1
|
Kim JC, Kim Y, Cho S, Park HS. Noncanonical Amino Acid Incorporation in Animals and Animal Cells. Chem Rev 2024; 124:12463-12497. [PMID: 39541258 DOI: 10.1021/acs.chemrev.3c00955] [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: 11/16/2024]
Abstract
Noncanonical amino acids (ncAAs) are synthetic building blocks that, when incorporated into proteins, confer novel functions and enable precise control over biological processes. These small yet powerful tools offer unprecedented opportunities to investigate and manipulate various complex life forms. In particular, ncAA incorporation technology has garnered significant attention in the study of animals and their constituent cells, which serve as invaluable model organisms for gaining insights into human physiology, genetics, and diseases. This review will provide a comprehensive discussion on the applications of ncAA incorporation technology in animals and animal cells, covering past achievements, current developments, and future perspectives.
Collapse
Affiliation(s)
- Joo-Chan Kim
- Department of Chemistry, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - YouJin Kim
- Department of Chemistry, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Suho Cho
- Department of Chemistry, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Hee-Sung Park
- Department of Chemistry, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| |
Collapse
|
2
|
Jin WM, Zhu Y, Cai ZQ, He N, Yu ZQ, Li S, Yang JY. Progress of Clinical Studies Targeting Claudin18.2 for the Treatment of Gastric Cancer. Dig Dis Sci 2024; 69:2631-2647. [PMID: 38769225 DOI: 10.1007/s10620-024-08435-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 04/10/2024] [Indexed: 05/22/2024]
Abstract
Claudin18.2 is a tight junction protein, highly selective, generally expressed only in normal gastric mucosal epithelial cells, which can effectively maintain the polarity of epithelial and endothelial cells, thus effectively regulating the permeability and conductance of the paracellular pathway. Abnormal expression of Claudin18.2 can occur in various primary malignant tumors, especially gastrointestinal tumors, and even in metastatic foci. It regulates its expression by activating the aPKC/MAPK/AP-1 pathway, and therefore, the Claudin18.2 protein is a pan-cancer target expressed in primary and metastatic lesions in human cancer types. Zolbetuximab (IMAB362), an antibody specific for Claudin18.2, has been successfully tested in a phase III clinical trial, and the results of the study showed that combining Zolbetuximab with chemotherapy notably extends patients' survival and is expected to be a potential first-line treatment for patients with Claudin18.2(+)/HER-2(-) gastric cancer. Here, we systematically describe the biological properties and oncogenic effects of Claudin18.2, centering on its clinical-pathological aspects and the progress of drug studies in gastric cancer, which can help to further explore its clinical value.
Collapse
Affiliation(s)
- Wu-Mei Jin
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, People's Republic of China
| | - Yan Zhu
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, People's Republic of China
| | - Zhi-Qiang Cai
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, People's Republic of China
| | - Na He
- Department of General, First Affiliated Hospital of Yangtze University, Jingzhou, People's Republic of China
| | - Zhi-Qiong Yu
- Department of Respiratory, First Affiliated Hospital of Yangtze University, Jingzhou, People's Republic of China
| | - Shuang Li
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, People's Republic of China
| | - Ji-Yuan Yang
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, People's Republic of China.
| |
Collapse
|
3
|
Mitsiades CS. Proteasome Inhibitors in Multiple Myeloma: Biological Insights on Mechanisms of Action or Resistance Informed by Functional Genomics. Hematol Oncol Clin North Am 2024; 38:321-336. [PMID: 38278626 DOI: 10.1016/j.hoc.2023.12.016] [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] [Indexed: 01/28/2024]
Abstract
During the last 20 years, proteasome inhibitors have been a cornerstone for the therapeutic management of multiple myeloma (MM). This review highlights how MM research has evolved over time in terms of our understanding of the mechanistic basis for the pronounced clinical activity of proteasome inhibitors in MM, compared with the limited clinical applications of this drug class outside the setting of plasma cell dyscrasias.
Collapse
Affiliation(s)
- Constantine S Mitsiades
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA; Ludwig Center at Harvard, Boston, MA, USA.
| |
Collapse
|
4
|
Liu J, Xing L, Li J, Wen K, Liu N, Liu Y, Wu G, Wang S, Ogiya D, Song TY, Kurata K, Penailillo J, Morelli E, Wang T, Hong X, Gulla A, Tai YT, Munshi N, Richardson P, Carrasco R, Hideshima T, Anderson KC. Epigenetic regulation of CD38/CD48 by KDM6A mediates NK cell response in multiple myeloma. Nat Commun 2024; 15:1367. [PMID: 38355622 PMCID: PMC10866908 DOI: 10.1038/s41467-024-45561-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 01/29/2024] [Indexed: 02/16/2024] Open
Abstract
Anti-CD38 monoclonal antibodies like Daratumumab (Dara) are effective in multiple myeloma (MM); however, drug resistance ultimately occurs and the mechanisms behind this are poorly understood. Here, we identify, via two in vitro genome-wide CRISPR screens probing Daratumumab resistance, KDM6A as an important regulator of sensitivity to Daratumumab-mediated antibody-dependent cellular cytotoxicity (ADCC). Loss of KDM6A leads to increased levels of H3K27me3 on the promoter of CD38, resulting in a marked downregulation in CD38 expression, which may cause resistance to Daratumumab-mediated ADCC. Re-introducing CD38 does not reverse Daratumumab-mediated ADCC fully, which suggests that additional KDM6A targets, including CD48 which is also downregulated upon KDM6A loss, contribute to Daratumumab-mediated ADCC. Inhibition of H3K27me3 with an EZH2 inhibitor resulted in CD38 and CD48 upregulation and restored sensitivity to Daratumumab. These findings suggest KDM6A loss as a mechanism of Daratumumab resistance and lay down the proof of principle for the therapeutic application of EZH2 inhibitors, one of which is already FDA-approved, in improving MM responsiveness to Daratumumab.
Collapse
Affiliation(s)
- Jiye Liu
- Jerome Lipper Multiple Myeloma Center, Lebow Institute for Myeloma Therapeutics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Lijie Xing
- Department of Hematology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Jiang Li
- Clinical Big Data Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, 518107, China
| | - Kenneth Wen
- Jerome Lipper Multiple Myeloma Center, Lebow Institute for Myeloma Therapeutics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Ning Liu
- Jerome Lipper Multiple Myeloma Center, Lebow Institute for Myeloma Therapeutics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
| | - Yuntong Liu
- Jerome Lipper Multiple Myeloma Center, Lebow Institute for Myeloma Therapeutics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Gongwei Wu
- Center for Functional Cancer Epigenetics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Su Wang
- Vertex pharmaceuticals, Boston, MA, 02210, USA
| | - Daisuke Ogiya
- Department of Hematology and Oncology, School of Medicine, Tokai University, Isehara, 259-1193, Japan
| | - Tian-Yu Song
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Keiji Kurata
- Jerome Lipper Multiple Myeloma Center, Lebow Institute for Myeloma Therapeutics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Johany Penailillo
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Eugenio Morelli
- Jerome Lipper Multiple Myeloma Center, Lebow Institute for Myeloma Therapeutics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Tingjian Wang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Xiaoning Hong
- Clinical Big Data Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, 518107, China
| | - Annamaria Gulla
- Jerome Lipper Multiple Myeloma Center, Lebow Institute for Myeloma Therapeutics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), 10060, Italy
| | - Yu-Tzu Tai
- Jerome Lipper Multiple Myeloma Center, Lebow Institute for Myeloma Therapeutics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Nikhil Munshi
- Jerome Lipper Multiple Myeloma Center, Lebow Institute for Myeloma Therapeutics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Paul Richardson
- Jerome Lipper Multiple Myeloma Center, Lebow Institute for Myeloma Therapeutics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Ruben Carrasco
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02215, USA
| | - Teru Hideshima
- Jerome Lipper Multiple Myeloma Center, Lebow Institute for Myeloma Therapeutics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Kenneth C Anderson
- Jerome Lipper Multiple Myeloma Center, Lebow Institute for Myeloma Therapeutics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.
| |
Collapse
|
5
|
Yan X, Liu X, Zhao C, Chen GQ. Applications of synthetic biology in medical and pharmaceutical fields. Signal Transduct Target Ther 2023; 8:199. [PMID: 37169742 PMCID: PMC10173249 DOI: 10.1038/s41392-023-01440-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 03/15/2023] [Accepted: 03/24/2023] [Indexed: 05/13/2023] Open
Abstract
Synthetic biology aims to design or assemble existing bioparts or bio-components for useful bioproperties. During the past decades, progresses have been made to build delicate biocircuits, standardized biological building blocks and to develop various genomic/metabolic engineering tools and approaches. Medical and pharmaceutical demands have also pushed the development of synthetic biology, including integration of heterologous pathways into designer cells to efficiently produce medical agents, enhanced yields of natural products in cell growth media to equal or higher than that of the extracts from plants or fungi, constructions of novel genetic circuits for tumor targeting, controllable releases of therapeutic agents in response to specific biomarkers to fight diseases such as diabetes and cancers. Besides, new strategies are developed to treat complex immune diseases, infectious diseases and metabolic disorders that are hard to cure via traditional approaches. In general, synthetic biology brings new capabilities to medical and pharmaceutical researches. This review summarizes the timeline of synthetic biology developments, the past and present of synthetic biology for microbial productions of pharmaceutics, engineered cells equipped with synthetic DNA circuits for diagnosis and therapies, live and auto-assemblied biomaterials for medical treatments, cell-free synthetic biology in medical and pharmaceutical fields, and DNA engineering approaches with potentials for biomedical applications.
Collapse
Affiliation(s)
- Xu Yan
- School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Xu Liu
- PhaBuilder Biotech Co. Ltd., Shunyi District, Zhaoquan Ying, 101309, Beijing, China
| | - Cuihuan Zhao
- School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Guo-Qiang Chen
- School of Life Sciences, Tsinghua University, 100084, Beijing, China.
- Center for Synthetic and Systems Biology, Tsinghua University, 100084, Beijing, China.
- MOE Key Lab for Industrial Biocatalysis, Dept Chemical Engineering, Tsinghua University, 100084, Beijing, China.
| |
Collapse
|
6
|
Sibuh BZ, Gahtori R, Al-Dayan N, Pant K, Far BF, Malik AA, Gupta AK, Sadhu S, Dohare S, Gupta PK. Emerging trends in immunotoxin targeting cancer stem cells. Toxicol In Vitro 2022; 83:105417. [PMID: 35718257 DOI: 10.1016/j.tiv.2022.105417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/10/2022] [Accepted: 06/11/2022] [Indexed: 12/30/2022]
Abstract
Cancer stem cells (CSCs) are self-renewing multipotent cells that play a vital role in the development of cancer drug resistance conditions. Various therapies like conventional, targeted, and radiotherapies have been broadly used in targeting and killing these CSCs. Among these, targeted therapy selectively targets CSCs and leads to overcoming disease recurrence conditions in cancer patients. Immunotoxins (ITs) are protein-based therapeutics with selective targeting capabilities. These chimeric molecules are composed of two functional moieties, i.e., a targeting moiety for cell surface binding and a toxin moiety that induces the programmed cell death upon internalization. Several ITs have been constructed recently, and their preclinical and clinical efficacies have been evaluated. In this review, we comprehensively discussed the recent preclinical and clinical advances as well as significant challenges in ITs targeting CSCs, which might reduce the burden of drug resistance conditions in cancer patients from bench to bedside.
