWue-1 × CD3, induces T-cell-mediated cytotoxicity towards human multiple myeloma cells. Leukemia 2004; 18:636-44. [PMID: 14737072 DOI: 10.1038/sj.leu.2403264] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The development of antibody-based strategies for the treatment of multiple myeloma (MM) has been hampered so far by the fact that suitable plasma cell-specific surface antigens have been missing. However, recently a novel monoclonal antibody, designated Wue-1, has been generated that specifically recognizes normal and malignant human plasma cells. Therefore, Wue-1 is an interesting and promising candidate to develop novel immunotherapeutic strategies for the treatment of MM. One variant for an antibody-based strategy is the bispecific antibody approach. Recombinant bispecific single-chain (bsc) antibodies are especially interesting candidates because they show exceptional biological properties. We have generated a novel MM-directed recombinant bsc antibody, bscWue-1 x CD3, and analyzed the biological properties of this antibody using the MM cell line NCI-H929 and primary cells from the bone marrow of patients with MM. We were able to show that bscWue-1 x CD3 induces efficient and selective T-cell-mediated cell death of NCI-H929 cells and primary myeloma cells in nine out of 11 cases. The bscWue-1 x CD3 Ab is efficacious even at low E:T ratios, and with or without additional T-cell pre- or costimulation. Target cell lyses were specific for Wue-1 antigen-positive cells and could be blocked by the Wue-1 monoclonal antibody.
Collapse
Affiliation(s)
- D Hönemann
- Department of Hematology, Oncology and Tumor-Immunology, Helios Clinics, Robert-Rössle Cancer Center, University Medical Center Charité, Berlin, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Affiliation(s)
- George J Weiner
- Holden Comprehensive Cancer Center, Department of Internal Medicine, University of Iowa, Iowa City, Iowa 52242, USA
| | | |
Collapse
|
34
|
|
35
|
Watanabe K, Watanabe R, Shioda A, Mizoguchi K, Sugimoto T, Terao K. Investigation of the mechanism of drug-induced autoimmune hemolytic anemia in cynomolgus monkeys elicited by a repeated-dose of a humanized monoclonal antibody drug. J Toxicol Sci 2003; 28:123-38. [PMID: 12974605 DOI: 10.2131/jts.28.123] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
We investigated the mechanism of hemolytic anemia detected in a repeated-dose toxicity study using cynomolgus monkeys that were treated with a humanized antibody drug. This drug was an IgG1 monoclonal antibody (MoAb) that binds to the human HM1.24 antigen named anti-HM1.24 MoAb. The presence of the HM1.24 antigen on the erythrocyte membranes and the erythrocyte agglutination following the addition of anti-HM1.24 MoAb was examined. In addition, an indirect Coombs' test, a hemolysis assay and the measurement of anti-single stranded-DNA antibodies were performed using test animal serum or plasma. The specific binding of FITC- and 125I-labeled anti-HM1.24 MoAb to the erythrocyte membrane was not observed. HM1.24 antigen was not identified on the erythrocyte membranes. However, a high concentration (more than 713 microg/mL) of anti-HM1.24 MoAb hemagglutinated the erythrocyte suspensions. The cause of this agglutination was unclear, but it is assumed that the non-specific binding and/or adhesion caused the direct agglutination. In the examination using test serum from the anemic monkeys, a positive reaction in the indirect Coombs' test was noted. Moreover, in these Coombs' test-positive animals, the production of anti-single stranded-DNA antibodies was sequentially increased. In the female monkey sacrificed in extremis due to severe anemia, an in vitro hemolytic reaction was detected attributable to complement activation. From these results, the hemolytic anemia detected in the repeated-dose toxicity study was diagnosed as a drug-induced autoimmune hemolytic anemia (AIHA) and the primary cause was assumed to be production of IgG class anti-erythrocyte autoantibodies.
Collapse
Affiliation(s)
- Kazuto Watanabe
- Fuji Gotemba Laboratories, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba-shi, Shizuoka, 412-8513, Japan
| | | | | | | | | | | |
Collapse
|
36
|
Abstract
Over the past decade the potential for delivering targeted therapy against malignant disease by the use of monoclonal antibodies (MAbs) has begun to be realised. Haematological malignancies, because of the relative accessibility of the malignant cell in blood and bone marrow and the understanding of haemopoietic lineage-specific antigens, have provided a successful testing ground for this therapy. There have been many technical developments which have allowed the safe delivery of more potent antibody constructs. The development of human or chimeric antibodies has largely overcome the problems associated with host immune responses to rodent-derived MAbs. Protein engineering to combine MAbs with other biologically active molecules such as radioisotopes, toxins, chemotherapy and cytokines, has made available a new range of agents with clinical activity. The purpose of this review is not to give a catalogue of all therapeutic antibodies but rather to outline the principles of this approach, the current state of knowledge, and possible directions for future development. First, the general requirements and strategies for use of both unmodified and conjugated MAbs are discussed, followed by a summary of the trial data in specific lymphoid and myeloid haemopoietic malignancies. The focus is on MAbs that now have an accepted use in clinical practice, with some discussion of newer MAbs under development. Vaccination strategies and the role of MAbs in bone marrow transplantation are not discussed in detail. The trials of the next decade will address issues such as: whether clinical activity translates into improved survival; the optimal strategies and timing for clinical use; whether increasing potency of MAbs (as in radio- and immunoconjugates) will increase toxicity and, finally, what other potential molecules, such as those influencing cell growth and death, may be targeted.
