1
|
Liu X, Song Y, Cheng P, Liang B, Xing D. Targeting HER2 in solid tumors: Unveiling the structure and novel epitopes. Cancer Treat Rev 2024; 130:102826. [PMID: 39270365 DOI: 10.1016/j.ctrv.2024.102826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024]
Abstract
Human epidermal growth factor receptor-2 (HER2) is overexpressed in various solid tumor types, acting as an established therapeutic target. Over the last three decades, the fast-paced development of diverse HER2-targeted agents, notably marked by the introduction of the antibody-drug conjugate (ADC), yielding substantial improvements in survival rates. However, resistance to anti-HER2 treatments continues to pose formidable challenges. The complex structure and dynamic dimerization properties of HER2 create significant hurdles in the development of novel targeted therapeutics. In this review, we synthesize the latest insights into the structural intricacies of HER2 and present an unprecedented overview of the epitope characteristics of HER2-targeted antibodies and their derivatives. Furthermore, we delve into the correlation between anti-HER2 antibody binding epitopes and their respective functions, with a particular focus on their efficacy against resistant tumors. In addition, we highlight the potential of emerging anti-HER2 agents that target specific sites or non-overlapping epitopes, poised to transform the therapeutic landscape for HER2-positive tumors in the foreseeable future.
Collapse
Affiliation(s)
- Xinlin Liu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Qingdao Cancer Institute, Qingdao 266071, China
| | - Yunlong Song
- Qingdao Municipal Center for Disease Control and Prevention, Qingdao Institute of Preventive Medicine, Qingdao 266033, China
| | - Panpan Cheng
- Qingdao Municipal Center for Disease Control and Prevention, Qingdao Institute of Preventive Medicine, Qingdao 266033, China
| | - Bing Liang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Qingdao Cancer Institute, Qingdao 266071, China.
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Qingdao Cancer Institute, Qingdao 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
2
|
Dong W, Wang W, Cao C. The Evolution of Antibody-Drug Conjugates: Toward Accurate DAR and Multi-specificity. ChemMedChem 2024; 19:e202400109. [PMID: 38758596 DOI: 10.1002/cmdc.202400109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/15/2024] [Accepted: 05/15/2024] [Indexed: 05/18/2024]
Abstract
Antibody-drug conjugates (ADCs) consist of antibodies, linkers and payloads. They offer targeted delivery of potent cytotoxic drugs to tumor cells, minimizing off-target effects. However, the therapeutic efficacy of ADCs is compromised by heterogeneity in the drug-to-antibody ratio (DAR), which impacts both cytotoxicity and pharmacokinetics (PK). Additionally, the emergence of drug resistance poses significant challenges to the clinical advancement of ADCs. To overcome these limitations, a variety of strategies have been developed, including the design of multi-specific drugs with accurate DAR. This review critically summarizes the current challenges faced by ADCs, categorizing key issues and evaluating various innovative solutions. We provide an in-depth analysis of the latest methodologies for achieving homogeneous DAR and explore design strategies for multi-specific drugs aimed at combating drug resistance. Our discussion offers a current perspective on the advancements made in refining ADC technologies, with an emphasis on enhancing therapeutic outcomes.
Collapse
Affiliation(s)
- Wenge Dong
- State Key Laboratory of Elemento-Organic Chemistry and Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Wanqi Wang
- State Key Laboratory of Elemento-Organic Chemistry and Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Chan Cao
- State Key Laboratory of Elemento-Organic Chemistry and Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| |
Collapse
|
3
|
Xing Y, Zhang F, Yang T, Yin C, Yang A, Yan B, Zhao J. Augmented antitumor immune responses of HER2-targeted pyroptotic induction by long-lasting recombinant immunopyroptotins. Heliyon 2024; 10:e30444. [PMID: 38737283 PMCID: PMC11088320 DOI: 10.1016/j.heliyon.2024.e30444] [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: 12/02/2023] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/14/2024] Open
Abstract
Pyroptosis is a well-documented form of programmed cell death caused by the gasdermin-driven perforation of cell membranes. Selective induction of pyroptosis in tumor cells represents a promising antitumor strategy to enhance the efficacy of immunotherapy. In this study, we established a recombinant protein-based immunopyroptotin strategy that led to the intratumoral induction of pyroptosis for HER2-directed therapy. Long-lasting immunopyroptotins were constructed by sequentially fusing the humanized anti-HER2 single-chain antibody P1h3, albumin-binding peptide (ABD035 or dAb7h8), cathepsin B-cleavable peptide B2, endosome-disruptive peptide E5C3, and active pyroptotic effector gasdermin D-N fragment (GN). After purification, we evaluated the cytotoxicity and antitumor immune responses primarily induced by the immunopyroptotins in HER2-overexpressing breast cancer cells. The resulting ABD035-immunoGN and dAb7h8-immunoGN showed improved in vitro cytotoxicity in HER2-overexpressing cancer cells compared with that in the immunotBid that we previously generated to induce tumor cell apoptosis. The binding of long-lasting immunopyroptotins to albumin increased the half-life by approximately 7-fold in nude mice. The enhanced antitumor efficacy of long-lasting immunopyroptotins was confirmed in both N87 tumor-bearing T cell-deficient mice and 4T1-hHER2 bilateral tumor-bearing immunocompetent mice. Immunopyroptotin treatment elicited systemic antitumor immune responses involving CD8+ T cells and mature dendritic cells and upregulated the expression of proinflammatory cytokines, leading to sustained remission of non-injected distant tumors. This study extends the repertoire of antibody-based therapeutics through the tumor-targeted delivery of a constitutively active pore-forming gasdermin-N fragment, which shows great potential for pyroptosis-based antitumor therapy.