Collapse
Affiliation(s)
- Belay Zeleke Sibuh
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Knowledge Park III, Greater Noida 201310, Uttar Pradesh, India
| | - Rekha Gahtori
- Department of Biotechnology, Sir J.C. Bose Technical Campus, Kumaun University, Bhimtal, Nainital 263136, Uttarakhand, India
| | - Noura Al-Dayan
- Department of Medical Lab Sciences, Prince Sattam bin Abdulaziz University, Alkharj 16278, Saudi Arabia
| | - Kumud Pant
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun 248002, Uttarakhand, India
| | - Bahareh Farasati Far
- Department of Chemistry, Iran University of Science and Technology, Tehran, Iran
| | - Asrar Ahmad Malik
- Department of Life Sciences, School of Basic Sciences and Research (SBSR), Sharda University, Knowledge Park III, Greater Noida 201310, Uttar Pradesh, India
| | - Ashish Kumar Gupta
- Department of Life Sciences, J.C. Bose University of Science and Technology, YMCA, Faridabad 121006, Haryana, India
| | - Soumi Sadhu
- Department of Life Sciences, School of Basic Sciences and Research (SBSR), Sharda University, Knowledge Park III, Greater Noida 201310, Uttar Pradesh, India
| | - Sushil Dohare
- Department of Epidemiology, Faculty of Public Health & Tropical Medicine, Jazan University, Jazan, Saudi Arabia
| | - Piyush Kumar Gupta
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun 248002, Uttarakhand, India; Department of Life Sciences, School of Basic Sciences and Research (SBSR), Sharda University, Knowledge Park III, Greater Noida 201310, Uttar Pradesh, India.
| |
Collapse
|
7
|
Lateef OM, Akintubosun MO, Olaoba OT, Samson SO, Adamczyk M. Making Sense of "Nonsense" and More: Challenges and Opportunities in the Genetic Code Expansion, in the World of tRNA Modifications. Int J Mol Sci 2022; 23:938. [PMID: 35055121 PMCID: PMC8779196 DOI: 10.3390/ijms23020938] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/10/2022] [Accepted: 01/12/2022] [Indexed: 01/09/2023] Open
Abstract
The evolutional development of the RNA translation process that leads to protein synthesis based on naturally occurring amino acids has its continuation via synthetic biology, the so-called rational bioengineering. Genetic code expansion (GCE) explores beyond the natural translational processes to further enhance the structural properties and augment the functionality of a wide range of proteins. Prokaryotic and eukaryotic ribosomal machinery have been proven to accept engineered tRNAs from orthogonal organisms to efficiently incorporate noncanonical amino acids (ncAAs) with rationally designed side chains. These side chains can be reactive or functional groups, which can be extensively utilized in biochemical, biophysical, and cellular studies. Genetic code extension offers the contingency of introducing more than one ncAA into protein through frameshift suppression, multi-site-specific incorporation of ncAAs, thereby increasing the vast number of possible applications. However, different mediating factors reduce the yield and efficiency of ncAA incorporation into synthetic proteins. In this review, we comment on the recent advancements in genetic code expansion to signify the relevance of systems biology in improving ncAA incorporation efficiency. We discuss the emerging impact of tRNA modifications and metabolism in protein design. We also provide examples of the latest successful accomplishments in synthetic protein therapeutics and show how codon expansion has been employed in various scientific and biotechnological applications.
Collapse
Affiliation(s)
- Olubodun Michael Lateef
- Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland; (O.M.L.); (M.O.A.); (S.O.S.)
| | | | - Olamide Tosin Olaoba
- Laboratory of Functional and Structural Biochemistry, Federal University of Sao Carlos, Sao Carlos 13565-905, SP, Brazil;
| | - Sunday Ocholi Samson
- Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland; (O.M.L.); (M.O.A.); (S.O.S.)
| | - Malgorzata Adamczyk
- Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland; (O.M.L.); (M.O.A.); (S.O.S.)
| |
Collapse
|
8
|
Tang H, Zhang P, Luo X. Recent Technologies for Genetic Code Expansion and their Implications on Synthetic Biology Applications. J Mol Biol 2021; 434:167382. [PMID: 34863778 DOI: 10.1016/j.jmb.2021.167382] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 11/18/2021] [Accepted: 11/24/2021] [Indexed: 02/03/2023]
Abstract
Genetic code expansion (GCE) enables the site-specific incorporation of non-canonical amino acids as novel building blocks for the investigation and manipulation of proteins. The advancement of genetic code expansion has been benefited from the development of synthetic biology, while genetic code expansion also helps to create more synthetic biology tools. In this review, we summarize recent advances in genetic code expansion brought by synthetic biology progresses, including engineering of the translation machinery, genome-wide codon reassignment, and the biosynthesis of non-canonical amino acids. We highlight the emerging application of this technology in construction of new synthetic biology parts, circuits, chassis, and products.
Collapse
Affiliation(s)
- Hongting Tang
- Center for Synthetic Biochemistry, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Pan Zhang
- Center for Synthetic Biochemistry, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xiaozhou Luo
- Center for Synthetic Biochemistry, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| |
Collapse
|
9
|
Abstract
Multiple myeloma is the second most common hematological malignancy in adults, accounting for 2% of all cancer-related deaths in the UK. Current chemotherapy-based regimes are insufficient, as most patients relapse and develop therapy resistance. This review focuses on current novel antibody- and aptamer-based therapies aiming to overcome current therapy limitations, as well as their respective limitations and areas of improvement. The use of computer modeling methods, as a tool to study and improve ligand-receptor alignments for the use of novel therapy development will also be discussed, as it has become a rapid, reliable and comparatively inexpensive method of investigation.
Collapse
|
10
|
Facile Generation of Potent Bispecific Fab via Sortase A and Click Chemistry for Cancer Immunotherapy. Cancers (Basel) 2021; 13:cancers13184540. [PMID: 34572769 PMCID: PMC8467688 DOI: 10.3390/cancers13184540] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/04/2021] [Accepted: 09/06/2021] [Indexed: 12/18/2022] Open
Abstract
Simple Summary The formats of bispecific antibody have been investigated for many years to enhance the stability of the structure and anti-tumor efficacy. One of the formats combining two Fabs at their C termini provides unmodified variable region and comparable activity to other fragment-based bispecific antibodies that are usually combined in a head-to-tail manner. However, the current strategy to produce the BiFab molecule is limited to a semisynthetic method that introduces unnatural amino acid to antibodies’ sequences during production. To improve the application of BiFab format in investigational biodrugs, we have applied sortase A-mediated “bio-click” chemistry to generate BiFab, for facile assembly of Fab molecules that have been expressed and stored as BiFab module candidates. The BiFabs made by our method stimulate T cell proliferation and activation with favorable in vitro and in vivo anti-tumor activit. Our results indicate that BiFab made by sortase A-mediated click chemistry could be used to efficiently generate various BiFabs with high potency, which further supports personalized tumor immunotherapy in the future. Abstract Bispecific antibodies (BsAbs) for T cell engagement have shown great promise in cancer immunotherapy, and their clinical applications have been proven in treating hematological malignance. Bispecific antibody binding fragment (BiFab) represents a promising platform for generating non-Fc bispecific antibodies. However, the generation of BiFab is still challenging, especially by means of chemical conjugation. More conjugation strategies, e.g., enzymatic conjugation and modular BiFab preparation, are needed to improve the robustness and flexibility of BiFab preparation. We successfully used chemo-enzymatic conjugation approach to generate bispecific antibody (i.e., BiFab) with Fabs from full-length antibodies. Paired click handles (e.g., N3 and DBCO) was introduced to the C-terminal LPETG tag of Fabs via sortase A mediated transpeptidation, followed by site-specific conjugation between two click handle-modified Fabs for BiFab generation. Both BiFabCD20/CD3 (EC50 = 0.26 ng/mL) and BiFabHer2/CD3 exhibited superior efficacy in mediating T cells, from either PBMC or ATC, to kill target tumor cell lines while spared antigen-negative tumor cells in vitro. The BiFabCD20/CD3 also efficiently inhibited CD20-positive tumor growth in mouse xenograft model. We have established a facile sortase A-mediated click handle installation to generate homogeneous and functional BiFabs. The exemplary BiFabs against different targets showed superior efficacy in redirecting and activating T cells to specifically kill target tumor cells, demonstrating the robustness of sortase A-mediated “bio-click” chemistry in generating various potent BiFabs. This approach also holds promise for further efficient construction of a Fab derivative library for personalized tumor immunotherapy in the future.
Collapse
|
11
|
Manandhar M, Chun E, Romesberg FE. Genetic Code Expansion: Inception, Development, Commercialization. J Am Chem Soc 2021; 143:4859-4878. [DOI: 10.1021/jacs.0c11938] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Miglena Manandhar
- Synthorx, a Sanofi Company, La Jolla, California 92037, United States
| | - Eugene Chun
- Synthorx, a Sanofi Company, La Jolla, California 92037, United States
| | | |
Collapse
|
12
|
Fang Y, Hou J. Immunotherapeutic strategies targeting B cell maturation antigen in multiple myeloma. Mil Med Res 2021; 8:9. [PMID: 33504363 PMCID: PMC7839214 DOI: 10.1186/s40779-021-00302-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 01/19/2021] [Indexed: 12/31/2022] Open
Abstract
Multiple myeloma (MM) is the second most common hematologic malignancy, and is characterized by the clonal expansion of malignant plasma cells. Despite the recent improvement in patient outcome due to the use of novel therapeutic agents and stem cell transplantation, all patients eventually relapse due to clone evolution. B cell maturation antigen (BCMA) is highly expressed in and specific for MM cells, and has been implicated in the pathogenesis as well as treatment development for MM. In this review, we will summarize representative anti-BCMA immune therapeutic strategies, including BCMA-targeted vaccines, anti-BCMA antibodies and BCMA-targeted CAR cells. Combination of different immunotherapeutic strategies of targeting BCMA, multi-target immune therapeutic strategies, and adding immune modulatory agents to normalize anti-MM immune system in minimal residual disease (MRD) negative patients, will also be discussed.
Collapse
Affiliation(s)
- Yi Fang
- Department of Hematology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Jian Hou
- Department of Hematology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China.
| |
Collapse
|
13
|
Verkleij CP, Frerichs KA, Broekmans M, Absalah S, Maas-Bosman PW, Kruyswijk S, Nijhof IS, Mutis T, Zweegman S, van de Donk NW. T-cell redirecting bispecific antibodies targeting BCMA for the treatment of multiple myeloma. Oncotarget 2020; 11:4076-4081. [PMID: 33227097 PMCID: PMC7665238 DOI: 10.18632/oncotarget.27792] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 10/13/2020] [Indexed: 12/13/2022] Open
Abstract
B-cell maturation antigen (BCMA)-targeting bispecific antibodies and bispecific T-cell engagers (BiTEs) redirect T-cells to BCMA-expressing multiple myeloma (MM) cells. These MM cells are subsequently eliminated via various mechanisms of action including the release of granzymes and perforins. Several phase 1, dose-escalation studies show pronounced activity of BCMA-targeting bispecific antibodies, including teclistamab, AMG420 and CC-93269, in heavily pretreated MM patients. Cytokine release syndrome is the most common adverse event, which can be adequately managed with tocilizumab or steroids. Several clinical trials are currently evaluating combination therapy with a BCMA-specific bispecific antibody, based on preclinical findings showing that immunomodulatory drugs or CD38-targeting antibodies enhance the activity of bispecific antibodies. In addition, bispecific antibodies, targeting other MM cell surface antigens (i. e. GPRC5D, CD38 and FcRH5), are also evaluated in early phase clinical trials. Such bispecific antibodies, targeting other antigens, may be given to patients with low baseline BCMA expression, disease with substantial heterogeneity in BCMA expression, following prior BCMA-targeted therapy, or combined with BCMA bispecific antibodies to prevent development of antigen escape.