Collapse
|
37
|
Peggs KS, Mackinnon S. Adoptive cellular therapy: a therapeutic reality? Hematology 2002; 7:127-36. [PMID: 12243974 DOI: 10.1080/1024533021000013915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
The past decade has seen numerous lines of evidence emerging that suggest human malignancies may be sensitive to the effects of cellular immunotherapy. An increasing understanding of the nature of the effector cells and their target antigens is now leading to more focused efforts to harness these responses for therapeutic benefit. However, clinical application has proven more challenging than initially envisaged. Advances in the setting of allogeneic stem cell transplantation now allow attempts to augment both immunological recovery and anti-tumour activity. Some of the most attractive targets here are allospecific rather than truly tumour-specific. Application outside of this setting is based on attempts to delineate further tumour-specific or, increasingly, tumour-selective targets. This review summarizes these developments and highlights some of the issues that remain to be resolved.
Collapse
Affiliation(s)
- Karl S Peggs
- Department of Haematology, University College Hospital, London, UK.
| | | |
Collapse
|
38
|
Role for macrophage inflammatory protein (MIP)-1α and MIP-1β in the development of osteolytic lesions in multiple myeloma. Blood 2002. [DOI: 10.1182/blood.v100.6.2195.h81802002195_2195_2202] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Multiple myeloma (MM) cells cause devastating bone destruction by activating osteoclasts in the bone marrow milieu. However, the mechanism of enhanced bone resorption in patients with myeloma is poorly understood. In the present study, we investigated a role of C-C chemokines, macrophage inflammatory protein (MIP)–1α and MIP-1β, in MM cell-induced osteolysis. These chemokines were produced and secreted by a majority of MM cell lines as well as primary MM cells from patients. Secretion of MIP-1α and MIP-1β correlated well with the ability of myeloma cells to enhance osteoclastic bone resorption both in vitro and in vivo as well as in MM patients. In osteoclastogenic cultures of rabbit bone cells, cocultures with myeloma cells as well as addition of myeloma cell-conditioned media enhanced both formation of osteoclastlike cells and resorption pits to an extent comparable to the effect of recombinant MIP-1α and MIP-1β. Importantly, these effects were mostly reversed by neutralizing antibodies against MIP-1α and MIP-1β, or their cognate receptor, CCR5, suggesting critical roles of these chemokines. We also demonstrated that stromal cells express CCR5 and that recombinant MIP-1α and MIP-1β induce expression of receptor activator of nuclear factor-κB (RANK) ligand by stromal cells, thereby stimulating osteoclast differentiation of preosteoclastic cells. These results suggest that MIP-1α and MIP-1β may be major osteoclast-activating factors produced by MM cells.
Collapse
|
39
|
Liso A, Benedetti R, Flenghi L, Falini B. Vaccine therapy of B cell malignancies: different strategies for a novel approach. Leuk Lymphoma 2001; 42:881-9. [PMID: 11697643 DOI: 10.3109/10428190109097707] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
This review deals with the theoretical principles and experimental results of immunotherapy for B cell malignancies, namely for non-Hodgkin lymphomas (NHLs) and multiple myeloma. Its focus is the use of vaccines in clinical practice with particular emphasis on the most recent developments and therapeutic opportunities arising from combination therapies. Previous studies will be reviewed and the present status of vaccine technology summarized.
Collapse
Affiliation(s)
- A Liso
- University of Perugia, Institute of Hematology, Italy.
| | | | | | | |
Collapse
|
40
|
Abstract
The failure of chemotherapy to cure a significant proportion of multiple myeloma (MM) patients and increasing knowledge of tumor immunology and MM biology have generated considerable interest in immunotherapy for this lethal disease. Immunotherapy for MM can be divided into three broad categories: passive antibody-mediated immunotherapy, active specific immunization (vaccination), and adoptive T-cell immunotherapy. Early clinical trials using anti-CD20 monoclonal antibodies (mAbs) have met with limited success so far but have also suggested that selected patient subgroups may benefit from this treatment. The availability of a truly tumor-specific antigen such as immunoglobulin idiotype, the recent demonstration that MM cells process and present idiotype to T lymphocytes, and formal evidence of an antitumor effect of idiotypic vaccination in follicular lymphoma provide the framework for applying idiotypic vaccination in MM. The ability to generate ex vivo functional dendritic cells has made it possible to fuse them with patients' MM cells, thus producing a different type of customized vaccine. Dendritic cells are also a pivotal reagent to generate ex vivo MM-specific cytotoxic T lymphocytes (CTLs) to be reinfused into the patient for adoptive immunotherapy. This review summarizes achievements in MM immunotherapy based on data reported since 1998.
Collapse
Affiliation(s)
- P A Ruffini
- Department of Experimental Transplantation and Immunology, Medicine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | |
Collapse
|