1
|
Davé E, Durrant O, Dhami N, Compson J, Broadbridge J, Archer S, Maroof A, Whale K, Menochet K, Bonnaillie P, Barry E, Wild G, Peerboom C, Bhatta P, Ellis M, Hinchliffe M, Humphreys DP, Heywood SP. TRYBE®: an Fc-free antibody format with three monovalent targeting arms engineered for long in vivo half-life. MAbs 2023; 15:2160229. [PMID: 36788124 PMCID: PMC9937000 DOI: 10.1080/19420862.2022.2160229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023] Open
Abstract
TrYbe® is an Fc-free therapeutic antibody format, capable of engaging up to three targets simultaneously, with long in vivo half-life conferred by albumin binding. This format is shown by small-angle X-ray scattering to be conformationally flexible with favorable 'reach' properties. We demonstrate the format's broad functionality by co-targeting of soluble and cell surface antigens. The benefit of monovalent target binding is illustrated by the lack of formation of large immune complexes when co-targeting multivalent antigens. TrYbes® are manufactured using standard mammalian cell culture and protein A affinity capture processes. TrYbes® have been formulated at high concentrations and have favorable drug-like properties, including stability, solubility, and low viscosity. The unique functionality and inherent developability of the TrYbe® makes it a promising multi-specific antibody fragment format for antibody therapy.
Collapse
Affiliation(s)
- Emma Davé
- Early Solutions, UCB Biopharma UK, Slough, UK
| | | | - Neha Dhami
- Early Solutions, UCB Biopharma UK, Slough, UK
| | | | | | | | | | - Kevin Whale
- Early Solutions, UCB Biopharma UK, Slough, UK
| | | | | | - Emily Barry
- Early Solutions, UCB Biopharma UK, Slough, UK
| | - Gavin Wild
- PV Supply and Technology Solutions, UCB Biopharma UK, Slough, UK
| | - Claude Peerboom
- PV Supply and Technology Solutions, UCB Biopharma SRL, Braine-l'Alleud, Belgium, EU
| | | | - Mark Ellis
- Early Solutions, UCB Biopharma UK, Slough, UK
| | | | | | - Sam P. Heywood
- Early Solutions, UCB Biopharma UK, Slough, UK,CONTACT Sam P. Heywood Early Solutions, UCB Biopharma UK, 208 Bath Road, Slough, SL1 3XE, Slough, UK
| |
Collapse
|
2
|
Mossenta M, Busato D, Dal Bo M, Macor P, Toffoli G. Novel Nanotechnology Approaches to Overcome Drug Resistance in the Treatment of Hepatocellular Carcinoma: Glypican 3 as a Useful Target for Innovative Therapies. Int J Mol Sci 2022; 23:10038. [PMID: 36077433 PMCID: PMC9456072 DOI: 10.3390/ijms231710038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/30/2022] [Accepted: 08/30/2022] [Indexed: 11/23/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the second most lethal tumor, with a 5-year survival rate of 18%. Early stage HCC is potentially treatable by therapies with curative intent, whereas chemoembolization/radioembolization and systemic therapies are the only therapeutic options for intermediate or advanced HCC. Drug resistance is a critical obstacle in the treatment of HCC that could be overcome by the use of targeted nanoparticle-based therapies directed towards specific tumor-associated antigens (TAAs) to improve drug delivery. Glypican 3 (GPC3) is a member of the glypican family, heparan sulfate proteoglycans bound to the cell surface via a glycosylphosphatidylinositol anchor. The high levels of GPC3 detected in HCC and the absence or very low levels in normal and non-malignant liver make GPC3 a promising TAA candidate for targeted nanoparticle-based therapies. The use of nanoparticles conjugated with anti-GPC3 agents may improve drug delivery, leading to a reduction in severe side effects caused by chemotherapy and increased drug release at the tumor site. In this review, we describe the main clinical features of HCC and the common treatment approaches. We propose the proteoglycan GPC3 as a useful TAA for targeted therapies. Finally, we describe nanotechnology approaches for anti-GPC3 drug delivery systems based on NPs for HCC treatment.
Collapse
Affiliation(s)
- Monica Mossenta
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Davide Busato
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Paolo Macor
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| |
Collapse
|
3
|
A HER2 Tri-Specific NK Cell Engager Mediates Efficient Targeting of Human Ovarian Cancer. Cancers (Basel) 2021; 13:cancers13163994. [PMID: 34439149 PMCID: PMC8394622 DOI: 10.3390/cancers13163994] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/29/2021] [Accepted: 08/04/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary HER2 is a marker known to be over-expressed on breast cancer, rendering it one of the most useful solid tumor targets for antibody-based therapies. Despite expression on ovarian cancer, results targeting HER2 in this setting have been disappointing, thus requiring more aggressive approaches. Natural killer (NK) cells are known as principal mediators of cancer cell killing, but cancer cells find ways to deter them. We devised a tri-specific biological drug containing antibody fragments that simultaneously binds NK cells and cancer cells and at the same time delivers a natural cytokine signal that triggers robust NK cell expansion. In vitro studies show the drug augments NK cell killing of a number of HER2-positive human cell lines, while enhancing NK cell activation and proliferation. Studies in mice engrafted with human ovarian cancer showed the drug has anti-tumor efficacy, clearly demonstrating its ability to bolster NK cells in their ability to contain tumor cell growth. Abstract Clinical studies validated antibodies directed against HER2, trastuzumab, and pertuzumab, as useful methodology to target breast cancer cases where HER2 is expressed. The hope was that HER2 targeting using these antibodies in ovarian cancer patients would prove useful as well, but clinical studies have shown lackluster results in this setting, indicating a need for a more comprehensive approach. Immunotherapy approaches stimulating the innate immune system show great promise, although enhancing natural killer (NK) function is not an established mainstream immunotherapy. This study focused on a new nanobody platform technology in which the bispecific antibody was altered to incorporate a cytokine. Herein we describe bioengineered CAM1615HER2 consisting of a camelid VHH antibody fragment recognizing CD16 and a single chain variable fragment (scFv) recognizing HER2 cross-linked by the human interleukin-15 (IL-15) cytokine. This tri-specific killer engager (TriKETM) showed in vitro prowess in its ability to kill ovarian cancer human cell lines. In addition, we demonstrated its efficacy in inducing potent anti-cancer effects in an in vivo xenograft model of human ovarian cancer engrafting both cancer cells and human NK cells. While previous approaches with trastuzumab and pertuzumab faltered in ovarian cancer, the hope is incorporating targeting and cytokine priming within the same molecule will enhance efficacy in this setting.
Collapse
|
4
|
Park S, Nguyen MQ, Ta HKK, Nguyen MT, Lee G, Kim CJ, Jang YJ, Choe H. Soluble Cytoplasmic Expression and Purification of Immunotoxin HER2(scFv)-PE24B as a Maltose Binding Protein Fusion. Int J Mol Sci 2021; 22:6483. [PMID: 34204265 PMCID: PMC8234717 DOI: 10.3390/ijms22126483] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/04/2021] [Accepted: 06/10/2021] [Indexed: 01/30/2023] Open
Abstract
Human epidermal growth factor receptor 2 (HER-2) is overexpressed in many malignant tumors. The anti-HER2 antibody trastuzumab has been approved for treating HER2-positive early and metastatic breast cancers. Pseudomonas exotoxin A (PE), a bacterial toxin of Pseudomonas aeruginosa, consists of an A-domain with enzymatic activity and a B-domain with cell binding activity. Recombinant immunotoxins comprising the HER2(scFv) single-chain Fv from trastuzumab and the PE24B catalytic fragment of PE display promising cytotoxic effects, but immunotoxins are typically insoluble when expressed in the cytoplasm of Escherichia coli, and thus they require solubilization and refolding. Herein, a recombinant immunotoxin gene was fused with maltose binding protein (MBP) and overexpressed in a soluble form in E. coli. Removal of the MBP yielded stable HER2(scFv)-PE24B at 91% purity; 0.25 mg of pure HER2(scFv)-PE24B was obtained from a 500 mL flask culture. Purified HER2(scFv)-PE24B was tested against four breast cancer cell lines differing in their surface HER2 level. The immunotoxin showed stronger cytotoxicity than HER2(scFv) or PE24B alone. The IC50 values for HER2(scFv)-PE24B were 28.1 ± 2.5 pM (n = 9) and 19 ± 1.4 pM (n = 9) for high HER2-positive cell lines SKBR3 and BT-474, respectively, but its cytotoxicity was lower against MDA-MB-231 and MCF7. Thus, fusion with MBP can facilitate the soluble expression and purification of scFv immunotoxins.
Collapse
Affiliation(s)
- Sangsu Park
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea; (S.P.); (M.Q.N.); (H.K.K.T.); (M.T.N.); (Y.J.J.)
| | - Minh Quan Nguyen
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea; (S.P.); (M.Q.N.); (H.K.K.T.); (M.T.N.); (Y.J.J.)
| | - Huynh Kim Khanh Ta
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea; (S.P.); (M.Q.N.); (H.K.K.T.); (M.T.N.); (Y.J.J.)
| | - Minh Tan Nguyen
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea; (S.P.); (M.Q.N.); (H.K.K.T.); (M.T.N.); (Y.J.J.)
- NTT Hi-Tech Institute, Nguyen Tat Thanh University, Ho Chi Minh City 70000, Vietnam
| | - Gunsup Lee
- R&D Center, Fatiabgen Co., Ltd., Seoul 05855, Korea;
| | - Chong Jai Kim
- Department of Pathology, Asan-Minnesota Institute for Innovating Transplantation, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea;
| | - Yeon Jin Jang
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea; (S.P.); (M.Q.N.); (H.K.K.T.); (M.T.N.); (Y.J.J.)
| | - Han Choe
- Department of Physiology, Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea; (S.P.); (M.Q.N.); (H.K.K.T.); (M.T.N.); (Y.J.J.)
| |
Collapse
|
5
|
Shilova O, Shramova E, Proshkina G, Deyev S. Natural and Designed Toxins for Precise Therapy: Modern Approaches in Experimental Oncology. Int J Mol Sci 2021; 22:ijms22094975. [PMID: 34067057 PMCID: PMC8124712 DOI: 10.3390/ijms22094975] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 04/29/2021] [Accepted: 05/05/2021] [Indexed: 02/08/2023] Open
Abstract
Cancer cells frequently overexpress specific surface receptors providing tumor growth and survival which can be used for precise therapy. Targeting cancer cell receptors with protein toxins is an attractive approach widely used in contemporary experimental oncology and preclinical studies. Methods of targeted delivery of toxins to cancer cells, different drug carriers based on nanosized materials (liposomes, nanoparticles, polymers), the most promising designed light-activated toxins, as well as mechanisms of the cytotoxic action of the main natural toxins used in modern experimental oncology, are discussed in this review. The prospects of the combined therapy of tumors based on multimodal nanostructures are also discussed.
Collapse
Affiliation(s)
- Olga Shilova
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia; (E.S.); (G.P.)
- Correspondence: (O.S.); (S.D.)
| | - Elena Shramova
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia; (E.S.); (G.P.)
| | - Galina Proshkina
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia; (E.S.); (G.P.)
| | - Sergey Deyev
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia; (E.S.); (G.P.)
- Center of Biomedical Engineering, Sechenov University, 119991 Moscow, Russia
- Research Centrum for Oncotheranostics, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia
- Correspondence: (O.S.); (S.D.)
| |
Collapse
|
6
|
Ou-Yang Q, Ren JL, Yan B, Feng JN, Yang AG, Zhao J. Syngeneic homograft of framework regions enhances the affinity of the mouse anti-human epidermal receptor 2 single-chain antibody e23sFv. Exp Ther Med 2020; 21:136. [PMID: 33456503 PMCID: PMC7791966 DOI: 10.3892/etm.2020.9568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 10/30/2020] [Indexed: 12/03/2022] Open
Abstract
e23sFv is a HER2-targeted single-chain variable fragment (scFV) that was characterized as the targeting portion of a HER2-targeted tumour proapoptotic molecule in our previous study. In vitro antibody affinity maturation is a method to enhance antibody affinity either by complementarity-determining region (CDR) mutagenesis or by framework region (FR) engraftment. In the present study, the affinity of e23sFv was enhanced using two strategies. In one approach, site-directed mutations were introduced into the FRs of e23sFv (designated EMEY), and in the other approach e23sFv FRs were substituted with FRs from the most homologous screened antibodies (designated EX1 and EX2). Notably, EX1 derived from the FR engraftment strategy demonstrated a 4-fold higher affinity for HER2 compared with e23sFv and was internalized into HER2-overexpressing cells; however, EMEY and EX2 exhibited reduced affinity for HER2 and decreased internalization potential compared with EX1. The 3D structure of EX1 and the HER2-EX1 complex was acquired using molecular homology modelling and docking and the HER2 epitopes of EX1 and the molecular interaction energy of the EX1-HER2 complex were predicted. In the present study, it was demonstrated that scFv affinity improvement based on sequence alignment was feasible and effective. Moreover, the FR grafting strategy was indicated to be more effective and simple compared with site-directed mutagenesis to improve e23sFv affinity. In conclusion, it was indicated that the affinity-improved candidate EX1 may present a great potential for the diagnosis and treatment of HER2-overexpressing tumours.
Collapse
Affiliation(s)
- Qing Ou-Yang
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China.,State Key Laboratory of Kidney Diseases, Department of Nephrology, Chinese PLA General Hospital & Chinese PLA Medical School, Beijing 100853, P.R. China
| | - Jun-Lin Ren
- Department of Infectious Diseases, PLA Navy General Hospital, Beijing 100142, P.R. China
| | - Bo Yan
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Jian-Nan Feng
- Department of Immunology, Beijing Institute of Basic Medical Sciences, Beijing 100850, P.R. China
| | - An-Gang Yang
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Jing Zhao
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
7
|
Busato D, Mossenta M, Baboci L, Di Cintio F, Toffoli G, Dal Bo M. Novel immunotherapeutic approaches for hepatocellular carcinoma treatment. Expert Rev Clin Pharmacol 2019; 12:453-470. [PMID: 30907177 DOI: 10.1080/17512433.2019.1598859] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION The introduction of immune checkpoint inhibitors has been lately proposed for the treatment of hepatocellular carcinoma (HCC) with respect to other cancer types. Several immunotherapeutic approaches are now under evaluation for HCC treatment including: i) antibodies acting as immune checkpoint inhibitors; ii) antibodies targeting specific tumor-associated antigens; iii) chimeric antigen receptor redirected T (CAR-T) cells targeting specific tumor-associated antigens; iv) vaccination strategies with tumor-specific epitopes. Areas covered: The review provides a wide description of the clinical trials investigating the efficacy of the main immunotherapeutic approaches proposed for the treatment of patients affected by HCC. Expert opinion: The balancing between immunostimulative and immunosuppressive factors in the context of HCC tumor microenvironment results in heterogeneous response rates to immunotherapeutic approaches such as checkpoint inhibitors, among HCC patients. In this context, it becomes crucial the identification of predictive factors determining the treatment response. A multiple approach using different biomarkers could be useful to identify the subgroup of HCC patients responsive to the treatment with a checkpoint inhibitor (as an example, nivolumab) as single agent, and to identify those patients in which other treatment regimens, such as the combination with sorafenib, or with locoregional therapies, could be more effective.
Collapse
Affiliation(s)
- Davide Busato
- a Experimental and Clinical Pharmacology Unit , Centro di Riferimento Oncologico di Aviano (CRO), IRCCS , Aviano (PN) , Italy.,b Department of Life Sciences , University of Trieste , Trieste , Italy
| | - Monica Mossenta
- a Experimental and Clinical Pharmacology Unit , Centro di Riferimento Oncologico di Aviano (CRO), IRCCS , Aviano (PN) , Italy.,b Department of Life Sciences , University of Trieste , Trieste , Italy
| | - Lorena Baboci
- a Experimental and Clinical Pharmacology Unit , Centro di Riferimento Oncologico di Aviano (CRO), IRCCS , Aviano (PN) , Italy
| | - Federica Di Cintio
- a Experimental and Clinical Pharmacology Unit , Centro di Riferimento Oncologico di Aviano (CRO), IRCCS , Aviano (PN) , Italy.,b Department of Life Sciences , University of Trieste , Trieste , Italy
| | - Giuseppe Toffoli
- a Experimental and Clinical Pharmacology Unit , Centro di Riferimento Oncologico di Aviano (CRO), IRCCS , Aviano (PN) , Italy
| | - Michele Dal Bo
- a Experimental and Clinical Pharmacology Unit , Centro di Riferimento Oncologico di Aviano (CRO), IRCCS , Aviano (PN) , Italy
| |
Collapse
|
8
|
Eggers R, Philippi A, Altmeyer MO, Breinig F, Schmitt MJ. Primary T cells for mRNA-mediated immunotoxin delivery. Gene Ther 2018; 25:47-53. [PMID: 28937681 DOI: 10.1038/gt.2017.87] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 08/23/2017] [Accepted: 09/18/2017] [Indexed: 12/30/2022]
Abstract
Immune cells become increasingly attractive as delivery system for immunotoxins in cancer therapy to reduce the intrinsic toxicity and severe side effects of chimeric protein toxins. In this study, we investigated the potential of human primary T cells to deliver a secreted immunotoxin through transient messenger RNA (mRNA) transfection. The chimeric protein toxin was directed toward the neovasculature of cancer cells by fusing a truncated version of Pseudomonas exotoxin A (PE38) to human vascular endothelial growth factor (VEGF) and to the single chain variable fragment (scFv) of anti-Her2/neu. Protocols for the transient transfection of human embryonic kidney cells (HEK293) as well as activated primary human T cells were established. Transient transfection with mRNA coding for the immunotoxins e23-PE38, VEGF-PE38 and its attenuated variant VEGF-PE38D yielded efficient expression and secretion. Mass spectrometry analysis endorsed that a fraction of VEGF-PE38D was properly translocated into the endoplasmic reticulum. Furthermore, cytotoxic activity of immunotoxin secreting T cells toward cancer cells was confirmed in co-culture with ovarian adenocarcinoma cells in the presence of a bispecific antibody (bsAb), highlighting the potential of primary T cells for mRNA-mediated immunotoxin delivery.
Collapse
Affiliation(s)
- R Eggers
- Environmental safety group, Korea Institute of Science and Technology (KIST Europe), Saarbrücken, Germany
| | - A Philippi
- Environmental safety group, Korea Institute of Science and Technology (KIST Europe), Saarbrücken, Germany
| | - M O Altmeyer
- Environmental safety group, Korea Institute of Science and Technology (KIST Europe), Saarbrücken, Germany
| | - F Breinig
- Department of Biosciences, Molecular and Cell Biology and Center of Human and Molecular Biology (ZHMB), Saarland University, Saarbrücken, Germany
| | - M J Schmitt
- Department of Biosciences, Molecular and Cell Biology and Center of Human and Molecular Biology (ZHMB), Saarland University, Saarbrücken, Germany
| |
Collapse
|
9
|
Brok MWJD, de Gast GC, Schellens JHM, Beijnen JH. Targeted toxins. J Oncol Pharm Pract 2016. [DOI: 10.1177/107815529900500401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective. Current modalities used in the treatment of cancer often cause unacceptable damage to normal tissue. Toxins targeted toward tumor cells by antibodies or growth factors have the potential to selectively kill tumor cells while leaving normal tissue intact. The purpose of this review is to provide background information on targeted toxins and current clinical studies for this new class of anti-cancer compounds. Data sources. A MEDLINE search was conducted using the term “immunotoxins.” Relevant articles were also obtained by the systematic examination of article references. Data synthesis. The toxins Pseudomonas exotoxin, diphtheria toxin, and ricin toxin are often used as targeted toxins. Deletion or mutation of the binding domains of these toxins decreased binding of the toxins to normal tissues. Antibodies or growth factors can be used as targeting moiety, and the resulting agents are called immunotoxins or fusion proteins, respectively. DNA technology and chemical modifications of the toxin as well as the antibody moiety led to smaller and less immunogenic targeted toxins. Smaller targeted toxins are less toxic and penetrate further into the tumor. The summary of several targeted toxins elicited during clinical trials in this review makes it clear that several targeted toxins are potential agents for the treatment of various cancers, although some problems still need to be overcome. These problems include toxicity, immunogenicity, cross-reactivity of the targeted toxin with life-sustaining tissue, heterogenicity of tumor cells, and limited tumor penetration.
Collapse
Affiliation(s)
- M W J den Brok
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute/Slotervaart Hospital, Amsterdam, The Netherlands
| | - G C de Gast
- Department of Medical Oncology, The Netherlands Cancer Institute/Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - J H M Schellens
- Department of Medical Oncology, The Netherlands Cancer Institute/Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands, Division of Drug Toxicology, Faculty of Pharmacy, Utrecht University, Utrecht, The Netherlands
| | - J H Beijnen
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute/Slotervaart Hospital, Amsterdam, The Netherlands, Department of Medical Oncology, The Netherlands Cancer Institute/Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands, Division of Drug Toxicology, Faculty of Pharmacy, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
10
|
Hassan R, Alewine C, Pastan I. New Life for Immunotoxin Cancer Therapy. Clin Cancer Res 2016; 22:1055-8. [PMID: 26463707 PMCID: PMC4775389 DOI: 10.1158/1078-0432.ccr-15-1623] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 09/30/2015] [Indexed: 01/16/2023]
Abstract
Immunotoxins are targeted anticancer therapeutics that kill cancer cells using a cytotoxic bacterial toxin payload. Their development for use in solid tumor malignancies was delayed due to issues with their immunogenicity and limited therapeutic window. However, new research has rejuvenated the field. Coadministration with a lymphocyte-depleting regimen of pentostatin and cyclophosphamide can delay antidrug antibody formation, increasing the number of treatment cycles that patients can receive and resulting in durable responses in heavily pretreated patients. In addition, a new generation of immunotoxin molecules with reduced immunogenicity and nonspecific toxicity has been developed through protein engineering techniques, and one has recently entered the clinic. In preclinical studies in mouse models, these new agents are effective against many tumor types as single agents, and also produce synergistic antitumor responses in combination with chemotherapy. These new immunotoxins have renewed excitement in the field and may prove a promising addition to the targeted therapy repertoire.
Collapse
Affiliation(s)
- Raffit Hassan
- Laboratory of Molecular Biology, National Cancer Institute, NIH, Bethesda, Maryland. Thoracic and GI Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland.
| | - Christine Alewine
- Laboratory of Molecular Biology, National Cancer Institute, NIH, Bethesda, Maryland
| | - Ira Pastan
- Laboratory of Molecular Biology, National Cancer Institute, NIH, Bethesda, Maryland.
| |
Collapse
|
11
|
Spiess K, Jakobsen MH, Kledal TN, Rosenkilde MM. The future of antiviral immunotoxins. J Leukoc Biol 2016; 99:911-25. [PMID: 26729815 DOI: 10.1189/jlb.2mr1015-468r] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 12/03/2015] [Indexed: 01/06/2023] Open
Abstract
There is a constant need for new therapeutic interventions in a wide range of infectious diseases. Over the past few years, the immunotoxins have entered the stage as promising antiviral treatments. Immunotoxins have been extensively explored in cancer treatment and have achieved FDA approval in several cases. Indeed, the design of new anticancer immunotoxins is a rapidly developing field. However, at present, several immunotoxins have been developed targeting a variety of different viruses with high specificity and efficacy. Rather than blocking a viral or cellular pathway needed for virus replication and dissemination, immunotoxins exert their effect by killing and eradicating the pool of infected cells. By targeting a virus-encoded target molecule, it is possible to obtain superior selectivity and drastically limit the side effects, which is an immunotoxin-related challenge that has hindered the success of immunotoxins in cancer treatment. Therefore, it seems beneficial to use immunotoxins for the treatment of virus infections. One recent example showed that targeting of virus-encoded 7 transmembrane (7TM) receptors by immunotoxins could be a future strategy for designing ultraspecific antiviral treatment, ensuring efficient internalization and hence efficient eradication of the pool of infected cells, both in vitro and in vivo. In this review, we provide an overview of the mechanisms of action of immunotoxins and highlight the advantages of immunotoxins as future anti-viral therapies.
Collapse
Affiliation(s)
- Katja Spiess
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Science, University of Copenhagen, Denmark; and
| | - Mette Høy Jakobsen
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Science, University of Copenhagen, Denmark; and
| | - Thomas N Kledal
- Section for Virology, Veterinary Institute, The Danish Technical University, Denmark
| | - Mette M Rosenkilde
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Science, University of Copenhagen, Denmark; and
| |
Collapse
|
12
|
Zdobnova T, Sokolova E, Stremovskiy O, Karpenko D, Telford W, Turchin I, Balalaeva I, Deyev S. A novel far-red fluorescent xenograft model of ovarian carcinoma for preclinical evaluation of HER2-targeted immunotoxins. Oncotarget 2015; 6:30919-28. [PMID: 26436696 PMCID: PMC4741577 DOI: 10.18632/oncotarget.5130] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 08/24/2015] [Indexed: 12/12/2022] Open
Abstract
We have created a novel fluorescent model of a human ovarian carcinoma xenograft overexpressing receptor HER2, a promising molecular target of solid tumors. The model is based on a newly generated SKOV-kat cell line stably expressing far-red fluorescent protein Katushka. Katushka is most suitable for the in vivo imaging due to an optimal combination of high brightness and emission in the "window of tissue transparency". The relevance of the fluorescent model for the in vivo monitoring of tumor growth and response to treatment was demonstrated using a newly created HER2-targeted recombinant immunotoxin based on the 4D5scFv antibody and a fragment of the Pseudomonas exotoxin A.
Collapse
Affiliation(s)
- Tatiana Zdobnova
- Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603950, Russia
| | - Evgeniya Sokolova
- Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603950, Russia
| | - Oleg Stremovskiy
- Institute of Bioorganic Chemistry of The Russian Academy of Sciences, Moscow 117997, Russia
| | - Dmitry Karpenko
- Institute of Bioorganic Chemistry of The Russian Academy of Sciences, Moscow 117997, Russia
| | - William Telford
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ilya Turchin
- Institute of Applied Physics of The Russian Academy of Sciences, Nizhny Novgorod, 603950 Russia
| | - Irina Balalaeva
- Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603950, Russia
| | - Sergey Deyev
- Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603950, Russia
- Institute of Bioorganic Chemistry of The Russian Academy of Sciences, Moscow 117997, Russia
| |
Collapse
|
13
|
Sokolova EA, Zdobnova TA, Stremovskiy OA, Balalaeva IV, Deyev SM. Novel recombinant anti-HER2/neu immunotoxin: design and antitumor efficiency. BIOCHEMISTRY. BIOKHIMIIA 2014; 79:1376-81. [PMID: 25716732 DOI: 10.1134/s0006297914120128] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The novel HER2/neu-specific recombinant immunotoxin 4D5scFv-PE40 consisting of 4D5scFv antibody (targeting module) and Pseudomonas exotoxin A fragment (effector module) combined in a single polypeptide chain via a flexible linker has been expressed and purified. This immunotoxin conserves specificity and affinity that are characteristics of the parental antibody 4D5scFv and exhibits selective and strong cytotoxic effect against cancer cells overexpressing HER2/neu receptor. The results of the experiments both in vitro (in cell cultures) and in vivo (in tumor-bearing animals) demonstrate high potential of 4D5scFv-PE40 for targeted therapy of tumors overexpressing HER2/neu.
Collapse
Affiliation(s)
- E A Sokolova
- Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, 603950, Russia.
| | | | | | | | | |
Collapse
|
14
|
HER2/neu: an increasingly important therapeutic target. Part 1: basic biology & therapeutic armamentarium. ACTA ACUST UNITED AC 2014. [DOI: 10.4155/cli.14.57] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
15
|
Lu Y, Ngo Ndjock Mbong G, Liu P, Chan C, Cai Z, Weinrich D, Boyle AJ, Reilly RM, Winnik MA. Synthesis of Polyglutamide-Based Metal-Chelating Polymers and Their Site-Specific Conjugation to Trastuzumab for Auger Electron Radioimmunotherapy. Biomacromolecules 2014; 15:2027-37. [DOI: 10.1021/bm500174p] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Yijie Lu
- Department
of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
| | - Ghislaine Ngo Ndjock Mbong
- Department
of Pharmaceutical Sciences, University of Toronto, 144 College
Street, Toronto, Ontario M5S 3M2, Canada
| | - Peng Liu
- Department
of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
| | - Conrad Chan
- Department
of Pharmaceutical Sciences, University of Toronto, 144 College
Street, Toronto, Ontario M5S 3M2, Canada
| | - Zhongli Cai
- Department
of Pharmaceutical Sciences, University of Toronto, 144 College
Street, Toronto, Ontario M5S 3M2, Canada
| | - Dirk Weinrich
- Department
of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
| | - Amanda J. Boyle
- Department
of Pharmaceutical Sciences, University of Toronto, 144 College
Street, Toronto, Ontario M5S 3M2, Canada
| | - Raymond M. Reilly
- Department
of Pharmaceutical Sciences, University of Toronto, 144 College
Street, Toronto, Ontario M5S 3M2, Canada
- Department
of Medical Imaging, University of Toronto, 263 McCaul Street, Toronto, Ontario M5T 1W7, Canada
- Toronto
General Research Institute, University Health Network, Toronto, Ontario M5G 2M9, Canada
| | - Mitchell A. Winnik
- Department
of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
| |
Collapse
|
16
|
Liu P, Cai Z, Kang JW, Boyle AJ, Adams J, Lu Y, Ngo Ndjock Mbong G, Sidhu S, Reilly RM, Winnik MA. Intracellular routing in breast cancer cells of streptavidin-conjugated trastuzumab Fab fragments linked to biotinylated doxorubicin-functionalized metal chelating polymers. Biomacromolecules 2014; 15:715-25. [PMID: 24506198 DOI: 10.1021/bm401483a] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
We describe the synthesis of a heterotelechelic metal-chelating polymer (Bi-MCP-Dox), a polyacrylamide with a number average degree of polymerization DPn = 50 (PDI = 1.2), with biotin (Bi) and doxorubicin (Dox) as functional chain ends and diethylenetriaminepentaacetic acid (DTPA) pendant groups as the binding sites for metal ions. We compared its behavior in cell-uptake experiments with a similar polymer (Bi-MCP) without Dox. These MCPs were complexed with trastuzumab Fab (tmFab) fragments covalently linked to streptavidin (SAv) to form tmFab-SAv-Bi-MCP-Dox and tmFab-SAv-Bi-MCP via the strong affinity between Bi and SAv. tmFab targets human epidermal growth factor receptor-2 (HER2), which is overexpressed on certain human breast cancer cells. Surface plasmon resonance (SPR) experiments with the extracellular domain (ECD) of HER2 showed that incorporation of the MCPs in these complexes had no significant effect on the association or dissociation rate with the HER2 ECD and the dissociation constants. The tmFab-complexed MCPs were subsequently labeled with (111)In (an Auger electron emitting radionuclide). Auger electrons can cause lethal DNA double strand breaks (DSBs) but only if they are emitted intracellularly and especially, in close proximity to the nucleus. To evaluate the cellular and nuclear uptake of tmFab-SAv-Bi-MCP-Dox, we incubated HER2+ SK-BR-3 human breast cancer cells with the complexes saturated with stable In(3+) and visualized their distribution by confocal fluorescence microscopy, monitoring the fluorescence of Dox. In parallel, we carried out cell fractionation studies on tmFab-SAv-Bi-MCP-Dox and on tmFab-SAv-Bi-MCP labeled with (111)In. Both radiolabeled complexes showed cell internalization and nuclear localization. We conclude that metal-chelating polymers with this composition appear to encourage internalization, nuclear uptake, and chromatin (DNA) binding of trastuzumab fragments modified with streptavidin in human breast cancer cells expressing HER2. Further study is needed to understand the impact of polymer charge on cellular uptake and distribution to intracellular compartments.
Collapse
Affiliation(s)
- Peng Liu
- Department of Chemistry, University of Toronto , 80 St. George Street, Toronto, Ontario, Canada , M5S 3H6
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
An update in the use of antibodies to treat glioblastoma multiforme. Autoimmune Dis 2013; 2013:716813. [PMID: 24294521 PMCID: PMC3835613 DOI: 10.1155/2013/716813] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 09/09/2013] [Indexed: 11/17/2022] Open
Abstract
Glioblastoma is a deadly brain disease and modest improvement in survival has been made. At initial diagnosis, treatment consists of maximum safe surgical resection, followed by temozolomide and chemoirradiation or adjuvant temozolomide alone. However, these treatments do not improve the prognosis and survival of patients. New treatment strategies are being sought according to the biology of tumors. The epidermal growth factor receptor has been considered as the hallmark in glioma tumors; thereby, some antibodies have been designed to bind to this receptor and block the downstream signaling pathways. Also, it is known that vascularization plays an important role in supplying new vessels to the tumor; therefore, new therapy has been guided to inhibit angiogenic growth factors in order to limit tumor growth. An innovative strategy in the treatment of glial tumors is the use of toxins produced by bacteria, which may be coupled to specific carrier-ligands and used for tumoral targeting. These carrier-ligands provide tumor-selective properties by the recognition of a cell-surface receptor on the tumor cells and promote their binding of the toxin-carrier complex prior to entry into the cell. Here, we reviewed some strategies to improve the management and treatment of glioblastoma and focused on the use of antibodies.
Collapse
|
18
|
Zhou BG, Liu MY, Qiu XC, Xu YM, Fan QY, Yang AG, Zhang Y, Xia H. A novel recombinant immunocasp-6 fusion gene specifically and efficiently suppresses HER2-overexpressing osteosarcoma. Oncol Rep 2013; 29:276-82. [PMID: 23135254 DOI: 10.3892/or.2012.2122] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 09/25/2012] [Indexed: 11/06/2022] Open
Abstract
Osteosarcoma is the most common primary malignant tumor of bone for adolescent or children. The poor prognosis of patients, due to its remote metastasis, has led to the exploration of more effective and less toxic treatments. Immunotherapy is a promising strategy for the treatment of human epidermal growth factor receptor 2 (HER2)-overexpressing tumors. Herein, we describe experiments conducted with a fusion gene, immunocasp-6, which was generated by fusing a HER2-specific single-chain Ab, a single-chain Pseudomonas exotoxin A and an active caspase-6 which can directly cleave lamin A leading to nucleus damage inducing programmed cell death. We demonstrated that immunocasp-6 can specifically and efficiently recognize and induce apoptosis in HER2-overexpressing osteosarcoma cells in vitro. The immunocasp-6 was transferred into BALB/c athymic mice bearing human osteosarcoma by i.m. injection of liposome-encapsulated pCMV-immunocap-6. Expression of immunocasp-6 not only strongly inhibited tumor growth and significantly prolonged animal survival, but also greatly prevented tumor metastasis. Our data showed that the immuno-casp-6 can specifically recognize HER2-overexpressing osteosarcoma cells, can also promptly attack their nucleus and induce apoptotic death, suggesting the potential of this strategy for the treatment of human HER2-overexpressing tumors.
Collapse
Affiliation(s)
- Ben-Gen Zhou
- Graduate School of Southern Medical University, Department of Orthopaedics, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, Guangdong, PR China
| | | | | | | | | | | | | | | |
Collapse
|
19
|
|
20
|
Li YM, Hall WA. Targeted toxins in brain tumor therapy. Toxins (Basel) 2010; 2:2645-62. [PMID: 22069569 PMCID: PMC3153175 DOI: 10.3390/toxins2112645] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Revised: 10/20/2010] [Accepted: 10/27/2010] [Indexed: 11/30/2022] Open
Abstract
Targeted toxins, also known as immunotoxins or cytotoxins, are recombinant molecules that specifically bind to cell surface receptors that are overexpressed in cancer and the toxin component kills the cell. These recombinant proteins consist of a specific antibody or ligand coupled to a protein toxin. The targeted toxins bind to a surface antigen or receptor overexpressed in tumors, such as the epidermal growth factor receptor or interleukin-13 receptor. The toxin part of the molecule in all clinically used toxins is modified from bacterial or plant toxins, fused to an antibody or carrier ligand. Targeted toxins are very effective against cancer cells resistant to radiation and chemotherapy. They are far more potent than any known chemotherapy drug. Targeted toxins have shown an acceptable profile of toxicity and safety in early clinical studies and have demonstrated evidence of a tumor response. Currently, clinical trials with some targeted toxins are complete and the final results are pending. This review summarizes the characteristics of targeted toxins and the key findings of the important clinical studies with targeted toxins in malignant brain tumor patients. Obstacles to successful treatment of malignant brain tumors include poor penetration into tumor masses, the immune response to the toxin component and cancer heterogeneity. Strategies to overcome these limitations are being pursued in the current generation of targeted toxins.
Collapse
Affiliation(s)
- Yan Michael Li
- Department of Neurosurgery, State University of New York Upstate Medical University, Syracuse, New York 13210, NY, USA.
| | | |
Collapse
|
21
|
Farzadnia M, Meibodi NT, Shandiz FH, Mahmoudi M, Bahar MM, Memar B, Amoian S, Maroozi F, Moheghi N. Evaluation of HER2/neu oncoprotein in serum and tissue samples of women with breast cancer: correlation with clinicopathological parameters. Breast 2010; 19:489-92. [PMID: 20675140 DOI: 10.1016/j.breast.2010.05.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2009] [Revised: 05/03/2010] [Accepted: 05/24/2010] [Indexed: 10/19/2022] Open
Abstract
BACKGROUND HER2/neu (HER2) is a proto-oncogen of the EGF Receptor family. The assessment of serum HER2 level is useful for predicting the patients' response to chemotherapy or hormonal therapy and selection of proper patients for treatment with Herceptin. We aimed to compare serum HER2 levels with immunohistochemistry in tumoral tissues and investigate correlation between these levels and various prognostic factors. MATERIALS AND METHODS This cross-sectional study was conducted on 75 patients with breast carcinoma referred to surgical ward of Mashhad Imam Reza's hospital from November 2008 to February 2009. Pre-operative serum samples were collected and stored in -20°C. Surgical samples were investigated for the type of carcinoma, tumor size, lymph node metastasis, stage as well as grade of the tumor. Tissue HER2 over-expression was evaluated by immunohistochemistry (IHC) staining and HER2 levels were studied by ELISA method. Statistical analysis was performed by SPSS software. RESULTS Serum HER2 cut-off level was 18.4 ng/ml; 46.7% of patients were serum HER2-positive and 43% were IHC positive. There was a high statistical correlation between these two parameters (P=0.018). Statistically, there was no significant correlation between serum HER2 and age, tumor size, stage, grade and metastatic lymph nodes (P>0.05). CONCLUSION Serum HER2 level assay can be considered as a complementary method besides tissue methods.
Collapse
Affiliation(s)
- Mehdi Farzadnia
- Department of Pathology, Faculty of Medicine, Emam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Wang F, Ren J, Qiu XC, Wang LF, Zhu Q, Zhang YQ, Huan Y, Meng YL, Yao LB, Chen SY, Xu YM, Yang AG. Selective Cytotoxicity to HER2-Positive Tumor Cells by a Recombinant e23sFv-TD-tBID Protein Containing a Furin Cleavage Sequence. Clin Cancer Res 2010; 16:2284-94. [DOI: 10.1158/1078-0432.ccr-09-2367] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
23
|
Colombo M, Corsi F, Foschi D, Mazzantini E, Mazzucchelli S, Morasso C, Occhipinti E, Polito L, Prosperi D, Ronchi S, Verderio P. HER2 targeting as a two-sided strategy for breast cancer diagnosis and treatment: Outlook and recent implications in nanomedical approaches. Pharmacol Res 2010; 62:150-65. [PMID: 20117211 DOI: 10.1016/j.phrs.2010.01.013] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2009] [Revised: 01/19/2010] [Accepted: 01/19/2010] [Indexed: 02/06/2023]
Abstract
At present, mammary carcinoma is the second most common type of malignant tumor in adult women after lung cancer, as more than one million women are diagnosed with breast cancer every year. Despite advances in diagnosis and treatment, which have resulted in a decrease in mortality in recent decades, breast cancer remains a major public health problem. One of the most significant unresolved clinical and scientific problems is the occurrence of resistance to clinical treatments and their toxicity (and how to predict, prevent and overcome them). However, the heterogeneity of human breast cancer in terms of genetic features, molecular profiles and clinical behavior represents a constraint obstructing the discovery of a solution to the disease. It is currently considered that the chances of success of therapy may increase if the tumor cells are selectively removed before they can evolve to their mature stages up to metastases production. Therefore, novel and more sensitive diagnostic tools are being developed, with the aim of improving the early and noninvasive detection of rising malignancies and the accuracy of tumor tissue localization. Meanwhile, there is an emerging use of targeted therapies in oncology, depending on the expression of specific proteins or genes present in tumor cells. Among the molecular targets considered for the treatment of breast cancer cells so far, we chose to focus on examples involving overexpression and/or gene amplification of "Human Epidermal growth factor Receptor 2" (HER2) protein. In current studies, various types of nanoparticles conjugated with the anti-HER2 monoclonal antibody, the so-called "trastuzumab", are investigated extensively due to promising results in biological and preclinical applications aimed at improving the treatment of breast cancer. In this paper, we present a critical review of the preparation and use of different kinds of trastuzumab-functionalized nanoparticles, with an emphasis on the therapeutic and diagnostic (theranostic) potential of this generation of hybrid nanoparticles, exploiting the multifaceted mechanisms of action of trastuzumab against malignant cells.
Collapse
Affiliation(s)
- Miriam Colombo
- Dipartimento di Biotecnologie e Bioscienze, Università di Milano-Bicocca, 20126 Milano, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Zielinski R, Lyakhov I, Jacobs A, Chertov O, Kramer-Marek G, Francella N, Stephen A, Fisher R, Blumenthal R, Capala J. Affitoxin--a novel recombinant, HER2-specific, anticancer agent for targeted therapy of HER2-positive tumors. J Immunother 2009; 32:817-25. [PMID: 19752752 PMCID: PMC3402039 DOI: 10.1097/cji.0b013e3181ad4d5d] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Expression of the human epidermal growth factor receptor 2 (HER2) is amplified in 25% to 30% of breast cancers and has been associated with an unfavorable prognosis. Here we report the construction, purification, and characterization of Affitoxin-a novel class of HER2-specific cytotoxic molecules combining HER2-specific Affibody molecule as a targeting moiety and PE38KDEL, which is a truncated version of Pseudomonas exotoxin A, as a cell killing agent. It is highly soluble and does not require additional refolding, oxidation, or reduction steps during its purification. Using surface plasmon resonance technology and competitive binding assays, we have shown that Affitoxin binds specifically to HER2 with nanomolar affinity. We have also observed a high correlation between HER2 expression and retention of Affitoxin bound to the cell surface. Affitoxin binding and internalization is followed by Pseudomonas exotoxin A activity domain-mediated ADP-ribosylation of translation elongation factor 2 and, consequently, inhibition of protein synthesis as shown by protein expression analysis of HER2-positive cells treated with Affitoxin. Measured IC50 value for HER2-negative cells MDA-MB468 (65+/-2.63 pM) was more than 20 times higher than the value for low HER2 level-expressing MCF7 cells (2.56+/-0.1 pM), and almost 3 orders of magnitude higher for its HER2-overexpressing derivative MCF7/HER2 (62.7+/-5.9 fM). These studies suggest that Affitoxin is an attractive PE38-based candidate for treatment of HER2-positive tumors.
Collapse
Affiliation(s)
- Rafal Zielinski
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Ilya Lyakhov
- SAIC-Frederick, Inc., NCI-Frederick, Frederick, MD
| | - Amy Jacobs
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Oleg Chertov
- SAIC-Frederick, Inc., NCI-Frederick, Frederick, MD
| | - Gabriela Kramer-Marek
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Nicholas Francella
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | | | | | - Robert Blumenthal
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jacek Capala
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
25
|
Vallera DA, Chen H, Sicheneder AR, Panoskaltsis-Mortari A, Taras EP. Genetic alteration of a bispecific ligand-directed toxin targeting human CD19 and CD22 receptors resulting in improved efficacy against systemic B cell malignancy. Leuk Res 2009; 33:1233-42. [PMID: 19327829 DOI: 10.1016/j.leukres.2009.02.006] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2008] [Revised: 02/03/2009] [Accepted: 02/05/2009] [Indexed: 12/28/2022]
Abstract
A bispecific ligand-directed toxin (BLT) called DT2219ARL consisting of two scFv ligands recognizing CD19 and CD22 and catalytic DT390 was genetically enhanced for superior in vivo anti-leukemia activity. Genetic alterations included reverse orienting VH-VL domains and adding aggregation reducing/stabilizing linkers. In vivo, these improvements resulted in previously unseen long-term tumor-free survivors measured in a bioluminescent xenograft imaging model in which the progression of human Raji Burkitt's lymphoma could be tracked in real time and in a Daudi model as well. Studies showed DT2219ARL was potent (IC50s 0.06-0.2 nM range) and selectively blockable. Imaging studies indicated the highly invasive nature of this B cell malignancy model and showed it likely induced pre-terminal hind limb paralysis because of metastasis to spinal regions prevented by DT2219ARL. DT2219ARL represents a new class of bispecific biological that can be continually improved by genetic mutation.
Collapse
Affiliation(s)
- Daniel A Vallera
- University of Minnesota Cancer Center, Section on Molecular Cancer Therapeutics, Department of Therapeutic Radiology-Radiation Oncology, Minneapolis, MN 55455, USA.
| | | | | | | | | |
Collapse
|
26
|
Qiu XC, Xu YM, Wang F, Fan QY, Wang LF, Ma BA, Jia LT, Zhao J, Meng YL, Yao LB, Chen SY, Yang AG. Single-chain antibody/activated BID chimeric protein effectively suppresses HER2-positive tumor growth. Mol Cancer Ther 2008; 7:1890-9. [PMID: 18644999 DOI: 10.1158/1535-7163.mct-07-2235] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BH3-interacting domain death agonist (BID) is a crucial element in death signaling pathways and is recognized as an intracellular link connecting the intrinsic mitochondrial apoptotic and extrinsic death receptor-mediated apoptotic pathways. Herein, we describe experiments conducted with a fusion protein, which was generated by fusing a human epidermal growth factor receptor-2 (HER2)-specific single-chain antibody with domain II of Pseudomonas exotoxin A and the truncated active BID (tBID). These experiments extend our previous work on several other immuno-proapoptotic proteins. Specifically, by excluding cells with undetectable HER2, we showed that the secreted immuno-tBID molecule selectively recognized and killed HER2-overexpressing tumor cells in vitro by attacking their mitochondria and inducing their apoptotic death. This apoptosis could only be inhibited partially by caspase pan-inhibitor zVAD and mitochondrial protector TAT-BH4. Subsequently, we transferred the immuno-tbid gene into BALB/c athymic mice bearing HER2-positive tumors together with other immuno-proapoptotic proteins using i.m. injections of liposome-encapsulated vectors. The expression of the immuno-tbid gene suppressed tumor growth and prolonged animal survival significantly. We also shortened the translocation domain of Pseudomonas exotoxin A II to only 10-amino acid sequence, which were crucial for furin cleavage. The new recombinant molecule retained the translocation efficiency and the ability of specific killing HER2-positive tumor cells. Our data showed that, compared with the toxins employed before, the chimeric immuno-tBID molecule can not only specifically recognize HER2-positive tumor cells but also certainly induce apoptosis even in the presence of zVAD and TAT-BH4, thereby suggesting an alternative approach to treating HER2/neu-positive tumors.
Collapse
Affiliation(s)
- Xiu-Chun Qiu
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Xi'an, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
|
28
|
Wang T, Zhao J, Ren JL, Zhang L, Wen WH, Zhang R, Qin WW, Jia LT, Yao LB, Zhang YQ, Chen SY, Yang AG. Recombinant Immunoproapoptotic Proteins with Furin Site Can Translocate and Kill HER2-Positive Cancer Cells. Cancer Res 2007; 67:11830-9. [DOI: 10.1158/0008-5472.can-07-1160] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
29
|
Stish BJ, Chen H, Shu Y, Panoskaltsis-Mortari A, Vallera DA. Increasing anticarcinoma activity of an anti-erbB2 recombinant immunotoxin by the addition of an anti-EpCAM sFv. Clin Cancer Res 2007; 13:3058-67. [PMID: 17505009 DOI: 10.1158/1078-0432.ccr-06-2454] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE erbB2, the product of the Her2-neu gene, is a well-established therapeutic target for antibody-based biologicals, but anti-erbB2 antibody-toxin fusion proteins are limited in their activity. The goal of this study was to determine if genetically adding an sFv targeting epithelial cell adhesion molecule (EpCAM) to an anti-Her2 sFv immunotoxin would result in enhanced antitumor activity. EXPERIMENTAL DESIGN In vitro studies were done in which the new bispecific immunotoxin DTEpCAM23 was compared with monospecific immunotoxins (DTEpCAM and DT23) to quantitate immunotoxin activity. Mixtures of monospecific immunotoxins were tested to determine if they were as effective as the bispecific immunotoxin. Binding and internalization studies were also done. In vivo, bispecific immunotoxins were given i.t. to athymic nude mice bearing HT-29 human colon cancer flank tumors and i.p. to mice with i.p. tumors. RESULTS DTEpCAM23 bispecific immunotoxins showed far greater activity than monospecific immunotoxin (sometimes over 2,000-fold) against most tumor lines. Bispecific immunotoxin was superior and selective in its activity against different carcinoma cell lines. Bispecific immunotoxin had greater activity than monospecific immunotoxin indicating an advantage of having both sFv on the same single-chain molecule. Binding and internalization studies did not explain the differences between bispecific immunotoxin and monospecific immunotoxin activity. Orientation of the sFvs on the molecule had a significant effect on in vitro and in vivo properties. The bispecific immunotoxins were more effective than the monospecific immunotoxin in the flank tumor mouse model. CONCLUSIONS The synthesis of bispecific immunotoxin created a new biological agent with superior in vitro and in vivo activity (over monospecific immunotoxin), more broad reactivity, more efficacy against tumors in vivo, and diminished toxic effects in mice.
Collapse
Affiliation(s)
- Brad J Stish
- Department of Therapeutic Radiology-Radiation Oncology, University of Minnesota Cancer Center, Minneapolis, Minnesota 55455, USA
| | | | | | | | | |
Collapse
|
30
|
Valabrega G, Montemurro F, Aglietta M. Trastuzumab: mechanism of action, resistance and future perspectives in HER2-overexpressing breast cancer. Ann Oncol 2007; 18:977-84. [PMID: 17229773 DOI: 10.1093/annonc/mdl475] [Citation(s) in RCA: 431] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Trastuzumab is a humanized mAb directed against the extracellular domain of the tyrosine kinase receptor HER2. Trastuzumab has shown clinical activity in HER2-overexpressing breast cancers and, at present, is currently approved for patients whose tumours have this abnormality, in both the metastatic and the adjuvant setting. Several issues about its optimal use, however, are still unresolved. One of the reasons for these uncertainties lies in the absence of conclusive data about its mechanism of action and possible primary or acquired resistance mechanisms. Therefore, clinical questions such as how to optimize patient selection, how to prevent resistance to trastuzumab, or what is the optimal management of those patients whose tumours progress during treatment still await convincing answers. This review summarises the current knowledge on the preclinical and clinical evidence about the mechanism of action of trastuzumab and on the mechanisms underlying the development of resistance and also briefly discusses their possible clinical implications.
Collapse
Affiliation(s)
- G Valabrega
- University Division of Medical Oncology and Haematology, Institute for Cancer Research and Treatment, Strada Provinciale 142, 10060 Candiolo, Torino, Italy.
| | | | | |
Collapse
|
31
|
Nahta R, Yu D, Hung MC, Hortobagyi GN, Esteva FJ. Mechanisms of disease: understanding resistance to HER2-targeted therapy in human breast cancer. ACTA ACUST UNITED AC 2006; 3:269-80. [PMID: 16683005 DOI: 10.1038/ncponc0509] [Citation(s) in RCA: 622] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2005] [Accepted: 02/07/2006] [Indexed: 12/13/2022]
Abstract
Trastuzumab is a monoclonal antibody targeted against the human epidermal growth factor receptor (HER) 2 tyrosine kinase receptor, which is overexpressed in approximately 25% of invasive breast cancers. The majority of patients with metastatic breast cancer who initially respond to trastuzumab, however, demonstrate disease progression within 1 year of treatment initiation. Preclinical studies have indicated several molecular mechanisms that could contribute to the development of trastuzumab resistance. Increased signaling via the phosphatidylinositol 3-kinase/Akt pathway could contribute to trastuzumab resistance because of activation of multiple receptor pathways that include HER2-related receptors or non-HER receptors such as the insulin-like growth factor 1 receptor, which appears to be involved in a cross-talk with HER2 in resistant cells. Additionally, loss of function of the tumor suppressor PTEN gene, the negative regulator of Akt, results in heightened Akt signaling that leads to decreased sensitivity to trastuzumab. Decreased interaction between trastuzumab and its target receptor HER2, which is due to steric hindrance of HER2 by cell surface proteins such as mucin-4 (MUC4), may block the inhibitory actions of trastuzumab. Novel therapies targeted against these aberrant molecular pathways offer hope that the effectiveness and duration of response to trastuzumab can be greatly improved.
Collapse
Affiliation(s)
- Rita Nahta
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston 77030-4009, USA.
| | | | | | | | | |
Collapse
|
32
|
Abstract
Immunotoxins are proteins that contain a toxin along with an antibody or growth factor that binds specifically to target cells. Nearly all protein toxins work by enzymatically inhibiting protein synthesis. For the immunotoxin to work, it must bind to and be internalized by the target cells, and the enzymatic fragment of the toxin must translocate to the cytosol. Once in the cytosol, 1 molecule is capable of killing a cell, making immunotoxins some of the most potent killing agents. Various plant and bacterial toxins have been genetically fused or chemically conjugated to ligands that bind to cancer cells. Among the most active clinically are those that bind to hematologic tumors. At present, only 1 agent, which contains human interleukin-2 and truncated diphtheria toxin, is approved for use in cutaneous T-cell lymphoma. Another, containing an anti-CD22 Fv and truncated Pseudomonas exotoxin, has induced complete remissions in a high proportion of cases of hairy-cell leukemia. Refinement of existing immunotoxins and development of new immunotoxins are underway to improve the treatment of cancer.
Collapse
Affiliation(s)
- Robert J Kreitman
- Clinical Immunotherapy Section, Laboratory of Molecular Biology, Centers for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 37, Room 5124B, Bethesda, MD 20892-4255, USA.
| |
Collapse
|
33
|
Nahta R, Esteva FJ. Herceptin: mechanisms of action and resistance. Cancer Lett 2006; 232:123-38. [PMID: 16458110 DOI: 10.1016/j.canlet.2005.01.041] [Citation(s) in RCA: 317] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2005] [Accepted: 01/30/2005] [Indexed: 11/28/2022]
Abstract
HER-2 is overexpressed in 20-25% of invasive breast cancers and is associated with an aggressive tumor phenotype and reduced survival rate. The HER-2 status of a tumor is the critical determinant of response to the HER-2-targeted antibody Herceptin. Thus, accurate assessment of HER-2 expression levels is essential for identifying breast cancer patients who will benefit from HER-2-targeted therapy. Herceptin combined with chemotherapy increases response rates, time to disease progression, and survival. However, the majority of cancers that initially respond to Herceptin begin to progress again within 1 year. This review describes mechanisms by which Herceptin inhibits cell growth in breast cancers that overexpress HER-2 and highlights possible mechanisms contributing to Herceptin resistance.
Collapse
Affiliation(s)
- Rita Nahta
- Department of Breast Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030-4009, USA
| | | |
Collapse
|
34
|
Heng CK, Othman RY. Bioinformatics in molecular immunology laboratories demonstrated: Modeling an anti-CMV scFv antibody. Bioinformation 2006; 1:118-20. [PMID: 17597869 PMCID: PMC1891672 DOI: 10.6026/97320630001118] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2006] [Accepted: 03/20/2006] [Indexed: 11/23/2022] Open
Abstract
A scFv (single chain variable fragment) antibody clone from anti-CMV (anti-cucumber mosaic virus) was successfully constructed from immunized mouse and the DNA sequence was submitted to GenBank (AY337618 and AY337619). The expression of a 32 kDa recombinant antibody in bacteria was verified using ELISA (enzyme-linked immunoassay) and western blot. However, elucidation of specific anti-CMV scFv function requires detailed and time consuming immuno-assays. Alternatively, useful functional information on anti-CMV scFV antibody can be obtained using available Bioinformatics tools and techniques without performing tedious assays. Here, we use the commonly used Bioinformatics tools and databases such as BLAST (basic local alignment search tool), GenBank, PDB (protein databank), KABAT numbering, SWISS-MODEL and Insight II to gain specific functional insights into anti-CMV scFv.
Collapse
Affiliation(s)
- Chua Kek Heng
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Malaysia
| | - Rofina Yasmin Othman
- Institute of Biological Sciences, Faculty of science, University of Malaya, Malaysia
| |
Collapse
|
35
|
|
36
|
Yu CJ, Jia LT, Meng YL, Zhao J, Zhang Y, Qiu XC, Xu YM, Wen WH, Yao LB, Fan DM, Jin BQ, Chen SY, Yang AG. Selective proapoptotic activity of a secreted recombinant antibody/AIF fusion protein in carcinomas overexpressing HER2. Gene Ther 2006; 13:313-20. [PMID: 16267568 DOI: 10.1038/sj.gt.3302672] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2005] [Revised: 07/19/2005] [Accepted: 08/25/2005] [Indexed: 11/09/2022]
Abstract
Apoptosis-inducing factor (AIF) represents a caspase-independent apoptotic pathway in the cell, and a mitochondrial localization sequence-truncated AIF (AIFDelta1-120) can be relocated from the cytoplasm to the nucleus and exhibit a constitutive proapoptotic activity. Here, we generated a chimeric immuno-AIF protein, which comprised an HER2 antibody, a Pseudomonas exotoxin translocation domain and AIFDelta1-120. Human Jurkat cells transfected with the immuno-AIF gene could express and secrete the chimeric protein, which selectively recognized HER2-overexpressing tumor cells and was endocytosed. Subsequent cleavage of truncated AIF from immuno-AIF and its release from the internalized vesicles resulted in apoptosis of tumor cells. Intramuscular injection of the immuno-AIF gene caused significant suppression of tumors and substantially prolonged mice survival in an HER2-overexpressing xenograft tumor model. Our study demonstrates the feasibility of the immuno-AIF gene as a novel approach to treating cancers that overexpress HER2.
Collapse
Affiliation(s)
- C-J Yu
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Vallera DA, Todhunter DA, Kuroki DW, Shu Y, Sicheneder A, Chen H. A bispecific recombinant immunotoxin, DT2219, targeting human CD19 and CD22 receptors in a mouse xenograft model of B-cell leukemia/lymphoma. Clin Cancer Res 2005; 11:3879-88. [PMID: 15897589 DOI: 10.1158/1078-0432.ccr-04-2290] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A novel bispecific single-chain fusion protein, DT2219, was assembled consisting of the catalytic and translocation domains of diphtheria toxin (DT(390)) fused to two repeating sFv subunits recognizing CD19 and CD22 and expressed in Escherichia coli. Problems with yield, purity, and aggregation in the refolding step were solved by incorporating a segment of human muscle aldolase and by using a sodium N-lauroyl-sarcosine detergent-based refolding procedure. Problems with reduced efficacy were addressed by combining the anti-CD19 and anti-CD22 on the same single-chain molecule. DT2219 had greater anticancer activity than monomeric or bivalent immunotoxins made with anti-CD19 and anti-CD22 sFv alone and it showed a higher level of binding to patient leukemia cells and to CD19(+)CD22(+) Daudi or Raji cells than did anti-CD19 and anti-CD22 parental monoclonal antibodies. The resulting DT2219, mutated to enhance its avidity, was cytotoxic to Daudi cells in vitro (IC(50) = 0.3 nmol/L). In vivo, DT2219 was effective in a flank tumor therapy model in which it significantly inhibited tumor growth (P < 0.05) and in a systemic model in which it significantly prolonged survival of severe combined immunodeficient mice with established Daudi (P < 0.008) compared with controls. DT2219 has broader reactivity in recognizing B-cell malignancies, has more killing power, and requires less toxin than using individual immunotoxin, which warrants further investigation as a new drug for treating B leukemia/lymphoma.
Collapse
MESH Headings
- Animals
- Antibodies
- Antigens, CD/immunology
- Antigens, CD19/immunology
- Antigens, Differentiation, B-Lymphocyte/immunology
- Cell Adhesion Molecules/immunology
- Cell Death
- Diphtheria Toxin/immunology
- Escherichia coli/genetics
- Escherichia coli/immunology
- Female
- Immunotoxins/immunology
- Immunotoxins/pharmacology
- Lectins/immunology
- Leukemia, B-Cell
- Lymphoma, B-Cell
- Mice
- Mice, Nude
- Mice, SCID
- Molecular Conformation
- Sialic Acid Binding Ig-like Lectin 2
- Survival Analysis
- Transplantation, Heterologous
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Daniel A Vallera
- Department of Therapeutic Radiology-Radiation Oncology, Section on Molecular Cancer Therapeutics, University of Minnesota Cancer Center, Minneapolis, Minnesota, USA.
| | | | | | | | | | | |
Collapse
|
38
|
Xia YJ, Wen WH, Huang WQ, Huang BC. Development of a phage displayed disulfide-stabilized Fv fragment vaccine against Vibrio anguillarum. Vaccine 2005; 23:3174-80. [PMID: 15837217 DOI: 10.1016/j.vaccine.2004.12.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2004] [Accepted: 12/15/2004] [Indexed: 11/22/2022]
Abstract
Anti-idiotype monoclonal antibody 1E10 can mimic the protective epitope of Vibrio anguillarum and be used as vaccine to prevent fish infection of V. anguillarum. In this study, the variable heavy (V(H)) domain and variable light (V(L)) domain of mAb1E10 were cloned by RT-PCR and were linked to each other by a disulfide bond engineered at position 44 of V(H) and position 105 of V(L) that lie between structurally conserved framework positions. Mutated V(H) 44 and V(L) 105 were inserted into phagemid pCANTAB5E. When co-transfected by recombinant pCANTAB5E and helper phage M13KO7, the host Escherichia coli cells secreted disulfide-stabilized Fv fragment (dsFv) which displayed on the surface of filamentous phage. The binding specificity of the phage-displayed dsFv was proved by ELISA method. Protection experiment showed that Japanese flounders can develop high titer of antibody against the dsFv and survival ratio of vaccinated group was significantly different from control groups. Thus, this phage-displayed dsFv may be used as vaccine against V. anguillarum in fishery.
Collapse
Affiliation(s)
- Yong-juan Xia
- Department of Histology and Embryology, Fourth Military Medical University, Xi'an 710032, China
| | | | | | | |
Collapse
|
39
|
Abstract
The c-erbB-2 proto-oncogene encodes a 185 kDa transmembrane Type 1 tyrosine kinase receptor whose amplification and/or overexpression has been linked with poor prognosis in a variety of cancers. The oncoprotein has been suggested to play a key role in tumour cell invasion, motility and metastasis, and in responsiveness to therapeutic agents. Over-expression of c-erbB-2 therefore identifies an important subset of patients with a high probability of relapse, but low probability of response to certain conventional therapies. The cell surface location of the oncoprotein, its stability of expression and low levels in normal adult tissues render it an attractive target for immunotherapeutic intervention. Although a 'self' antigen, there is evidence that c-erbB-2 p185 can induce both humoral and cell-mediated immune responses in cancer patients. Approaches to exploit p185 as an immunotherapeutic target include vaccination with peptides, plasmid DNA or vectors (viruses/bacteria) carrying the gene; with cytokines, co-stimulatory factors and superantigens being evaluated as adjuvants. Many monoclonal antibody (mAb)-based strategies are also in clinical development. Monoclonal antibodies can serve multiple functions; direct inhibition of c-erbB-2 activity, recruitment of host effector mechanisms and direct or indirect delivery of toxic payloads. Clinical trials in patients with late stage disease have shown that many of these approaches are safe, feasible and relatively non-toxic, and, in some cases, objective responses have been seen. As with all immunotherapy, the greatest benefit is likely to be obtained in patients with minimal residual disease in an adjuvant setting; such studies are awaited with interest.
Collapse
Affiliation(s)
- S Eccles
- Section of Cancer Therapeutics, McElwain Laboratories, Institute of Cancer Research, Cotswold Rd., Belmont, Sutton, Surrey, SM2 5NG, UK
| |
Collapse
|
40
|
Kreitman RJ. Recombinant toxins in haematologic malignancies and solid tumours. Expert Opin Investig Drugs 2005; 7:1405-27. [PMID: 15992040 DOI: 10.1517/13543784.7.9.1405] [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
Recombinant toxins constitute a new modality for the treatment of cancer, since they target cells displaying specific surface-receptors or antigens. They are fusion proteins, which contain toxin and ligand regions, and are produced in Escherichia coli. The ligand may be a growth factor or a fragment of an antibody, and the toxin is usually one of the two bacterial toxins: Pseudomonas exotoxin and diphtheria toxin. Compared to the earlier generation chemical conjugates of ligands and toxins, recombinant toxins have many advantages, including homogeneity with respect to the connection between the ligand and toxin, ease and yield of production and small size. A variety of chemotherapy-resistant haematologic and solid tumours have been targeted with recombinant toxins, and clinical trials with many of them have recently demonstrated their effectiveness. Moreover, their unwanted toxic effects are different from those of most chemotherapeutic agents, supporting the expectation that they can be combined with existing modalities to improve the clinical resources available to treat cancer in humans.
Collapse
Affiliation(s)
- R J Kreitman
- Division of Cancer Biology, National Cancer Institute, National Institutes of Health, 37/4B27, 37 Convent Drive, MSC 4255, Bethesda, MD 20892, USA
| |
Collapse
|
41
|
von Minckwitz G, Harder S, Hövelmann S, Jäger E, Al-Batran SE, Loibl S, Atmaca A, Cimpoiasu C, Neumann A, Abera A, Knuth A, Kaufmann M, Jäger D, Maurer AB, Wels WS. Phase I clinical study of the recombinant antibody toxin scFv(FRP5)-ETA specific for the ErbB2/HER2 receptor in patients with advanced solid malignomas. Breast Cancer Res 2005; 7:R617-26. [PMID: 16168106 PMCID: PMC1242130 DOI: 10.1186/bcr1264] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2005] [Revised: 04/06/2005] [Accepted: 05/04/2005] [Indexed: 11/25/2022] Open
Abstract
INTRODUCTION ScFv(FRP5)-ETA is a recombinant antibody toxin with binding specificity for ErbB2 (HER2). It consists of an N-terminal single-chain antibody fragment (scFv), genetically linked to truncated Pseudomonas exotoxin A (ETA). Potent antitumoral activity of scFv(FRP5)-ETA against ErbB2-overexpressing tumor cells was previously demonstrated in vitro and in animal models. Here we report the first systemic application of scFv(FRP5)-ETA in human cancer patients. METHODS We have performed a phase I dose-finding study, with the objective to assess the maximum tolerated dose and the dose-limiting toxicity of intravenously injected scFv(FRP5)-ETA. Eighteen patients suffering from ErbB2-expressing metastatic breast cancers, prostate cancers, head and neck cancer, non small cell lung cancer, or transitional cell carcinoma were treated. Dose levels of 2, 4, 10, 12.5, and 20 microg/kg scFv(FRP5)-ETA were administered as five daily infusions each for two consecutive weeks. RESULTS No hematologic, renal, and/or cardiovascular toxicities were noted in any of the patients treated. However, transient elevation of liver enzymes was observed, and considered dose limiting, in one of six patients at the maximum tolerated dose of 12.5 microg/kg, and in two of three patients at 20 microg/kg. Fifteen minutes after injection, peak concentrations of more than 100 ng/ml scFv(FRP5)-ETA were obtained at a dose of 10 microg/kg, indicating that predicted therapeutic levels of the recombinant protein can be applied without inducing toxic side effects. Induction of antibodies against scFv(FRP5)-ETA was observed 8 days after initiation of therapy in 13 patients investigated, but only in five of these patients could neutralizing activity be detected. Two patients showed stable disease and in three patients clinical signs of activity in terms of signs and symptoms were observed (all treated at doses > or = 10 microg/kg). Disease progression occurred in 11 of the patients. CONCLUSION Our results demonstrate that systemic therapy with scFv(FRP5)-ETA can be safely administered up to a maximum tolerated dose of 12.5 microg/kg in patients with ErbB2-expressing tumors, justifying further clinical development.
Collapse
Affiliation(s)
| | - Sebastian Harder
- Institute for Clinical Pharmacology, University Hospital Frankfurt, Germany
| | | | - Elke Jäger
- Medizinische Klinik II, Hämatologie-Onkologie, Krankenhaus Nordwest, Frankfurt, Germany
| | - Salah-Eddin Al-Batran
- Medizinische Klinik II, Hämatologie-Onkologie, Krankenhaus Nordwest, Frankfurt, Germany
| | - Sibylle Loibl
- Department of Gynecology and Obstetrics, University Hospital Frankfurt, Germany
| | - Akin Atmaca
- Medizinische Klinik II, Hämatologie-Onkologie, Krankenhaus Nordwest, Frankfurt, Germany
| | - Christian Cimpoiasu
- Department of Gynecology and Obstetrics, University Hospital Frankfurt, Germany
| | - Antje Neumann
- Medizinische Klinik II, Hämatologie-Onkologie, Krankenhaus Nordwest, Frankfurt, Germany
| | | | - Alexander Knuth
- Medizinische Klinik II, Hämatologie-Onkologie, Krankenhaus Nordwest, Frankfurt, Germany
- Department of Oncology, University Hospital Zürich, Switzerland
| | - Manfred Kaufmann
- Department of Gynecology and Obstetrics, University Hospital Frankfurt, Germany
| | - Dirk Jäger
- Medizinische Klinik II, Hämatologie-Onkologie, Krankenhaus Nordwest, Frankfurt, Germany
- Department of Oncology, University Hospital Zürich, Switzerland
| | | | - Winfried S Wels
- Chemotherapeutisches Forschungsinstitut Georg-Speyer-Haus, Frankfurt, Germany
| |
Collapse
|
42
|
Vallera DA, Todhunter D, Kuroki DW, Shu Y, Sicheneder A, Panoskaltsis-Mortari A, Vallera VD, Chen H. Molecular modification of a recombinant, bivalent anti-human CD3 immunotoxin (Bic3) results in reduced in vivo toxicity in mice. Leuk Res 2005; 29:331-41. [PMID: 15661270 DOI: 10.1016/j.leukres.2004.08.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2004] [Accepted: 08/17/2004] [Indexed: 10/26/2022]
Abstract
A novel bivalent single chain fusion protein, Bic3, was assembled consisting of the catalytic and translocation domains of diphtheria toxin (DT(390)) fused to two repeating sFv molecules recognizing human CD3 epsilon of the human T-cell receptor. Historically, problems with these constructs include low yield, toxicity, and reduced efficacy. Instead of using conventional Gly(4)Ser linkers to connect heavy/light chains, aggregation reducing linkers (ARL) were used which when combined with a new SLS-based refolding method reduced aggregation and enhanced the yield of final product. Toxicity was reduced at least 25-fold by repeating the two sFv molecules and adding a portion of the hinge-CH2-CH3 human constant regions. The resulting Bic3 was just as cytotoxic to HPB-MLT.UM T leukemia cells in vitro (IC(50)=4 pmol) as a monovalent construct made with the same DT and sFv. In vivo, Bic3 was effective in a new and aggressive therapy model in which it significantly prolonged survival of scid mice with established human T-cell leukemia (p<0.0001 compared to controls). Importantly, no toxicity measured by weight loss, enzyme function, or histology was observed at the highest dose of Bic3 tested (2000 ug/kg). Bic3 warrants investigation as a new drug for treating T-cell malignancy and other T-cell related disorders.
Collapse
Affiliation(s)
- Daniel A Vallera
- Department of Therapeutic Radiology-Radiation Oncology, University of Minnesota Cancer Center, Section on Molecular Cancer Therapeutics, MMC: 367, Minneapolis, MN 55455, USA.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Joshi BH, Puri RK. Optimization of expression and purification of two biologically active chimeric fusion proteins that consist of human interleukin-13 and Pseudomonas exotoxin in Escherichia coli. Protein Expr Purif 2005; 39:189-98. [PMID: 15642470 DOI: 10.1016/j.pep.2004.10.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2004] [Revised: 10/20/2004] [Indexed: 10/26/2022]
Abstract
We have previously reported that a variety of solid human tumor cell lines express a large number of receptors for interleukin-13 (IL-13). These receptors could be targeted with a chimeric fusion protein consisting of human IL-13 and a truncated form of Pseudomonas exotoxin (PE). We describe here optimization of critical steps involved in high yield expression of two recombinant chimeric fusion proteins for obtaining highly purified and biologically active cytotoxins in Escherichia coli. The chimeric constructs of human IL-13 and two 38 kDa truncated PEs: (i) PE38 and (ii) PE38QQR, (three lysine residues in PE38 at 590, 606, and 613 substituted with two glutamine and one arginine) were used for protein expression in pET prokaryotic expression vector system with kanamycin as a selection antibiotic. Our results suggest that fresh transformation of E. coli and induction by isopropyl-beta-D-thiogalactopyranoside (IPTG) for 6 h resulted in maximum protein expression. To further improve the yield, we used a genetically modified E. coli strain, BL21(DE3)pLysS, which carries a plasmid for lysozyme with a weak promoter that inhibits T7 RNA polymerase and minimizes protein production in the absence of IPTG. Use of this strain eliminated the need for lysozyme digestion of the induced bacteria to release inclusion bodies, which resulted in expression of purer protein as compared to the conventional BL21(DE3) strain. Additional protocol optimizations included 16 h solubilization of inclusion bodies, constitution of refolding buffer, and timing of dialysis. These proteins were finally purified by Q-Sepharose, mono-Q, and gel filtration chromatography. Between 14-22 and 21-28 mg highly purified and biologically active protein was obtained from 1L of BL21 (DE3) and BL21 (DE3) pLysS bacteria culture, respectively. As IL-13R targeting for brain tumor therapy offers an exciting treatment option, optimization of production of IL-13PE will enhance production of clinical grade material for Phase III clinical trials.
Collapse
Affiliation(s)
- Bharat H Joshi
- Laboratory of Molecular Tumor Biology, Division of Cellular and Gene Therapies, Office of Cellular, Tissue and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD, USA
| | | |
Collapse
|
44
|
Xu YM, Wang LF, Jia LT, Qiu XC, Zhao J, Yu CJ, Zhang R, Zhu F, Wang CJ, Jin BQ, Chen SY, Yang AG. A caspase-6 and anti-human epidermal growth factor receptor-2 (HER2) antibody chimeric molecule suppresses the growth of HER2-overexpressing tumors. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2004; 173:61-7. [PMID: 15210759 DOI: 10.4049/jimmunol.173.1.61] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Clinical studies have suggested that human epidermal growth factor receptor-2 (HER2) provide a useful target for antitumor therapy. We previously described the generation of a chimeric HER2-targeted immunocasp-3 protein. In this study, we extend the repertoire of chimeric proapoptotic proteins with immunocasp-6, a construct that comprises a HER2-specific single-chain Ab, a single-chain Pseudomonas exotoxin A, and an active caspase-6, which can directly cleave lamin A leading to nucleus damage and inducing programmed cell death. We demonstrate that the secreted immunocasp-6 molecule selectively recognizes and induces apoptosis in HER2-overexpressing tumor cells in vitro, but not in cells with undetectable HER2. The immunocasp-6 gene was next transferred into BALB/c athymic mice bearing human breast SK-BR-3 tumors by i.m. injection of liposome-encapsulated vectors, by intratumor injection of adenoviral vectors, or by i.v. injection of PBMC modified by retroviral infection. Regardless of the method used, expression of immunocasp-6 suppressed tumor growth and prolonged animal survival significantly. Our data show that the chimeric immunocasp-6 molecule can recognize HER2-positive tumor cells, promptly attack their nucleus, and induce their apoptotic death, suggesting the potential of this strategy for the treatment of human cancers that overexpress HER2.
Collapse
Affiliation(s)
- Yan-Ming Xu
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, 17 Changle West Road, 710-032 Xi'an, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Zhao J, Zhang LH, Jia LT, Zhang L, Xu YM, Wang Z, Yu CJ, Peng WD, Wen WH, Wang CJ, Chen SY, Yang AG. Secreted Antibody/Granzyme B Fusion Protein Stimulates Selective Killing of HER2-overexpressing Tumor Cells. J Biol Chem 2004; 279:21343-8. [PMID: 15004021 DOI: 10.1074/jbc.m312648200] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Targeted cell killing is required for effective treatment of cancers. We previously described the generation of a chimeric immunocasp-3 protein and its potent selective antitumor activity (Jia, L. T., Zhang, L. H., Yu, C. J., Zhao, J., Xu, Y. M., Gui, J. H., Jin, M., Ji, Z. L., Wen, W. H., Wang, C. J., Chen, S. Y., and Yang, A. G. (2003) Cancer Res. 63, 3257-3262). Here we extend the repertoire of another chimeric pro-apoptotic protein immunoGrB, which comprises an anti-HER2 single-chain antibody, a Pseudomonas exotoxin A translocation domain and active granzyme B. Human lymphoma Jurkat cells transfected with the immunoGrB gene expression vector were able to produce and secrete the chimeric protein. The immunoGrB molecule selectively recognized and destroyed HER2-overexpressing tumor cells both in vitro and in nude mouse after intramuscular injection of the immunoGrB expression plasmid. Further in vivo study showed that intravenous administration of immunoGrB gene-modified lymphocytes led to suppression of HER2-overexpressing tumor growth and prolonged animal survival because of continuous secretion of immunoGrB molecules into blood and lymph fluid. These results demonstrate that the chimeric immunoGrB molecule, which is capable of antibody-directed targeting and granzyme B-mediated killing, has therapeutic potential against HER2 tumors, especially in cases in which caspase-dependent apoptosis is inhibited.
Collapse
Affiliation(s)
- Jing Zhao
- Departments of Biochemistry and Molecular Biology and Immunology, the Fourth Military Medical University, Xi'an 710032, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Onda M, Wang QC, Guo HF, Cheung NKV, Pastan I. In vitro and in vivo cytotoxic activities of recombinant immunotoxin 8H9(Fv)-PE38 against breast cancer, osteosarcoma, and neuroblastoma. Cancer Res 2004; 64:1419-24. [PMID: 14973056 DOI: 10.1158/0008-5472.can-03-0570] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The 8H9 monoclonal antibody (MAb) is highly reactive with a cell surface glycoprotein expressed on human breast cancers, childhood sarcomas, and neuroblastomas but is not reactive with the cell surface of normal human tissues. This specific reactivity suggests that MAb 8H9 may be useful for targeted cancer therapy. To explore this possibility, we generated two recombinant immunotoxins (ITs) using the single-chain Fv (scFv) of MAb 8H9. Initially the 8H9(scFv) cDNA was fused to a DNA encoding a 38-kDa truncated form of Pseudomonas exotoxin (PE38) to generate the IT 8H9(scFv)-PE38. The fusion gene was expressed in Escherichia coli, and the IT was purified to near homogeneity from inclusion bodies. The purified IT showed specific cytotoxicity on nine different cancer cell lines derived from breast cancer, osteosarcoma, and neuroblastomas, known to react with MAb 8H9. The cytotoxic activity was inhibited by MAb 8H9, showing the cytotoxic activity is specific. The antitumor activity of 8H9(scFv)-PE38 was evaluated in severe combined immunodeficient mice bearing MCF-7 breast cancers or OHS-M1 osteosarcomas. The IT showed a specific dose-dependent antitumor activity at 0.075 and 0.15 mg/kg. Next, a more stable disulfide-linked IT, 8H9(dsFv)-PE38, was constructed. It was produced in high yield (16%) and showed cytotoxic and antitumor activities similar to those of 8H9(scFv)-PE38. 8H9(dsFv)-PE38 was given to two cynomolgus monkeys at doses of 0.1 and 0.2 mg/kg i.v. QOD x 3 and was well tolerated. This shows that a dose that causes significant tumor regressions in mice is well tolerated by monkeys. These results make 8H9(dsFv)-PE38 a candidate for further development as a therapeutic agent for breast cancers, osteosarcomas, and neuroblastomas.
Collapse
Affiliation(s)
- Masanori Onda
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute/NIH, 37 Convent Drive, Bethesda, MD 20892-4264, USA
| | | | | | | | | |
Collapse
|
47
|
Rampaul RS, Pinder SE, Gullick WJ, Robertson JFR, Ellis IO. HER-2 in breast cancer--methods of detection, clinical significance and future prospects for treatment. Crit Rev Oncol Hematol 2003; 43:231-44. [PMID: 12270780 DOI: 10.1016/s1040-8428(01)00207-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The Human Epidermal Growth Factor (HER-2) oncogene encodes a transmembrane tyrosine kinase receptor with extensive homology to the Epidermal Growth Factor Receptor (EGFR) which is the prototypal member of this family of receptor tyrosine kinases. HER-2 gene amplification is found in 20-30% of breast cancers. Various methods such as immunohistochemistry, southern and slot blotting, enzyme immunoassays and fluorescence in situ hybridization have all been employed to evaluate HER-2 gene and protein abnormalities. Of these immunohistochemistry is the most frequently employed but there are valid indications for the other avaliable methods. However, it is prudent that whichever methods employed are standardized, especially those that possess may have a degree of subjectivity in their assesment.
Collapse
Affiliation(s)
- R S Rampaul
- Department of Surgery, Nottingham City Hospital, Nottingham, UK
| | | | | | | | | |
Collapse
|
48
|
Nahta R, Hortobagyi GN, Esteva FJ. Novel pharmacological approaches in the treatment of breast cancer. Expert Opin Investig Drugs 2003; 12:909-21. [PMID: 12783596 DOI: 10.1517/13543784.12.6.909] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Breast cancer is the most common malignancy among women. Novel pharmacological agents, including hormonal, cytotoxic and biological therapies, are currently being developed and tested in clinical trials and may offer patients more treatment options and an improved chance of long-term survival. Signal transduction inhibitors that block endocrine or growth factor pathways have demonstrated exciting antitumour effects in clinical trials. In addition to new chemotherapeutic drugs, numerous biological agents including growth factor receptor-directed monoclonal antibodies and tyrosine kinase inhibitors that target specific molecular lesions are being examined as potential breast cancer treatments.
Collapse
Affiliation(s)
- Rita Nahta
- Department of Molecular Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas, 77030, USA
| | | | | |
Collapse
|
49
|
Spooner RA, Friedlos F, Maycroft K, Stribbling SM, Roussel J, Brueggen J, Stolz B, O'Reilly T, Wood J, Matter A, Marais R, Springer CJ. A novel vascular endothelial growth factor-directed therapy that selectively activates cytotoxic prodrugs. Br J Cancer 2003; 88:1622-30. [PMID: 12771932 PMCID: PMC2377106 DOI: 10.1038/sj.bjc.6600911] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
We have generated fusion proteins between vascular endothelial growth factor (VEGF) and the bacterial enzyme carboxypeptidase G2 (CPG2) that can activate the prodrug 4-[(2-chloroethyl)(2-mesyloxyethyl)amino]benzoyl-L-glutamic acid (CMDA). Three asparagine residues of CPG2 were mutated to glutamine (CPG2(Q)3) to prevent glycosylation during secretion, and truncations of VEGF(165) were fused to either the C- or N-terminal of CPG2. The K(m) of the fusion proteins (37.5 microM) was similar to that of secreted CPG2(Q)3 (29.5 microM) but greater than that of wild-type CPG2 (8 microM). The affinity of the fusion proteins for VEGF receptor-2 (VEGFR2) (K(d)=0.5-1.1 nM) was similar to that of [(125)I]VEGF (K(d)=0.5 nM) (ELISA) or slightly higher (K(d)=1.3-9.6 nM) (competitive RIA). One protein, VEGF(115)-CPG2(Q)3-H(6), possessed 140% of the enzymic activity of secreted CPG2(Q)3, and had a faster half-maximal binding time for VEGFR2 (77 s), than the other candidates (330 s). In vitro, VEGF(115)-CPG2(Q)3-H(6) targeted CMDA cytotoxicity only towards VEGFR-expressing cells. The plasma half-life of VEGF(115)-CPG2(Q)3-H(6) in vivo was 3 h, comparable to equivalent values observed in ADEPT. We conclude that enzyme prodrug therapy using VEGF as a targeting moiety represents a promising novel antitumour therapy, with VEGF(115)-CPG2(Q)3-H(6) being a lead candidate.
Collapse
Affiliation(s)
- R A Spooner
- Cancer Research UK Centre for Cancer Therapeutics at the Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
- Cancer Research UK Centre for Cell and Molecular Biology at the Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - F Friedlos
- Cancer Research UK Centre for Cancer Therapeutics at the Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - K Maycroft
- Cancer Research UK Centre for Cell and Molecular Biology at the Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - S M Stribbling
- Cancer Research UK Centre for Cancer Therapeutics at the Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - J Roussel
- Novartis Pharmaceutical AG, Pharmaceutical Research, CH-4002, Basel, Switzerland
| | - J Brueggen
- Novartis Pharmaceutical AG, Pharmaceutical Research, CH-4002, Basel, Switzerland
| | - B Stolz
- Novartis Pharmaceutical AG, Pharmaceutical Research, CH-4002, Basel, Switzerland
| | - T O'Reilly
- Novartis Pharmaceutical AG, Pharmaceutical Research, CH-4002, Basel, Switzerland
| | - J Wood
- Novartis Pharmaceutical AG, Pharmaceutical Research, CH-4002, Basel, Switzerland
| | - A Matter
- Novartis Pharmaceutical AG, Pharmaceutical Research, CH-4002, Basel, Switzerland
| | - R Marais
- Cancer Research UK Centre for Cell and Molecular Biology at the Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - C J Springer
- Cancer Research UK Centre for Cancer Therapeutics at the Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
- Cancer Research UK Centre for Cancer Therapeutics at the Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK. E-mail:
| |
Collapse
|
50
|
Nahta R, Hortobágyi GN, Esteva FJ. Growth factor receptors in breast cancer: potential for therapeutic intervention. Oncologist 2003; 8:5-17. [PMID: 12604728 DOI: 10.1634/theoncologist.8-1-5] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Increased expression and activation of receptor tyrosine kinases occurs frequently in human breast carcinomas. Several therapies targeting these receptors are currently in clinical trials. Therapeutic strategies include blockade of individual receptors with monoclonal antibodies and inhibition of tyrosine kinase function. Trastuzumab is the first of these biologic therapies to be approved for patients with human epidermal growth factor receptor 2 (HER2)-overexpressing metastatic breast cancer. Novel trastuzumab-based combinations are being investigated in patients with advanced breast cancer. Large clinical trials have also been launched in the adjuvant setting. Small molecules that inhibit specific tyrosine kinases (e.g., epidermal growth factor receptor, HER2) are in phase I and phase II clinical trials. Other growth-factor-targeted drugs that have reached clinical development include STI571 and antibodies directed at the insulin-like growth factor pathway. Biologic therapies directed against these important receptors are promising. In this review we discuss challenges and opportunities for the development of growth-factor-targeted approaches for the treatment of breast cancer.
Collapse
Affiliation(s)
- Rita Nahta
- Department of Breast Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | |
Collapse
|