Collapse
Affiliation(s)
- Christie P.M. Verkleij
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Shared first authors
| | - Kristine A. Frerichs
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
- Shared first authors
| | - Marloes Broekmans
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
| | - Saida Absalah
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
| | | | - Sandy Kruyswijk
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
| | - Inger S. Nijhof
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
| | - Tuna Mutis
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
| | - Sonja Zweegman
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
| | - Niels W.C.J. van de Donk
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, The Netherlands
| |
Collapse
|
14
|
Bera TK. Anti-BCMA Immunotoxins: Design, Production, and Preclinical Evaluation. Biomolecules 2020; 10:biom10101387. [PMID: 33003418 PMCID: PMC7600380 DOI: 10.3390/biom10101387] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/21/2020] [Accepted: 09/24/2020] [Indexed: 11/16/2022] Open
Abstract
Multiple myeloma (MM) is a B-cell malignancy that is incurable for a majority of patients. B-cell maturation antigen (BCMA) is a lineage-restricted differentiation protein highly expressed in multiple myeloma cells but not in other normal tissues except normal plasma B cells. Due to the restricted expression and being a cell surface membrane protein, BCMA is an ideal target for immunotherapy approaches in MM. Recombinant immunotoxins (RITs) are a novel class of protein therapeutics that are composed of the Fv or Fab portion of an antibody fused to a cytotoxic agent. RITs were produced by expressing plasmids encoding the components of the anti-BCMA RITs in E. coli followed by inclusion body preparation, solubilization, renaturation, and purification by column chromatography. The cytotoxic activity of RITs was tested in vitro by WST-8 assays using BCMA expressing cell lines and on cells isolated from MM patients. The in vivo efficacy of RITs was tested in a xenograft mouse model using BCMA expressing multiple myeloma cell lines. Anti-BCMA recombinant immunotoxins are very effective in killing myeloma cell lines and cells isolated from myeloma patients expressing BCMA. Two mouse models of myeloma showed that the anti-BCMA immunotoxins can produce a long-term complete response and warrant further preclinical development.
Collapse
Affiliation(s)
- Tapan K Bera
- Laboratory of Molecular Biology, Center for Cancer Research, NCI, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
15
|
Soekojo CY, Ooi M, de Mel S, Chng WJ. Immunotherapy in Multiple Myeloma. Cells 2020; 9:E601. [PMID: 32138182 PMCID: PMC7140529 DOI: 10.3390/cells9030601] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 02/27/2020] [Accepted: 02/27/2020] [Indexed: 12/15/2022] Open
Abstract
Multiple myeloma is a complex disease and immune dysfunction has been known to play an important role in the disease pathogenesis, progression, and drug resistance. Recent efforts in drug development have been focused on immunotherapies to modify the MM disease process. Here, we summarize the emerging immunotherapies in the MM treatment landscape.
Collapse
Affiliation(s)
| | | | | | - Wee Joo Chng
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, National University Health System, 1E Kent Ridge Road, Singapore 119228, Singapore; (C.Y.S.); (M.O.); (S.d.M.)
| |
Collapse
|
16
|
Frerichs KA, Broekmans MEC, Marin Soto JA, van Kessel B, Heymans MW, Holthof LC, Verkleij CPM, Boominathan R, Vaidya B, Sendecki J, Axel A, Gaudet F, Pillarisetti K, Zweegman S, Adams HC, Mutis T, van de Donk NWCJ. Preclinical Activity of JNJ-7957, a Novel BCMA×CD3 Bispecific Antibody for the Treatment of Multiple Myeloma, Is Potentiated by Daratumumab. Clin Cancer Res 2020; 26:2203-2215. [PMID: 31969333 DOI: 10.1158/1078-0432.ccr-19-2299] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 12/03/2019] [Accepted: 01/17/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Multiple myeloma (MM) patients with disease refractory to all available drugs have a poor outcome, indicating the need for new agents with novel mechanisms of action. EXPERIMENTAL DESIGN We evaluated the anti-MM activity of the fully human BCMA×CD3 bispecific antibody JNJ-7957 in cell lines and bone marrow (BM) samples. The impact of several tumor- and host-related factors on sensitivity to JNJ-7957 therapy was also evaluated. RESULTS We show that JNJ-7957 has potent activity against 4 MM cell lines, against tumor cells in 48 of 49 BM samples obtained from MM patients, and in 5 of 6 BM samples obtained from primary plasma cell leukemia patients. JNJ-7957 activity was significantly enhanced in patients with prior daratumumab treatment, which was partially due to enhanced killing capacity of daratumumab-exposed effector cells. BCMA expression did not affect activity of JNJ-7957. High T-cell frequencies and high effector:target ratios were associated with improved JNJ-7957-mediated lysis of MM cells. The PD-1/PD-L1 axis had a modest negative impact on JNJ-7957 activity against tumor cells from daratumumab-naïve MM patients. Soluble BCMA impaired the ability of JNJ-7957 to kill MM cells, although higher concentrations were able to overcome this negative effect. CONCLUSIONS JNJ-7957 effectively kills MM cells ex vivo, including those from heavily pretreated MM patients, whereby several components of the immunosuppressive BM microenvironment had only modest effects on its killing capacity. Our findings support the ongoing trial with JNJ-7957 as single agent and provide the preclinical rationale for evaluating JNJ-7957 in combination with daratumumab in MM.
Collapse
Affiliation(s)
- Kristine A Frerichs
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Marloes E C Broekmans
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Jhon A Marin Soto
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Berris van Kessel
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Martijn W Heymans
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Epidemiology and Biostatistics, Amsterdam, the Netherlands
| | - Lisa C Holthof
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Christie P M Verkleij
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | | | - Bhavesh Vaidya
- Janssen Research and Development, LLC, Spring House, Pennsylvania
| | - Jocelyn Sendecki
- Janssen Research and Development, LLC, Spring House, Pennsylvania
| | - Amy Axel
- Janssen Research and Development, LLC, Spring House, Pennsylvania
| | - Francois Gaudet
- Janssen Research and Development, LLC, Spring House, Pennsylvania
| | | | - Sonja Zweegman
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Homer C Adams
- Janssen Research and Development, LLC, Spring House, Pennsylvania
| | - Tuna Mutis
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Niels W C J van de Donk
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
17
|
Pont MJ, Hill T, Cole GO, Abbott JJ, Kelliher J, Salter AI, Hudecek M, Comstock ML, Rajan A, Patel BKR, Voutsinas JM, Wu Q, Liu L, Cowan AJ, Wood BL, Green DJ, Riddell SR. γ-Secretase inhibition increases efficacy of BCMA-specific chimeric antigen receptor T cells in multiple myeloma. Blood 2019; 134:1585-1597. [PMID: 31558469 PMCID: PMC6871311 DOI: 10.1182/blood.2019000050] [Citation(s) in RCA: 225] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 08/20/2019] [Indexed: 12/15/2022] Open
Abstract
B-cell maturation antigen (BCMA) is a validated target for chimeric antigen receptor (CAR) T-cell therapy in multiple myeloma (MM). Despite promising objective response rates, most patients relapse, and low levels of BCMA on a subset of tumor cells has been suggested as a probable escape mechanism. BCMA is actively cleaved from the tumor cell surface by the ubiquitous multisubunit γ-secretase (GS) complex, which reduces ligand density on tumor cells for CAR T-cell recognition and releases a soluble BCMA (sBCMA) fragment capable of inhibiting CAR T-cell function. Sufficient sBCMA can accumulate in the bone marrow of MM patients to inhibit CAR T-cell recognition of tumor cells, and potentially limit efficacy of BCMA-directed adoptive T-cell therapy. We investigated whether blocking BCMA cleavage by small-molecule GS inhibitors (GSIs) could augment BCMA-targeted CAR T-cell therapy. We found that exposure of myeloma cell lines and patient tumor samples to GSIs markedly increased surface BCMA levels in a dose-dependent fashion, concurrently decreased sBCMA concentrations, and improved tumor recognition by CAR T cells in vitro. GSI treatment of MM tumor-bearing NOD/SCID/γc-/- mice increased BCMA expression on tumor cells, decreased sBCMA in peripheral blood, and improved antitumor efficacy of BCMA-targeted CAR T-cell therapy. Importantly, short-term GSI administration to MM patients markedly increases the percentage of BCMA+ tumor cells, and the levels of BCMA surface expression in vivo. Based on these data, a US Food and Drug Administration (FDA)-approved clinical trial has been initiated, combining GSI with concurrent BCMA CAR T-cell therapy. This trial was registered at www.clinicaltrials.gov as #NCT03502577.
Collapse
Affiliation(s)
- Margot J Pont
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Tyler Hill
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Gabriel O Cole
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Joe J Abbott
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Jessica Kelliher
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Alexander I Salter
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Michael Hudecek
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany; and
| | - Melissa L Comstock
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Anusha Rajan
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA
| | | | - Jenna M Voutsinas
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Qian Wu
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Lingfeng Liu
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Andrew J Cowan
- Department of Medicine, University of Washington, Seattle, WA
| | - Brent L Wood
- Department of Medicine, University of Washington, Seattle, WA
| | - Damian J Green
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Stanley R Riddell
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
18
|
Gu W, An J, Meng H, Yu N, Zhong Y, Meng F, Xu Y, Cornelissen JJLM, Zhong Z. CD44-Specific A6 Short Peptide Boosts Targetability and Anticancer Efficacy of Polymersomal Epirubicin to Orthotopic Human Multiple Myeloma. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1904742. [PMID: 31560141 DOI: 10.1002/adma.201904742] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 09/11/2019] [Indexed: 06/10/2023]
Abstract
Chemotherapy is widely used in the clinic though its benefits are controversial owing to low cancer specificity. Nanovehicles capable of selectively transporting drugs to cancer cells have been energetically pursued to remodel cancer treatment. However, no active targeting nanomedicines have succeeded in clinical translation to date, partly due to either modest targetability or complex fabrication. CD44-specific A6 short peptide (KPSSPPEE) functionalized polymersomal epirubicin (A6-PS-EPI), which boosts targetability and anticancer efficacy toward human multiple myeloma (MM) in vivo, is described. A6-PS-EPI encapsulating 11 wt% EPI is small (≈55 nm), robust, reduction-responsive, and easy to fabricate. Of note, A6 decoration markedly augments the uptake and anticancer activity of PS-EPI in CD44-overexpressing LP-1 MM cells. A6-PS-EPI displays remarkable targeting ability to orthotopic LP-1 MM, causing depleted bone damage and striking survival benefits compared to nontargeted PS-EPI. Overall, A6-PS-EPI, as a simple and intelligent nanotherapeutic, demonstrates high potential for clinical translation.
Collapse
Affiliation(s)
- Wenxing Gu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
- Department of Biomolecular Nanotechnology, MESA+ Institute for Nanotechnology, University of Twente, 7500 AE, Enschede, The Netherlands
| | - Jingnan An
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215123, P. R. China
| | - Hao Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Na Yu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Yinan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Yang Xu
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215123, P. R. China
| | - Jeroen J L M Cornelissen
- Department of Biomolecular Nanotechnology, MESA+ Institute for Nanotechnology, University of Twente, 7500 AE, Enschede, The Netherlands
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| |
Collapse
|
19
|
Sanchez E, Smith EJ, Yashar MA, Patil S, Li M, Porter AL, Tanenbaum EJ, Schlossberg RE, Soof CM, Hekmati T, Tang G, Wang CS, Chen H, Berenson JR. The Role of B-Cell Maturation Antigen in the Biology and Management of, and as a Potential Therapeutic Target in, Multiple Myeloma. Target Oncol 2019; 13:39-47. [PMID: 29230672 DOI: 10.1007/s11523-017-0538-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
B-cell maturation antigen (BCMA) was originally identified as a cell membrane receptor, expressed exclusively on late stage B-cells and plasma cells (PCs). Investigations of BCMA as a target for therapeutic intervention in multiple myeloma (MM) were initiated in 2007, using cSG1 as a naked antibody (Ab) as well as an Ab-drug conjugate (ADC) targeting BCMA, ultimately leading to ongoing clinical studies for previously treated MM patients. Since then, multiple companies have developed anti-BCMA-directed ADCs. Additionally, there are now three bispecific antibodies in development, which bind to both BCMA and CD3ε on T-cells. This latter binding results in T-cell recruitment and activation, causing target cell lysis. More recently, T-cells have been genetically engineered to recognize BCMA-expressing cells and, in 2013, the first report of anti-BCMA-chimeric antigen receptor T-cells showed that these killed MM cell lines and human MM xenografts in mice. BCMA is also solubilized in the blood (soluble BCMA [sBCMA]) and MM patients with progressive disease have significantly higher sBCMA levels than those responding to treatment. sBCMA circulating in the blood may limit the efficacy of these anti-BCMA-directed therapies. When sBCMA binds to B-cell activating factor (BAFF), BAFF is unable to perform its major biological function of inducing B-cell proliferation and differentiation into Ab-secreting PC. However, the use of γ-secretase inhibitors, which prevent shedding of BCMA from PCs, may improve the efficacy of these BCMA-directed therapies.
Collapse
Affiliation(s)
- Eric Sanchez
- Institute for Myeloma & Bone Cancer Research, 9201 W Sunset Blvd, Suite 300, West Hollywood, CA, 90069, USA
| | - Emily J Smith
- Institute for Myeloma & Bone Cancer Research, 9201 W Sunset Blvd, Suite 300, West Hollywood, CA, 90069, USA
| | - Moryel A Yashar
- Institute for Myeloma & Bone Cancer Research, 9201 W Sunset Blvd, Suite 300, West Hollywood, CA, 90069, USA
| | - Saurabh Patil
- Institute for Myeloma & Bone Cancer Research, 9201 W Sunset Blvd, Suite 300, West Hollywood, CA, 90069, USA
| | - Mingjie Li
- Institute for Myeloma & Bone Cancer Research, 9201 W Sunset Blvd, Suite 300, West Hollywood, CA, 90069, USA
| | - Autumn L Porter
- Institute for Myeloma & Bone Cancer Research, 9201 W Sunset Blvd, Suite 300, West Hollywood, CA, 90069, USA
| | - Edward J Tanenbaum
- Institute for Myeloma & Bone Cancer Research, 9201 W Sunset Blvd, Suite 300, West Hollywood, CA, 90069, USA
| | - Remy E Schlossberg
- Institute for Myeloma & Bone Cancer Research, 9201 W Sunset Blvd, Suite 300, West Hollywood, CA, 90069, USA
| | - Camilia M Soof
- Institute for Myeloma & Bone Cancer Research, 9201 W Sunset Blvd, Suite 300, West Hollywood, CA, 90069, USA
| | - Tara Hekmati
- Institute for Myeloma & Bone Cancer Research, 9201 W Sunset Blvd, Suite 300, West Hollywood, CA, 90069, USA
| | - George Tang
- Institute for Myeloma & Bone Cancer Research, 9201 W Sunset Blvd, Suite 300, West Hollywood, CA, 90069, USA
| | - Cathy S Wang
- Institute for Myeloma & Bone Cancer Research, 9201 W Sunset Blvd, Suite 300, West Hollywood, CA, 90069, USA
| | - Haiming Chen
- Institute for Myeloma & Bone Cancer Research, 9201 W Sunset Blvd, Suite 300, West Hollywood, CA, 90069, USA
| | - James R Berenson
- Institute for Myeloma & Bone Cancer Research, 9201 W Sunset Blvd, Suite 300, West Hollywood, CA, 90069, USA.
| |
Collapse
|
20
|
Panowski SH, Kuo TC, Zhang Y, Chen A, Geng T, Aschenbrenner L, Kamperschroer C, Pascua E, Chen W, Delaria K, Farias S, Bateman M, Dushin RG, Chin SM, Van Blarcom TJ, Yeung YA, Lindquist KC, Chunyk AG, Kuang B, Han B, Mirsky M, Pardo I, Buetow B, Martin TG, Wolf JL, Shelton D, Rajpal A, Strop P, Chaparro-Riggers J, Sasu BJ. Preclinical Efficacy and Safety Comparison of CD3 Bispecific and ADC Modalities Targeting BCMA for the Treatment of Multiple Myeloma. Mol Cancer Ther 2019; 18:2008-2020. [DOI: 10.1158/1535-7163.mct-19-0007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 06/03/2019] [Accepted: 08/12/2019] [Indexed: 11/16/2022]
|
21
|
Zhu G, Foletti D, Liu X, Ding S, Melton Witt J, Hasa-Moreno A, Rickert M, Holz C, Aschenbrenner L, Yang AH, Kraynov E, Evering W, Obert L, Lee C, Sai T, Mistry T, Lindquist KC, Van Blarcom T, Strop P, Chaparro-Riggers J, Liu SH. Targeting CLDN18.2 by CD3 Bispecific and ADC Modalities for the Treatments of Gastric and Pancreatic Cancer. Sci Rep 2019; 9:8420. [PMID: 31182754 PMCID: PMC6557842 DOI: 10.1038/s41598-019-44874-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 05/27/2019] [Indexed: 02/07/2023] Open
Abstract
Human CLDN18.2 is highly expressed in a significant proportion of gastric and pancreatic adenocarcinomas, while normal tissue expression is limited to the epithelium of the stomach. The restricted expression makes it a potential drug target for the treatment of gastric and pancreatic adenocarcinoma, as evidenced by efforts to target CLDN18.2 via naked antibody and CAR-T modalities. Herein we describe CLDN18.2-targeting via a CD3-bispecific and an antibody drug conjugate and the characterization of these potential therapeutic molecules in efficacy and preliminary toxicity studies. Anti-hCLDN18.2 ADC, CD3-bispecific and diabody, targeting a protein sequence conserved in rat, mouse and monkey, exhibited in vitro cytotoxicity in BxPC3/hCLDN18.2 (IC50 = 1.52, 2.03, and 0.86 nM) and KATO-III/hCLDN18.2 (IC50 = 1.60, 0.71, and 0.07 nM) respectively and inhibited tumor growth of pancreatic and gastric patient-derived xenograft tumors. In a rat exploratory toxicity study, the ADC was tolerated up to 10 mg/kg. In a preliminary assessment of tolerability, the anti-CLDN18.2 diabody (0.34 mg/kg) did not produce obvious signs of toxicity in the stomach of NSG mice 4 weeks after dosing. Taken together, our data indicate that targeting CLDN18.2 with an ADC or bispecific modality could be a valid therapeutic approach for the treatment of gastric and pancreatic cancer.
Collapse
Affiliation(s)
- Guoyun Zhu
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.
| | - Davide Foletti
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,23 and Me, 349 Oyster Point Blvd, South San Francisco, CA, 94080, USA
| | - Xiaohui Liu
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA
| | - Sheng Ding
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,Gilead Sciences, 333 Lakeside Drive, Foster City, CA, 94404, USA
| | - Jody Melton Witt
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,Grifols Diagnostic Solutions, 6455 Christie Ave B-334C, Emeryville, CA, 94608, USA
| | - Adela Hasa-Moreno
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,Kodiak Sciences Inc., 2631 Hanover St, Palo Alto, CA, 94304, USA
| | - Mathias Rickert
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,Applied Molecular Transport, 1 Tower Place, Suite 850, South San Francisco, CA, 94080, USA
| | - Charles Holz
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,Grifols Diagnostic Solutions, 6455 Christie Ave B-334C, Emeryville, CA, 94608, USA
| | - Laura Aschenbrenner
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, 10646 Science Center Dr., San Diego, CA, 92121, USA.,Covance Inc. Early Phase Development Solutions, 3301 Kinsman Blvd, Madison, WI, 53704, USA
| | - Amy H Yang
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, 10646 Science Center Dr., San Diego, CA, 92121, USA
| | - Eugenia Kraynov
- BioMedicine Design, Pfizer Worldwide Research and Development, 10646 Science Center Dr., San Diego, CA, 92121, USA
| | - Winston Evering
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, 10646 Science Center Dr., San Diego, CA, 92121, USA
| | - Leslie Obert
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, 280 Shennecossett Rd, Groton, CT, 06340, USA.,GSK, 1250 South Collegeville Road, Collegeville, PA, 19426, USA
| | - Chenyu Lee
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,Alector, 151 Oyster Point Blvd #300, South San Francisco, CA, 94080, USA
| | - Tao Sai
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA
| | - Tina Mistry
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA
| | - Kevin C Lindquist
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA
| | - Thomas Van Blarcom
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,Allogene Therapeutics, 210 E. Grand Avenue, South San Francisco, CA, 94080, USA
| | - Pavel Strop
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,Bristol-Myers Squibb, 700 Bay Rd suite A, Redwood City, CA, 94063, USA
| | - Javier Chaparro-Riggers
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA
| | - Shu-Hui Liu
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,Multitude Therapeutics, Abmart, 3698 Haven Avenue Suite A, Redwood City, CA, 94063, USA
| |
Collapse
|
22
|
Ghobrial I, Cruz CH, Garfall A, Shah N, Munshi N, Kaufman J, Boise LH, Morgan G, Adalsteinsson VA, Manier S, Pillai R, Malavasi F, Lonial S. Immunotherapy in Multiple Myeloma: Accelerating on the Path to the Patient. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2019; 19:332-344. [DOI: 10.1016/j.clml.2019.02.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 01/11/2019] [Accepted: 02/11/2019] [Indexed: 11/16/2022]
|
23
|
Iftikhar A, Hassan H, Iftikhar N, Mushtaq A, Sohail A, Rosko N, Chakraborty R, Razzaq F, Sandeep S, Valent JN, Kanate AS, Anwer F. Investigational Monoclonal Antibodies in the Treatment of Multiple Myeloma: A Systematic Review of Agents under Clinical Development. Antibodies (Basel) 2019; 8:E34. [PMID: 31544840 PMCID: PMC6640719 DOI: 10.3390/antib8020034] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 05/12/2019] [Accepted: 05/13/2019] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Immunotherapy for multiple myeloma (MM) has been the focus in recent years due to its myeloma-specific immune responses. We reviewed the literature on non-Food and Drug Administration (FDA) approved monoclonal antibodies (mAbs) to highlight future perspectives. We searched PubMed, EMBASE, Web of Science, Cochrane Library and ClinicalTrials.gov to include phase I/II clinical trials. Data from 39 studies (1906 patients) were included. Of all the agents, Isatuximab (Isa, anti-CD38) and F50067 (anti-CXCR4) were the only mAbs to produce encouraging results as monotherapy with overall response rates (ORRs) of 66.7% and 32% respectively. Isa showed activity when used in combination with lenalidomide (Len) and dexamethasone (Dex), producing a clinical benefit rate (CBR) of 83%. Additionally, Isa used in combination with pomalidomide (Pom) and Dex resulted in a CBR of 73%. Indatuximab Ravtansine (anti-CD138 antibody-drug conjugate) produced an ORR of 78% and 79% when used in combination with Len-Dex and Pom-Dex, respectively. CONCLUSIONS Combination therapy using mAbs such as indatuximab, pembrolizumab, lorvotuzumab, siltuximab or dacetuzumab with chemotherapy agents produced better outcomes as compared to monotherapies. Further clinical trials investigating mAbs targeting CD38 used in combination therapy are warranted.