Collapse
Affiliation(s)
- Yuqi Xing
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Feiyu Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Tian Yang
- Department of Intensive Care Medicine, Bethune International Peace Hospital, Hebei, 050082, China
| | - Chunhui Yin
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Angang Yang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Immunology, Fourth Military Medical University, Xi'an, 710032, China
| | - Bo Yan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Jing Zhao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| |
Collapse
|
4
|
Lewis GD, Li G, Guo J, Yu SF, Fields CT, Lee G, Zhang D, Dragovich PS, Pillow T, Wei B, Sadowsky J, Leipold D, Wilson T, Kamath A, Mamounas M, Lee MV, Saad O, Choeurng V, Ungewickell A, Monemi S, Crocker L, Kalinsky K, Modi S, Jung KH, Hamilton E, LoRusso P, Krop I, Schutten MM, Commerford R, Sliwkowski MX, Cho E. The HER2-directed antibody-drug conjugate DHES0815A in advanced and/or metastatic breast cancer: preclinical characterization and phase 1 trial results. Nat Commun 2024; 15:466. [PMID: 38212321 PMCID: PMC10784567 DOI: 10.1038/s41467-023-44533-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 12/14/2023] [Indexed: 01/13/2024] Open
Abstract
Approved antibody-drug conjugates (ADCs) for HER2-positive breast cancer include trastuzumab emtansine and trastuzumab deruxtecan. To develop a differentiated HER2 ADC, we chose an antibody that does not compete with trastuzumab or pertuzumab for binding, conjugated to a reduced potency PBD (pyrrolobenzodiazepine) dimer payload. PBDs are potent cytotoxic agents that alkylate and cross-link DNA. In our study, the PBD dimer is modified to alkylate, but not cross-link DNA. This HER2 ADC, DHES0815A, demonstrates in vivo efficacy in models of HER2-positive and HER2-low cancers and is well-tolerated in cynomolgus monkey safety studies. Mechanisms of action include induction of DNA damage and apoptosis, activity in non-dividing cells, and bystander activity. A dose-escalation study (ClinicalTrials.gov: NCT03451162) in patients with HER2-positive metastatic breast cancer, with the primary objective of evaluating the safety and tolerability of DHES0815A and secondary objectives of characterizing the pharmacokinetics, objective response rate, duration of response, and formation of anti-DHES0815A antibodies, is reported herein. Despite early signs of anti-tumor activity, patients at higher doses develop persistent, non-resolvable dermal, ocular, and pulmonary toxicities, which led to early termination of the phase 1 trial.
Collapse
Affiliation(s)
- Gail D Lewis
- Discovery Oncology, Genentech, South San Francisco, CA, USA.
| | - Guangmin Li
- Discovery Oncology, Genentech, South San Francisco, CA, USA
| | - Jun Guo
- Discovery Oncology, Genentech, South San Francisco, CA, USA
| | - Shang-Fan Yu
- Translational Oncology, Genentech, South San Francisco, CA, USA
| | | | - Genee Lee
- Translational Oncology, Genentech, South San Francisco, CA, USA
| | | | | | - Thomas Pillow
- Discovery Chemistry, Genentech, South San Francisco, CA, USA
| | - BinQing Wei
- Computational Chemistry, Genentech, South San Francisco, CA, USA
| | - Jack Sadowsky
- Protein Chemistry, Genentech, South San Francisco, CA, USA
- Carmot Therapeutics, Berkeley, CA, USA
| | - Douglas Leipold
- Preclinical and Translational Pharmacokinetics, Genentech, South San Francisco, CA, USA
| | - Tim Wilson
- Oncology Biomarker Development, Genentech, South San Francisco, CA, USA
| | - Amrita Kamath
- Preclinical and Translational Pharmacokinetics, Genentech, South San Francisco, CA, USA
| | - Michael Mamounas
- Project Team Leadership, Oncology, Genentech, South San Francisco, CA, USA
| | - M Violet Lee
- Bioanalytical Sciences, Genentech, South San Francisco, CA, USA
| | - Ola Saad
- Bioanalytical Sciences, Genentech, South San Francisco, CA, USA
| | | | | | - Sharareh Monemi
- Early Clinical Development, Oncology, Genentech, South San Francisco, CA, USA
| | - Lisa Crocker
- Translational Oncology, Genentech, South San Francisco, CA, USA
| | - Kevin Kalinsky
- Winship Cancer Institute at Emory University, Atlanta, GA, USA
| | - Shanu Modi
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kyung Hae Jung
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Erika Hamilton
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN, USA
| | | | - Ian Krop
- Yale Cancer Center, Yale University, New Haven, CT, USA
| | - Melissa M Schutten
- Safety Assessment Pathology, Genentech, South San Francisco, CA, USA
- SeaGen, South San Francisco, CA, USA
| | - Renee Commerford
- Early Clinical Development, Oncology, Genentech, South San Francisco, CA, USA
- Gilead Sciences, Foster City, CA, USA
| | | | - Eunpi Cho
- Early Clinical Development, Oncology, Genentech, South San Francisco, CA, USA
| |
Collapse
|
5
|
Liu X, Luan L, Liu X, Jiang D, Deng J, Xu J, Yuan Y, Xing J, Chen B, Xing D, Huang H. A novel nanobody-based HER2-targeting antibody exhibits potent synergistic antitumor efficacy in trastuzumab-resistant cancer cells. Front Immunol 2023; 14:1292839. [PMID: 37954614 PMCID: PMC10634241 DOI: 10.3389/fimmu.2023.1292839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 10/16/2023] [Indexed: 11/14/2023] Open
Abstract
Human epithelial growth factor receptor-2 (HER2) plays an oncogenic role in numerous tumors, including breast, gastric, and various other solid tumors. While anti-HER2 therapies are approved for the treatment of HER2-positive tumors, a necessity persists for creating novel HER2-targeted agents to resolve therapeutic resistance. Utilizing a synthetic nanobody library and affinity maturation, our study identified four anti-HER2 nanobodies that exhibited high affinity and specificity. These nanobodies recognized three distinct epitopes of HER2-ECD. Additionally, we constructed VHH-Fc and discovered that they facilitated superior internalization and showed moderate growth inhibition. Compared to the combination of trastuzumab and pertuzumab, the VHH-Fc combos or their combination with trastuzumab demonstrated greater or comparable antitumor activity in both ligand-independent and ligand-driven tumors. Most remarkably, A9B5-Fc, which targeted domain I of HER2-ECD, displayed significantly enhanced trastuzumab-synergistic antitumor efficacy compared to pertuzumab under trastuzumab-resistant conditions. Our findings offer anti-HER2 nanobodies with high affinity and non-overlapping epitope recognition. The novel nanobody-based HER2-targeted antibody, A9B5-Fc, binding to HER2-ECD I, mediates promising receptor internalization. It possesses the potential to serve as a potent synergistic partner with trastuzumab, contributing to overcoming acquired resistance.
Collapse
Affiliation(s)
- Xinlin Liu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
| | - Linli Luan
- Noventi Biopharmaceuticals Co., Ltd, Shanghai, China
| | - Xi Liu
- Bioworkshops (Suzhou) Limited, Souzhou, China
| | - Dingwen Jiang
- Noventi Biopharmaceuticals Co., Ltd, Shanghai, China
| | - Junwen Deng
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
| | - Jiazhen Xu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
| | - Yang Yuan
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
| | - Jiyao Xing
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
| | - Bingguan Chen
- Noventi Biopharmaceuticals Co., Ltd, Shanghai, China
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Haiming Huang
- Noventi Biopharmaceuticals Co., Ltd, Shanghai, China
| |
Collapse
|
6
|
Peng K, Zou Z, Li J, Xie Y, Ming Z, Jiang T, Luo W, Hu X, Nie Y, Chen L, Luo T, Peng T, Ma D, Liu S, Luo ZY. Spinosyn A and Its Derivative Inhibit Colorectal Cancer Cell Growth via the EGFR Pathway. JOURNAL OF NATURAL PRODUCTS 2023; 86:2111-2121. [PMID: 37682035 DOI: 10.1021/acs.jnatprod.3c00276] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
Spinosyn A (SPA), derived from a soil microorganism, Saccharopolyspora spinosa, and its derivative, LM2I, has potential inhibitory effects on a variety of cancer cells. However, the effects of SPA and LM2I in inhibiting the growth of human colorectal cancer cells and the molecular mechanisms underlying these effects are not fully understood. Cell viability was tested by using a 3-(4,5-dimethylthiazol-2-yl-)-2,5-diphenyltetrazolium bromide (MTT) assay and a colony formation assay. On the basis of the IC50 values of SPA and LM2I in seven colorectal cancer (CRC) cell lines, sensitive (HT29 and SW480) and insensitive (SW620 and RKO) cell lines were screened. The GSE2509 and GSE10843 data sets were used to identify 69 differentially expressed genes (DEGs) between sensitive and insensitive cell lines. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, and protein-protein interactions (PPI) were performed to elucidate the molecular mechanisms of the DEGs. The hub gene of the DEGs was detected by Western blot analysis and verified using the CRISPR/Cas9 system. Our data indicate that SPA and its derivative LM2I have significant antiproliferative activity in seven colorectal cancer cell lines and colorectal xenograft tumors. On the basis of bioinformatics analysis, it was demonstrated that epidermal growth factor receptor (EGFR) was the hub gene of the DEGs and was associated with the inhibitory effects of SPA and LM2I in CRC cell lines. The study also revealed that SPA and LM2I inhibited the EGFR pathway in vitro and in vivo.
Collapse
Affiliation(s)
- Kunjian Peng
- Department of Biochemistry and Molecular Biology, Hunan Province Key Laboratory of Basic and Applied Hematology, Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Xiangya School of Medicine, Central South University, Changsha, 410008 Hunan, China
| | - Zizheng Zou
- Department of Biochemistry and Molecular Biology, Hunan Province Key Laboratory of Basic and Applied Hematology, Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Xiangya School of Medicine, Central South University, Changsha, 410008 Hunan, China
- Department of Science and Education, Yiyang Central Hospital, Yiyang, 413099 Hunan, China
| | - Jijia Li
- Hunan Key Laboratory of Oral Health Research & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, 410008 Hunan, China
| | - Yuanzhu Xie
- Department of Biochemistry and Molecular Biology, Hunan Province Key Laboratory of Basic and Applied Hematology, Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Xiangya School of Medicine, Central South University, Changsha, 410008 Hunan, China
| | - Zhengnan Ming
- Department of Biochemistry and Molecular Biology, Hunan Province Key Laboratory of Basic and Applied Hematology, Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Xiangya School of Medicine, Central South University, Changsha, 410008 Hunan, China
| | - Ting Jiang
- Department of Biochemistry and Molecular Biology, Hunan Province Key Laboratory of Basic and Applied Hematology, Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Xiangya School of Medicine, Central South University, Changsha, 410008 Hunan, China
| | - Wensong Luo
- Department of Biochemistry and Molecular Biology, Hunan Province Key Laboratory of Basic and Applied Hematology, Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Xiangya School of Medicine, Central South University, Changsha, 410008 Hunan, China
| | - Xiyuan Hu
- Department of Biochemistry and Molecular Biology, Hunan Province Key Laboratory of Basic and Applied Hematology, Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Xiangya School of Medicine, Central South University, Changsha, 410008 Hunan, China
| | - Yuan Nie
- Department of Biochemistry and Molecular Biology, Hunan Province Key Laboratory of Basic and Applied Hematology, Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Xiangya School of Medicine, Central South University, Changsha, 410008 Hunan, China
| | - Ling Chen
- Department of Biochemistry and Molecular Biology, Hunan Province Key Laboratory of Basic and Applied Hematology, Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Xiangya School of Medicine, Central South University, Changsha, 410008 Hunan, China
| | - Tiao Luo
- Hunan Key Laboratory of Oral Health Research & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, 410008 Hunan, China
| | - Ting Peng
- Department of Biochemistry and Molecular Biology, Hunan Province Key Laboratory of Basic and Applied Hematology, Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Xiangya School of Medicine, Central South University, Changsha, 410008 Hunan, China
| | - Dayou Ma
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008 Hunan, China
| | - Suyou Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008 Hunan, China
| | - Zhi-Yong Luo
- Department of Biochemistry and Molecular Biology, Hunan Province Key Laboratory of Basic and Applied Hematology, Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Xiangya School of Medicine, Central South University, Changsha, 410008 Hunan, China
| |
Collapse
|
7
|
Choi Y, Cho BK, Seok SH, Kim C, Ryu JH, Kwon IC. Controlled spatial characteristics of ligands on nanoparticles: Determinant of cellular functions. J Control Release 2023; 360:672-686. [PMID: 37437847 DOI: 10.1016/j.jconrel.2023.07.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/27/2023] [Accepted: 07/08/2023] [Indexed: 07/14/2023]
Abstract
Interactions of various ligands and receptors have been extensively investigated because they regulate a series of signal transduction leading to various functional cellular outcomes. The receptors on cell membrane recognize their specific ligands, resulting in specific binding between ligands and receptors. Accumulating evidence reveals that the receptors recognize the difference on the spatial characteristics of ligands as well as the types of ligands. Thus, control on spatial characteristics of multiple ligands presented on therapeutic nanoparticles is believed to impact the cellular functions. Specifically, the localized and multivalent distribution of ligands on nanoparticles can induce receptor oligomerization and receptor clustering, controlling intensity or direction of signal transduction cascades. Here, we will introduce recent studies on the use of material-based nanotechnology to control spatial characteristics of ligands and their effect on cellular functions. These therapeutic nanoparticles with controlled spatial characteristics of ligands may be a promising strategy for maximized therapeutic outcome.
Collapse
Affiliation(s)
- Youngjin Choi
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Bo Kyung Cho
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Su Hyun Seok
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Chansoo Kim
- Computational Science Centre & ASSIST, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; AI-Robot Department, University of Science and Technology, Seoul 02792, Republic of Korea
| | - Ju Hee Ryu
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.
| | - Ick Chan Kwon
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
8
|
Rabia E, Garambois V, Dhommée C, Larbouret C, Lajoie L, Buscail Y, Jimenez-Dominguez G, Choblet-Thery S, Liaudet-Coopman E, Cerutti M, Jarlier M, Ravel P, Gros L, Pirot N, Thibault G, Zhukovsky EA, Gérard PE, Pèlegrin A, Colinge J, Chardès T. Design and selection of optimal ErbB-targeting bispecific antibodies in pancreatic cancer. Front Immunol 2023; 14:1168444. [PMID: 37153618 PMCID: PMC10157173 DOI: 10.3389/fimmu.2023.1168444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/06/2023] [Indexed: 05/10/2023] Open
Abstract
The ErbB family of receptor tyrosine kinases is a primary target for small molecules and antibodies for pancreatic cancer treatment. Nonetheless, the current treatments for this tumor are not optimal due to lack of efficacy, resistance, or toxicity. Here, using the novel BiXAb™ tetravalent format platform, we generated bispecific antibodies against EGFR, HER2, or HER3 by considering rational epitope combinations. We then screened these bispecific antibodies and compared them with the parental single antibodies and antibody pair combinations. The screen readouts included measuring binding to the cognate receptors (mono and bispecificity), intracellular phosphorylation signaling, cell proliferation, apoptosis and receptor expression, and also immune system engagement assays (antibody-dependent cell-mediated cytotoxicity and complement-dependent cytotoxicity). Among the 30 BiXAbs™ tested, we selected 3Patri-1Cetu-Fc, 3Patri-1Matu-Fc and 3Patri-2Trastu-Fc as lead candidates. The in vivo testing of these three highly efficient bispecific antibodies against EGFR and HER2 or HER3 in pre-clinical mouse models of pancreatic cancer showed deep antibody penetration in these dense tumors and robust tumor growth reduction. Application of such semi-rational/semi-empirical approach, which includes various immunological assays to compare pre-selected antibodies and their combinations with bispecific antibodies, represents the first attempt to identify potent bispecific antibodies against ErbB family members in pancreatic cancer.
Collapse
Affiliation(s)
- Emilia Rabia
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Véronique Garambois
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Christine Dhommée
- GICC, Groupe Innovation et Ciblage Cellulaire, Université de Tours, Tours, France
| | - Christel Larbouret
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Laurie Lajoie
- GICC, Groupe Innovation et Ciblage Cellulaire, Université de Tours, Tours, France
| | - Yoan Buscail
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
- Réseau d’Histologie Expérimentale de Montpellier, BioCampus, Université de Montpellier, UAR3426 CNRS-US09 INSERM, Montpellier, France
| | - Gabriel Jimenez-Dominguez
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Sylvie Choblet-Thery
- Plateforme Bacfly, Baculovirus et Thérapie, BioCampus, UAR3426 CNRS-US09 INSERM, Saint-Christol-Lèz Alès, France
| | - Emmanuelle Liaudet-Coopman
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Martine Cerutti
- Plateforme Bacfly, Baculovirus et Thérapie, BioCampus, UAR3426 CNRS-US09 INSERM, Saint-Christol-Lèz Alès, France
| | - Marta Jarlier
- ICM, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Patrice Ravel
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Laurent Gros
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
- CNRS, Centre National de la Recherche Scientifique, Paris, France
| | - Nelly Pirot
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
- Réseau d’Histologie Expérimentale de Montpellier, BioCampus, Université de Montpellier, UAR3426 CNRS-US09 INSERM, Montpellier, France
| | - Gilles Thibault
- GICC, Groupe Innovation et Ciblage Cellulaire, Université de Tours, Tours, France
| | - Eugene A. Zhukovsky
- Biomunex Pharmaceuticals, Incubateur Paris Biotech santé, Hopital Cochin, Paris, France
| | | | - André Pèlegrin
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Jacques Colinge
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Thierry Chardès
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
- Plateforme Bacfly, Baculovirus et Thérapie, BioCampus, UAR3426 CNRS-US09 INSERM, Saint-Christol-Lèz Alès, France
- CNRS, Centre National de la Recherche Scientifique, Paris, France
- *Correspondence: Thierry Chardès,
| |
Collapse
|
9
|
Domain-level epitope mapping of polyclonal antibodies against HER-1 and HER-2 receptors using phage display technology. Sci Rep 2022; 12:12268. [PMID: 35851313 PMCID: PMC9293994 DOI: 10.1038/s41598-022-16411-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 07/11/2022] [Indexed: 11/09/2022] Open
Abstract
HER-1 and HER-2 are tumor-associated antigens overexpressed in several epithelial tumors, and successfully targeted by therapeutic approaches against cancer. Vaccination with their recombinant extracellular domains has had encouraging results in the pre-clinical setting. As complex humoral responses targeting multiple epitopes within each antigen are the ultimate goal of such active immunotherapy strategies, molecular dissection of the mixture of antibody specificities is required. The current work exploits phage display of antigenic versions of HER-1 and HER-2 domains to accomplish domain-level epitope mapping. Recognition of domains I, III and IV of both antigens by antibodies of immunized mice was shown, indicating diverse responses covering a broad range of antigenic regions. The combination of phage display and site-directed mutagenesis allowed mutational screening of antigen surface, showing polyclonal antibodies' recognition of mutated receptor escape variants known to arise in patients under the selective pressure of the anti-HER-1 antibody cetuximab. Phage-displayed HER domains have thus the potential to contribute to fine specificity characterization of humoral responses during future development of anti-cancer vaccines.
Collapse
|
10
|
Kast F, Schwill M, Stüber JC, Pfundstein S, Nagy-Davidescu G, Rodríguez JMM, Seehusen F, Richter CP, Honegger A, Hartmann KP, Weber TG, Kroener F, Ernst P, Piehler J, Plückthun A. Engineering an anti-HER2 biparatopic antibody with a multimodal mechanism of action. Nat Commun 2021; 12:3790. [PMID: 34145240 PMCID: PMC8213836 DOI: 10.1038/s41467-021-23948-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 05/25/2021] [Indexed: 02/07/2023] Open
Abstract
The receptor tyrosine kinase HER2 acts as oncogenic driver in numerous cancers. Usually, the gene is amplified, resulting in receptor overexpression, massively increased signaling and unchecked proliferation. However, tumors become frequently addicted to oncogenes and hence are druggable by targeted interventions. Here, we design an anti-HER2 biparatopic and tetravalent IgG fusion with a multimodal mechanism of action. The molecule first induces HER2 clustering into inactive complexes, evidenced by reduced mobility of surface HER2. However, in contrast to our earlier binders based on DARPins, clusters of HER2 are thereafter robustly internalized and quantitatively degraded. This multimodal mechanism of action is found only in few of the tetravalent constructs investigated, which must target specific epitopes on HER2 in a defined geometric arrangement. The inhibitory effect of our antibody as single agent surpasses the combination of trastuzumab and pertuzumab as well as its parental mAbs in vitro and it is effective in a xenograft model.
Collapse
Affiliation(s)
- Florian Kast
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Martin Schwill
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
- TOLREMO therapeutics AG, Muttenz, Switzerland
| | - Jakob C Stüber
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
- Roche Innovation Center Munich, Penzberg, Germany
| | - Svende Pfundstein
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
- Zurich Integrative Rodent Physiology (ZIRP), University of Zurich, Zurich, Switzerland
| | | | - Josep M Monné Rodríguez
- Laboratory for Animal Model Pathology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Frauke Seehusen
- Laboratory for Animal Model Pathology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Christian P Richter
- Department of Biology/Chemistry and Center for Cellular Nanoanalytics, Osnabrück University, Osnabrück, Germany
| | | | | | | | | | - Patrick Ernst
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
- Dean's Office and Coordination Office of the Academic Medicine Zurich, University of Zurich, Zurich, Switzerland
| | - Jacob Piehler
- Department of Biology/Chemistry and Center for Cellular Nanoanalytics, Osnabrück University, Osnabrück, Germany
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
11
|
Guleria M, Sharma R, Amirdhanayagam J, Sarma HD, Rangarajan V, Dash A, Das T. Formulation and clinical translation of [ 177Lu]Lu-trastuzumab for radioimmunotheranostics of metastatic breast cancer. RSC Med Chem 2021; 12:263-277. [PMID: 34046615 PMCID: PMC8128050 DOI: 10.1039/d0md00319k] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 10/14/2020] [Indexed: 11/21/2022] Open
Abstract
Trastuzumab (Herceptin®) is an approved immunotherapeutic agent used for the treatment of metastatic breast cancer over-expressing HER2 antigen receptors. The aim of the present work is to standardize the formulation protocol of [177Lu]Lu-trastuzumab addressing various reaction parameters, evaluating the efficacy of the radiolabeled product by in vitro investigations, scaling-up the preparation for administration in patients and performing preliminary clinical studies in patients suffering from metastatic breast cancer. Trastuzumab was conjugated with a suitable bi-functional chelating agent namely, p-NCS-benzyl-DOTA. On average 6.15 ± 0.92 p-NCS-benzyl-DOTA molecules were observed to be attached to each trastuzumab moiety. [177Lu]Lu-trastuzumab could be prepared with >95% radiochemical purity (% RCP) employing the optimized radiolabeling procedure. In vitro studies revealed the affinity of [177Lu]Lu-trastuzumab towards HER2 +ve cancer cell lines as well as against HER2 protein (K d = 13.61 nM and 11.36 nM, respectively). The value for percentage immunoreactive fraction (% IRF) for [177Lu]Lu-trastuzumab was observed to be 76.92 ± 2.80. Bio-distribution studies in Swiss mice revealed non-specific uptake in the blood, liver, lungs and heart followed by gradual clearance of activity predominantly through the hepatobiliary route. Preliminary clinical studies carried out in 8 cancer patients with immunohistochemically proven HER2 positive metastatic breast cancer revealed preferential localization of [177Lu]Lu-trastuzumab in breast cancer lesions, which was in concordance with [18F]FDG-PET scans recorded earlier in the same patient indicating the potential of the agent towards radioimmunotheranostic applications.
Collapse
Affiliation(s)
- Mohini Guleria
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre Trombay Mumbai - 400085 India +91 22 2550 5151 +91 22 2559 0613
| | - Rohit Sharma
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre Trombay Mumbai - 400085 India +91 22 2550 5151 +91 22 2559 0613
| | - Jeyachitra Amirdhanayagam
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre Trombay Mumbai - 400085 India +91 22 2550 5151 +91 22 2559 0613
| | - Haladhar D Sarma
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre Trombay Mumbai - 400085 India
| | - Venkatesh Rangarajan
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital Parel Mumbai - 400012 India
| | - Ashutosh Dash
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre Trombay Mumbai - 400085 India +91 22 2550 5151 +91 22 2559 0613
- Homi Bhabha National Institute Anushaktinagar Mumbai - 400094 India
| | - Tapas Das
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre Trombay Mumbai - 400085 India +91 22 2550 5151 +91 22 2559 0613
- Homi Bhabha National Institute Anushaktinagar Mumbai - 400094 India
| |
Collapse
|
12
|
Ma P, Ren P, Zhang C, Tang J, Yu Z, Zhu X, Fan K, Li G, Zhu W, Sang W, Min C, Chen W, Huang X, Yang G, Lerner RA. Avidity-Based Selection of Tissue-Specific CAR-T Cells from a Combinatorial Cellular Library of CARs. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003091. [PMID: 33747727 PMCID: PMC7967050 DOI: 10.1002/advs.202003091] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/21/2020] [Indexed: 05/08/2023]
Abstract
Using T-cell chimeric antigen receptors (CAR-T) to activate and redirect T cells to tumors expressing the cognate antigen represents a powerful approach in cancer therapy. However, normal tissues with low expression of tumor-associated antigens (TAAs) can be mistargeted, resulting in severe side effects. An approach using a collection of T cells expressing a diverse, 106-member combinatorial cellular library of CARs, in which members can be specifically enriched based on avidity for cell membrane antigens, is reported. Using CD38 as the target antigen, an efficient and effective selection of CARs specifically recognizing CD38+ tumor cells is demonstrated. These selected CAR-T's produce cytokines known to be associated with T cell activation in a CD38 expression-dependent manner. This avidity-based selection endows the engineered T cells with minimal off-tumor effects, while retaining robust antitumor efficacy both in vitro and in vivo. The described method may facilitate the application of CAR-T therapy to TAAs previously considered undruggable.
Collapse
Affiliation(s)
- Peixiang Ma
- Shanghai Institute for Advanced Immunochemical StudiesShanghaiTech UniversityShanghai201210China
| | - Ping Ren
- Shanghai Institute for Advanced Immunochemical StudiesShanghaiTech UniversityShanghai201210China
| | - Chuyue Zhang
- Shanghai Institute for Advanced Immunochemical StudiesShanghaiTech UniversityShanghai201210China
- School of Life Science and TechnologyShanghaiTech UniversityShanghai201210China
- Institute of Biochemistry and Cell BiologyShanghai Institutes for Biological SciencesChinese Academy of SciencesShanghai200031China
- University of Chinese Academy of SciencesBeijing100049China
| | - Jiaxing Tang
- Shanghai Institute for Advanced Immunochemical StudiesShanghaiTech UniversityShanghai201210China
- School of Life Science and TechnologyShanghaiTech UniversityShanghai201210China
- Institute of Biochemistry and Cell BiologyShanghai Institutes for Biological SciencesChinese Academy of SciencesShanghai200031China
- University of Chinese Academy of SciencesBeijing100049China
| | - Zheng Yu
- Shanghai Institute for Advanced Immunochemical StudiesShanghaiTech UniversityShanghai201210China
| | - Xuekai Zhu
- Shanghai Institute for Advanced Immunochemical StudiesShanghaiTech UniversityShanghai201210China
| | - Kun Fan
- Shanghai Institute for Advanced Immunochemical StudiesShanghaiTech UniversityShanghai201210China
- School of Life Science and TechnologyShanghaiTech UniversityShanghai201210China
- Institute of Biochemistry and Cell BiologyShanghai Institutes for Biological SciencesChinese Academy of SciencesShanghai200031China
- University of Chinese Academy of SciencesBeijing100049China
| | - Guanglei Li
- School of Life Science and TechnologyShanghaiTech UniversityShanghai201210China
| | - Wei Zhu
- Shanghai Institute for Advanced Immunochemical StudiesShanghaiTech UniversityShanghai201210China
| | - Wei Sang
- Department of HematologyThe Affiliated Hospital of Xuzhou Medical UniversityInstitute of HematologyXuzhou Medical UniversityXuzhou221000China
| | - Chenyu Min
- Velox PharmaceuticalsChangzhou213000China
| | - Wenzhang Chen
- Shanghai Institute for Advanced Immunochemical StudiesShanghaiTech UniversityShanghai201210China
| | - Xingxu Huang
- School of Life Science and TechnologyShanghaiTech UniversityShanghai201210China
| | - Guang Yang
- Shanghai Institute for Advanced Immunochemical StudiesShanghaiTech UniversityShanghai201210China
| | | |
Collapse
|
13
|
Benedetti F, Stracke F, Stadlmayr G, Stadlbauer K, Rüker F, Wozniak-Knopp G. Bispecific antibodies with Fab-arms featuring exchanged antigen-binding constant domains. Biochem Biophys Rep 2021; 26:100959. [PMID: 33718630 PMCID: PMC7920882 DOI: 10.1016/j.bbrep.2021.100959] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/16/2020] [Accepted: 02/16/2021] [Indexed: 12/27/2022] Open
Abstract
Monoclonal antibodies can acquire the property of engagement of a second antigen via fusion methods or modification of their CDR loops, but also by modification of their constant domains, such as in the mAb2 format where a set of mutated amino acid residues in the CH3 domains enables a high-affinity specific interaction with the second antigen. We tested the possibility of introducing multiple binding sites for the second antigen by replacing the Fab CH1/CL domain pair with a pair of antigen-binding CH3 domains in a model scaffold with trastuzumab variable domains and VEGF-binding CH3 domains. Such bispecific molecules were produced in a “Fab-like” format and in a full-length antibody format. Novel constructs were of expected molecular composition using mass spectrometry. They were expressed at a high level in standard laboratory conditions, purified as monomers with Protein A and gel filtration and were of high thermostability. Their high-affinity binding to both target antigens was retained. Finally, the Her2/VEGF binding domain-exchanged bispecific antibody was able to mediate a potentiated surface Her2-internalization effect on the Her2-overexpressing cell line SK-BR-3 due to improved level of cross-linking with the endogenously secreted cytokine. To conclude, bispecific antibodies with Fabs featuring exchanged antigen-binding CH3 domains offer an alternative solution in positioning and valency of antigen binding sites. Fab constant domains can be efficiently exchanged for antigen-binding CH3 domains. Such mutagenesis results in bispecific antibodies with correct chain pairing. Domain-exchanged bispecific Fab- and IgG-like formats are of favorable biophysical properties. Resulting bispecific antibodies show high-affinity binding to both target antigens.
Collapse
Key Words
- Ab, antibody
- BLI, biolayer interferometry
- BSA, bovine serum albumin
- Bispecific antibody
- CDR, complementarity determining region
- DSC, differential scanning calorimetry
- Domain-exchanged antibody
- EC50, half-maximal effective concentration
- FBS, fetal bovine serum
- FITC, fluorescein isothiocyanate
- Fab constant domain exchange
- Fab, fragment antigen binding
- Fc, fragment crystallizable
- Fcab, Fc with antigen binding properties
- HPLC-SEC, high pressure liquid chromatography-size exclusion chromatography
- Her2 internalization
- IgG, immunoglobulin G
- LC-ESI-MS, liquid chromatography-electrospray ionization-mass spectrometry
- PBS, phosphate buffered saline
- PE, phycoerythrin
- PEI, polyethylenimine
- PNGase F, Peptide:N-glycosidase F
- RMSD, root mean square deviation
- TRA, trastuzumab
- Tm, melting temperature
- VEGF, vascular endothelial growth factor
- “Knobs-into-holes” heterodimerization
Collapse
Affiliation(s)
- Filippo Benedetti
- CD Laboratory for Innovative Immunotherapeutics, Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Muthgasse 18, 1190, Vienna, