1
|
Williams L, Li L, Yazaki PJ, Wong P, Miller A, Hong T, Poku EK, Bhattacharya S, Shively JE, Kujawski M. Generation of IL-2-Fc-antibody conjugates by click chemistry. Biotechnol J 2023; 18:e2300115. [PMID: 37300381 DOI: 10.1002/biot.202300115] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/10/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND Immunocytokines (ICKs) are antibody directed cytokines produced by genetic fusion of an antibody to a cytokine. METHODS We now show that antibodies conjugated by click chemistry to interleukin-2 (IL-2)-Fc form fully active conjugates, and in one example, equivalent activity to a genetically produced ICK. RESULTS An IL-2-Fc fusion protein was optimized for click chemistry at hinge cysteines using protein stabilizing IL-2 mutations at Lys35 and Cys125 and Fc hinge mutations at Cys142 and Cys148. The IL-2-Fc fusion protein with K35E and C125S mutations with 3 intact hinge cysteines, designated as IL-2-Fc Par, was selected based on its minimal tendency to aggregate. IL-2-Fc-antibody clicked conjugates retained high IL-2 activity and bound target antigens comparable to parent antibodies. An IL-2-Fc-anti-CEA click conjugate showed comparable anti-tumor activity to an anti-CEA-IL-2 ICK in immunocompetent CEA transgenic mice bearing CEA positive orthotopic breast tumors. Significant increases in IFNγ+ /CD8+ and decreases in FoxP3+ /CD4+ T-cells were found for the clicked conjugate and ICK therapies, suggesting a common mechanism of tumor reduction. CONCLUSION The production of antibody targeted IL-2 therapy via a click chemistry approach is feasible with comparable activity to genetically produced ICKs with the added advantage of multiplexing with other monoclonal antibodies.
Collapse
Affiliation(s)
- Lindsay Williams
- Department of Immunology and Theranostics, Riggs Diabetes, Metabolism, and Research Institute, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Lin Li
- Department of Immunology and Theranostics, Riggs Diabetes, Metabolism, and Research Institute, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Paul J Yazaki
- Department of Immunology and Theranostics, Riggs Diabetes, Metabolism, and Research Institute, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Patty Wong
- Department of Immunology and Theranostics, Riggs Diabetes, Metabolism, and Research Institute, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Aaron Miller
- Department of Immunology and Theranostics, Riggs Diabetes, Metabolism, and Research Institute, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Teresa Hong
- Department of Immunology and Theranostics, Riggs Diabetes, Metabolism, and Research Institute, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Erasmus K Poku
- Radiopharmacy, City of Hope, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Supriyo Bhattacharya
- Department of Computational and Quantitative Medicine, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - John E Shively
- Department of Immunology and Theranostics, Riggs Diabetes, Metabolism, and Research Institute, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Maciej Kujawski
- Department of Immunology and Theranostics, Riggs Diabetes, Metabolism, and Research Institute, Beckman Research Institute of City of Hope, Duarte, California, USA
| |
Collapse
|
2
|
Faber ML, Oldham RAA, Thakur A, Rademacher MJ, Kubicka E, Dlugi TA, Gifford SA, McKillop WM, Schloemer NJ, Lum LG, Medin JA. Novel anti-CD30/CD3 bispecific antibodies activate human T cells and mediate potent anti-tumor activity. Front Immunol 2023; 14:1225610. [PMID: 37646042 PMCID: PMC10461807 DOI: 10.3389/fimmu.2023.1225610] [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: 05/19/2023] [Accepted: 07/12/2023] [Indexed: 09/01/2023] Open
Abstract
CD30 is expressed on Hodgkin lymphomas (HL), many non-Hodgkin lymphomas (NHLs), and non-lymphoid malignancies in children and adults. Tumor expression, combined with restricted expression in healthy tissues, identifies CD30 as a promising immunotherapy target. An anti-CD30 antibody-drug conjugate (ADC) has been approved by the FDA for HL. While anti-CD30 ADCs and chimeric antigen receptors (CARs) have shown promise, their shortcomings and toxicities suggest that alternative treatments are needed. We developed novel anti-CD30 x anti-CD3 bispecific antibodies (biAbs) to coat activated patient T cells (ATCs) ex vivo prior to autologous re-infusions. Our goal is to harness the dual specificity of the biAb, the power of cellular therapy, and the safety of non-genetically modified autologous T cell infusions. We present a comprehensive characterization of the CD30 binding and tumor cell killing properties of these biAbs. Five unique murine monoclonal antibodies (mAbs) were generated against the extracellular domain of human CD30. Resultant anti-CD30 mAbs were purified and screened for binding specificity, affinity, and epitope recognition. Two lead mAb candidates with unique sequences and CD30 binding clusters that differ from the ADC in clinical use were identified. These mAbs were chemically conjugated with OKT3 (an anti-CD3 mAb). ATCs were armed and evaluated in vitro for binding, cytokine production, and cytotoxicity against tumor lines and then in vivo for tumor cell killing. Our lead mAb was subcloned to make a Master Cell Bank (MCB) and screened for binding against a library of human cell surface proteins. Only huCD30 was bound. These studies support a clinical trial in development employing ex vivo-loading of autologous T cells with this novel biAb.