Collapse
Affiliation(s)
- Ahmad Iftikhar
- Department of Internal Medicine, The University of Arizona, Tucson, AZ 85721, USA.
| | - Hamza Hassan
- Department of Internal Medicine, Rochester General Hospital, Rochester, NY 14621, USA.
| | - Nimra Iftikhar
- Dow University of Health Sciences, Karachi 74200, Pakistan.
| | - Adeela Mushtaq
- Department of Internal Medicine, University of Pittsburgh Medical Center, McKeesport, PA 16148, USA.
| | - Atif Sohail
- Department of Internal Medicine, Rochester General Hospital, Rochester, NY 14621, USA.
| | - Nathaniel Rosko
- Taussig Cancer Center, Cleveland Clinic, Cleveland, OH 44106, USA.
| | | | - Faryal Razzaq
- Foundation University Medical College, Islamabad 44000, Pakistan.
| | - Sonia Sandeep
- Department of Pathology, Wilson Medical Center, Wilson, NC 27893, USA.
| | | | | | - Faiz Anwer
- Taussig Cancer Center, Cleveland Clinic, Cleveland, OH 44106, USA.
| |
Collapse
|
24
|
Friedman KM, Garrett TE, Evans JW, Horton HM, Latimer HJ, Seidel SL, Horvath CJ, Morgan RA. Effective Targeting of Multiple B-Cell Maturation Antigen-Expressing Hematological Malignances by Anti-B-Cell Maturation Antigen Chimeric Antigen Receptor T Cells. Hum Gene Ther 2019; 29:585-601. [PMID: 29641319 DOI: 10.1089/hum.2018.001] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
B-cell maturation antigen (BCMA) expression has been proposed as a marker for the identification of malignant plasma cells in patients with multiple myeloma (MM). Nearly all MM tumor cells express BCMA, while normal tissue expression is restricted to plasma cells and a subset of mature B cells. Consistent BCMA expression was confirmed on MM biopsies (29/29 BCMA+), and it was further demonstrated that BCMA is expressed in a substantial number of lymphoma samples, as well as primary chronic lymphocytic leukemia B cells. To target BCMA using redirected autologous T cells, lentiviral vectors (LVV) encoding chimeric antigen receptors (CARs) were constructed with four unique anti-BCMA single-chain variable fragments, fused to the CD137 (4-1BB) co-stimulatory and CD3ζ signaling domains. One LVV, BB2121, was studied in detail, and BB2121 CAR-transduced T cells (bb2121) exhibited a high frequency of CAR + T cells and robust in vitro activity against MM cell lines, lymphoma cell lines, and primary chronic lymphocytic leukemia peripheral blood. Based on receptor quantification, bb2121 recognized tumor cells expressing as little as 222 BCMA molecules per cell. The in vivo pharmacology of anti-BCMA CAR T cells was studied in NSG mouse models of human MM, Burkitt lymphoma, and mantle cell lymphoma, where mice received a single intravenous administration of vehicle, control vector-transduced T cells, or anti-BCMA CAR-transduced T cells. In all models, the vehicle and control CAR T cells failed to inhibit tumor growth. In contrast, treatment with bb2121 resulted in rapid and sustained elimination of the tumors and 100% survival in all treatment models. Together, these data support the further development of anti-BCMA CAR T cells as a potential treatment for not only MM but also some lymphomas.
Collapse
|
25
|
Eckhert E, Hewitt R, Liedtke M. B-cell maturation antigen directed monoclonal antibody therapies for multiple myeloma. Immunotherapy 2019; 11:801-811. [PMID: 31094254 DOI: 10.2217/imt-2018-0199] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Multiple myeloma affects 30,000 new patients in the USA yearly, with 5-year median overall survival rates of 82, 62 and 40% for patients in groups I, II and III of the revised international staging system. Novel therapeutic and prognostic tools are changing the way we treat patients with this historically difficult to manage condition. B-cell maturation antigen (BCMA) represents an ideal therapeutic target in myeloma because of its high expression rate and high specificity for myeloma cells. Preclinical data indicate that anti-BCMA monoclonal antibody therapies are highly potent, and initial data from Phase I clinical trials indicate that these drugs are well tolerated. Numerous ongoing Phase I and II clinical trials of anti-BCMA monoclonal antibodies are currently under way.
Collapse
Affiliation(s)
- Erik Eckhert
- Department of Medicine, Stanford University Hospital, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - Rhonda Hewitt
- Department of Medicine, Division of Hematology/Oncology, Stanford University Hospital, 875 Blake Wilbur Drive, Stanford, CA 94305, USA
| | - Michaela Liedtke
- Department of Medicine, Division of Hematology/Oncology, Stanford University Hospital, 875 Blake Wilbur Drive, Stanford, CA 94305, USA
| |
Collapse
|
26
|
Gavriatopoulou M, Ntanasis-Stathopoulos I, Dimopoulos MA, Terpos E. Anti-BCMA antibodies in the future management of multiple myeloma. Expert Rev Anticancer Ther 2019; 19:319-326. [PMID: 30810049 DOI: 10.1080/14737140.2019.1586539] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
INTRODUCTION B-cell maturation antigen (BCMA) belongs to the tumor necrosis factor receptor family and is expressed on late B-cells and plasma cells. Serum BCMA is elevated in patients with multiple myeloma (MM) and chronic lymphocytic leukemia (CLL), and might represent a novel prognostic and monitoring tool. Serum BCMA levels can predict both progression free survival (PFS) and overall survival (OS). Several therapeutic strategies are currently under investigation including BCMA-directed monoclonal Abs (either naked or with drug conjugates, and bispecific Abs) and cellular T-cell therapies (chimeric antigen receptor T-cells) with impressive clinical results. Areas covered: This review aims to present the mechanisms of action and the available data on efficacy and safety of therapies targeting BCMA. Expert opinion: The preliminary preclinical and clinical results from the phase 1 and 2 studies have demonstrated significant activity of the anti-BCMA therapeutic strategies. The main toxicities induced include Cytokine Release Syndrome (CRS) and ocular toxicity. The management of these adverse events remains currently an issue of controversy.
Collapse
Affiliation(s)
- Maria Gavriatopoulou
- a Oncology Department, Department of Therapeutics, Alexandra Hospital , National and Kapodistrian University of Athens , Athens , Greece
| | - Ioannis Ntanasis-Stathopoulos
- a Oncology Department, Department of Therapeutics, Alexandra Hospital , National and Kapodistrian University of Athens , Athens , Greece
| | - Meletios Athanasios Dimopoulos
- a Oncology Department, Department of Therapeutics, Alexandra Hospital , National and Kapodistrian University of Athens , Athens , Greece
| | - Evangelos Terpos
- a Oncology Department, Department of Therapeutics, Alexandra Hospital , National and Kapodistrian University of Athens , Athens , Greece
| |
Collapse
|
27
|
Immunotherapy: A Novel Era of Promising Treatments for Multiple Myeloma. Int J Mol Sci 2018; 19:ijms19113613. [PMID: 30445802 PMCID: PMC6274949 DOI: 10.3390/ijms19113613] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/05/2018] [Accepted: 11/13/2018] [Indexed: 01/24/2023] Open
Abstract
Multiple myeloma (MM) remains an incurable hematological malignancy characterized by clonal proliferation of malignant plasma cells in bone marrow. In the last 20 years, the introduction of autologous stem cell transplantation, followed by proteasome inhibitors and immunomodulatory agents, increased the survival of MM patients by 50%. However, still a high proportion of patients relapse and become refractory, especially, high-risk patients with adverse cytogenetics where these treatment combinations have shown limited benefit. Therefore, novel strategies, such as immunotherapy, have been developed in the last few years to help improve the survival of these patients. Immunotherapy treatments include a high number of different strategies used to attack the tumor cells by using the immune system. Here, we will review the most successful immunotherapy strategies published up to date in patients with relapsed or refractory (R/R) MM, including monoclonal antibodies targeting specific antigens on the tumor cells, antibodies combined with cytotoxic drugs or Antibodies Drug Conjugates, immune checkpoint inhibitors which eliminate the barriers that damper immune cells and prevent them from attacking tumor cells, bi-specific T-cell engagers antibodies (BiTEs), bi-specific antibodies and the infusion of chimeric antigen receptor-modified T cells. We overview the results of clinical studies that have been presented up to date and also review pre-clinical studies describing potential novel treatments for MM.
Collapse
|
28
|
Huang Y, Liu T. Therapeutic applications of genetic code expansion. Synth Syst Biotechnol 2018; 3:150-158. [PMID: 30345400 PMCID: PMC6190509 DOI: 10.1016/j.synbio.2018.09.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 09/16/2018] [Accepted: 09/18/2018] [Indexed: 12/05/2022] Open
Abstract
In nature, a limited, conservative set of amino acids are utilized to synthesize proteins. Genetic code expansion technique reassigns codons and incorporates noncanonical amino acids (ncAAs) through orthogonal aminoacyl-tRNA synthetase (aaRS)/tRNA pairs. The past decade has witnessed the rapid growth in diversity and scope for therapeutic applications of this technology. Here, we provided an update on the recent progress using genetic code expansion in the following areas: antibody-drug conjugates (ADCs), bispecific antibodies (BsAb), immunotherapies, long-lasting protein therapeutics, biosynthesized peptides, engineered viruses and cells, as well as other therapeutic related applications, where the technique was used to elucidate the mechanisms of biotherapeutics and drug targets.
Collapse
Affiliation(s)
| | - Tao Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| |
Collapse
|
29
|
Thakur A, Huang M, Lum LG. Bispecific antibody based therapeutics: Strengths and challenges. Blood Rev 2018; 32:339-347. [PMID: 29482895 DOI: 10.1016/j.blre.2018.02.004] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 01/31/2018] [Accepted: 02/16/2018] [Indexed: 01/13/2023]
Abstract
Monoclonal antibody-based targeted therapy has greatly improved treatment options for patients. However, long-term efficacy of such antibodies is limited by resistance mechanisms. New insights into the mechanisms by which tumors evade immune control have driven innovative therapeutic strategies to eliminate cancer by re-directing immune cells to tumors. Advances in protein engineering technology have generated multiple bispecific antibody (BsAb) formats capable of targeting multiple antigens as a single agent. Approval of two BsAb and three check point blocking mAbs represent a paradigm shift in the use of antibody constructs. Since BsAbs can directly target immune cells to tumors, drug resistance and severe adverse effects are much reduced. The wave of next generation "bispecific or multispecific antibodies" has advanced multiple candidates into ongoing clinical trials. In this review, we focus on preclinical and clinical studies in hematological malignancies as well as discuss reasons for the limited success of BsAbs against solid tumors.
Collapse
Affiliation(s)
- Archana Thakur
- Department of Medicine, Division of Hematology/Oncology, University of Virginia Cancer Center, Charlottesville, VA, USA.
| | - Manley Huang
- Department of Medicine, Division of Hematology/Oncology, University of Virginia Cancer Center, Charlottesville, VA, USA
| | - Lawrence G Lum
- Department of Medicine, Division of Hematology/Oncology, University of Virginia Cancer Center, Charlottesville, VA, USA
| |
Collapse
|
30
|
Morgan GJ, Rasche L. Maintaining therapeutic progress in multiple myeloma by integrating genetic and biological advances into the clinic. Expert Rev Hematol 2018; 11:513-523. [PMID: 29944024 DOI: 10.1080/17474086.2018.1489718] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Utilizing advances in genetic and immunologic analysis to segment and direct treatment is potentially a way of maintaining therapeutic progress toward cure in multiple myeloma (MM). This approach works well using clinical segments but can be optimized using recent genetic and immunologic technologies, which have opened the possibility of enhancing risk stratification and disease subclassification. Areas covered: This position paper discusses strategies to segment myeloma into subgroups with distinct risk profiles and distinct targetable lesions are presented. Expert commentary: Risk stratified treatment of MM is already a clinical reality that can be enhanced by the developmental of unified segmentation and testing approaches. Mutation-targeted treatment has proven to be effective against the RAS pathway, but is compromised by intra-clonal and spatiotemporal heterogeneity. Identifying new disease segments based on tumor biology or immunological content of the microenvironment offers an exciting new way to control and even eradicate myeloma clones. Going forward, risk and biologically stratified therapy for myeloma is a promising way of maintaining therapeutic progress, as is precision immunotherapy directed by the cellular context of the bone marrow.
