1
|
Morgenstern-Kaplan D, Kareff SA, Trabolsi A, Rodriguez E, Krause H, Ribeiro JR, Tan H, Antonarakis ES, Lou E, Nagasaka M, Algaze S, Lenz HJ, Liu SV, Halmos B, Hoon DSB, Seeber A, Ma PC, El-Deiry WS, Vanderwalde AM, Lopes G. Genomic, immunologic, and prognostic associations of TROP2 (TACSTD2) expression in solid tumors. Oncologist 2024; 29:e1480-e1491. [PMID: 38986529 PMCID: PMC11546728 DOI: 10.1093/oncolo/oyae168] [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: 03/25/2024] [Accepted: 06/13/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND TROP2 (TACSTD2) expression is associated with decreased overall survival (OS) in some solid tumors, and the TROP2-targeting antibody-drug conjugate (ADC) sacituzumab govitecan has been approved in breast and urothelial carcinomas. We aimed to explore the multi-omic landscape associated with TACSTD2 gene expression in various solid tumors to identify patients most likely to benefit from this approach. METHODS Breast (N = 11 246), colorectal (N = 15 425), hepatocellular (N = 433), pancreatic (N = 5488), and urothelial (N = 4125) tumors were stratified into quartiles by TACSTD2 gene expression, analyzed by next-generation DNA sequencing, whole transcriptome sequencing, and immunohistochemistry at Caris Life Sciences (Phoenix, AZ). Survival data were obtained from insurance claims, and Kaplan-Meier estimates were calculated for molecularly defined cohorts. RESULTS Several pathogenic mutations were associated with TACSTD2-high tumors, including TP53 in breast, colorectal (CRC), pancreatic, and hepatocellular cancers; KRAS in pancreatic and CRC cancers; ARID1A and FGFR3 in urothelial cancer; and CTNNB1 in hepatocellular cancer. TACSTD2-low breast tumors were enriched for copy number amplifications in CCND1 and FGF/R family member genes. TACSTD2 high was generally associated with more immune cell infiltration and greater T-cell inflammation scores. Patients with TACSTD2-high breast, CRC, and pancreatic cancers demonstrated a significantly shorter OS than TACSTD2-low tumors. This was restricted to CRC with microsatellite stable tumors and patients with pancreatic cancer with KRAS-mutant tumors. Patients with breast cancer with TACSTD2-high tumors also experienced significantly worse OS following immune checkpoint inhibitors. CONCLUSIONS TACSTD2 expression is associated with key driver alterations and a more active immune microenvironment, suggesting possible combinatorial strategies with TROP2-targeting ADCs plus immunotherapy in various solid tumors.
Collapse
Affiliation(s)
- Dan Morgenstern-Kaplan
- Department of Medicine, Division of Medical Oncology, University of Miami Sylvester Comprehensive Cancer Center/Jackson Memorial Hospital, Miami, FL 33131, United States
| | - Samuel A Kareff
- Department of Medicine, Division of Medical Oncology, University of Miami Sylvester Comprehensive Cancer Center/Jackson Memorial Hospital, Miami, FL 33131, United States
| | - Asaad Trabolsi
- Department of Medicine, Division of Medical Oncology, University of Miami Sylvester Comprehensive Cancer Center/Jackson Memorial Hospital, Miami, FL 33131, United States
| | - Estelamari Rodriguez
- Department of Medicine, Division of Medical Oncology, University of Miami Sylvester Comprehensive Cancer Center/Jackson Memorial Hospital, Miami, FL 33131, United States
| | - Harris Krause
- Caris Life Sciences, Phoenix, AZ 85040, United States
| | | | - Heng Tan
- Department of Medicine, Division of Medical Oncology, University of Miami Sylvester Comprehensive Cancer Center/Jackson Memorial Hospital, Miami, FL 33131, United States
| | | | - Emil Lou
- University of Minnesota Masonic Cancer Center, Minneapolis, MN 55455, United States
| | - Misako Nagasaka
- Division of Hematology/Oncology, University of California Irvine School of Medicine, Orange, CA 92617, United States
| | - Sandra Algaze
- Division of Medical Oncology, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033, United States
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033, United States
| | - Stephen V Liu
- Georgetown Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20007, United States
| | - Balazs Halmos
- Montefiore Einstein Comprehensive Cancer Center, Bronx, NY 10461, United States
| | - Dave S B Hoon
- Saint John’s Cancer Institute, Providence Health System, Santa Monica, CA 90404, United States
| | - Andreas Seeber
- Tyrolean Cancer Research Institute, Innsbruck Medical University, Innsbruck 6020, Austria
| | - Patrick C Ma
- Division of Hematology/Oncology, Penn State Cancer Institute, Hershey, PA 17033, United States
| | - Wafik S El-Deiry
- Legorreta Cancer Center, Warren Alpert Medical School of Brown University, Providence, RI 02912, United States
| | | | - Gilberto Lopes
- Department of Medicine, Division of Medical Oncology, University of Miami Sylvester Comprehensive Cancer Center/Jackson Memorial Hospital, Miami, FL 33131, United States
| |
Collapse
|
2
|
van Diest E, Nicolasen MJT, Kramer L, Zheng J, Hernández-López P, Beringer DX, Kuball J. The making of multivalent gamma delta TCR anti-CD3 bispecific T cell engagers. Front Immunol 2023; 13:1052090. [PMID: 36685546 PMCID: PMC9851377 DOI: 10.3389/fimmu.2022.1052090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 12/15/2022] [Indexed: 01/07/2023] Open
Abstract
Introduction We have recently developed a novel T cell engager concept by utilizing γ9δ2TCR as tumor targeting domain, named gamma delta TCR anti-CD3 bispecific molecule (GAB), targeting the phosphoantigen-dependent orchestration of BTN2A1 and BTN3A1 at the surface of cancer cells. GABs are made by the fusion of the ectodomains of a γδTCR to an anti-CD3 single chain variable fragment (scFv) (γδECTO-αCD3), here we explore alternative designs with the aim to enhance GAB effectivity. Methods The first alternative design was made by linking the variable domains of the γ and δ chain to an anti-CD3 scFv (γδVAR-αCD3). The second alternative design was multimerizing γδVAR-αCD3 proteins to increase the tumor binding valency. Both designs were expressed and purified and the potency to target tumor cells by T cells of the alternative designs was compared to γδECTO-αCD3, in T cell activation and cytotoxicity assays. Results and discussion The γδVAR-αCD3 proteins were poorly expressed, and while the addition of stabilizing mutations based on finding for αβ single chain formats increased expression, generation of meaningful amounts of γδVAR-αCD3 protein was not possible. As an alternative strategy, we explored the natural properties of the original GAB design (γδECTO-αCD3), and observed the spontaneous formation of γδECTO-αCD3-monomers and -dimers during expression. We successfully enhanced the fraction of γδECTO-αCD3-dimers by shortening the linker length between the heavy and light chain in the anti-CD3 scFv, though this also decreased protein yield by 50%. Finally, we formally demonstrated with purified γδECTO-αCD3-dimers and -monomers, that γδECTO-αCD3-dimers are superior in function when compared to similar concentrations of monomers, and do not induce T cell activation without simultaneous tumor engagement. In conclusion, a γδECTO-αCD3-dimer based GAB design has great potential, though protein production needs to be further optimized before preclinical and clinical testing.
Collapse
Affiliation(s)
- Eline van Diest
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Mara J. T. Nicolasen
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Lovro Kramer
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Jiali Zheng
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Patricia Hernández-López
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Dennis X. Beringer
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Jürgen Kuball
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands,Department of Hematology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands,*Correspondence: Jürgen Kuball,
| |
Collapse
|
3
|
Cheal SM, Chung SK, Vaughn BA, Cheung NKV, Larson SM. Pretargeting: A Path Forward for Radioimmunotherapy. J Nucl Med 2022; 63:1302-1315. [PMID: 36215514 DOI: 10.2967/jnumed.121.262186] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/07/2022] [Indexed: 12/19/2022] Open
Abstract
Pretargeted radioimmunodiagnosis and radioimmunotherapy aim to efficiently combine antitumor antibodies and medicinal radioisotopes for high-contrast imaging and high-therapeutic-index (TI) tumor targeting, respectively. As opposed to conventional radioimmunoconjugates, pretargeted approaches separate the tumor-targeting step from the payload step, thereby amplifying tumor uptake while reducing normal-tissue exposure. Alongside contrast and TI, critical parameters include antibody immunogenicity and specificity, availability of radioisotopes, and ease of use in the clinic. Each of the steps can be optimized separately; as modular systems, they can find broad applications irrespective of tumor target, tumor type, or radioisotopes. Although this versatility presents enormous opportunity, pretargeting is complex and presents unique challenges for clinical translation and optimal use in patients. The purpose of this article is to provide a brief historical perspective on the origins and development of pretargeting strategies in nuclear medicine, emphasizing 2 protein delivery systems that have been extensively evaluated (i.e., biotin-streptavidin and hapten-bispecific monoclonal antibodies), as well as radiohaptens and radioisotopes. We also highlight recent innovations, including pretargeting with bioorthogonal chemistry and novel protein vectors (such as self-assembling and disassembling proteins and Affibody molecules). We caution the reader that this is by no means a comprehensive review of the past 3 decades of pretargeted radioimmunodiagnosis and pretargeted radioimmunotherapy. But we do aim to highlight major developmental milestones and to identify benchmarks for success with regard to TI and toxicity in preclinical models and clinically. We believe this approach will lead to the identification of key obstacles to clinical success, revive interest in the utility of radiotheranostics applications, and guide development of the next generation of pretargeted theranostics.
Collapse
Affiliation(s)
- Sarah M Cheal
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York;
| | - Sebastian K Chung
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brett A Vaughn
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Steven M Larson
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
4
|
Monterrey DT, Ayuso-Fernández I, Oroz-Guinea I, García-Junceda E. Design and biocatalytic applications of genetically fused multifunctional enzymes. Biotechnol Adv 2022; 60:108016. [PMID: 35781046 DOI: 10.1016/j.biotechadv.2022.108016] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 06/27/2022] [Accepted: 06/27/2022] [Indexed: 01/01/2023]
Abstract
Fusion proteins, understood as those created by joining two or more genes that originally encoded independent proteins, have numerous applications in biotechnology, from analytical methods to metabolic engineering. The use of fusion enzymes in biocatalysis may be even more interesting due to the physical connection of enzymes catalyzing successive reactions into covalently linked complexes. The proximity of the active sites of two enzymes in multi-enzyme complexes can make a significant contribution to the catalytic efficiency of the reaction. However, the physical proximity of the active sites does not guarantee this result. Other aspects, such as the nature and length of the linker used for the fusion or the order in which the enzymes are fused, must be considered and optimized to achieve the expected increase in catalytic efficiency. In this review, we will relate the new advances in the design, creation, and use of fused enzymes with those achieved in biocatalysis over the past 20 years. Thus, we will discuss some examples of genetically fused enzymes and their application in carbon‑carbon bond formation and oxidative reactions, generation of chiral amines, synthesis of carbohydrates, biodegradation of plant biomass and plastics, and in the preparation of other high-value products.