Collapse
Affiliation(s)
- Mary L. Faber
- Department of Pediatrics, Medical College of Wisconsin (MCW), Milwaukee, WI, United States
| | - Robyn A. A. Oldham
- Department of Pediatrics, Medical College of Wisconsin (MCW), Milwaukee, WI, United States
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Archana Thakur
- Department of Medicine, Division of Hematology/Oncology, University of Virginia Cancer Center, Charlottesville, VA, United States
| | - Mary Jo Rademacher
- Department of Pediatrics, Medical College of Wisconsin (MCW), Milwaukee, WI, United States
| | - Ewa Kubicka
- Department of Medicine, Division of Hematology/Oncology, University of Virginia Cancer Center, Charlottesville, VA, United States
| | - Theresa A. Dlugi
- Department of Pediatrics, Medical College of Wisconsin (MCW), Milwaukee, WI, United States
| | - Steven A. Gifford
- Department of Pediatrics, Medical College of Wisconsin (MCW), Milwaukee, WI, United States
| | - William M. McKillop
- Department of Pediatrics, Medical College of Wisconsin (MCW), Milwaukee, WI, United States
| | - Nathan J. Schloemer
- Department of Pediatrics, Medical College of Wisconsin (MCW), Milwaukee, WI, United States
| | - Lawrence G. Lum
- Department of Medicine, Division of Hematology/Oncology, University of Virginia Cancer Center, Charlottesville, VA, United States
| | - Jeffrey A. Medin
- Department of Pediatrics, Medical College of Wisconsin (MCW), Milwaukee, WI, United States
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Department of Biochemistry, MCW, Milwaukee, WI, United States
| |
Collapse
|
3
|
Vaishampayan UN, Thakur A, Chen W, Deol A, Patel M, Dobson K, Dickow B, Schalk D, Schienshang A, Whitaker S, Polend A, Fontana JA, Heath EI, Lum. LG. Phase II Trial of Pembrolizumab and Anti-CD3 x Anti-HER2 Bispecific Antibody-Armed Activated T Cells in Metastatic Castration-Resistant Prostate Cancer. Clin Cancer Res 2023; 29:122-133. [PMID: 36255393 PMCID: PMC9812860 DOI: 10.1158/1078-0432.ccr-22-1601] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/03/2022] [Accepted: 10/14/2022] [Indexed: 02/06/2023]
Abstract
PURPOSE A phase II study was conducted to evaluate the safety and efficacy of the combination of HER2 bispecific antibody (HER2Bi)-armed activated T cells (HER2 BAT) and programmed death 1 inhibitor, pembrolizumab. PATIENTS AND METHODS Patients with metastatic castration-resistant prostate cancer (mCRPC) with 0 to 1 performance status and normal liver, kidney, and marrow function, pre- or post-docetaxel chemotherapy were eligible. Primary endpoint was 6-month progression-free survival (PFS). Peripheral blood mononuclear cells were obtained by a single apheresis, shipped to University of Virginia, activated with OKT3 and expanded for 14 days in IL2, harvested, and armed with HER2Bi and cryopreserved. HER2 BATs were infused twice weekly for 4 weeks and pembrolizumab was administered every 21 days for a maximum duration of 6 months starting 1 to 3 weeks prior to HER2 BATs infusion. RESULTS Fourteen patients were enrolled with a median age of 69 (range 57-82 years) and median PSA of 143.4 (range 8.2-4210 ng/dL). Two patients had peritoneal metastases, 1 had lymph node (LN) only metastases and 11 had bone metastases of which 7 had bone and LN metastases. All were pretreated with androgen receptor axis targeted agents and 7 (50%) had prior docetaxel chemotherapy. The toxicities were grade1-2 infusion reactions with fever, chills, headaches, nausea and/or myalgias. Primary endpoint of 6 month PFS was achieved in 5 of 14 patients (38.5%; 95% confidence interval, 19.5%-76.5%). Median PFS was 5 months and median survival was 31.6 months. CONCLUSIONS The safety and promising efficacy makes this combination worthy of future investigation in mCRPC.
Collapse
Affiliation(s)
- Ulka N. Vaishampayan
- Karmanos Cancer Center/Wayne State University, Detroit, MI/University of Michigan
| | - Archana Thakur
- Emily Couric Cancer Center, University of Virginia, Charlottesville VA
| | - Wei Chen
- Karmanos Cancer Center/Wayne State University, Detroit, MI
| | - Abhinav Deol
- Karmanos Cancer Center/Wayne State University, Detroit, MI
| | - Meera Patel
- Karmanos Cancer Center/Wayne State University, Detroit, MI
| | | | - Brenda Dickow
- Karmanos Cancer Center/Wayne State University, Detroit, MI
| | - Dana Schalk
- Emily Couric Cancer Center, University of Virginia, Charlottesville VA
| | - Amy Schienshang
- Emily Couric Cancer Center, University of Virginia, Charlottesville VA
| | - Sarah Whitaker
- Emily Couric Cancer Center, University of Virginia, Charlottesville VA
| | - Amanda Polend
- Emily Couric Cancer Center, University of Virginia, Charlottesville VA
| | | | | | - Lawrence G. Lum.
- Emily Couric Cancer Center, University of Virginia, Charlottesville VA
| |
Collapse
|
4
|
Kujawski M, Li L, Li H, Yazaki PJ, Swiderski P, Shively JE. T-cell surface generation of dual bivalent, bispecific T-cell engaging, RNA duplex cross-linked antibodies (dbBiTERs) for re-directed tumor cell lysis. Biotechnol J 2022; 17:e2100389. [PMID: 34773368 PMCID: PMC9177045 DOI: 10.1002/biot.202100389] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 11/06/2021] [Accepted: 11/09/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND Genetic engineered Bispecific T-cell engagers (BiTEs) generate potent cytotoxic effects. METHODS Alternately, click chemistry engineered, dual specific bivalent Bispecific T-cell engaging antibodies (dbBiTEs) on T-cell surfaces can be generated from parent monoclonal antibodies. RESULTS We show the formation of dbBiTEs on the surface of T-cells along with the introduction of complementary 2'-OMe RNA 32-mer oligonucleotides allowing duplex formation between antibodies, designated as dbBiTERs. dbBiTERs generated in solution from anti-CEA and anti-CD3 OKT3 antibodies retained specific binding to CEA positive versus CEA negative cancer cells and to CD3 positive T-cells comparable to dbBiTEs. When T-cells were precoated with dbBiTEs or dbBiTERs and mixed with CEA positive versus CEA negative cancer cells, similar dose dependent and specific cytotoxicity were observed in redirected cell lysis assays. On-cell generated dbBiTERs exerted potent cytotoxic responses against CEA positive targets and were localized at the cell surface by immuno-gold EM. In addition, we demonstrate that target and T-cells, each coated separately with complementary 2'OMe-RNA-linked antibodies can be cross-linked by RNA duplex formation in vitro to generate redirected cell lysis. CONCLUSION The facile generation of dbBiTERs with specific cytolytic activity from intact antibodies and their generation on-cell offers a new avenue for antigen specific T-cell therapy.