Collapse
Affiliation(s)
- Gareth J Morgan
- a Myeloma Institute , The University of Arkansas for Medical Sciences , Little Rock , AR , USA
| | - Leo Rasche
- a Myeloma Institute , The University of Arkansas for Medical Sciences , Little Rock , AR , USA
| |
Collapse
|
31
|
Liu P, Jin Y, Sattar H, Liu H, Xie W, Zhou F. Natural killer cell immunotherapy against multiple myeloma: Progress and possibilities. J Leukoc Biol 2018; 103:821-828. [PMID: 29733502 DOI: 10.1002/jlb.2ru0517-176rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 01/06/2018] [Accepted: 01/07/2018] [Indexed: 12/29/2022] Open
Affiliation(s)
- Pan Liu
- Department of Hematology; Zhongnan Hospital; Wuhan University; Wuhan P.R. China
| | - Yanxia Jin
- Department of Hematology; Zhongnan Hospital; Wuhan University; Wuhan P.R. China
| | - Haseeb Sattar
- Department of Clinical Pharmacy; Wuhan Union Hospital; affiliated Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan P.R. China
| | - Hailing Liu
- Department of Clinical Hematology; Second Affiliated Hospital; Xi'an Jiao Tong University; Xi'an P.R. China
| | - Weiling Xie
- Department of Hematology; Zhongnan Hospital; Wuhan University; Wuhan P.R. China
| | - Fuling Zhou
- Department of Hematology; Zhongnan Hospital; Wuhan University; Wuhan P.R. China
- Hubei Key Laboratory of Tumor Biological Behavior; Wuhan P.R. China
| |
Collapse
|
32
|
Sanchez E, Tanenbaum EJ, Patil S, Li M, Soof CM, Vidisheva A, Waterman GN, Hekmati T, Tang G, Wang CS, Chen H, Berenson J. The clinical significance of B-cell maturation antigen as a therapeutic target and biomarker. Expert Rev Mol Diagn 2018; 18:319-329. [PMID: 29504446 DOI: 10.1080/14737159.2018.1448269] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Eric Sanchez
- Institute for Myeloma and Bone Cancer Research (IMBCR), West Hollywood, CA, USA
| | - Edward J. Tanenbaum
- Institute for Myeloma and Bone Cancer Research (IMBCR), West Hollywood, CA, USA
| | - Saurabh Patil
- Institute for Myeloma and Bone Cancer Research (IMBCR), West Hollywood, CA, USA
| | - Mingjie Li
- Institute for Myeloma and Bone Cancer Research (IMBCR), West Hollywood, CA, USA
| | - Camilia M. Soof
- Institute for Myeloma and Bone Cancer Research (IMBCR), West Hollywood, CA, USA
| | | | - Gabriel N. Waterman
- Keck School of Medicine, University of Southern California - Los Angeles, Los Angeles, CA, USA
| | - Tara Hekmati
- Institute for Myeloma and Bone Cancer Research (IMBCR), West Hollywood, CA, USA
| | - George Tang
- Institute for Myeloma and Bone Cancer Research (IMBCR), West Hollywood, CA, USA
| | - Cathy S. Wang
- Institute for Myeloma and Bone Cancer Research (IMBCR), West Hollywood, CA, USA
| | - Haiming Chen
- Institute for Myeloma and Bone Cancer Research (IMBCR), West Hollywood, CA, USA
| | - James Berenson
- Institute for Myeloma and Bone Cancer Research (IMBCR), West Hollywood, CA, USA
| |
Collapse
|
33
|
Lee L, Draper B, Chaplin N, Philip B, Chin M, Galas-Filipowicz D, Onuoha S, Thomas S, Baldan V, Bughda R, Maciocia P, Kokalaki E, Neves MP, Patel D, Rodriguez-Justo M, Francis J, Yong K, Pule M. An APRIL-based chimeric antigen receptor for dual targeting of BCMA and TACI in multiple myeloma. Blood 2018; 131:746-758. [PMID: 29284597 PMCID: PMC5922275 DOI: 10.1182/blood-2017-05-781351] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 12/13/2017] [Indexed: 12/15/2022] Open
Abstract
B-cell maturation antigen (BCMA) is a promising therapeutic target for multiple myeloma (MM), but expression is variable, and early reports of BCMA targeting chimeric antigen receptors (CARs) suggest antigen downregulation at relapse. Dual-antigen targeting increases targetable tumor antigens and reduces the risk of antigen-negative disease escape. "A proliferation-inducing ligand" (APRIL) is a natural high-affinity ligand for BCMA and transmembrane activator and calcium-modulator and cyclophilin ligand (TACI). We quantified surface tumor expression of BCMA and TACI on primary MM cells (n = 50). All cases tested expressed BCMA, and 39 (78%) of them also expressed TACI. We engineered a third-generation APRIL-based CAR (ACAR), which killed targets expressing either BCMA or TACI (P < .01 and P < .05, respectively, cf. control, effector-to-target [E:T] ratio 16:1). We confirmed cytolysis at antigen levels similar to those on primary MM, at low E:T ratios (56.2% ± 3.9% killing of MM.1s at 48 h, E:T ratio 1:32; P < .01) and of primary MM cells (72.9% ± 12.2% killing at 3 days, E:T ratio 1:1; P < .05, n = 5). Demonstrating tumor control in the absence of BCMA, we maintained cytolysis of primary tumor expressing both BCMA and TACI in the presence of a BCMA-targeting antibody. Furthermore, using an intramedullary myeloma model, ACAR T cells caused regression of an established tumor within 2 days. Finally, in an in vivo model of tumor escape, there was complete ACAR-mediated tumor clearance of BCMA+TACI- and BCMA-TACI+ cells, and a single-chain variable fragment CAR targeting BCMA alone resulted in outgrowth of a BCMA-negative tumor. These results support the clinical potential of this approach.
Collapse
Affiliation(s)
- Lydia Lee
- Department of Haematology, University College London (UCL) Cancer Institute, London, United Kingdom
| | - Benjamin Draper
- Department of Haematology, University College London (UCL) Cancer Institute, London, United Kingdom
| | - Neil Chaplin
- Department of Haematology, University College London (UCL) Cancer Institute, London, United Kingdom
| | - Brian Philip
- Department of Haematology, University College London (UCL) Cancer Institute, London, United Kingdom
| | - Melody Chin
- Department of Haematology, University College London (UCL) Cancer Institute, London, United Kingdom
| | - Daria Galas-Filipowicz
- Department of Haematology, University College London (UCL) Cancer Institute, London, United Kingdom
| | | | | | | | | | - Paul Maciocia
- Department of Haematology, University College London (UCL) Cancer Institute, London, United Kingdom
| | - Eva Kokalaki
- Department of Haematology, University College London (UCL) Cancer Institute, London, United Kingdom
| | - Margarida P Neves
- Department of Haematology, University College London (UCL) Cancer Institute, London, United Kingdom
| | - Dominic Patel
- Department of Histopathology, UCL, London, United Kingdom
| | | | | | - Kwee Yong
- Department of Haematology, University College London (UCL) Cancer Institute, London, United Kingdom
| | - Martin Pule
- Department of Haematology, University College London (UCL) Cancer Institute, London, United Kingdom
- Autolus Ltd., London, United Kingdom; and
| |
Collapse
|
34
|
Abstract
Harnessing the power of the human immune system to treat cancer is the essence of immunotherapy. Monoclonal antibodies engage the innate immune system to destroy targeted cells. For the last 30years, antibody-dependent cell-mediated cytotoxicity and complement-dependent cytotoxicity have been the main mechanisms of anti-tumor action of unconjugated antibody drugs. Efforts to exploit the potentials of other immune cells, in particular T cells, culminated in the recent approval of two T cell engaging bispecific antibody (T-BsAb) drugs, thereby stimulating new efforts to accelerate similar platforms through preclinical and clinical trials. In this review, we have compiled the worldwide effort in exploring T cell engaging bispecific antibodies. Our special emphasis is on the lessons learned, with the hope to derive insights in this fast evolving field with tremendous clinical potential.
Collapse
Affiliation(s)
- Z Wu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - N V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States.
| |
Collapse
|
35
|
Velasquez MP, Bonifant CL, Gottschalk S. Redirecting T cells to hematological malignancies with bispecific antibodies. Blood 2018; 131:30-38. [PMID: 29118005 PMCID: PMC5755042 DOI: 10.1182/blood-2017-06-741058] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 08/30/2017] [Indexed: 12/13/2022] Open
Abstract
There is a need to improve outcomes for patients with recurrent and/or refractory hematological malignancies. Immunotherapy holds the promise to meet this need, because it does not rely on the cytotoxic mechanism of conventional therapies. Among different forms of immunotherapy, redirecting T cells to hematological malignancies with bispecific antibodies (BsAbs) is an attractive strategy. BsAbs are an "off-the-shelf" product that is easily scalable in contrast to adoptive T-cell therapies. Among these, the bispecific T-cell engager blinatumomab has emerged as the most successful BsAb to date. It consists of 2 single-chain variable fragments specific for CD19 present on B-cell malignancies and CD3 expressed on almost all T cells. Blinatumomab has shown potent antitumor activity as a single agent, particularly for acute lymphoblastic leukemia, resulting in its US Food and Drug Administration approval. However, although successful in inducing remissions, these are normally short-lived, with median response durations of <1 year. Nevertheless, the success of blinatumomab has reinvigorated the BsAb field, which is bustling with preclinical and clinical studies for not only B-cell-derived lymphoblastic leukemia and lymphoma but also acute myeloid leukemia and multiple myeloma. Here, we will review the successes and challenges of T-cell-targeted BsAbs for the immunotherapy of hematological malignancies with special focus on conducted clinical studies and strategies to improve their efficacy.
Collapse
Affiliation(s)
- Mireya Paulina Velasquez
- Department of Bone Marrow Transplant and Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN; and
| | - Challice L Bonifant
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI
| | - Stephen Gottschalk
- Department of Bone Marrow Transplant and Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN; and
| |
Collapse
|
36
|
Bera TK, Abe Y, Ise T, Oberle A, Gallardo D, Liu XF, Nagata S, Binder M, Pastan I. Recombinant immunotoxins targeting B-cell maturation antigen are cytotoxic to myeloma cell lines and myeloma cells from patients. Leukemia 2017; 32:569-572. [PMID: 29149102 PMCID: PMC5808081 DOI: 10.1038/leu.2017.315] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- T K Bera
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Cancer Biology Research, Bethesda, MD, USA
| | - Y Abe
- Center, Sanford Research, Sioux Falls, SD, USA
| | - T Ise
- Center, Sanford Research, Sioux Falls, SD, USA
| | - A Oberle
- Klinik für Onkologie, Hämatologie und KMT mit Sektion Pneumologie Universitätsklinikum Hamburg Eppendorf, Hamburg, Germany
| | - D Gallardo
- Leidos Biomedical Research, Inc., National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - X-F Liu
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Cancer Biology Research, Bethesda, MD, USA
| | - S Nagata
- Center, Sanford Research, Sioux Falls, SD, USA
| | - M Binder
- Klinik für Onkologie, Hämatologie und KMT mit Sektion Pneumologie Universitätsklinikum Hamburg Eppendorf, Hamburg, Germany
| | - I Pastan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Cancer Biology Research, Bethesda, MD, USA
| |
Collapse
|
37
|
Cho SF, Lin L, Xing L, Yu T, Wen K, Anderson KC, Tai YT. Monoclonal Antibody: A New Treatment Strategy against Multiple Myeloma. Antibodies (Basel) 2017; 6:antib6040018. [PMID: 31548533 PMCID: PMC6698817 DOI: 10.3390/antib6040018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/09/2017] [Accepted: 11/10/2017] [Indexed: 12/22/2022] Open
Abstract
2015 was a groundbreaking year for the multiple myeloma community partly due to the breakthrough approval of the first two monoclonal antibodies in the treatment for patients with relapsed and refractory disease. Despite early disappointments, monoclonal antibodies targeting CD38 (daratumumab) and signaling lymphocytic activation molecule F7 (SLAMF7) (elotuzumab) have become available for patients with multiple myeloma in the same year. Specifically, phase 3 clinical trials of combination therapies incorporating daratumumab or elotuzumab indicate both efficacy and a very favorable toxicity profile. These therapeutic monoclonal antibodies for multiple myeloma can kill target cells via antibody-dependent cell-mediated cytotoxicity, complement-dependent cytotoxicity, and antibody-dependent phagocytosis, as well as by direct blockade of signaling cascades. In addition, their immunomodulatory effects may simultaneously inhibit the immunosuppressive bone marrow microenvironment and restore the key function of immune effector cells. In this review, we focus on monoclonal antibodies that have shown clinical efficacy or promising preclinical anti-multiple myeloma activities that warrant further clinical development. We summarize mechanisms that account for the in vitro and in vivo anti-myeloma effects of these monoclonal antibodies, as well as relevant preclinical and clinical results. Monoclonal antibody-based immunotherapies have already and will continue to transform the treatment landscape in multiple myeloma.
Collapse
Affiliation(s)
- Shih-Feng Cho
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| | - Liang Lin
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| | - Lijie Xing
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, No. 324, Jingwu Road, Jinan 250021, China.
| | - Tengteng Yu
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| | - Kenneth Wen
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| | - Kenneth C Anderson
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| | - Yu-Tzu Tai
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
38
|
Patterson JT, Isaacson J, Kerwin L, Atassi G, Duggal R, Bresson D, Zhu T, Zhou H, Fu Y, Kaufmann GF. PSMA-targeted bispecific Fab conjugates that engage T cells. Bioorg Med Chem Lett 2017; 27:5490-5495. [PMID: 29126850 DOI: 10.1016/j.bmcl.2017.09.065] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 09/29/2017] [Accepted: 09/30/2017] [Indexed: 01/06/2023]
Abstract
Bioconjugate formats provide alternative strategies for antigen targeting with bispecific antibodies. Here, PSMA-targeted Fab conjugates were generated using different bispecific formats. Interchain disulfide bridging of an αCD3 Fab enabled installation of either the PSMA-targeting small molecule DUPA (SynFab) or the attachment of an αPSMA Fab (BisFab) by covalent linkage. Optimization of the reducing conditions was critical for selective interchain disulfide reduction and good bioconjugate yield. Activity of αPSMA/CD3 Fab conjugates was tested by in vitro cytotoxicity assays using prostate cancer cell lines. Both bispecific formats demonstrated excellent potency and antigen selectivity.
Collapse
Affiliation(s)
- James T Patterson
- Sorrento Therapeutics, Inc., 4955 Directors Place, San Diego, CA 92121, USA.
| | - Jason Isaacson
- Sorrento Therapeutics, Inc., 4955 Directors Place, San Diego, CA 92121, USA
| | - Lisa Kerwin
- Sorrento Therapeutics, Inc., 4955 Directors Place, San Diego, CA 92121, USA
| | - Ghazi Atassi
- Sorrento Therapeutics, Inc., 4955 Directors Place, San Diego, CA 92121, USA
| | - Rohit Duggal
- Sorrento Therapeutics, Inc., 4955 Directors Place, San Diego, CA 92121, USA
| | - Damien Bresson
- Sorrento Therapeutics, Inc., 4955 Directors Place, San Diego, CA 92121, USA
| | - Tong Zhu
- Sorrento Therapeutics, Inc., 4955 Directors Place, San Diego, CA 92121, USA
| | - Heyue Zhou
- Sorrento Therapeutics, Inc., 4955 Directors Place, San Diego, CA 92121, USA
| | - Yanwen Fu
- Sorrento Therapeutics, Inc., 4955 Directors Place, San Diego, CA 92121, USA
| | - Gunnar F Kaufmann
- Sorrento Therapeutics, Inc., 4955 Directors Place, San Diego, CA 92121, USA.
| |
Collapse
|
39
|
Sherbenou DW, Mark TM, Forsberg P. Monoclonal Antibodies in Multiple Myeloma: A New Wave of the Future. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2017; 17:545-554. [PMID: 28734795 DOI: 10.1016/j.clml.2017.06.030] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 06/08/2017] [Indexed: 11/17/2022]
Abstract
In 2015, 2 monoclonal antibodies were approved for the treatment of relapsed or refractory multiple myeloma (RRMM), elotuzumab and daratumumab. Elotuzumab is a monoclonal IgG-κ antibody directed against SLAMF7 (signaling lymphocytic activation molecule F7), a cell surface receptor involved in natural killer cell activation. Daratumumab is a monoclonal IgG-κ antibody that binds to CD38, a transmembrane protein found on the surface of myeloma cells and responsible for cellular adhesion and ectoenzymatic activity. Both elotuzumab and daratumumab act through recruitment of the immune system to enhance cellular cytotoxicity directed against myeloma cells. Elotuzumab requires lenalidomide and dexamethasone combined to enhance progression-free survival in patients with RRMM, and daratumumab has both single-agent and combination activity with either lenalidomide or the proteasome inhibitor bortezomib in RRMM. The adverse effect profile of both agents mainly consists of allergic-type infusion reactions. Other considerations for monoclonal antibody use in the treatment of MM include the potential for interference in serum protein electrophoresis testing and cross-reactivity of daratumumab with CD38 present on red blood cells. In the present report, we discussed the clinical development of daratumumab and elotuzumab and newer immunologic approaches to the treatment of MM.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antineoplastic Agents, Immunological/administration & dosage
- Antineoplastic Agents, Immunological/adverse effects
- Antineoplastic Agents, Immunological/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Clinical Trials as Topic
- Combined Modality Therapy
- Drug Evaluation, Preclinical
- Humans
- Immunotherapy/methods
- Molecular Targeted Therapy
- Multiple Myeloma/drug therapy
- Multiple Myeloma/metabolism
- Multiple Myeloma/pathology
- Treatment Outcome
Collapse
Affiliation(s)
- Daniel W Sherbenou
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO.
| | - Tomer M Mark
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Peter Forsberg
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
40
|
Rasche L, Weinhold N, Morgan GJ, van Rhee F, Davies FE. Immunologic approaches for the treatment of multiple myeloma. Cancer Treat Rev 2017; 55:190-199. [PMID: 28431262 DOI: 10.1016/j.ctrv.2017.03.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 03/27/2017] [Accepted: 03/28/2017] [Indexed: 12/11/2022]
Abstract
The FDA approval of two monoclonal antibodies in 2015has heralded a new era of targeted immunotherapies for multiple myeloma (MM). In this review we discuss the recent approaches using different immunological components to treat MM. In particular, we review current monoclonal antibody based therapies, engineered T- and NK cell products, 'off-target' immunomodulation, and strategies utilizing allogeneic cell transplantation in MM. We discuss how an immunologic approach offers promise for the treatment of this genetically heterogeneous disease, and how patients with acquired drug resistance may particularly benefit from these therapies. We also describe some of the limitations of the current strategies and speculate on the future of personalized immunotherapies for MM.
Collapse
Affiliation(s)
- Leo Rasche
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| | - Niels Weinhold
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Gareth J Morgan
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Frits van Rhee
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Faith E Davies
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
41
|
Abstract
In this issue of Cancer Cell, Li et al. and Seckinger et al. describe promising results of two T-cell-dependent bi-specific antibodies for the treatment of multiple myeloma: one targets FcRH5 expressed on B cells, whereas the other targets the B cell maturation antigen expressed on plasma cells.
Collapse
Affiliation(s)
- Marta Chesi
- Mayo Clinic in Arizona, Phoenix, AZ 85054, USA
| | | |
Collapse
|
42
|
Seckinger A, Delgado JA, Moser S, Moreno L, Neuber B, Grab A, Lipp S, Merino J, Prosper F, Emde M, Delon C, Latzko M, Gianotti R, Lüoend R, Murr R, Hosse RJ, Harnisch LJ, Bacac M, Fauti T, Klein C, Zabaleta A, Hillengass J, Cavalcanti-Adam EA, Ho AD, Hundemer M, San Miguel JF, Strein K, Umaña P, Hose D, Paiva B, Vu MD. Target Expression, Generation, Preclinical Activity, and Pharmacokinetics of the BCMA-T Cell Bispecific Antibody EM801 for Multiple Myeloma Treatment. Cancer Cell 2017; 31:396-410. [PMID: 28262554 DOI: 10.1016/j.ccell.2017.02.002] [Citation(s) in RCA: 241] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 10/07/2016] [Accepted: 01/31/2017] [Indexed: 01/12/2023]
Abstract
We identified B cell maturation antigen (BCMA) as a potential therapeutic target in 778 newly diagnosed and relapsed myeloma patients. We constructed an IgG-based BCMA-T cell bispecific antibody (EM801) and showed that it increased CD3+ T cell/myeloma cell crosslinking, followed by CD4+/CD8+ T cell activation, and secretion of interferon-γ, granzyme B, and perforin. This effect is CD4 and CD8 T cell mediated. EM801 induced, at nanomolar concentrations, myeloma cell death by autologous T cells in 34 of 43 bone marrow aspirates, including those from high-risk patients and patients after multiple lines of treatment, tumor regression in six of nine mice in a myeloma xenograft model, and depletion of BCMA+ cells in cynomolgus monkeys. Pharmacokinetics and pharmacodynamics indicate weekly intravenous/subcutaneous administration.
Collapse
Affiliation(s)
- Anja Seckinger
- Universitätsklinikum Heidelberg, Labor für Myelomforschung and Medizinische Klinik V, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Jose Antonio Delgado
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada, IDISNA, Avda. Pío XII, 55, 31008 Pamplona, Spain
| | - Samuel Moser
- Roche Innovation Center Zurich, 8952 Schlieren, Switzerland
| | - Laura Moreno
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada, IDISNA, Avda. Pío XII, 55, 31008 Pamplona, Spain
| | - Brigitte Neuber
- Universitätsklinikum Heidelberg, Medizinische Klinik V, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Anna Grab
- Universitätsklinikum Heidelberg, Labor für Myelomforschung and Medizinische Klinik V, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Susanne Lipp
- Universitätsklinikum Heidelberg, Labor für Myelomforschung and Medizinische Klinik V, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Juana Merino
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada, IDISNA, Avda. Pío XII, 55, 31008 Pamplona, Spain
| | - Felipe Prosper
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada, IDISNA, Avda. Pío XII, 55, 31008 Pamplona, Spain
| | - Martina Emde
- Universitätsklinikum Heidelberg, Labor für Myelomforschung and Medizinische Klinik V, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Camille Delon
- Roche Innovation Center Zurich, 8952 Schlieren, Switzerland
| | - Melanie Latzko
- Roche Innovation Center Zurich, 8952 Schlieren, Switzerland
| | - Reto Gianotti
- Roche Innovation Center Zurich, 8952 Schlieren, Switzerland
| | - Remo Lüoend
- Roche Innovation Center Zurich, 8952 Schlieren, Switzerland
| | - Ramona Murr
- Roche Innovation Center Zurich, 8952 Schlieren, Switzerland
| | - Ralf J Hosse
- Roche Innovation Center Zurich, 8952 Schlieren, Switzerland
| | | | - Marina Bacac
- Roche Innovation Center Zurich, 8952 Schlieren, Switzerland
| | - Tanja Fauti
- Roche Innovation Center Zurich, 8952 Schlieren, Switzerland
| | | | - Aintzane Zabaleta
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada, IDISNA, Avda. Pío XII, 55, 31008 Pamplona, Spain
| | - Jens Hillengass
- Universitätsklinikum Heidelberg, Medizinische Klinik V, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Elisabetta Ada Cavalcanti-Adam
- University of Heidelberg, Institute for Physical Chemistry and Max Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Anthony D Ho
- Universitätsklinikum Heidelberg, Medizinische Klinik V, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Michael Hundemer
- Universitätsklinikum Heidelberg, Medizinische Klinik V, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Jesus F San Miguel
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada, IDISNA, Avda. Pío XII, 55, 31008 Pamplona, Spain
| | | | - Pablo Umaña
- Roche Innovation Center Zurich, 8952 Schlieren, Switzerland
| | - Dirk Hose
- Universitätsklinikum Heidelberg, Labor für Myelomforschung and Medizinische Klinik V, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.
| | - Bruno Paiva
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada, IDISNA, Avda. Pío XII, 55, 31008 Pamplona, Spain.
| | | |
Collapse
|
43
|
Wang F, Wang H, Shen YD, Li YJ, Dong JX, Xu ZL, Yang JY, Sun YM, Xiao ZL. Bispecific Monoclonal Antibody-Based Multianalyte ELISA for Furaltadone Metabolite, Malachite Green, and Leucomalachite Green in Aquatic Products. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:8054-8061. [PMID: 27706938 DOI: 10.1021/acs.jafc.6b03233] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
A new multianalyte immunoassay was designed to screen furaltadone metabolite 5-morpholinomethyl-3-amino-2-oxazolidone (AMOZ), malachite green (MG), and leucomalachite green (LMG) in aquatic products using a bispecific monoclonal antibody (BsMAb). Gradient drug mutagenesis methods were separately used to prepare an anti-3-nitrobenzaldehyde-derivatized AMOZ (3-NPAMOZ) hybridoma cell line that was hypoxanthine-guanine-phosphoribosyltransferase (HGRPT) deficient and an anti-LMG hybridoma cell line that was thymidine kinase (TK) deficient. BsMAb recognizing 3-NPAMOZ and LMG was generated using hybrid-hybridomas of HGRPT and TK deficient cell lines. For AMOZ and LMG, respectively, the BsMAb-based indirect competitive ELSIA (ic-ELISA) values of 1.7 ng/mL and 45.3 ng/mL and detection limits of 0.2 ng/mL and 4.8 ng/mL. To establish the ic-ELISA, 3-NPAMOZ derivatized from AMOZ with 3-nitrobenzaldehyde and LMG reduced from MG by potassium borohydride was recognized by BsMAb. Recoveries of AMOZ, MG, and LMG in aquatic products were satisfactory and correlated with HPLC analysis. Thus, the multianalyte ic-ELISA is suitable for rapid quantification of AMOZ, MG, and LMG in aquatic products.
Collapse
Affiliation(s)
- Feng Wang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University , Guangzhou 510642, China
| | - Hong Wang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University , Guangzhou 510642, China
| | - Yu-Dong Shen
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University , Guangzhou 510642, China
| | - Yong-Jun Li
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University , Guangzhou 510642, China
- Zhongshan Quality Supervision & Inspection Institute of Agricultural Products , Zhongshan 528403, China
| | - Jie-Xian Dong
- Department of Entomology and Nematology and UCD Comprehensive Cancer Center, University of California , Davis, California 95616, United States
| | - Zhen-Lin Xu
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University , Guangzhou 510642, China
| | - Jin-Yi Yang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University , Guangzhou 510642, China
| | - Yuan-Ming Sun
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University , Guangzhou 510642, China
| | - Zhi-Li Xiao
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University , Guangzhou 510642, China
| |
Collapse
|
44
|
Schieferdecker A, Shoshani O, Westner B, Zipori D, Fehse B, Kröger N, Ayuk F. Potent in vitro and in vivo effects of polyclonal anti-human-myeloma globulins. Oncotarget 2016; 7:67061-67070. [PMID: 27563813 PMCID: PMC5341857 DOI: 10.18632/oncotarget.11489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 08/10/2016] [Indexed: 11/25/2022] Open
Abstract
Introduction Multiple myeloma is still incurable in most cases. Polyclonal anti T lymphocyte globulins (ATG) have been reported to kill human myeloma cells in vitro and in mouse models. Methods Anti-human-myeloma globulins (AMG) were produced by immunizing rabbits with human myeloma cell lines RPMI-8226 (AMG-8226) or KMS-12-BM (AMG-12-BM). Cytotoxicity of the polyclonal antibodies was analyzed in vitro and in a xenograft NOD-SCID mouse model. Results Both AMG had stronger cytotoxicity against myeloma cells compared to ATG. In primary T cells, AMG-8226 showed greater complement-dependent cytotoxicity (CDC) than ATG, whereas complement-independent cytotoxicity did not differ. Effects on non-hematopoietic cell lines were also similar. Competitive blocking assays revealed fourfold more antibodies against CD38 in AMG-8226 compared to ATG. Low concentrations of AMG-8226 and ATG increased ADCC. At higher concentrations, ATG inhibited ADCC more potently than AMG-8226. Combinations of ATG and AMG-8226 with melphalan or bortezomib showed additive to synergistic cytotoxicity on myeloma cells. The cytotoxic effects of AMG and ATG were confirmed in the xenograft NOD-SCID mouse model. Conclusion Our data show more potent antimyeloma effects of AMG compared to ATG. These results lay the ground for the development of polyclonal antibodies for the treatment of multiple myeloma.
Collapse
Affiliation(s)
- Aneta Schieferdecker
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Current Affiliation: Department of Oncology and Hematology with Section Pneumology, Hubertus Wald Tumorzentrum/ UCCH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ofer Shoshani
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.,Current Affiliation: San Diego Branch, Ludwig Institute for Cancer Research, La Jolla, CA, USA
| | - Benedikt Westner
- Neovii (formerly Fresenius) Biotech GmbH, Gräfelfing, Germany.,Current Affiliation: Acino AG, Miesbach, Germany
| | - Dov Zipori
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Boris Fehse
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Francis Ayuk
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
45
|
Geyer MB, Brentjens RJ. Review: Current clinical applications of chimeric antigen receptor (CAR) modified T cells. Cytotherapy 2016; 18:1393-1409. [PMID: 27592405 DOI: 10.1016/j.jcyt.2016.07.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 06/27/2016] [Accepted: 07/15/2016] [Indexed: 12/19/2022]
Abstract
The past several years have been marked by extraordinary advances in clinical applications of immunotherapy. In particular, adoptive cellular therapy utilizing chimeric antigen receptor (CAR)-modified T cells targeted to CD19 has demonstrated substantial clinical efficacy in children and adults with relapsed or refractory B-cell acute lymphoblastic leukemia (B-ALL) and durable clinical benefit in a smaller subset of patients with relapsed or refractory chronic lymphocytic leukemia (CLL) or B-cell non-Hodgkin lymphoma (B-NHL). Early-phase clinical trials are currently assessing CAR T-cell safety and efficacy in additional malignancies. Here, we discuss clinical results from the largest series to date investigating CD19-targeted CAR T cells in B-ALL, CLL, and B-NHL, including discussion of differences in CAR T-cell design and production and treatment approach, as well as clinical efficacy, nature of severe cytokine release syndrome and neurologic toxicities, and CAR T-cell expansion and persistence. We additionally review the current and forthcoming use of CAR T cells in multiple myeloma and several solid tumors and highlight challenges and opportunities afforded by the current state of CAR T-cell therapies, including strategies to overcome inhibitory aspects of the tumor microenvironment and enhance antitumor efficacy.
Collapse
Affiliation(s)
- Mark B Geyer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Renier J Brentjens
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
46
|
A transplant “immunome” screening platform defines a targetable epitope fingerprint of multiple myeloma. Blood 2016; 127:3202-14. [DOI: 10.1182/blood-2015-10-676536] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 03/22/2016] [Indexed: 12/13/2022] Open
Abstract
Key Points
The myeloma transplant B-cell immunome is predictive for response to treatment. It may be exploited by immunosequencing and library technology as a source for unique target structures and antibodies for immunotherapy.
Collapse
|
47
|
Lee L, Bounds D, Paterson J, Herledan G, Sully K, Seestaller-Wehr LM, Fieles WE, Tunstead J, McCahon L, Germaschewski FM, Mayes PA, Craigen JL, Rodriguez-Justo M, Yong KL. Evaluation of B cell maturation antigen as a target for antibody drug conjugate mediated cytotoxicity in multiple myeloma. Br J Haematol 2016; 174:911-22. [DOI: 10.1111/bjh.14145] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 02/29/2016] [Indexed: 01/25/2023]
Affiliation(s)
- Lydia Lee
- Haematology; UCL Cancer Institute; London UK
| | | | | | | | | | | | | | - James Tunstead
- Platform Technology Sciences; GlaxoSmithKline; Collegeville PA USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Wang JZ, Zhang YH, Guo XH, Zhang HY, Zhang Y. The double-edge role of B cells in mediating antitumor T-cell immunity: Pharmacological strategies for cancer immunotherapy. Int Immunopharmacol 2016; 36:73-85. [PMID: 27111515 DOI: 10.1016/j.intimp.2016.04.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Revised: 04/10/2016] [Accepted: 04/13/2016] [Indexed: 12/22/2022]
Abstract
Emerging evidence reveals the controversial role of B cells in antitumor immunity, but the underlying mechanisms have to be explored. Three latest articles published in the issue 521 of Nature in 2015 reconfirmed the puzzling topic and put forward some explanations of how B cells regulate antitumor T-cell responses both positively and negatively. This paper attempts to demonstrate that different B-cell subpopulations have distinct immunological properties and that they are involved in either antitumor responses or immunosuppression. Recent studies supporting the positive and negative roles of B cells in tumor development were summarized comprehensively. Several specific B-cell subpopulations, such as IgG(+), IgA(+), IL-10(+), and regulatory B cells, were described in detail. The mechanisms underlying the controversial B-cell effects were mainly attributed to different B-cell subpopulations, different B-cell-derived cytokines, direct B cell-T cell interaction, different cancer categories, and different malignant stages, and the immunological interaction between B cells and T cells is mediated by dendritic cells. Promising B-cell-based antitumor strategies were proposed and novel B-cell regulators were summarized to present interesting therapeutic targets. Future investigations are needed to make sure that B-cell-based pharmacological strategies benefit cancer immunotherapy substantially.
Collapse
Affiliation(s)
- Jing-Zhang Wang
- Department of Medical Technology, College of Medicine, Affiliated Hospital, Hebei University of Engineering, Handan 056002, PR China.
| | - Yu-Hua Zhang
- Department of Library, Hebei University of Engineering, Handan 056038, PR China
| | - Xin-Hua Guo
- Department of Medicine, College of Medicine, Hebei University of Engineering, Handan 056002, PR China
| | - Hong-Yan Zhang
- Department of Medical Technology, College of Medicine, Affiliated Hospital, Hebei University of Engineering, Handan 056002, PR China
| | - Yuan Zhang
- Department of Medical Technology, College of Medicine, Affiliated Hospital, Hebei University of Engineering, Handan 056002, PR China
| |
Collapse
|
49
|
Wu D, Cheung S, O'Sullivan CJ, Gao Y, Chen ZL, O'Shea DF. Strained alkyne substituted near infrared BF2azadipyrromethene fluorochrome. RSC Adv 2016. [DOI: 10.1039/c6ra19843k] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Synthesis and evaluation of a strained alkyne substituted NIR BF2-azadipyrromethene (NIR-AZA) shows potential for translation to clinical imaging.
Collapse
Affiliation(s)
- Dan Wu
- Department of Pharmaceutical and Medicinal Chemistry
- Royal College of Surgeons in Ireland
- Ireland
| | - Shane Cheung
- Department of Pharmaceutical and Medicinal Chemistry
- Royal College of Surgeons in Ireland
- Ireland
| | - Corry James O'Sullivan
- Department of Pharmaceutical and Medicinal Chemistry
- Royal College of Surgeons in Ireland
- Ireland
| | - Yinghua Gao
- Department of Pharmaceutical Science & Technology
- College of Chemistry and Biology
- Donghua University
- Shanghai 201620
- China
| | - Zhi-long Chen
- Department of Pharmaceutical Science & Technology
- College of Chemistry and Biology
- Donghua University
- Shanghai 201620
- China
| | - Donal F. O'Shea
- Department of Pharmaceutical and Medicinal Chemistry
- Royal College of Surgeons in Ireland
- Ireland
| |
Collapse
|
50
|
Patterson DM, Prescher JA. Orthogonal bioorthogonal chemistries. Curr Opin Chem Biol 2015; 28:141-9. [DOI: 10.1016/j.cbpa.2015.07.006] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Revised: 06/20/2015] [Accepted: 07/17/2015] [Indexed: 01/20/2023]
|