Collapse
Affiliation(s)
- Dianelis T Monterrey
- Departamento de Química Bioorgánica, Instituto de Química Orgánica General (IQOG), CSIC, Juan de la Cierva 3, 28006 Madrid, Spain.
| | - Iván Ayuso-Fernández
- Departamento de Química Bioorgánica, Instituto de Química Orgánica General (IQOG), CSIC, Juan de la Cierva 3, 28006 Madrid, Spain.
| | - Isabel Oroz-Guinea
- Departamento de Química Bioorgánica, Instituto de Química Orgánica General (IQOG), CSIC, Juan de la Cierva 3, 28006 Madrid, Spain.
| | - Eduardo García-Junceda
- Departamento de Química Bioorgánica, Instituto de Química Orgánica General (IQOG), CSIC, Juan de la Cierva 3, 28006 Madrid, Spain.
| |
Collapse
|
5
|
Wei Q, Wang Y, Liu Z, Liu M, Cao S, Jiang H, Xia J. Multienzyme Assembly on Caveolar Membranes In Cellulo. ACS Catal 2022. [DOI: 10.1021/acscatal.2c01906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Qixin Wei
- Department of Chemistry and Center for Cell & Developmental Biology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Yue Wang
- Department of Chemistry and Center for Cell & Developmental Biology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Zhenjun Liu
- Department of Chemistry and Center for Cell & Developmental Biology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Min Liu
- Department of Chemistry and Center for Cell & Developmental Biology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Sheng Cao
- Department of Chemistry and Center for Cell & Developmental Biology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Hao Jiang
- Department of Chemistry and Center for Cell & Developmental Biology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Jiang Xia
- Department of Chemistry and Center for Cell & Developmental Biology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| |
Collapse
|
6
|
An albumin scaffold grafted with an alpha-helical motif delivers therapeutic payloads by modular coiled-coil assembly. Int J Biol Macromol 2022; 205:376-384. [PMID: 35157904 DOI: 10.1016/j.ijbiomac.2022.02.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 02/02/2022] [Accepted: 02/08/2022] [Indexed: 11/22/2022]
Abstract
A short in vivo half-life of protein-based therapeutics often restricts successful clinical translation despite their promising efficacy in vitro. As a biocompatible half-life extender, human serum albumin (HSA) has proven effective in some cases. While genetic fusion is well-established for interlinking HSA and a protein payload, it is limited to structurally simple proteins, necessitating new strategies to expand the utility of HSA for delivery of therapeutic proteins. Here, we report a novel HSA variant (eHSA) as a modular and long-acting carrier compatible with any protein payload of interest. The assembly between eHSA and a payload was driven by a heterodimeric coiled-coil interaction in which a short α-helix grafted onto HSA specifically bound to a complementary α-helix genetically fused to a payload. We showed various proteins including tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), single-chain TRAIL, or green fluorescent protein could piggyback onto eHSA via simple mixing without losing native activity. Additionally, either in presence or absence of a payload, eHSA was found to retain the pH-dependent FcRn-binding behavior - a critical attribute for prolonged survival in the systemic circulation. These results demonstrate eHSA would serve as a modular platform capable of delivering various therapeutic proteins with potentially long in vivo half-lives.
Collapse
|
7
|
Modular Assembly of Phosphite Dehydrogenase and Phenylacetone Monooxygenase for Tuning Cofactor Regeneration. Biomolecules 2021; 11:biom11060905. [PMID: 34204515 PMCID: PMC8234031 DOI: 10.3390/biom11060905] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/10/2021] [Accepted: 06/13/2021] [Indexed: 02/07/2023] Open
Abstract
The use of multienzyme complexes can facilitate biocatalytic cascade reactions by employing fusion enzymes or protein tags. In this study, we explored the use of recently developed peptide tags that promote complex formation of the targeted proteins: the dimerization-docking and anchoring domain (RIDD–RIAD) system. These peptides allow self-assembly based on specific protein–protein interactions between both peptides and allow tuning of the ratio of the targeted enzymes as the RIAD peptide binds to two RIDD peptides. Each of these tags were added to the C-terminus of a NADPH-dependent Baeyer–Villiger monooxygenase (phenylacetone monooxygenase, PAMO) and a NADPH-regenerating enzyme (phosphite dehydrogenase, PTDH). Several RIDD/RIAD-tagged PAMO and PTDH variants were successfully overproduced in E. coli and subsequently purified. Complementary tagged enzymes were mixed and analyzed for their oligomeric state, stability, and activity. Complexes were formed in the case of some specific combinations (PAMORIAD–PTDHRIDD and PAMORIAD/RIAD–PTDHRIDD). These enzyme complexes displayed similar catalytic activity when compared with the PTDH–PAMO fusion enzyme. The thermostability of PAMO in these complexes was retained while PTDH displayed somewhat lower thermostability. Evaluation of the biocatalytic performance by conducting conversions revealed that with a self-assembled PAMO–PTDH complex less PTDH was required for the same performance when compared with the PTDH–PAMO fusion enzyme.
Collapse
|
8
|
Fernandes CSM, Pina AS, Roque ACA. Affinity-triggered hydrogels: Developments and prospects in biomaterials science. Biomaterials 2020; 268:120563. [PMID: 33276200 DOI: 10.1016/j.biomaterials.2020.120563] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 01/05/2023]
Affiliation(s)
- Cláudia S M Fernandes
- UCIBIO, Chemistry Department, School of Science and Technology, NOVA University of Lisbon, Campus Caparica, 2829-516, Caparica, Portugal
| | - Ana Sofia Pina
- UCIBIO, Chemistry Department, School of Science and Technology, NOVA University of Lisbon, Campus Caparica, 2829-516, Caparica, Portugal
| | - Ana Cecília A Roque
- UCIBIO, Chemistry Department, School of Science and Technology, NOVA University of Lisbon, Campus Caparica, 2829-516, Caparica, Portugal.
| |
Collapse
|
9
|
Hosseini SS, Khalili S, Baradaran B, Bidar N, Shahbazi MA, Mosafer J, Hashemzaei M, Mokhtarzadeh A, Hamblin MR. Bispecific monoclonal antibodies for targeted immunotherapy of solid tumors: Recent advances and clinical trials. Int J Biol Macromol 2020; 167:1030-1047. [PMID: 33197478 DOI: 10.1016/j.ijbiomac.2020.11.058] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/04/2020] [Accepted: 11/09/2020] [Indexed: 02/07/2023]
Abstract
Bispecific antibodie (BsAbs) combine two or more epitope-recognizing sequences into a single protein molecule. The first therapeutic applications of BsAbs were focused on cancer therapy. However, these antibodies have grown to cover a wider disease spectrum, including imaging, diagnosis, prophylaxis, and therapy of inflammatory and autoimmune diseases. BsAbs can be categorized into IgG-like formats and non-IgG-like formats. Different technologies have been used for the construction of BsAbs including "CrossMAb", "Quadroma", "knobs-into-holes" and molecular cloning. The mechanism of action for BsAbs includes the induction of CDC, ADCC, ADCP, apoptosis, and recruitment of cell surface receptors, as well as activation or inhibition of signaling pathways. The first clinical trials included mainly leukemia and lymphoma, but solid tumors are now being investigated. The BsAbs bind to a tumor-specific antigen using one epitope, while the second epitope binds to immune cell receptors such as CD3, CD16, CD64, and CD89, with the goal of stimulating the immune response against cancer cells. Currently, over 20 different commercial methods have been developed for the construction of BsAbs. Three BsAbs are currently clinically approved and marketed, and more than 85 clinical trials are in progress. In the present review, we discuss recent trends in the design, engineering, clinical applications, and clinical trials of BsAbs in solid tumors.
Collapse
Affiliation(s)
- Seyed Samad Hosseini
- Department of Biotechnology, Higher Education Institute of Rab-Rashid, Tabriz, Iran
| | - Saeed Khalili
- Department of Biology Sciences, Faculty of Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Negar Bidar
- Department of Biotechnology, Higher Education Institute of Rab-Rashid, Tabriz, Iran
| | - Mohammad-Ali Shahbazi
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Jafar Mosafer
- Nanotechnology Research center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Biotechnology, School of Paramedical Science, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Mahmoud Hashemzaei
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Zabol University of Medical Sciences, Zabol, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, 02114, USA; Department of Dermatology, Harvard Medical School, Boston, MA, 02115, USA; Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa.
| |
Collapse
|
10
|
Sutherland AR, Alam MK, Geyer CR. Post‐translational Assembly of Protein Parts into Complex Devices by Using SpyTag/SpyCatcher Protein Ligase. Chembiochem 2018; 20:319-328. [DOI: 10.1002/cbic.201800538] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Indexed: 01/21/2023]
Affiliation(s)
- Ashley R. Sutherland
- Department of BiochemistryUniversity of Saskatchewan Saskatoon SK S7N 5E5 Canada
| | - Md. Kausar Alam
- Donnelly Centre for Cellular and Biomolecular ResearchUniversity of Toronto Toronto ON M5S3E1 Canada
| | - C. Ronald Geyer
- Department of Pathology and Laboratory MedicineUniversity of Saskatchewan Saskatoon SK S7N 5E5 Canada
| |
Collapse
|
11
|
Goldenberg DM, Stein R, Sharkey RM. The emergence of trophoblast cell-surface antigen 2 (TROP-2) as a novel cancer target. Oncotarget 2018; 9:28989-29006. [PMID: 29989029 PMCID: PMC6034748 DOI: 10.18632/oncotarget.25615] [Citation(s) in RCA: 169] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 05/31/2018] [Indexed: 12/31/2022] Open
Abstract
TROP-2 is a glycoprotein first described as a surface marker of trophoblast cells, but subsequently shown to be increased in many solid cancers, with lower expression in certain normal tissues. It regulates cancer growth, invasion and spread by several signaling pathways, and has a role in stem cell biology and other diseases. This review summarizes TROP-2's properties, especially in cancer, and particularly its role as a target for antibody-drug conjugates (ADC) or immunotherapy. When the irinotecan metabolite, SN-38, is conjugated to a humanized anti-TROP-2 antibody (sacituzumab govitecan), it shows potent broad anticancer activity in human cancer xenografts and in patients with advanced triple-negative breast, non-small cell and small-cell lung, as well as urothelial cancers.
Collapse
Affiliation(s)
- David M. Goldenberg
- Center for Molecular Medicine and Immunology, Belleville, NJ, USA
- IBC Pharmaceuticals, Inc., Morris Plains, NJ, USA
| | - Rhona Stein
- Center for Molecular Medicine and Immunology, Belleville, NJ, USA
| | - Robert M. Sharkey
- Center for Molecular Medicine and Immunology, Belleville, NJ, USA
- Immunomedics, Inc., Morris Plains, NJ, USA
| |
Collapse
|
12
|
Jin J, Park G, Park JB, Kim S, Kim H, Chung J. An anti-EGFR × cotinine bispecific antibody complexed with cotinine-conjugated duocarmycin inhibits growth of EGFR-positive cancer cells with KRAS mutations. Exp Mol Med 2018; 50:1-14. [PMID: 29795377 PMCID: PMC5967348 DOI: 10.1038/s12276-018-0096-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 03/10/2018] [Accepted: 03/16/2018] [Indexed: 11/30/2022] Open
Abstract
Antibody-drug conjugates (ADCs) can selectively deliver cytotoxic agents to tumor cells and are frequently more potent than naked antibodies. However, optimization of the conjugation process between antibodies and cytotoxic agents and characterization of ADCs are laborious and time-consuming processes. Here, we describe a novel ADC platform using a tetravalent bispecific antibody that simultaneously binds to the tumor-associated antigen and a hapten conjugated to a cytotoxic agent. We selected cotinine as the hapten because it is not present in biological systems and is inert and nontoxic. We prepared an anti-epidermal growth factor receptor (EGFR) × cotinine bispecific antibody and mixed it with an equimolar amount of cotinine-conjugated duocarmycin to form the ADC. This ADC showed significant in vitro and in vivo antitumor activity against EGFR-positive, cetuximab-refractory lung adenocarcinoma cells with KRAS mutations. A new method for producing antibody-drug conjugates (ADCs), pairing anti-cancer drugs with antibodies that deliver them specifically to cancer cells, may help to develop therapies for hard-to-treat cancers. ADCs show promise for treating several cancers, but are difficult to produce. Junho Chung at the Seoul National University College of Medicine in South Korea and co-workers tested a new way of forming the conjugates, using cotinine, a non-toxic molecule related to nicotine, as a link. They bound cotinine to the anti-cancer drug duocarmycin, then formulated an antibody with two binding sites, one for cotinine, one for tumor cells. Combining the drug−cotinine complex with the antibody resulted in a drug–cotinine-antibody ADC that delivered duocarmycin exclusively to cancer cells and inhibited tumor growth in mice. This simplified method may help develop treatments for other drug-resistant cancers.
Collapse
Affiliation(s)
- Junyeong Jin
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, 00380, Republic of Korea.,Department of Biomedical Science, Seoul National University College of Medicine, Seoul, 00380, Republic of Korea
| | - Gunwoo Park
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, 00380, Republic of Korea.,Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang, 10408, Republic of Korea
| | - Jong Bae Park
- Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang, 10408, Republic of Korea.,Department of Cancer Biomedical Science, National Cancer Center, Graduate School of Cancer Science and Policy, 323 Ilsan-ro, Ilsandong-gu, Goyang, 10408, Republic of Korea
| | - Soohyun Kim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, 00380, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, 00380, Republic of Korea
| | - Hyori Kim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, 00380, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, 00380, Republic of Korea.,Asan Institute for Life Sciences, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Junho Chung
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, 00380, Republic of Korea. .,Department of Biomedical Science, Seoul National University College of Medicine, Seoul, 00380, Republic of Korea. .,Cancer Research Institute, Seoul National University College of Medicine, Seoul, 00380, Republic of Korea.
| |
Collapse
|
13
|
Nastase MV, Zeng-Brouwers J, Wygrecka M, Schaefer L. Targeting renal fibrosis: Mechanisms and drug delivery systems. Adv Drug Deliv Rev 2018; 129:295-307. [PMID: 29288033 DOI: 10.1016/j.addr.2017.12.019] [Citation(s) in RCA: 168] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 11/10/2017] [Accepted: 12/22/2017] [Indexed: 12/21/2022]
Abstract
Renal fibrosis is the common outcome of many chronic kidney diseases (CKD) independent of the underlying etiology. Despite a host of promising experimental data, currently available strategies only ameliorate or delay the progression of CKD but do not reverse fibrosis. One of the major impediments of translating novel antifibrotic strategies from bench to bedside is due to the intricacies of the drug delivery process. In this review, we briefly describe mechanisms of renal fibrosis and methods of drug transfer into the kidney. Various tools used in gene therapy to administer nucleic acids in vivo are discussed. Furthermore, we review the modes of action of protein- or peptide-based drugs with target-specific antibodies and cytokines incorporated in hydrogels. Additionally, we assess an intriguing new method to deliver drugs specifically to tubular epithelial cells via conjugation with ligands binding to the megalin receptor. Finally, plant-derived compounds with antifibrotic properties are also summarized.
Collapse
Affiliation(s)
- Madalina V Nastase
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe-Universität, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; National Institute for Chemical-Pharmaceutical Research and Development, 112 Vitan Avenue, 031299 Bucharest, Romania
| | - Jinyang Zeng-Brouwers
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe-Universität, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Malgorzata Wygrecka
- Department of Biochemistry, Faculty of Medicine, Universities of Giessen and Marburg Lung Center, Friedrichstrasse 24, 35392 Giessen, Germany.
| | - Liliana Schaefer
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe-Universität, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| |
Collapse
|
14
|
Sedykh SE, Prinz VV, Buneva VN, Nevinsky GA. Bispecific antibodies: design, therapy, perspectives. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:195-208. [PMID: 29403265 PMCID: PMC5784585 DOI: 10.2147/dddt.s151282] [Citation(s) in RCA: 190] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Antibodies (Abs) containing two different antigen-binding sites in one molecule are called bispecific. Bispecific Abs (BsAbs) were first described in the 1960s, the first monoclonal BsAbs were generated in the 1980s by hybridoma technology, and the first article describing the therapeutic use of BsAbs was published in 1992, but the number of papers devoted to BsAbs has increased significantly in the last 10 years. Particular interest in BsAbs is due to their therapeutic use. In the last decade, two BsAbs - catumaxomab in 2009 and blinatumomab in 2014, were approved for therapeutic use. Papers published in recent years have been devoted to various methods of BsAb generation by genetic engineering and chemical conjugation, and describe preclinical and clinical trials of these drugs in a variety of diseases. This review considers diverse BsAb-production methods, describes features of therapeutic BsAbs approved for medical use, and summarizes the prospects of practical application of promising new BsAbs.
Collapse
Affiliation(s)
- Sergey E Sedykh
- Laboratory of Repair Enzymes, Siberian Branch of Russian Academy of Sciences Institute of Chemical Biology and Fundamental Medicine, Novosibirsk State University, Novosibirsk, Russia
| | - Victor V Prinz
- Laboratory of Repair Enzymes, Siberian Branch of Russian Academy of Sciences Institute of Chemical Biology and Fundamental Medicine, Novosibirsk State University, Novosibirsk, Russia
| | - Valentina N Buneva
- Laboratory of Repair Enzymes, Siberian Branch of Russian Academy of Sciences Institute of Chemical Biology and Fundamental Medicine, Novosibirsk State University, Novosibirsk, Russia
| | - Georgy A Nevinsky
- Laboratory of Repair Enzymes, Siberian Branch of Russian Academy of Sciences Institute of Chemical Biology and Fundamental Medicine, Novosibirsk State University, Novosibirsk, Russia
| |
Collapse
|
15
|
Okesola BO, Mata A. Multicomponent self-assembly as a tool to harness new properties from peptides and proteins in material design. Chem Soc Rev 2018; 47:3721-3736. [DOI: 10.1039/c8cs00121a] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Nature is enriched with a wide variety of complex, synergistic and highly functional protein-based multicomponent assemblies.
Collapse
Affiliation(s)
- Babatunde O. Okesola
- School of Engineering and Materials Science
- Institute of Bioengineering
- Queen Mary University of London
- UK
| | - Alvaro Mata
- School of Engineering and Materials Science
- Institute of Bioengineering
- Queen Mary University of London
- UK
| |
Collapse
|
16
|
Chang CH, Goldenberg DM. Enhancing the antitumor potency of T cells redirected by bispecific antibodies. Oncoscience 2017; 4:120-121. [PMID: 29142899 PMCID: PMC5672892 DOI: 10.18632/oncoscience.366] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 10/03/2017] [Indexed: 01/13/2023] Open
Affiliation(s)
- Chien-Hsing Chang
- IBC Pharmaceuticals Inc., and Immunomedics Inc., Morris Plains, New Jersey, USA
| | - David M Goldenberg
- IBC Pharmaceuticals Inc., and Immunomedics Inc., Morris Plains, New Jersey, USA
| |
Collapse
|
17
|
Green DJ, Press OW. Whither Radioimmunotherapy: To Be or Not To Be? Cancer Res 2017; 77:2191-2196. [PMID: 28428282 DOI: 10.1158/0008-5472.can-16-2523] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 10/26/2016] [Accepted: 01/20/2017] [Indexed: 01/08/2023]
Abstract
Therapy of cancer with radiolabeled monoclonal antibodies has produced impressive results in preclinical experiments and in clinical trials conducted in radiosensitive malignancies, particularly B-cell lymphomas. Two "first-generation," directly radiolabeled anti-CD20 antibodies, 131iodine-tositumomab and 90yttrium-ibritumomab tiuxetan, were FDA-approved more than a decade ago but have been little utilized because of a variety of medical, financial, and logistic obstacles. Newer technologies employing multistep "pretargeting" methods, particularly those utilizing bispecific antibodies, have greatly enhanced the therapeutic efficacy of radioimmunotherapy and diminished its toxicities. The dramatically improved therapeutic index of bispecific antibody pretargeting appears to be sufficiently compelling to justify human clinical trials and reinvigorate enthusiasm for radioimmunotherapy in the treatment of malignancies, particularly lymphomas. Cancer Res; 77(9); 2191-6. ©2017 AACR.
Collapse
Affiliation(s)
- Damian J Green
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington. .,Department of Medicine, University of Washington, Seattle, Washington
| | - Oliver W Press
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Department of Medicine, University of Washington, Seattle, Washington.,Department of Bioengineering, University of Washington, Seattle, Washington
| |
Collapse
|
18
|
Thakur A, Lum LG. "NextGen" Biologics: Bispecific Antibodies and Emerging Clinical Results. Expert Opin Biol Ther 2016; 16:675-88. [PMID: 26848610 DOI: 10.1517/14712598.2016.1150996] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Bispecific antibodies (BsAb) are emerging as a novel approach for dual targeting strategies. Two bispecific antibodies are approved for therapy and >30 are in clinical development. The first generation of BsAb were produced by chemical cross-linking or hybridoma technology; with the recent advent of genetic and protein engineering technologies numerous formats of bispecific antibodies have emerged using either the fragments of IgG or whole IgG molecules. Further areas of development include dual blockade of different disease pathways, diagnosis and imaging. AREAS COVERED Biologics, including bi- or multi-specific antibodies and T cell-based approaches are rapidly changing the landscape of cancer therapeutics. New engineering platforms for bi- or multi-specific antibodies and scaffolds offer improved efficacy and reduced toxicities over IgG-based monoclonal antibodies. Preclinical and clinical studies using different formats of BsAbs are described in this review using PubMed as a literature search tool. EXPERT OPINION A comprehensive presentation of preclinical data and clinical trials evaluating the various formats of BsAbs indicate their safety and efficacy. However, a vast opportunity to fine tune physical properties and functional activity of biologics to improve the stability, engagement of cytotoxic CD8 T cells and multi-antigen targeting strategy through protein engineering holds a greater therapeutic potential.
Collapse
Affiliation(s)
- Archana Thakur
- a Department of Oncology , Wayne State University and Karmanos Cancer Institute , Detroit , MI , USA
| | - Lawrence G Lum
- a Department of Oncology , Wayne State University and Karmanos Cancer Institute , Detroit , MI , USA.,b Department of Medicine , Wayne State University and Karmanos Cancer Institute , Detroit , MI , USA.,c Department of Pediatrics , Wayne State University and Karmanos Cancer Institute , Detroit , MI , USA.,d Department of Immunology and Microbiology , Wayne State University and Karmanos Cancer Institute , Detroit , MI , USA
| |
Collapse
|
19
|
Spiess C, Zhai Q, Carter PJ. Alternative molecular formats and therapeutic applications for bispecific antibodies. Mol Immunol 2015; 67:95-106. [DOI: 10.1016/j.molimm.2015.01.003] [Citation(s) in RCA: 417] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 12/30/2014] [Accepted: 01/02/2015] [Indexed: 12/21/2022]
|
20
|
Xu Y, Lee J, Tran C, Heibeck TH, Wang WD, Yang J, Stafford RL, Steiner AR, Sato AK, Hallam TJ, Yin G. Production of bispecific antibodies in "knobs-into-holes" using a cell-free expression system. MAbs 2015; 7:231-42. [PMID: 25427258 PMCID: PMC4623329 DOI: 10.4161/19420862.2015.989013] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Bispecific antibodies have emerged in recent years as a promising field of research for therapies in oncology, inflammable diseases, and infectious diseases. Their capability of dual target recognition allows for novel therapeutic hypothesis to be tested, where traditional mono-specific antibodies would lack the needed mode of target engagement. Among extremely diverse architectures of bispecific antibodies, knobs-into-holes (KIHs) technology, which involves engineering CH3 domains to create either a “knob” or a “hole” in each heavy chain to promote heterodimerization, has been widely applied. Here, we describe the use of a cell-free expression system (Xpress CF) to produce KIH bispecific antibodies in multiple scaffolds, including 2-armed heterodimeric scFv-KIH and one-armed asymmetric BiTE-KIH with tandem scFv. Efficient KIH production can be achieved by manipulating the plasmid ratio between knob and hole, and further improved by addition of prefabricated knob or hole. These studies demonstrate the versatility of Xpress CF in KIH production and provide valuable insights into KIH construct design for better assembly and expression titer.
Collapse
Key Words
- BiTE, bispecific T-cell engager
- BiTE-KIH
- CHO, Chinese hamster ovary
- ELISA, enzyme-linked immunosorbent assay
- EpCAM, epithelial cell adhesion molecule
- FACS, fluorescence-activated cell sorting
- Fab, antigen-binding fragment
- Fc, fragment crystallizable
- FcR, Fc receptor
- HC, immunoglobulin heavy chain
- HER2, human epidermal growth factor receptor 2
- IgG, immunoglobulin G
- KIH, knob-into-hole
- LC, immunoglobulin light chain
- LC-MS, liquid chromatography-mass spectrometry
- PK, pharmacokinetics
- bispecific antibody
- cell-free protein expression
- knob-into-hole
- prefabrication
- scFv, single-chain fragment variable
- scFv-KIH
Collapse
Affiliation(s)
- Yiren Xu
- a Sutro Biopharma, Inc. ; South San Francisco , CA USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Wang H, Heilshorn SC. Adaptable hydrogel networks with reversible linkages for tissue engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2015; 27:3717-36. [PMID: 25989348 PMCID: PMC4528979 DOI: 10.1002/adma.201501558] [Citation(s) in RCA: 434] [Impact Index Per Article: 48.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 04/18/2015] [Indexed: 05/19/2023]
Abstract
Adaptable hydrogels have recently emerged as a promising platform for three-dimensional (3D) cell encapsulation and culture. In conventional, covalently crosslinked hydrogels, degradation is typically required to allow complex cellular functions to occur, leading to bulk material degradation. In contrast, adaptable hydrogels are formed by reversible crosslinks. Through breaking and re-formation of the reversible linkages, adaptable hydrogels can be locally modified to permit complex cellular functions while maintaining their long-term integrity. In addition, these adaptable materials can have biomimetic viscoelastic properties that make them well suited for several biotechnology and medical applications. In this review, an overview of adaptable-hydrogel design considerations and linkage selections is presented, with a focus on various cell-compatible crosslinking mechanisms that can be exploited to form adaptable hydrogels for tissue engineering.
Collapse
Affiliation(s)
- Huiyuan Wang
- Department of Materials Science & Engineering, Stanford University, Stanford, CA 94305, USA
| | - Sarah C. Heilshorn
- Department of Materials Science & Engineering, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
22
|
Liu T, Du J, Luo X, Schultz PG, Wang F. Homogeneously modified immunoglobulin domains for therapeutic application. Curr Opin Chem Biol 2015; 28:66-74. [PMID: 26117722 DOI: 10.1016/j.cbpa.2015.06.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Revised: 06/02/2015] [Accepted: 06/09/2015] [Indexed: 11/28/2022]
Abstract
The field of therapeutic antibodies has been revolutionized over the past decade, led by the development of novel antibody-modification technologies. Besides the huge success achieved by therapeutic monoclonal antibodies, a diversity of antibody derivatives have emerged with hope to outperform their parental antibodies. Here we review the recent development of methodologies to modify immunoglobulin domains and their therapeutic applications. The innovative genetic and chemical approaches enable novel and controllable modifications on immunoglobulin domains, producing homogeneous therapeutics with new functionalities or enhanced therapeutic profiles. Such therapeutics, including antibody-drug conjugates, bispecific antibodies, and antibody/Fc fusion proteins, have demonstrated great prospects in the treatment of cancer, auto-immune diseases, infectious diseases, and many other disorders.
Collapse
Affiliation(s)
- Tao Liu
- California Institute for Biomedical Research (Calibr), 11119 N. Torrey Pines Road, La Jolla, CA 92037, United States
| | - Juanjuan Du
- California Institute for Biomedical Research (Calibr), 11119 N. Torrey Pines Road, La Jolla, CA 92037, United States
| | - Xiaozhou Luo
- Department of Chemistry, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, United States
| | - Peter G Schultz
- California Institute for Biomedical Research (Calibr), 11119 N. Torrey Pines Road, La Jolla, CA 92037, United States; Department of Chemistry, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, United States
| | - Feng Wang
- California Institute for Biomedical Research (Calibr), 11119 N. Torrey Pines Road, La Jolla, CA 92037, United States.
| |
Collapse
|
23
|
Abstract
The eradication of cancer remains a vexing problem despite recent advances in our understanding of the molecular basis of neoplasia. One therapeutic approach that has demonstrated potential involves the selective targeting of radionuclides to cancer-associated cell surface antigens using monoclonal antibodies. Such radioimmunotherapy (RIT) permits the delivery of a high dose of therapeutic radiation to cancer cells, while minimizing the exposure of normal cells. Although this approach has been investigated for several decades, the cumulative advances in cancer biology, antibody engineering and radiochemistry in the past decade have markedly enhanced the ability of RIT to produce durable remissions of multiple cancer types.
Collapse
Affiliation(s)
- Steven M Larson
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA
| | - Jorge A Carrasquillo
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA
| | - Nai-Kong V Cheung
- 1] Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA. [2]
| | - Oliver W Press
- 1] Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, P.O. BOX 19024, Seattle, Washington 98109, USA. [2]
| |
Collapse
|
24
|
Abstract
Targeted treatment of cancer with monoclonal antibodies has added to the beneficial outcome of patients. In an attempt to improve anti-tumor activity of monoclonal antibodies, multi-specific antibodies have entered the research arena. To date, only a few multi-specific constructs have entered phase III clinical trials, in contrast to classical monoclonal antibodies, which are the standard first-line therapy in several tumor entities. In this review, we will assess selected multi-specific antibodies in pre-clinical and clinical development that may be new treatment options for cancer patients in the very near future. We will further evaluate therapy modalities including the timely distribution or the combination of various therapeutic approaches and assess the potential role of multi-specific antibodies in cancer treatment.
Collapse
Affiliation(s)
- Ron D Jachimowicz
- Department I of Internal Medicine, Innate Immunity Group, University Hospital Cologne, Joseph Stelzmann Str. 9, 50937, Cologne, Germany,
| | | | | |
Collapse
|
25
|
Shvartsur A, Bonavida B. Trop2 and its overexpression in cancers: regulation and clinical/therapeutic implications. Genes Cancer 2015; 6:84-105. [PMID: 26000093 PMCID: PMC4426947 DOI: 10.18632/genesandcancer.40] [Citation(s) in RCA: 206] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 10/19/2014] [Indexed: 12/13/2022] Open
Abstract
Trop2 is a transmembrane glycoprotein encoded by the Tacstd2 gene. It is an intracellular calcium signal transducer that is differentially expressed in many cancers. It signals cells for self-renewal, proliferation, invasion, and survival. It has stem cell-like qualities. Trop2 is expressed in many normal tissues, though in contrast, it is overexpressed in many cancers and the overexpression of Trop2 is of prognostic significance. Several ligands have been proposed that interact with Trop2. Trop2 signals the cells via different pathways and it is transcriptionally regulated by a complex network of several transcription factors. Trop2 expression in cancer cells has been correlated with drug resistance. Several strategies target Trop2 on cancer cells that include antibodies, antibody fusion proteins, chemical inhibitors, nanoparticles, etc. The in vitro studies and pre-clinical studies, using these various therapeutic treatments, have resulted in significant inhibition of tumor cell growth both in vitro and in vivo in mice. A clinical study is underway using IMMU-132 (hrS7 linked to SN38) in patients with epithelial cancers. This review describes briefly the various characteristics of cancer cells overexpressing Trop2 and the potential application of Trop2 as both a prognostic biomarker and as a therapeutic target to reverse resistance.
Collapse
Affiliation(s)
- Anna Shvartsur
- Department of Microbiology, Immunology and Molecular Genetics, Jonsson Comprehensive Cancer Center and David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Benjamin Bonavida
- Department of Microbiology, Immunology and Molecular Genetics, Jonsson Comprehensive Cancer Center and David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
26
|
Kim H, Yoon S, Chung J. In vitro and in vivo application of anti-cotinine antibody and cotinine-conjugated compounds. BMB Rep 2014; 47:130-4. [PMID: 24499668 PMCID: PMC4163880 DOI: 10.5483/bmbrep.2014.47.3.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 01/09/2014] [Accepted: 01/20/2014] [Indexed: 12/04/2022] Open
Abstract
The combination of a high-affinity antibody to a hapten, and hapten-conjugated compounds, can provide an alternative to the direct chemical cross-linking of the antibody and compounds. An optimal hapten for in vitro use is one that is absent in biological systems. For in vivo applications, additional characteristics such as pharmacological safety and physiological inertness would be beneficial. Additionally, methods for cross-linking the hapten to various chemical compounds should be available. Cotinine, a major metabolite of nicotine, is considered advantageous in these aspects. A high-affinity anti-cotinine recombinant antibody has recently become available, and can be converted into various formats, including a bispecific antibody. The bispecific anti-cotinine antibody was successfully applied to immunoblot, enzyme immunoassay, immunoaffinity purification, and pre-targeted in vivo radioimmunoimaging. The anti-cotinine IgG molecule could be complexed with aptamers to form a novel affinity unit, and extended the in vivo half-life of aptamers, opening up the possibility of applying the same strategy to therapeutic peptides and chemical compounds. [BMB Reports 2014; 47(3): 130-134]
Collapse
Affiliation(s)
- Hyori Kim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 110-799; Cancer Research Institute, Seoul National University, Seoul 110-799, Korea
| | - Soomin Yoon
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 110-799, Korea
| | - Junho Chung
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 110-799; Cancer Research Institute, Seoul National University, Seoul 110-799, Korea
| |
Collapse
|
27
|
van de Watering FCJ, Rijpkema M, Robillard M, Oyen WJG, Boerman OC. Pretargeted imaging and radioimmunotherapy of cancer using antibodies and bioorthogonal chemistry. Front Med (Lausanne) 2014; 1:44. [PMID: 25593917 PMCID: PMC4292049 DOI: 10.3389/fmed.2014.00044] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 10/22/2014] [Indexed: 01/08/2023] Open
Abstract
Selective delivery of radionuclides to tumors may be accomplished using a two-step approach, in which in the first step the tumor is pretargeted with an unlabeled antibody construct and in the second step the tumor is targeted with a radiolabeled small molecule. This results in a more rapid clearance of the radioactivity from normal tissues due to the fast pharmacokinetics of the small molecule as compared to antibodies. In the last decade, several pretargeting approaches have been tested, which have shown improved tumor-to-background ratios and thus improved imaging and therapy as compared to directly labeled antibodies. In this review, we will discuss the strategies and applications in (pre-)clinical studies of pretargeting concepts based on the use of bispecific antibodies, which are capable of binding to both a target antigen and a radiolabeled peptide. So far, three generations of the bispecific antibody-based pretargeting approach have been studied. The first clinical studies have shown the feasibility and potential for these pretargeting systems to detect and treat tumor lesions. However, to fully integrate the pretargeting approach in clinic, further research should focus on the best regime and pretargeting protocol. Additionally, recent developments in the use of bioorthogonal chemistry for pretargeting of tumors suggest that this chemical pretargeting approach is an attractive alternative strategy for the detection and treatment of tumor lesions.
Collapse
Affiliation(s)
- Floor C J van de Watering
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center , Nijmegen , Netherlands
| | - Mark Rijpkema
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center , Nijmegen , Netherlands
| | | | - Wim J G Oyen
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center , Nijmegen , Netherlands
| | - Otto C Boerman
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center , Nijmegen , Netherlands
| |
Collapse
|
28
|
Liu D, Cardillo TM, Wang Y, Rossi EA, Goldenberg DM, Chang CH. Trop-2-targeting tetrakis-ranpirnase has potent antitumor activity against triple-negative breast cancer. Mol Cancer 2014; 13:53. [PMID: 24606732 PMCID: PMC4015355 DOI: 10.1186/1476-4598-13-53] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 02/26/2014] [Indexed: 02/07/2023] Open
Abstract
Background Ranpirnase (Rap) is an amphibian ribonuclease with reported antitumor activity, minimal toxicity, and negligible immunogenicity in clinical studies, but the unfavorable pharmacokinetics and suboptimal efficacy hampered its further clinical development. To improve the potential of Rap-based therapeutics, we have used the DOCK-AND-LOCK™ (DNL™) method to construct a class of novel IgG-Rap immunoRNases. In the present study, a pair of these constructs, (Rap)2-E1-(Rap)2 and (Rap)2-E1*-(Rap)2, comprising four copies of Rap linked to the CH3 and CK termini of hRS7 (humanized anti-Trop-2), respectively, were evaluated as potential therapeutics for triple-negative breast cancer (TNBC). Methods The DNL-based immunoRNases, (Rap)2-E1-(Rap)2 and (Rap)2-E1*-(Rap)2, were characterized and tested for biological activities in vitro on a panel of breast cancer cell lines and in vivo in a MDA-MB-468 xenograft model. Results (Rap)2-E1-(Rap)2 was highly purified (>95%), exhibited specific cell binding and rapid internalization in MDA-MB-468, a Trop-2-expressing TNBC line, and displayed potent in vitro cytotoxicity (EC50 ≤ 1 nM) against diverse breast cancer cell lines with moderate to high expression of Trop-2, including MDA-MB-468, BT-20, HCC1806, SKBR-3, and MCF-7. In comparison, structural counterparts of (Rap)2-E1-(Rap)2, generated by substituting hRS7 with selective non-Trop-2-binding antibodies, such as epratuzumab (anti-CD22), were at least 50-fold less potent than (Rap)2-E1-(Rap)2 in MDA-MB-468 and BT-20 cells, both lacking the expression of the cognate antigen. Moreover, (Rap)2-E1-(Rap)2 was less effective (EC50 > 50 nM) in MDA-MB-231 (low Trop-2) or HCC1395 (no Trop-2), and did not show any toxicity to human peripheral blood mononuclear cells. In a mouse TNBC model, a significant survival benefit was achieved with (Rap)2-E1*-(Rap)2 when given the maximal tolerated dose. Conclusions A new class of immunoRNases was generated with enhanced potency for targeted therapy of cancer. The promising results from (Rap)2-E1-(Rap)2 and (Rap)2-E1*-(Rap)2 support their further investigation as a potential treatment option for TNBC and other Trop-2-expressing cancers.
Collapse
Affiliation(s)
- Donglin Liu
- IBC Pharmaceuticals, Inc,, Morris Plains 07950, NJ, USA.
| | | | | | | | | | | |
Collapse
|
29
|
Rossi DL, Rossi EA, Cardillo TM, Goldenberg DM, Chang CH. A new class of bispecific antibodies to redirect T cells for cancer immunotherapy. MAbs 2014; 6:381-91. [PMID: 24492297 PMCID: PMC3984327 DOI: 10.4161/mabs.27385] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 11/22/2013] [Accepted: 12/01/2013] [Indexed: 12/31/2022] Open
Abstract
Various constructs of bispecific antibodies (bsAbs) to redirect effector T cells for the targeted killing of tumor cells have shown considerable promise in both preclinical and clinical studies. The single-chain variable fragment (scFv)-based formats, including bispecific T-cell engager (BiTE) and dual-affinity re-targeting (DART), which provide monovalent binding to both CD3 on T cells and to the target antigen on tumor cells, can exhibit rapid blood clearance and neurological toxicity due to their small size (~55 kDa). Herein, we describe the generation, by the modular DOCK-AND-LOCK™) (DNL™) method, of novel T-cell redirecting bispecific antibodies, each comprising a monovalent anti-CD3 scFv covalently conjugated to a stabilized dimer of different anti-tumor Fabs. The potential advantages of this design include bivalent binding to tumor cells, a larger size (~130 kDa) to preclude renal clearance and penetration of the blood-brain barrier, and potent T-cell mediated cytotoxicity. These prototypes were purified to near homogeneity, and representative constructs were shown to provoke the formation of immunological synapses between T cells and their target tumor cells in vitro, resulting in T-cell activation and proliferation, as well as potent T-cell mediated anti-tumor activity. In addition, in vivo studies in NOD/SCID mice bearing Raji Burkitt lymphoma or Capan-1 pancreatic carcinoma indicated statistically significant inhibition of tumor growth compared with untreated controls.
Collapse
Affiliation(s)
| | - Edmund A Rossi
- Immunomedics, Inc; Morris Plains, NJ USA
- IBC Pharmaceuticals, Inc; Morris Plains, NJ USA
| | | | - David M Goldenberg
- Immunomedics, Inc; Morris Plains, NJ USA
- IBC Pharmaceuticals, Inc; Morris Plains, NJ USA
- Garden State Cancer Center; Center for Molecular Medicine and Immunology; Morris Plains, NJ USA
| | - Chien-Hsing Chang
- Immunomedics, Inc; Morris Plains, NJ USA
- IBC Pharmaceuticals, Inc; Morris Plains, NJ USA
| |
Collapse
|
30
|
Hess C, Venetz D, Neri D. Emerging classes of armed antibody therapeutics against cancer. MEDCHEMCOMM 2014. [DOI: 10.1039/c3md00360d] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
31
|
Bispecific Her2 × cotinine antibody in combination with cotinine-(histidine)2-iodine for the pre-targeting of Her2-positive breast cancer xenografts. J Cancer Res Clin Oncol 2013; 140:227-33. [PMID: 24292501 DOI: 10.1007/s00432-013-1548-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 11/11/2013] [Indexed: 10/26/2022]
Abstract
PURPOSE Cotinine has optimal characteristics as a hapten for pre-targeted radioimmunotherapy (PRIT). This study was performed to evaluate the applicability of cotinine/anti-cotinine antibody to PRIT. METHODS We developed and prepared a tandem, single-chain, variable fragment Fc fusion protein [tandem single-chain variable fragment (scFv) Fc fusion protein] that is reactive to both human epidermal growth factor receptor 2 (Her2) and cotinine. Its simultaneous reactivity to Her2 and cotinine was tested in an enzyme-linked immunosorbent assay (ELISA) and two radioimmunoassays (RIA) employing Her2-coated RIA tubes and a Her2-overexpressing cell line. For in vivo imaging, mice bearing Her2-positive tumors were injected with a mixture of tandem scFv Fc fusion and (125)I-cotinine-conjugated histidine dipeptide ((125)I-cotinine peptide). After a delay, (125)I-cotinine peptide was injected again. RESULTS ELISA and RIA results showed that tandem scFv Fc fusion protein successfully bound to both Her2 and cotinine. In single-photon emission computed tomography (SPECT), the complex of tandem scFv Fc fusion protein and (125)I-cotinine peptide was localized to Her2-positive tumor xenografts in mice 4 h after the first injection. Enhanced radioactivity at the site of the Her2-positive tumor lesion was monitored 1 h after the second injection. CONCLUSIONS With these findings, we conclude that the tandem scFv Fc fusion protein and cotinine hapten system have the potential to be applied in PRIT.
Collapse
|
32
|
The Development of Bispecific Hexavalent Antibodies as a Novel Class of DOCK-AND-LOCKTM (DNLTM) Complexes. Antibodies (Basel) 2013. [DOI: 10.3390/antib2020353] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
|
33
|
Interferon-λ1 linked to a stabilized dimer of Fab potently enhances both antitumor and antiviral activities in targeted cells. PLoS One 2013; 8:e63940. [PMID: 23696859 PMCID: PMC3655979 DOI: 10.1371/journal.pone.0063940] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Accepted: 04/08/2013] [Indexed: 01/09/2023] Open
Abstract
The type III interferons (IFNs), comprising IFN-λ1, IFN-λ2, and IFN-λ3, behave similarly to IFN-α in eliciting antiviral, antitumor, and immune-modulating activities. Due to their more restricted cellular targets, IFN-λs are attractive as potential alternatives to existing therapeutic regimens based on IFN-αs. We have applied the DOCK-AND-LOCK™ method to improve the anti-proliferative potency of IFN-λ1 up to 1,000-fold in targeted cancer cell lines by tethering stabilized Fab dimers, derived from hRS7 (humanized anti-Trop-2), hMN-15 (humanized anti-CEACAM6), hL243 (humanized anti-HLA-DR), and c225 (chimeric anti-EGFR), to IFN-λ1 site-specifically, resulting in novel immunocytokines designated (E1)-λ1, (15)-λ1, (C2)-λ1, and (c225)-λ1, respectively. Targeted delivery of IFN-λ1 via (15)-λ1 or (c225)-λ1 to respective antigen-expressing cells also significantly increased antiviral activity when compared with non-targeting (C2)-λ1, as demonstrated in human lung adenocarcinoma cell line A549 by (15)-λ1 against encephalomyocarditis virus (EC50 = 22.2 pM versus 223 pM), and in human hepatocarcinoma cell line Huh-7 by (c225)-λ1 against hepatitis C virus (EC50 = 0.56 pM versus 91.2 pM). These promising results, which are attributed to better localization and stronger binding of IFN-λ1 to antibody-targeted cells, together with the favorable pharmacokinetic profile of (E1)-λ1 in mice (T1/2 = 8.6 h), support further investigation of selective prototypes as potential antiviral and antitumor therapeutic agents.
Collapse
|
34
|
Cardillo TM, Trisal P, Arrojo R, Goldenberg DM, Chang CH. Targeting both IGF-1R and mTOR synergistically inhibits growth of renal cell carcinoma in vitro. BMC Cancer 2013; 13:170. [PMID: 23548153 PMCID: PMC3623828 DOI: 10.1186/1471-2407-13-170] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 03/19/2013] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Advanced or metastatic renal cell carcinoma (RCC) has a poor prognosis, because it is relatively resistant to conventional chemotherapy or radiotherapy. Treatments with human interferon-α2b alone or in combination with mammalian target of rapamycin (mTOR) inhibitors have led to only a modest improvement in clinical outcome. One observation made with mTOR inhibitors is that carcinomas can overcome these inhibitory effects by activating the insulin-like growth factor-I (IGF-I) signaling pathway. Clinically, there is an association of IGF-I receptor (IGF-IR) expression in RCC and poor long-term patient survival. We have developed a humanized anti-IGF-IR monoclonal antibody, hR1, which binds to RCC, resulting in effective down-regulation of IGF-IR and moderate inhibition of cell proliferation in vitro. In this work, we evaluate the anti-tumor activity of two novel IGF-1R-targeting agents against renal cell carcinoma given alone or in combination with an mTOR inhibitor. METHODS hR1 was linked by the DOCK-AND-LOCK™ (DNL™) method to four Fabs of hR1, generating Hex-hR1, or to four molecules of interferon-α2b, generating 1R-2b. Eight human RCC cell lines were screened for IGF-1R expression and sensitivity to treatment with hR1 in vitro. Synergy with an mTOR inhibitor, temsirolimus, was tested in a cell line (ACHN) with low sensitivity to hR1. RESULTS Hex-hR1 induced the down-regulation of IGF-IR at 10-fold lower concentrations compared to the parental hR1. Sensitivity to growth inhibition mediated by hR1 and Hex-hR1 treatments correlated with IGF-1R expression (higher expression was more sensitive). The potency of 1R-2b to inhibit the in vitro growth of RCC was also demonstrated in two human cell lines, ACHN and 786-O, with EC50-values of 63 and 48 pM, respectively. When combined with temsirolimus, a synergistic growth-inhibition with hR1, Hex-hR1, and 1R-2b was observed in ACHN cells at concentrations as low as 10 nM for hR1, 1 nM for Hex-hR1, and 2.6 nM for 1R-2b. CONCLUSIONS Both Hex-hR1 and 1R-2b proved to be more potent than parental hR1 in inhibiting growth of RCC in vitro. Synergy was achieved when each of the three hR1-based agents was combined with temsirolimus, suggesting a new approach for treating RCC.
Collapse
|
35
|
Klöhn PC, Wuellner U, Zizlsperger N, Zhou Y, Tavares D, Berger S, Zettlitz KA, Proetzel G, Yong M, Begent RH, Reichert JM. IBC's 23rd Annual Antibody Engineering, 10th Annual Antibody Therapeutics international conferences and the 2012 Annual Meeting of The Antibody Society: December 3-6, 2012, San Diego, CA. MAbs 2013; 5:178-201. [PMID: 23575266 PMCID: PMC3893229 DOI: 10.4161/mabs.23655] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 01/17/2013] [Indexed: 01/13/2023] Open
Abstract
The 23rd Annual Antibody Engineering, 10th Annual Antibody Therapeutics international conferences, and the 2012 Annual Meeting of The Antibody Society, organized by IBC Life Sciences with contributions from The Antibody Society and two Scientific Advisory Boards, were held December 3-6, 2012 in San Diego, CA. The meeting drew over 800 participants who attended sessions on a wide variety of topics relevant to antibody research and development. As a prelude to the main events, a pre-conference workshop held on December 2, 2012 focused on intellectual property issues that impact antibody engineering. The Antibody Engineering Conference was composed of six sessions held December 3-5, 2012: (1) From Receptor Biology to Therapy; (2) Antibodies in a Complex Environment; (3) Antibody Targeted CNS Therapy: Beyond the Blood Brain Barrier; (4) Deep Sequencing in B Cell Biology and Antibody Libraries; (5) Systems Medicine in the Development of Antibody Therapies/Systematic Validation of Novel Antibody Targets; and (6) Antibody Activity and Animal Models. The Antibody Therapeutics conference comprised four sessions held December 4-5, 2012: (1) Clinical and Preclinical Updates of Antibody-Drug Conjugates; (2) Multifunctional Antibodies and Antibody Combinations: Clinical Focus; (3) Development Status of Immunomodulatory Therapeutic Antibodies; and (4) Modulating the Half-Life of Antibody Therapeutics. The Antibody Society's special session on applications for recording and sharing data based on GIATE was held on December 5, 2012, and the conferences concluded with two combined sessions on December 5-6, 2012: (1) Development Status of Early Stage Therapeutic Antibodies; and (2) Immunomodulatory Antibodies for Cancer Therapy.
Collapse
Affiliation(s)
- Peter-Christian Klöhn
- MRC Prion Unit; Department of Neurodegenerative Diseases; UCL Institute of Neurology; London, UK
| | | | - Nora Zizlsperger
- Protein Engineering and Antibody Technologies; EMD Serono Research Institute, Inc.; Billerica MA USA
| | - Yu Zhou
- Department of Anesthesia; University of California, San Francisco; San Francisco, CA USA
| | | | - Sven Berger
- Institut de Recherche, Centre d'Immunologie Pierre Fabre; St Julien en Genevois, France
| | - Kirstin A. Zettlitz
- Crump Institute for Molecular Imaging; Department of Molecular and Medical Pharmacology; David Geffen School of Medicine at UCLA; California NanoSystems Institute; University of California Los Angeles; Los Angeles, CA USA
| | | | - May Yong
- UCL Cancer Institute; London, UK
| | | | | |
Collapse
|
36
|
Chang CH, Wang Y, Trisal P, Li R, Rossi DL, Nair A, Gupta P, Losman M, Cardillo TM, Rossi EA, Goldenberg DM. Evaluation of a novel hexavalent humanized anti-IGF-1R antibody and its bivalent parental IgG in diverse cancer cell lines. PLoS One 2012; 7:e44235. [PMID: 22952934 PMCID: PMC3432068 DOI: 10.1371/journal.pone.0044235] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 07/30/2012] [Indexed: 12/11/2022] Open
Abstract
A major mechanism of monoclonal antibodies that selectively target the insulin-like growth factor type 1 receptor (IGF-1R) to inhibit tumor growth is by downregulating the receptor, regardless whether they are capable (antagonistic) or incapable (agonistic) of blocking the binding of cognate ligands. We have developed and characterized a novel agonistic anti-IGF-1R humanized antibody, hR1, and used the Dock-and-Lock (DNL) method to construct Hex-hR1, the first multivalent antibody comprising 6 functional Fabs of hR1, with the aim of enhancing potency of hR1. Based on cross-blocking experiments, hR1 recognizes a region of cysteine-rich domain on the α-subunit, different from the epitopes mapped for existing anti-IGF-1R antibodies, yet hR1 is similar to other anti-IGF-1R antibodies in downregulating IGF-1R and inhibiting proliferation, colony formation, or invasion of selected cancer cell lines in vitro, as well as suppressing growth of the RH-30 rhabdomyosarcoma xenograft in nude mice when combined with the mTOR inhibitor, rapamycin. Hex-hR1 and hR1 are generally comparable in their bioactivities under the in-intro and in-vivo conditions investigated. Nevertheless, in selective experiments involving a direct comparison of potency, Hex-hR1 demonstrated a stronger effect on inhibiting cell proliferation stimulated by IGF-1 and could effectively downregulate IGF-1R at a concentration as low as 20 pM.
Collapse
Affiliation(s)
- Chien-Hsing Chang
- Immunomedics, Inc, Morris Plains, New Jersey, United States of America
- IBC Pharmaceuticals, Inc., Morris Plains, New Jersey, United States of America
| | - Yang Wang
- Immunomedics, Inc, Morris Plains, New Jersey, United States of America
| | - Preeti Trisal
- Immunomedics, Inc, Morris Plains, New Jersey, United States of America
| | - Rongxiu Li
- Immunomedics, Inc, Morris Plains, New Jersey, United States of America
| | - Diane L. Rossi
- Immunomedics, Inc, Morris Plains, New Jersey, United States of America
| | - Anju Nair
- Immunomedics, Inc, Morris Plains, New Jersey, United States of America
| | - Pankaj Gupta
- Immunomedics, Inc, Morris Plains, New Jersey, United States of America
| | - Michele Losman
- Immunomedics, Inc, Morris Plains, New Jersey, United States of America
| | | | - Edmund A. Rossi
- IBC Pharmaceuticals, Inc., Morris Plains, New Jersey, United States of America
| | - David M. Goldenberg
- Immunomedics, Inc, Morris Plains, New Jersey, United States of America
- IBC Pharmaceuticals, Inc., Morris Plains, New Jersey, United States of America
- Center of Molecular Medicine and Immunology, Garden State Cancer Center, Morris Plains, New Jersey, United States of America
| |
Collapse
|
37
|
Chang CH, Hinkula J, Loo M, Falkeborn T, Li R, Cardillo TM, Rossi EA, Goldenberg DM, Wahren B. A novel class of anti-HIV agents with multiple copies of enfuvirtide enhances inhibition of viral replication and cellular transmission in vitro. PLoS One 2012; 7:e41235. [PMID: 22844444 PMCID: PMC3402531 DOI: 10.1371/journal.pone.0041235] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 06/19/2012] [Indexed: 01/08/2023] Open
Abstract
We constructed novel HIV-1 fusion inhibitors that may overcome the current limitations of enfuvirtide, the first such therapeutic in this class. The three prototypes generated by the Dock-and-Lock (DNL) technology to comprise four copies of enfuvirtide tethered site-specifically to the Fc end of different humanized monoclonal antibodies potently neutralize primary isolates (both R5-tropic and X4-tropic), as well as T-cell-adapted strains of HIV-1 in vitro. All three prototypes show EC50 values in the subnanomolar range, which are 10- to 100-fold lower than enfuvirtide and attainable whether or not the constitutive antibody targets HIV-1. The potential of such conjugates to purge latently infected cells was also demonstrated in a cell-to-cell viral inhibition assay by measuring their efficacy to inhibit the spread of HIV-1LAI from infected human peripheral blood mononuclear cells to Jurkat T cells over a period of 30 days following viral activation with 100 nM SAHA (suberoylanilide hydroxamic acid). The IgG-like half-life was not significantly different from that of the parental antibody, as shown by the mean serum concentration of one prototype in mice at 72 h. These encouraging results provide a rationale to develop further novel anti-HIV agents by coupling additional antibodies of interest with alternative HIV-inhibitors via recombinantly-produced, self-assembling, modules.
Collapse
Affiliation(s)
- Chien-Hsing Chang
- Immunomedics, Inc., Morris Plains, New Jersey, United States of America
- IBC Pharmaceuticals, Inc., Morris Plains, New Jersey, United States of America
- * E-mail: (CHC); (BW)
| | - Jorma Hinkula
- Department of Microbiology and Tumor Biology, Karolinska Institutet, Stockholm, Sweden
- Department of Molecular Virology, Linkoping University, Linkoping, Sweden
| | - Meiyu Loo
- IBC Pharmaceuticals, Inc., Morris Plains, New Jersey, United States of America
| | - Tina Falkeborn
- Department of Molecular Virology, Linkoping University, Linkoping, Sweden
| | - Rongxiu Li
- Immunomedics, Inc., Morris Plains, New Jersey, United States of America
| | | | - Edmund A. Rossi
- IBC Pharmaceuticals, Inc., Morris Plains, New Jersey, United States of America
| | - David M. Goldenberg
- Immunomedics, Inc., Morris Plains, New Jersey, United States of America
- IBC Pharmaceuticals, Inc., Morris Plains, New Jersey, United States of America
- Center for Molecular Medicine and Immunology, Garden State Cancer Center, Morris Plains, New Jersey, United States of America
| | - Britta Wahren
- Department of Microbiology and Tumor Biology, Karolinska Institutet, Stockholm, Sweden
- * E-mail: (CHC); (BW)
| |
Collapse
|
38
|
Abstract
INTRODUCTION Over a half a century ago, radiolabeled antibodies were shown to localize selectively in tissues based on the expression of unique antigens. Antibodies have since become the de facto targeting agent, even inspiring the development of non-antibody compounds for targeting purposes. AREAS COVERED In this article, we review various aspects of how antibodies are transforming the way cancer is being detected and treated, with the growing demand for unconjugated and many new antibody conjugates. While unconjugated antibodies continue to garner most of the attention, interest in new antibody drug conjugates and immunotoxins has expanded over the past few years. However, there continues to be active research with new radioimmunoconjugates for imaging and therapy, particularly with α-emitters, as well as antibody-targeted cytokines and other biological response modifiers. EXPERT OPINION The increasing number of new agents being developed and tested clinically suggests that antibody-targeted compounds will have an expanding role in the future.
Collapse
Affiliation(s)
- David M Goldenberg
- Center for Molecular Medicine and Immunology, 300 The American Road, Morris Plains, NJ 07950, USA
| | | |
Collapse
|
39
|
Goldenberg DM, Chang CH, Rossi EA, McBride WJ, Sharkey RM, Sharkey RM. Pretargeted molecular imaging and radioimmunotherapy. Am J Cancer Res 2012; 2:523-40. [PMID: 22737190 PMCID: PMC3364558 DOI: 10.7150/thno.3582] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2011] [Accepted: 10/31/2011] [Indexed: 01/31/2023] Open
Abstract
Pretargeting is a multi-step process that first has an unlabeled bispecific antibody (bsMAb) localize within a tumor by virtue of its anti-tumor binding site(s) before administering a small, fast-clearing radiolabeled compound that then attaches to the other portion of the bsMAb. The compound's rapid clearance significantly reduces radiation exposure outside of the tumor and its small size permits speedy delivery to the tumor, creating excellent tumor/nontumor ratios in less than 1 hour. Haptens that bind to an anti-hapten antibody, biotin that binds to streptavidin, or an oligonucleotide binding to a complementary oligonucleotide sequence have all been radiolabeled for use by pretargeting. This review will focus on a highly flexible anti-hapten bsMAb platform that has been used to target a variety of radionuclides to image (SPECT and PET) as well as treat tumors.
Collapse
|
40
|
Dual-targeting immunotherapy of lymphoma: potent cytotoxicity of anti-CD20/CD74 bispecific antibodies in mantle cell and other lymphomas. Blood 2012; 119:3767-78. [DOI: 10.1182/blood-2011-09-381988] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
We describe the use of novel bispecific hexavalent Abs (HexAbs) to enhance anticancer immunotherapy. Two bispecific HexAbs [IgG-(Fab)4 constructed from veltuzumab (anti-CD20 IgG) and milatuzumab (anti-CD74 IgG)] show enhanced cytotoxicity in mantle cell lymphoma (MCL) and other lymphoma/leukemia cell lines, as well as patient tumor samples, without a crosslinking Ab, compared with their parental mAb counterparts, alone or in combination. The bispecific HexAbs have different properties from and are more potent than their parental mAbs in vitro. The juxtaposition of CD20 and CD74 on MCL cells by the HexAbs resulted in homotypic adhesion and triggered intracellular changes that include loss of mitochondrial transmembrane potential, production of reactive oxygen species, rapid and sustained phosphorylation of ERKs and JNK, down-regulation of pAkt and Bcl-xL, actin reorganization, and lysosomal membrane permeabilization, culminating in cell death. They also displayed different potencies in depleting lymphoma cells and normal B cells from whole blood ex vivo and significantly extended the survival of nude mice bearing MCL xenografts in a dose-dependent manner, thus indicating stability and antitumor activity in vivo. Such bispecific HexAbs may constitute a new class of therapeutic agents for improved cancer immunotherapy, as shown here for MCL and other CD20+/CD74+ malignancies.
Collapse
|
41
|
Unique approach for B lymphoma therapy. Blood 2012; 119:3647-8. [PMID: 22517869 DOI: 10.1182/blood-2012-02-409052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
42
|
Lu HD, Charati MB, Kim IL, Burdick JA. Injectable shear-thinning hydrogels engineered with a self-assembling Dock-and-Lock mechanism. Biomaterials 2012; 33:2145-53. [DOI: 10.1016/j.biomaterials.2011.11.076] [Citation(s) in RCA: 154] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 11/28/2011] [Indexed: 01/06/2023]
|
43
|
Ferrari E, Soloviev M, Niranjan D, Arsenault J, Gu C, Vallis Y, O'Brien J, Davletov B. Assembly of protein building blocks using a short synthetic peptide. Bioconjug Chem 2012; 23:479-84. [PMID: 22299630 PMCID: PMC3309608 DOI: 10.1021/bc2005208] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
![]()
Combining proteins or their defined domains offers new
enhanced
functions. Conventionally, two proteins are either fused into a single
polypeptide chain by recombinant means or chemically cross-linked.
However, these strategies can have drawbacks such as poor expression
(recombinant fusions) or aggregation and inactivation (chemical cross-linking),
especially in the case of large multifunctional proteins. We developed
a new linking method which allows site-oriented, noncovalent, yet
irreversible stapling of modified proteins at neutral pH and ambient
temperature. This method is based on two distinct polypeptide linkers
which self-assemble in the presence of a specific peptide staple allowing
on-demand and irreversible combination of protein domains. Here we
show that linkers can either be expressed or be chemically conjugated
to proteins of interest, depending on the source of the proteins.
We also show that the peptide staple can be shortened to 24 amino
acids still permitting an irreversible combination of functional proteins.
The versatility of this modular technique is demonstrated by stapling
a variety of proteins either in solution or to surfaces.
Collapse
Affiliation(s)
- Enrico Ferrari
- MRC Laboratory of Molecular Biology, Hills Road, CB2 0QH Cambridge, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Salcedo L, Sopko N, Jiang HH, Damaser M, Penn M, Zutshi M. Chemokine upregulation in response to anal sphincter and pudendal nerve injury: potential signals for stem cell homing. Int J Colorectal Dis 2011; 26:1577-81. [PMID: 21706136 DOI: 10.1007/s00384-011-1269-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/10/2011] [Indexed: 02/04/2023]
Abstract
PURPOSE Stromal derived factor-1 (SDF-1) and monocyte chemotactic protein-3 (MCP-3) are signals forcing the migration of bone marrow-derived stem cells to ischemic tissue. This study investigates SDF-1 and MCP-3 expression following direct injury to the anal sphincter and pudendal nerve and to determine if these same mechanisms have any role. METHODS Chemokine expression was studied after anal sphincter injury in female rats after either a sphincterotomy (n = 15), pudendal nerve crush (PNC; n = 15), sham pudendal nerve crush (n = 15), or acted as unmanipulated controls (n = 5). Analysis was done at 1 h and 10 and 21 days after injury. RESULTS After injury, SDF-1 expression increased 40.2 ± 6.42 (P = 0.01) at 1 h and 28.2 ± 2.37 (P = 0.01) at 10 days, respectively, compared to controls. Likewise, MCP-3 expression increased 40.8 ± 8.17 (P = 0.02) at the same intervals compared to controls. After PNC, SDF-1 expression increased 46.4 ± 6.01 (P = 0.02) and 50.6 ± 10.11 (P = 0.01), and MCP-3 expression increased 46.3 ± 7.76 (P = 0.03) and 190.8 ± 22.15 (P = 0.01), respectively, at the same time intervals compared to controls. However, when PNC was compared to sham injured, a significant increase was seen in SDF-1 and MCP-3 at 10 days. At 21 days, PNC compared to sham injured was significantly low in expression for both SDF-1 and MCP-3 (P < 0.05). CONCLUSIONS Direct anal sphincter injury results in higher levels of SDF-1 and MCP-3 expression soon after injury, whereas denervation via pudendal nerve crush results in greater SDF-1 and MCP-3 expression 10 days after injury. Chemokine overexpression suggests the potential for cell-based therapeutic strategies.
Collapse
Affiliation(s)
- Levilester Salcedo
- Department of Colorectal Surgery, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | | | | | | | | | | |
Collapse
|
45
|
Preclinical studies on targeted delivery of multiple IFNα2b to HLA-DR in diverse hematologic cancers. Blood 2011; 118:1877-84. [PMID: 21680794 DOI: 10.1182/blood-2011-03-343145] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The short circulating half-life and side effects of IFNα affect its dosing schedule and efficacy. Fusion of IFNα to a tumor-targeting mAb (mAb-IFNα) can enhance potency because of increased tumor localization and improved pharmacokinetics. We used the Dock-and-Lock method to generate C2-2b-2b, a mAb-IFNα comprising tetrameric IFNα2b site-specifically linked to hL243 (humanized anti-HLA-DR). In vitro, C2-2b-2b inhibited various B-cell lymphoma leukemia and myeloma cell lines. In most cases, this immunocytokine was more effective than CD20-targeted mAb-IFNα or a mixture comprising the parental mAb and IFNα. Our findings indicate that responsiveness depends on HLA-DR expression/density and sensitivity to IFNα and hL243. C2-2b-2b induced more potent and longer-lasting IFNα signaling compared with nontargeted IFNα. Phosphorylation of STAT1 was more robust and persistent than that of STAT3, which may promote apoptosis. C2-2b-2b efficiently depleted lymphoma and myeloma cells from whole human blood but also exhibited some toxicity to B cells, monocytes, and dendritic cells. C2-2b-2b showed superior efficacy compared with nontargeting mAb-IFNα, peginterferonalfa-2a, or a combination of hL243 and IFNα, using human lymphoma and myeloma xenografts. These results suggest that C2-2b-2b should be useful in the treatment of various hematopoietic malignancies.
Collapse
|
46
|
Sharkey RM, Rossi EA, Chang CH, Goldenberg DM. Improved cancer therapy and molecular imaging with multivalent, multispecific antibodies. Cancer Biother Radiopharm 2010; 25:1-12. [PMID: 20187791 DOI: 10.1089/cbr.2009.0690] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Antibodies are highly versatile proteins with the ability to be used to target diverse compounds, such as radionuclides for imaging and therapy, or drugs and toxins for therapy, but also can be used unconjugated to elicit therapeutically beneficial responses, usually with minimal toxicity. This update describes a new procedure for forming multivalent and/or multispecific proteins, known as the dock-and-lock (DNL) technique. Developed as a procedure for preparing bispecific antibodies capable of binding divalently to a tumor antigen and monovalently to a radiolabeled hapten-peptide for pretargeted imaging and therapy, this methodology has the flexibility to create a number of other biologic agents of therapeutic interest. A variety of constructs, based on anti-CD20 and CD22 antibodies, have been made, with results showing that multispecific antibodies have very different properties from the respective parental monospecific antibodies. The technique is not restricted to antibody combination, but other biologics, such as interferon-alpha2b, have been prepared. These types of constructs not only allow small biologics to be sustained in the blood longer, but also to be selectively targeted. Thus, DNL technology is a highly flexible platform that can be used to prepare many different types of agents that could further improve cancer detection and therapy.
Collapse
Affiliation(s)
- Robert M Sharkey
- Center for Molecular Medicine and Immunology (CMMI), Belleville, New Jersey, USA
| | | | | | | |
Collapse
|
47
|
Rossi EA, Rossi DL, Stein R, Goldenberg DM, Chang CH. A bispecific antibody-IFNalpha2b immunocytokine targeting CD20 and HLA-DR is highly toxic to human lymphoma and multiple myeloma cells. Cancer Res 2010; 70:7600-9. [PMID: 20876805 DOI: 10.1158/0008-5472.can-10-2126] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The short circulating half-life and side effects of IFNα affect its dosing schedule and efficacy. Fusion of IFNα to a tumor-targeting monoclonal antibody (MAb-IFNα) can enhance potency due to increased tumor localization and improved pharmacokinetics. We report the generation and characterization of the first bispecific MAb-IFNα, designated 20-C2-2b, which comprises two copies of IFNα2b and a stabilized F(ab)(2) of hL243 (humanized anti-HLA-DR; IMMU-114) site-specifically linked to veltuzumab (humanized anti-CD20). In vitro, 20-C2-2b inhibited each of four lymphoma and eight myeloma cell lines, and was more effective than monospecific CD20-targeted MAb-IFNα or a mixture comprising the parental antibodies and IFNα in all but one (HLA-DR(-)/CD20(-)) myeloma line, suggesting that 20-C2-2b should be useful in the treatment of various hematopoietic malignancies. 20-C2-2b displayed greater cytotoxicity against KMS12-BM (CD20(+)/HLA-DR(+) myeloma) compared with monospecific MAb-IFNα, which targets only HLA-DR or CD20, indicating that all three components in 20-C2-2b could contribute to toxicity. Our findings indicate that a given cell's responsiveness to MAb-IFNα depends on its sensitivity to IFNα and the specific antibodies, as well as the expression and density of the targeted antigens.
Collapse
Affiliation(s)
- Edmund A Rossi
- IBC Pharmaceuticals, Inc., Morris Plains, New Jersey, USA
| | | | | | | | | |
Collapse
|
48
|
Walter RB, Press OW, Pagel JM. Pretargeted radioimmunotherapy for hematologic and other malignancies. Cancer Biother Radiopharm 2010; 25:125-42. [PMID: 20423225 DOI: 10.1089/cbr.2010.0759] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Radioimmunotherapy (RIT) has emerged as one of the most promising treatment options, particularly for hematologic malignancies. However, this approach has generally been limited by a suboptimal therapeutic index (target-to-nontarget ratio) and an inability to deliver sufficient radiation doses to tumors selectively. Pretargeted RIT (PRIT) circumvents these limitations by separating the targeting vehicle from the subsequently administered therapeutic radioisotope, which binds to the tumor-localized antibody or is quickly excreted if unbound. A growing number of preclinical proof-of-principle studies demonstrate that PRIT is feasible and safe and provides improved directed radionuclide delivery to malignant cells compared with conventional RIT while sparing normal cells from nonspecific radiotoxicity. Early phase clinical studies corroborate these preclinical findings and suggest better efficacy and lesser toxicities in patients with hematologic and other malignancies. With continued research, PRIT-based treatment strategies promise to become cornerstones to improved outcomes for cancer patients despite their complexities.
Collapse
Affiliation(s)
- Roland B Walter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109-1024, USA.
| | | | | |
Collapse
|
49
|
Multiple signaling pathways induced by hexavalent, monospecific, anti-CD20 and hexavalent, bispecific, anti-CD20/CD22 humanized antibodies correlate with enhanced toxicity to B-cell lymphomas and leukemias. Blood 2010; 116:3258-67. [PMID: 20628151 DOI: 10.1182/blood-2010-03-276857] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We have generated hexavalent antibodies (HexAbs) comprising 6 Fabs tethered to one Fc of human IgG1. Three such constructs, 20-20, a monospecific HexAb comprising 6 Fabs of veltuzumab (humanized anti-CD20 immunoglobulin G1κ [IgG1κ]), 20-22, a bispecific HexAb comprising veltuzumab and 4 Fabs of epratuzumab (humanized anti-CD22 IgG1κ), and 22-20, a bispecific HexAb comprising epratuzumab and 4 Fabs of veltuzumab, were previously shown to inhibit pro-liferation of several lymphoma cell lines at nanomolar concentrations in the absence of a crosslinking antibody. We now report an in-depth analysis of the apoptotic and survival signals induced by the 3 HexAbs in Burkitt lymphomas and provide in vitro cytotoxicity data for additional lymphoma cell lines and also chronic lymphocytic leukemia patient specimens. Among the key findings are the significant increase in the levels of phosphorylated p38 and phosphatase and tensin homolog deleted on chromosome 10 (PTEN) by all 3 HexAbs and the notable differences in the signaling events triggered by the HexAbs from those incurred by crosslinking veltuzumab or rituximab with a secondary antibody. Thus, the greatly enhanced direct toxicity of these HexAbs correlates with their ability to alter the basal expression of various intracellular proteins involved in regulating cell growth, survival, and apoptosis, with the net outcome leading to cell death.
Collapse
|
50
|
Sharkey RM, Rossi EA, McBride WJ, Chang CH, Goldenberg DM. Recombinant bispecific monoclonal antibodies prepared by the dock-and-lock strategy for pretargeted radioimmunotherapy. Semin Nucl Med 2010; 40:190-203. [PMID: 20350628 DOI: 10.1053/j.semnuclmed.2009.12.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The selective delivery of therapeutic radionuclides is a promising approach for treating cancer. Antibody-targeted radionuclides are of particular interest, with 2 products approved for the treatment of certain forms of non-Hodgkin lymphoma. However, for many other cancers, radioimmunotherapy has been ineffective, being limited by prolonged exposure to the highly radiosensitive bone marrow. An alternative approach, known as pretargeting, separates radionuclide from the antibody, allowing the radiation to be delivered on a small molecule that can quickly and efficiently migrate into the tumor, and then rapidly clear from the body with minimal retention in tissues. Several pretargeting methods have been developed that differ in the way they selectively capture the radionuclide. This review focuses on the development of a novel form of bispecific monoclonal antibody (bsMAb) pretargeting that uses a unique radiolabeled hapten-peptide system that can be modified to bind numerous therapeutic and imaging radionuclides. Together with a specialized recombinant humanized bsMAb prepared with by a technique known as the Dock-and-Lock method, this pretargeting procedure has been examined in many different animal models, showing a high level of sensitivity and specificity for localizing tumors, and improved efficacy with less hematologic toxicity associated with directly radiolabeled IgG. The bsMAb is a tri-Fab structure, having 2 binding arms for the tumor antigen and 1 capable of binding a hapten-peptide. Preclinical studies were preformed to support the clinical use of a bsMAb and a hapten-peptide bearing a single DOTA moiety (IMP-288). A phase 0 trial found an (131)I-tri-Fab bsMAb, TF2, that targets carcinoembryonic antigen was stable in vivo, quickly clears from the blood, and localizes known tumors. The first-in-patient pretargeting experience with the (111)In-IMP-288 also observed rapid clearance and low tissue (kidney) retention, as well as localization of tumors, providing initial promising evidence for developing these materials for radioimmunotherapy.
Collapse
Affiliation(s)
- Robert M Sharkey
- Garden State Cancer Center, Center for Molecular Medicine and Immunology, Belleville, NJ, USA.
| | | | | | | | | |
Collapse
|