Collapse
Affiliation(s)
- Maciej Kujawski
- Department of Immunology and Theranostics, City of Hope, Duarte, California, USA
| | - Lin Li
- Department of Immunology and Theranostics, City of Hope, Duarte, California, USA
| | - Harry Li
- Department of Immunology and Theranostics, City of Hope, Duarte, California, USA
| | - Paul J. Yazaki
- Department of Immunology and Theranostics, City of Hope, Duarte, California, USA
| | - Piotr Swiderski
- Shared Resources-DNA/RNA/Peptide, City of Hope, Duarte, California, USA
| | - John E. Shively
- Department of Immunology and Theranostics, City of Hope, Duarte, California, USA
| |
Collapse
|
5
|
Kujawski M, Li L, Bhattacharya S, Wong P, Lee WH, Williams L, Li H, Chea J, Poku K, Bowles N, Vaidehi N, Yazaki P, Shively JE. Generation of dual specific bivalent BiTEs (dbBIspecific T-cell engaging antibodies) for cellular immunotherapy. BMC Cancer 2019; 19:882. [PMID: 31488104 PMCID: PMC6727398 DOI: 10.1186/s12885-019-6056-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 08/19/2019] [Indexed: 01/12/2023] Open
Abstract
Background Bispecific T-cell engaging antibodies (BiTES), comprising dual anti-CD3 and anti-tumor antigen scFv fragments, are important therapeutic agents for the treatment of cancer. The dual scFv construct for BiTES requires proper protein folding while their small molecular size leads to rapid kidney clearance. Methods An intact (150 kDa) anti-tumor antigen antibody to CEA was joined in high yield (ca. 30%) to intact (150 kDa) anti-murine and anti-human CD3 antibodies using hinge region specific Click chemistry to form dual-specific, bivalent BiTES (dbBiTES, 300 kDa). dbBiTEs were tested in vitro by EM, flow cytometry and cell cytoxicity and in vivo by PET tumor imaging and redirected T-cell therapy. Results The interlocked hinge regions are compatible with a structural model that fits the electron micrographs of 300 kDa particles. Compared to intact anti-CEA antibody, dbBiTES exhibit high in vitro cytotoxicity, high in vivo tumor targeting as demonstrated by PET imaging, and redirected dbBiTE coated T-cells (1 microgram/10 million cells) that kill CEA+ target cells in vivo in CEA transgenic mice. Conclusion dbBiTE redirected T-cell therapy is a promising, efficient approach for targeting and killing cancer cells. Electronic supplementary material The online version of this article (10.1186/s12885-019-6056-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Maciej Kujawski
- Department of Molecular Imaging and Therapy, Beckman Research Institute City of Hope, Duarte, CA, 91010, USA
| | - Lin Li
- Department of Molecular Imaging and Therapy, Beckman Research Institute City of Hope, Duarte, CA, 91010, USA
| | - Supriyo Bhattacharya
- Department of Computational and Quantitative Medicine, Beckman Research Institute City of Hope, Duarte, CA, 91010, USA
| | - Patty Wong
- Department of Molecular Imaging and Therapy, Beckman Research Institute City of Hope, Duarte, CA, 91010, USA
| | - Wen-Hui Lee
- Department of Molecular Imaging and Therapy, Beckman Research Institute City of Hope, Duarte, CA, 91010, USA
| | - Lindsay Williams
- Department of Molecular Imaging and Therapy, Beckman Research Institute City of Hope, Duarte, CA, 91010, USA
| | - Harry Li
- Department of Molecular Imaging and Therapy, Beckman Research Institute City of Hope, Duarte, CA, 91010, USA
| | - Junie Chea
- Radiopharmacy, Beckman Research Institute City of Hope, Duarte, CA, 91010, USA
| | - Kofi Poku
- Radiopharmacy, Beckman Research Institute City of Hope, Duarte, CA, 91010, USA
| | - Nicole Bowles
- Radiopharmacy, Beckman Research Institute City of Hope, Duarte, CA, 91010, USA
| | - Nagarajan Vaidehi
- Department of Computational and Quantitative Medicine, Beckman Research Institute City of Hope, Duarte, CA, 91010, USA
| | - Paul Yazaki
- Department of Molecular Imaging and Therapy, Beckman Research Institute City of Hope, Duarte, CA, 91010, USA
| | - John E Shively
- Department of Molecular Imaging and Therapy, Beckman Research Institute City of Hope, Duarte, CA, 91010, USA.
| |
Collapse
|
6
|
Abstract
Cancer stem cells (CSCs) are crucial for tumor recurrence and distant metastasis. Immunologically targeting CSCs represents a promising strategy to improve efficacy of multimodal cancer therapy. Modulating the innate immune response involving Toll-like receptors, macrophages, natural killer cells, and γδT cells has therapeutic effects on CSCs. Antigens expressed by CSCs provide specific targets for immunotherapy. CSC-primed dendritic cell-based vaccines have induced significant antitumor immunity as an adjuvant therapy in experimental models of established tumors. Targeting the tumor microenvironment CSC niche with cytokines or checkpoint blockade provides additional strategies to eliminate CSCs.
Collapse
Affiliation(s)
- Jing Zhang
- Division of Surgical Oncology, University of Michigan Rogel Cancer Center, Room 3410, 1150 East Medical Center Drive, Ann Arbor, MI 48109, USA; Department of the 2nd Thoracic Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 116 Zhuodaoquan South Road, Hongshan District, Wuhan, Hubei Province 430070, China
| | - Qiao Li
- Division of Surgical Oncology, University of Michigan Rogel Cancer Center, 3520B MSRB-1, 1150 West Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Alfred E Chang
- Division of Surgical Oncology, University of Michigan Rogel Cancer Center, Room 3304, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA.
| |
Collapse
|
7
|
Yu S, Liu Q, Han X, Qin S, Zhao W, Li A, Wu K. Development and clinical application of anti-HER2 monoclonal and bispecific antibodies for cancer treatment. Exp Hematol Oncol 2017; 6:31. [PMID: 29209558 PMCID: PMC5704598 DOI: 10.1186/s40164-017-0091-4] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 11/20/2017] [Indexed: 12/15/2022] Open
Abstract
HER2-targeted immunotherapy consists of monoclonal antibodies (e.g. trastuzumab, pertuzumab), bispecific antibodies (e.g. MM-111, ertumaxomab) and activated T cells armed with anti-HER2 bispecific antibody (HER2Bi-aATC). Trastuzumab is a classic drug for the treatment of HER2 positive metastatic breast cancer. The combined application of pertuzumab, trastuzumab and paclitaxel has been suggested as a standard therapy for HER2 positive advanced breast cancer. The resistance to anti-HER2 antibody has resulted in disease progression. HER2-directed bispecific antibody may be a promising therapeutic approach for these patients. Ertumaxomab enhanced the interaction of immune effector cells and tumor cells. MM-111 simultaneously binds to HER2 and HER3 and blocks downstream signaling. Besides, HER2Bi-aATC is also an alternative therapeutic approach for HER2 positive cancers. In this review, we summarized the recent advancement of HER2-targeted monoclonal antibodies (trastuzumab, pertuzumab and T-DM1) and bispecific antibodies (MM-111, ertumaxomab and HER2Bi-aATC), especially focus on clinical trial results.
Collapse
Affiliation(s)
- Shengnan Yu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 China
| | - Qian Liu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 China
| | - Xinwei Han
- Department of Interventional Radiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
| | - Shuang Qin
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 China
| | - Weiheng Zhao
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 China
| | - Anping Li
- Department of Interventional Radiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030 China
| |
Collapse
|
8
|
Abstract
PURPOSE OF REVIEW Adoptive T-cell therapy has become one of the most exciting fields of cancer therapy in the past few years. In this article, we describe a method which combines adoptive T-cell therapy with antibody therapy by arming T cells from cord blood, normal patients, and cancer patients with bispecific antibodies capable of binding to tumor-associated antigens on one side of the bispecific antibody construct and T cells on another side of the construct. This approach redirects T cells against tumor cells in a non-MHC-restricted manner. RECENT FINDINGS Various methods for manipulating the immune system including check-point inhibitors, chimeric antigen receptor T cells, and bispecific antibodies have shown promising activity in treating both hematological malignancies and solid tumors with excellent success. In recent studies, activated T cells armed with bispecific antibodies have shown good preclinical activity, safety, and promising efficacy in the clinical trials. SUMMARY Activated T cells armed with bispecific antibodies represent a promising treatment for cancer immunotherapy.
Collapse
|
9
|
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
|
10
|
Thakur A, Lum LG. In Situ immunization by bispecific antibody targeted T cell therapy in breast cancer. Oncoimmunology 2016; 5:e1055061. [PMID: 27141330 PMCID: PMC4839366 DOI: 10.1080/2162402x.2015.1055061] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 05/21/2015] [Indexed: 11/16/2022] Open
Abstract
Metastatic breast cancer remains an incurable disease. Most patients experience objective treatment responses associated with palliation of symptoms; however, progression inevitably occurs. Thus, there is an urgent need for innovative therapeutic strategies that may halt disease progression. Adoptive T-cell therapy is a potent and promising treatment option with the potential to harness the intrinsic antitumor power of the immune system and provide long-term immunity against cancer.
Collapse
Affiliation(s)
- Archana Thakur
- Department of Oncology; Wayne State University, and Karmanos Cancer Institute; Detroit, MI, USA
| | - Lawrence G. Lum
- Department of Oncology; Wayne State University, and Karmanos Cancer Institute; Detroit, MI, USA
- Department of Medicine; Wayne State University, and Karmanos Cancer Institute; Detroit, MI, USA
- Department of Pediatrics; Wayne State University, and Karmanos Cancer Institute; Detroit, MI, USA
- Department of Immunology and Microbiology; Wayne State University and Karmanos Cancer Institute; Detroit, MI, USA
| |
Collapse
|
11
|
Tang J, Shen D, Zhang J, Ligler FS, Cheng K. Bispecific antibodies, nanoparticles and cells: bringing the right cells to get the job done. Expert Opin Biol Ther 2015; 15:1251-5. [PMID: 26004388 DOI: 10.1517/14712598.2015.1049944] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Pre-arming therapeutic cells with bispecific antibodies (BiAbs) before infusion can home the cells to specific tissue antigens in the body. With the development of nanotechnology, we developed a novel strategy, namely magnetic bispecific cell engager (MagBICE), that combines BiAbs with biodegradable iron nanoparticles. Compared to conventional BiAbs, the latter enables magnetic targeting and imaging. This editorial discusses current knowledge of BiAbs and their applications in targeting activated T cells to cancerous tissues or targeting bone marrow-derived stem cells to myocardial infarction. We will also discuss the fabrication of MagBICE and its application in treating rodents with myocardial infarction.
Collapse
Affiliation(s)
- Junnan Tang
- North Carolina State University, College of Veterinary Medicine, Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research , Raleigh, NC , USA
| | | | | | | | | |
Collapse
|
12
|
Lum LG, Thakur A, Al-Kadhimi Z, Colvin GA, Cummings FJ, Legare RD, Dizon DS, Kouttab N, Maizel A, Colaiace W, Liu Q, Rathore R. Targeted T-cell Therapy in Stage IV Breast Cancer: A Phase I Clinical Trial. Clin Cancer Res 2015; 21:2305-14. [PMID: 25688159 PMCID: PMC4433762 DOI: 10.1158/1078-0432.ccr-14-2280] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 01/19/2015] [Indexed: 01/17/2023]
Abstract
PURPOSE This study reports a phase I immunotherapy trial in 23 women with metastatic breast cancer consisting of eight infusions of anti-CD3 × anti-HER2 bispecific antibody (HER2Bi) armed anti-CD3-activated T cells (ATC) in combination with low-dose IL-2 and granulocyte-macrophage colony-stimulating factor to determine safety, maximum tolerated dose (MTD), technical feasibility, T-cell trafficking, immune responses, time to progression, and overall survival (OS). EXPERIMENTAL DESIGN ATC were expanded from leukapheresis product using IL2 and anti-CD3 monoclonal antibody and armed with HER2Bi. In 3+3 dose escalation design, groups of 3 patients received 5, 10, 20, or 40 × 10(9) armed ATC (aATC) per infusion. RESULTS There were no dose-limiting toxicities and the MTD was not defined. It was technically feasible to grow 160 × 10(9) ATC from a single leukapheresis. aATC persisted in the blood for weeks and trafficked to tumors. Infusions of aATC induced anti-breast cancer responses and increases in immunokines. At 14.5 weeks after enrollment, 13 of 22 (59.1%) evaluable patients had stable disease and 9 of 22 (40.9%) had progressive disease. The median OS was 36.2 months for all patients, 57.4 months for HER2 3+ patients, and 27.4 months for HER2 0-2+ patients. CONCLUSIONS Targeting HER2(+) and HER2(-) tumors with aATC infusions induced antitumor responses, increases in Th1 cytokines, and IL12 serum levels that suggest that aATC infusions vaccinated patients against their own tumors. These results provide a strong rationale for conducting phase II trials.
Collapse
Affiliation(s)
- Lawrence G Lum
- Department of Oncology, Wayne State University and Karmanos Cancer Institute, Detroit, Michigan. Department of Medicine, Wayne State University and Karmanos Cancer Institute, Detroit, Michigan. Department of Immunology and Microbiology, Wayne State University and Karmanos Cancer Institute, Detroit, Michigan.
| | - Archana Thakur
- Department of Oncology, Wayne State University and Karmanos Cancer Institute, Detroit, Michigan.
| | - Zaid Al-Kadhimi
- Department of Oncology, Wayne State University and Karmanos Cancer Institute, Detroit, Michigan. Department of Medicine, Wayne State University and Karmanos Cancer Institute, Detroit, Michigan
| | | | - Francis J Cummings
- Division of Hematology and Oncology, Department of Medicine, Roger Williams Hospital, Providence, Rhode Island
| | | | - Don S Dizon
- Massachusetts General Hospital Cancer Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Nicola Kouttab
- Department of Pathology, Roger Williams Medical Center, Providence, Rhode Island
| | - Abby Maizel
- Women & Infants Hospital, Providence, Rhode Island
| | - William Colaiace
- Department of Nuclear Medicine, Roger Williams Medical Center, Providence, Rhode Island
| | - Qin Liu
- Department of Medicine, Wistar Institute, Philadelphia, Pennsylvania
| | - Ritesh Rathore
- Division of Hematology and Oncology, Department of Medicine, Roger Williams Hospital, Providence, Rhode Island
| |
Collapse
|
13
|
Pan Q, Li Q, Liu S, Ning N, Zhang X, Xu Y, Chang AE, Wicha MS. Concise Review: Targeting Cancer Stem Cells Using Immunologic Approaches. Stem Cells 2015; 33:2085-92. [PMID: 25873269 DOI: 10.1002/stem.2039] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 02/11/2015] [Indexed: 12/15/2022]
Abstract
Cancer stem cells (CSCs) represent a small subset of tumor cells which have the ability to self-renew and generate the diverse cells that comprise the tumor bulk. They are responsible for local tumor recurrence and distant metastasis. However, they are resistant to conventional radiotherapy and chemotherapy. Novel immunotherapeutic strategies that specifically target CSCs may improve the efficacy of cancer therapy. To immunologically target CSC phenotypes, innate immune responses to CSCs have been reported using Natural killer cells and γδ T cells. To target CSC specifically, in vitro CSC-primed T cells have been successfully generated and shown targeting of CSCs in vivo after adoptive transfer. Recently, CSC-based dendritic cell vaccine has demonstrated significant induction of anti-CSC immunity both in vivo in immunocompetent hosts and in vitro as evident by CSC reactivity of CSC vaccine-primed antibodies and T cells. In addition, identification of specific antigens or genetic alterations in CSCs may provide more specific targets for immunotherapy. ALDH, CD44, CD133, and HER2 have served as markers to isolate CSCs from a number of tumor types in animal models and human tumors. They might serve as useful targets for CSC immunotherapy. Finally, since CSCs are regulated by interactions with the CSC niche, these interactions may serve as additional targets for CSC immunotherapy. Targeting the tumor microenvironment, such as interrupting the immune cell, for example, myeloid-derived suppressor cells, and cytokines, for example, IL-6 and IL-8, as well as the immune checkpoint (PD1/PDL1, etc.) may provide additional novel strategies to enhance the immunological targeting of CSCs.
Collapse
Affiliation(s)
- Qin Pan
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA.,State Key Laboratory of Virology, Department of Immunology, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University School of Medicine, Wuhan, Hubei Province, People's Republic of China
| | - Qiao Li
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Shuang Liu
- Department of Neurosurgery, Navy General Hospital, Beijing, People's Republic of China
| | - Ning Ning
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA.,Department of General Surgery, General Hospital of PLA, Beijing, People's Republic of China
| | - Xiaolian Zhang
- State Key Laboratory of Virology, Department of Immunology, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University School of Medicine, Wuhan, Hubei Province, People's Republic of China
| | - Yingxin Xu
- Department of General Surgery, General Hospital of PLA, Beijing, People's Republic of China
| | - Alfred E Chang
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Max S Wicha
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| |
Collapse
|
14
|
Urbanska K, Lynn RC, Stashwick C, Thakur A, Lum LG, Powell DJ. Targeted cancer immunotherapy via combination of designer bispecific antibody and novel gene-engineered T cells. J Transl Med 2014; 12:347. [PMID: 25496493 PMCID: PMC4272781 DOI: 10.1186/s12967-014-0347-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 11/29/2014] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Redirection of T lymphocytes against tumor antigens can induce dramatic regression of advanced stage malignancy. The use of bispecific antibodies (BsAbs) that bind both the T-cell receptor (TCR) and a target antigen is one promising approach to T-cell redirection. However, BsAbs indiscriminately bind all CD3+ T-cells and trigger TCR activation in the absence of parallel costimulatory signals required to overcome T-cell unresponsiveness or anergy. METHODS To address these limitations, a combination platform was designed wherein a unique BsAb referred to as frBsAb exclusively engages T-cells engineered to express a novel chimeric receptor comprised of extracellular folate receptor fused to intracellular TCR and CD28 costimulatory signaling domains in tandem; a BsAb-binding immune receptor (BsAb-IR). As a surrogate TCR, the BsAb-IR allows for concomitant TCR and costimulatory signaling exclusively in transduced T-cells upon engagement with specific frBsAbs, and can therefore redirect T-cells on command to desired antigen. Human primary T-cells were transduced with lentiviral vector and expanded for 14-18 days. BsAb-IRs were harvested and armed with frBsAbs to test for redirected cytotoxicity against CD20 positive cancer cell lines. RESULTS Using frBsAbs specific for CD20 or HER2, the lytic activity of primary human T-cells expressing the BsAb-IR was specifically redirected against CD20+ leukemic cells or HER2+ epithelial cancer cells, respectively, while non-engineered T-cells were not activated. Notably, elimination of the CD28 costimulatory domain from the BsAb-IR construct significantly reduced frBsAb-redirected antitumor responses, confirming that frBsAbs are capable of delivering simultaneous TCR activation and costimulatory signals to BsAb-IR T-cells. CONCLUSION In summary, our results establish the proof of concept that the combination of BsAbs with optimized gene-engineered T-cells provides the opportunity to specify and augment tumor antigen-specific T-cell activation and may improve upon the early success of conventional BsAbs in cancer immunotherapy.
Collapse
Affiliation(s)
- Katarzyna Urbanska
- Department of Obstetrics and Gynecology, Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Rachel C Lynn
- Department of Obstetrics and Gynecology, Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Caitlin Stashwick
- Department of Obstetrics and Gynecology, Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Archana Thakur
- Department of Oncology, Wayne State University, Detroit, MI, USA.
| | - Lawrence G Lum
- Department of Oncology, Wayne State University, Detroit, MI, USA.
| | - Daniel J Powell
- Department of Pathology and Laboratory Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Bldg 421, Smilow CTR, Rm 08-103, Philadelphia, PA, 19104-5156, USA.
- Department of Obstetrics and Gynecology, Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
15
|
Han H, Ma J, Zhang K, Li W, Liu C, Zhang Y, Zhang G, Ma P, Wang L, Zhang G, Tao H, Gao B. Bispecific anti-CD3 x anti-HER2 antibody mediates T cell cytolytic activity to HER2-positive colorectal cancer in vitro and in vivo. Int J Oncol 2014; 45:2446-54. [PMID: 25242665 DOI: 10.3892/ijo.2014.2663] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 08/18/2014] [Indexed: 11/05/2022] Open
Abstract
Targeting HER2 overexpressed breast cancer cells with anti‑HER2 monoclonal antibodies inhibits tumor growth. Here we investigated whether HER2 can serve as a target for T cell-mediated immunotherapy of human colorectal carcinoma. Specific cytolytic activity of activated T cells (ATCs) armed with anti‑CD3 x anti‑HER2 bispecific antibody (HER2Bi-Ab) against HER2+ tumor cells was evaluated by bioluminescent signal generated by luciferase reporter on tumor cells in vitro and in vivo. In contrast to unarmed ATCs, increased cytotoxic activity of HER2Bi-armed ATCs against HER2+ tumor cells was observed. Moreover, HER2Bi-armed ATCs expressed higher level of activation marker CD69 and secreted significantly higher levels of IFN-γ than the unarmed ATC counterpart. In addition, compared with anti‑HER2 mAb (Herceptin®) or unarmed ATC, HER2Bi-armed ATCs showed significant suppression against colorectal carcinoma cells. In colorectal tumor cell xenograft mice, infusion of HER2Bi-armed ATCs successfully inhibited the growth of Colo205-luc cells. The HER2Bi-armed ATCs with anti-tumor effects may provide a promising immunotherapy for colorectal carcinoma in the future.
Collapse
Affiliation(s)
- Huamin Han
- CAS Key Laboratory of Pathogenic Microbiology and Immunology (CASPMI), Centre for Molecular Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P.R. China
| | - Juan Ma
- CAS Key Laboratory of Pathogenic Microbiology and Immunology (CASPMI), Centre for Molecular Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P.R. China
| | - Keming Zhang
- Department of Hepatobiliary Surgery, 302 Military Hospital of China, Beijing, P.R. China
| | - Wei Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology (CASPMI), Centre for Molecular Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P.R. China
| | - Changzhen Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology (CASPMI), Centre for Molecular Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P.R. China
| | - Yu Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology (CASPMI), Centre for Molecular Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P.R. China
| | - Ganlin Zhang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine affiliated to Capital Medical University, Beijing, P.R. China
| | - Pan Ma
- CAS Key Laboratory of Pathogenic Microbiology and Immunology (CASPMI), Centre for Molecular Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P.R. China
| | - Lei Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology (CASPMI), Centre for Molecular Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P.R. China
| | - Ge Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology (CASPMI), Centre for Molecular Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P.R. China
| | - Hua Tao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology (CASPMI), Centre for Molecular Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P.R. China
| | - Bin Gao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology (CASPMI), Centre for Molecular Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P.R. China
| |
Collapse
|
16
|
Thakur A, Vaishampayan U, Lum LG. Immunotherapy and immune evasion in prostate cancer. Cancers (Basel) 2013; 5:569-90. [PMID: 24216992 PMCID: PMC3730318 DOI: 10.3390/cancers5020569] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 04/16/2013] [Accepted: 05/08/2013] [Indexed: 12/27/2022] Open
Abstract
Metastatic prostate cancer remains to this day a terminal disease. Prostatectomy and radiotherapy are effective for organ-confined diseases, but treatment for locally advanced and metastatic cancer remains challenging. Although advanced prostate cancers treated with androgen deprivation therapy achieves debulking of disease, responses are transient with subsequent development of castration-resistant and metastatic disease. Since prostate cancer is typically a slowly progressing disease, use of immune-based therapies offers an advantage to target advanced tumors and to induce antitumor immunity. This review will discuss the clinical merits of various vaccines and immunotherapies in castrate resistant prostate cancer and challenges to this evolving field of immune-based therapies.
Collapse
Affiliation(s)
- Archana Thakur
- Department of Oncology, Wayne State University, Detroit, MI 48201, USA
| | - Ulka Vaishampayan
- Department of Oncology, Wayne State University, Detroit, MI 48201, USA
| | - Lawrence G. Lum
- Department of Oncology, Wayne State University, Detroit, MI 48201, USA
- Department of Medicine, Wayne State University, Detroit, MI 48201, USA
- Department of Immunology and Microbiology, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
17
|
Efficient generation of stable bispecific IgG1 by controlled Fab-arm exchange. Proc Natl Acad Sci U S A 2013; 110:5145-50. [PMID: 23479652 DOI: 10.1073/pnas.1220145110] [Citation(s) in RCA: 254] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The promise of bispecific antibodies (bsAbs) to yield more effective therapeutics is well recognized; however, the generation of bsAbs in a practical and cost-effective manner has been a formidable challenge. Here we present a technology for the efficient generation of bsAbs with normal IgG structures that is amenable to both antibody drug discovery and development. The process involves separate expression of two parental antibodies, each containing single matched point mutations in the CH3 domains. The parental antibodies are mixed and subjected to controlled reducing conditions in vitro that separate the antibodies into HL half-molecules and allow reassembly and reoxidation to form highly pure bsAbs. The technology is compatible with standard large-scale antibody manufacturing and ensures bsAbs with Fc-mediated effector functions and in vivo stability typical of IgG1 antibodies. Proof-of-concept studies with HER2×CD3 (T-cell recruitment) and HER2×HER2 (dual epitope targeting) bsAbs demonstrate superior in vivo activity compared with parental antibody pairs.
Collapse
|
18
|
Zitron IM, Thakur A, Norkina O, Barger GR, Lum LG, Mittal S. Targeting and killing of glioblastoma with activated T cells armed with bispecific antibodies. BMC Cancer 2013; 13:83. [PMID: 23433400 PMCID: PMC3599512 DOI: 10.1186/1471-2407-13-83] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 02/11/2013] [Indexed: 12/29/2022] Open
Abstract
Background Since most glioblastomas express both wild-type EGFR and EGFRvIII as well as HER2/neu, they are excellent targets for activated T cells (ATC) armed with bispecific antibodies (BiAbs) that target EGFR and HER2. Methods ATC were generated from PBMC activated for 14 days with anti-CD3 monoclonal antibody in the presence of interleukin-2 and armed with chemically heteroconjugated anti-CD3×anti-HER2/neu (HER2Bi) and/or anti-CD3×anti-EGFR (EGFRBi). HER2Bi- and/or EGFRBi-armed ATC were examined for in vitro cytotoxicity using MTT and 51Cr-release assays against malignant glioma lines (U87MG, U118MG, and U251MG) and primary glioblastoma lines. Results EGFRBi-armed ATC killed up to 85% of U87, U118, and U251 targets at effector:target ratios (E:T) ranging from 1:1 to 25:1. Engagement of tumor by EGFRBi-armed ATC induced Th1 and Th2 cytokine secretion by armed ATC. HER2Bi-armed ATC exhibited comparable cytotoxicity against U118 and U251, but did not kill HER2-negative U87 cells. HER2Bi- or EGFRBi-armed ATC exhibited 50—80% cytotoxicity against four primary glioblastoma lines as well as a temozolomide (TMZ)-resistant variant of U251. Both CD133– and CD133+ subpopulations were killed by armed ATC. Targeting both HER2Bi and EGFRBi simultaneously showed enhanced efficacy than arming with a single BiAb. Armed ATC maintained effectiveness after irradiation and in the presence of TMZ at a therapeutic concentration and were capable of killing multiple targets. Conclusion High-grade gliomas are suitable for specific targeting by armed ATC. These data, together with additional animal studies, may provide the preclinical support for the use of armed ATC as a valuable addition to current treatment regimens.
Collapse
Affiliation(s)
- Ian M Zitron
- Department of Neurosurgery, Wayne State University, Karmanos Cancer Institute, Detroit, MI, USA
| | | | | | | | | | | |
Collapse
|
19
|
Abstract
Bispecific antibodies (BiAbs) offer a unique opportunity to redirect immune effector cells to kill cancer cells. BiAbs combine the benefits of different binding specificities of two monoclonal antibodies (mAbs) into a single construct. This unique feature of BiAbs enables approaches that are not possible with single mAbs. Advances in antibody engineering and antigen profiling of malignant cells have led to the development of a number of BiAb formats and their combinations for redirecting effector cells to tumor targets. There have been significant advances in the design and application of BiAbs for intravenous and local injection.The initial barrier of cytokine storm has been partially overcome by more recent constructs that have improved clinical effectiveness without dose-limiting toxicities. Since the recent revival of BiAbs, there has been multiple, ongoing, phase I/II and III trials, and some promising clinical outcomes have been reported in completed clinical studies. This review focuses on arming T cells with BiAbs to create the 'poor man's cytotoxic lymphocyte'.
Collapse
Affiliation(s)
- Lawrence G Lum
- Department of Oncology, Wayne State University and Barbara Ann Karmanos Cancer Center, Detroit, MI, USA
| | | |
Collapse
|
20
|
Lum LG, Ramesh M, Thakur A, Mitra S, Deol A, Uberti JP, Pellett PE. Targeting cytomegalovirus-infected cells using T cells armed with anti-CD3 × anti-CMV bispecific antibody. Biol Blood Marrow Transplant 2012; 18:1012-22. [PMID: 22313635 DOI: 10.1016/j.bbmt.2012.01.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Accepted: 01/31/2012] [Indexed: 11/25/2022]
Abstract
Human cytomegalovirus (CMV) reactivation and infection can lead to poor outcomes after allogeneic stem cell transplantation. We hypothesized that anti-CD3 activated T cells (ATCs) armed with chemically heteroconjugated anti-CD3 × polyclonal anti-CMV bispecific antibody (CMVBi) will target and eliminate CMV-infected cells. Arming doses of CMVBi as low as 0.01 ng/10(6) ATCs was able to mediate specific cytotoxicity (SC) directed at CMV-infected target cells significant above unarmed ATCs at mutiplicities of infection (MOI) between 0.01 and 1. At effector-to-target ratios (E:T) of 25:1, 12.5:1, 6.25:1, and 3.125:1, armed ATCs significantly enhanced killing of CMV-infected targets compared with unarmed ATCs. At an MOI of 1.0, the mean % SC directed at CMV-infected targets cells for CMVBi-armed ATCs at E:T of 3.12, 6.25, and 12.5 were 79%, 81%, and 82%, respectively; whereas the mean % SC for unarmed ATCs at the same E:T were all <20%. ATCs, Cytogam(®), or CMVBi alone did not lyse uninfected or CMV-infected targets. Co-cultures of CMVBi-armed ATCs with CMV-infected targets induced cytokine and chemokine release from armed ATCs. This nonmajor histocompatibility complex restricted strategy for targeting CMV could be used to prevent or treat CMV infections after allogeneic stem cell transplantation or organ transplantation.
Collapse
Affiliation(s)
- Lawrence G Lum
- Bone Marrow Transplantation and Immunotherapy Program, Wayne State University and Karmanos Cancer Institute, Detroit, MI, USA.
| | | | | | | | | | | | | |
Collapse
|
21
|
Lum LG, Al-Kadhimi Z. Development and prospects for bispecific antibody-based therapeutics in cancer and other applications. Expert Opin Drug Discov 2008; 3:1081-97. [PMID: 23506181 DOI: 10.1517/17460441.3.9.1081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Progress in molecular biology for the production of bispecific antibodies (BiAbs) and the expanding knowledge on receptors on malignant and normal target cells provide the basis for developing new strategies using antibody- and/or receptor-based platform technology for the treatment of cancer and other diseases. OBJECTIVE This review provides a preclinical and clinical perspective on application of bispecific antibodies for the treatment of solid tumors, hematologic malignancies and other diseases. METHODS This review focuses on BiAb-based immunotherapy, clinical trials, alternative strategies, the challenges of technology and future applications. RESULTS/CONCLUSION The successful application of a particular BiAb will depend on a thorough evaluation of the expected functional application and thoughtful engineering of structure, affinity and number of binding sites based on the desired function.
Collapse
Affiliation(s)
- Lawrence G Lum
- Professor of Medicine and Professor of Immunology and Microbiology, Scientific Director of BMT and Immunotherapy Program, Wayne State University, Member Barbara Ann Karmanos Cancer Institute, Hudson-Webber Cancer Research Center, 4100 John R., 7th Floor, Rm 740.1, Detroit, Michigan, MI 48201, USA +1 313 576 8326 ; +1 313 576 8939 ;
| | | |
Collapse
|
22
|
Baxevanis CN. Antibody-based cancer therapy. Expert Opin Drug Discov 2008; 3:441-52. [PMID: 23489099 DOI: 10.1517/17460441.3.4.441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND The clinical efficacy of mAbs is generally ascribed to interference with signaling pathways leading to arrest of cell-cycle progression and inhibition of tumor growth. Furthermore, mAbs also have the capacity to activate effector functions of the innate immune system and facilitate the destruction of malignant cells. OBJECTIVES The induction of tumor-specific immunity is a desired outcome in cancer immunotherapy. The prevailing situation raises one major question that has to be addressed. This is the clear need for the induction of tumor-specific immunity by combining mAb treatment with other modalities of cancer immunotherapy. METHODS Through mAb treatment, recent efforts focus at initiating or enhancing active antitumor immune responses by i) potentiating co-stimulation and blocking co-inhibition; and ii) rendering tumors more immunogenic through increased tumor peptide expression. RESULTS/CONCLUSIONS In this review, the functional characteristics of mAbs, together with their mechanisms of action and clinical application, is summarized as is the potential of combination immunotherapies using mAbs for the augmentation of adaptive antitumor immunity. The results from preclinical and clinical studies demonstrate that mAbs can also promote tumor-specific active immunity.
Collapse
Affiliation(s)
- Constantin N Baxevanis
- St. Savas Cancer Hospital, Cancer Immunology and Immunotherapy Center, 171 Alexandras Ave, 11522 Athens, Greece +30 210 6409380 ;
| |
Collapse
|
23
|
Friedländer E, Barok M, Szöllősi J, Vereb G. ErbB-directed immunotherapy: Antibodies in current practice and promising new agents. Immunol Lett 2008; 116:126-40. [DOI: 10.1016/j.imlet.2007.12.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2007] [Accepted: 12/01/2007] [Indexed: 01/24/2023]
|
24
|
Lum LG, Davol PA, Lee RJ. The new face of bispecific antibodies: targeting cancer and much more. Exp Hematol 2006; 34:1-6. [PMID: 16413384 DOI: 10.1016/j.exphem.2005.07.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2005] [Accepted: 03/17/2005] [Indexed: 01/25/2023]
Abstract
The term magic bullet was first coined by bacteriologist Paul Ehrlich in the late 1800s to describe a chemical with the ability to specifically target microorganisms while sparing normal host cells. His concept was later expanded to include treatments for cancer, but it is only in recent decades, with development and improvements in monoclonal antibody (mAb) technology, that the full therapeutic implications of "magic bullet" strategies have been realized. Expanding on the success of mAb-targeting, linking the specificity of two mAbs into a single agent, called a bispecific antibody (BiAb), allows for targeting of a therapeutic biological agent or cell to specific tissue antigens. Classically, BiAbs have been used for several decades to redirect cytotoxic T cells or other effector cells to kill tumor cells. Here, we review preclinical models and ongoing phase I clinical trials in which arming polyclonally activated T cells with BiAbs may provide anti-tumor activity without dose-limiting toxicities. Additionally, we review findings from this novel strategy that merges magic bullet technology with hematopoietic stem cells to repair injured myocardium. Arming stem cells with BiAbs directed at injury-associated antigens enhances specific homing and engraftment to myocardial infarctions and may significantly improve cardiac function, strongly suggesting new paradigms for BiAb-targeting applications in tissue repair.
Collapse
Affiliation(s)
- Lawrence G Lum
- Immunotherapy Program, Adele R. Decof Cancer Center, Roger Williams Medical Center, Providence, RI 02908, USA.
| | | | | |
Collapse
|
25
|
Grabert RC, Cousens LP, Smith JA, Olson S, Gall J, Young WB, Davol PA, Lum LG. Human T cells armed with Her2/neu bispecific antibodies divide, are cytotoxic, and secrete cytokines with repeated stimulation. Clin Cancer Res 2006; 12:569-76. [PMID: 16428502 DOI: 10.1158/1078-0432.ccr-05-2005] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Cancer immunotherapy has been limited by anergy of patient T cells, inadequate numbers of precursor tumor-specific CTL, and difficulty in producing therapeutic doses of CTL. To overcome these limitations, bispecific antibodies have been used to create artificial antibody receptors that direct polyclonal activated T cells (ATC) to target tumor antigens. Studies reported herein were designed to characterize bispecific antibody-armed ATC functions during multiple rounds of targeted cell stimulation. EXPERIMENTAL DESIGN ATCs were generated from human peripheral blood mononuclear cells (PBMC) by culture with anti-CD3 and interleukin 2 for 14 days and armed with anti-CD3 x anti-Her2 bispecific antibody (Her2Bi). In vitro, Her2Bi-armed ATC were examined for a range of functions after repeated stimulation with the Her2/neu-expressing breast cancer cell line SK-BR-3. PBMC isolated from cancer patients treated with Her2Bi-armed ATC were tested ex vivo for cytotoxicity against SK-BR-3. RESULTS In vitro, armed ATC divided, maintained surface Her2Bi, and expressed a range of activities for extended periods of time. Perforin-mediated cytotoxic activity by armed ATC continued for at least 336 hours, and cytokines and chemokines (i.e., IFN-gamma and regulated on activation, normal T-cell expressed and secreted protein [RANTES]) were secreted during successive rounds of stimulation. Furthermore, PBMC isolated from patients over their courses of immunotherapy exhibited significant cytolytic activity against SK-BR-3 as a function of Her2Bi-armed ATC infusions. CONCLUSIONS These studies show that armed ATC are specific, durable, and highly functional T-cell populations in vitro. These previously unappreciated broad and long-term functions of armed ATC are encouraging for their therapeutic use in treating cancer.
Collapse
Affiliation(s)
- Ryan C Grabert
- Immunotherapy and Blood and Stem Cell Transplant Programs, Adele R. Decof Cancer Center and Division of Hematology/Oncology, Department of Medicine, Roger Williams Medical Center, 825 Chalkstone Avenue, Providence, RI 02908, USA
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Boulter JM, Jakobsen BK. Stable, soluble, high-affinity, engineered T cell receptors: novel antibody-like proteins for specific targeting of peptide antigens. Clin Exp Immunol 2005; 142:454-60. [PMID: 16297157 PMCID: PMC1809535 DOI: 10.1111/j.1365-2249.2005.02929.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2005] [Indexed: 11/29/2022] Open
Abstract
The recent development of T cell receptor phage display opens up the possibility of engineering human T cell receptors with antibody-like binding properties for cell-surface peptide antigens. In this review we briefly discuss recent developments in molecular targeting of peptide antigens. We then discuss potential clinical applications of engineered high-affinity T cell receptors in autoimmunity and cancer.
Collapse
Affiliation(s)
- J M Boulter
- Department of Medical Biochemistry and Immunology, Henry Wellcome Building, School of Medicine, Cardiff University, Heath Park, UK.
| | | |
Collapse
|
27
|
Gall JM, Davol PA, Grabert RC, Deaver M, Lum LG. T cells armed with anti-CD3 x anti-CD20 bispecific antibody enhance killing of CD20+ malignant B cells and bypass complement-mediated rituximab resistance in vitro. Exp Hematol 2005; 33:452-9. [PMID: 15781336 DOI: 10.1016/j.exphem.2005.01.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2004] [Revised: 12/22/2004] [Accepted: 01/11/2005] [Indexed: 10/25/2022]
Abstract
OBJECTIVE Resistance to rituximab, a chimeric monoclonal antibody that binds to CD20, is a major limitation for the successful treatment of patients with non-Hodgkin lymphoma and other CD20+ B-cell malignancies. To circumvent rituximab resistance in these patient populations, we have constructed a bispecific antibody (BiAb), anti-CD3 x anti-CD20 (CD20Bi), that combines rituximab targeting with non-major histocompatibility complex (non-MHC)-restricted cytotoxicity mediated by activated T cells (ATC). MATERIALS AND METHODS Activated T cells were obtained from anti-CD3 activated peripheral blood mononuclear cells (PBMC) of normal donors or the leukapheresis products of patients by culturing in the presence of interleukin-2 for 6-14 days. After ATC expansion, the cells were armed with CD20Bi. Killing activity was evaluated by 51Cr-release assay. RESULTS Arming ATC with as little as 5 ng CD20Bi/10(6) cells significantly increased cytotoxicity above unarmed ATC. CD20Bi-armed ATC (50 ng/10(6) cells) efficiently lysed CD20+ cell lines at E:T of 6.25-50, but not the nonhematologic, CD20- SK-BR-3 cell line. High levels of cytotoxicity mediated by CD20Bi-armed ATC (p < 0.05) could not be blocked by an 8000-fold excess of soluble rituximab. CD20Bi-armed ATC in the presence of complement killed ARH-77 cells, a rituximab-complement pathway-resistant multiple myeloma, significantly (p < 0.05) better than rituximab or unarmed ATC, suggesting that CD20Bi-armed ATC may be clinically effective for treatment of rituximab-resistant CD20+ hematologic malignancies. CONCLUSIONS Our findings demonstrate that CD20Bi-armed ATC enhance cytotoxicity against CD20+ B-cell lines and circumvent complement-mediated rituximab resistance, providing a strong rationale for this immune-based strategy for the treatment of rituximab-refractory CD20+ B-cell malignancies.
Collapse
Affiliation(s)
- Jonathan M Gall
- Cancer Immunotherapy Laboratory and Blood and Bone Marrow Transplant Programs, Adele R. Decof Cancer Center, Roger Williams Medical Center, Providence, RI 02908, USA
| | | | | | | | | |
Collapse
|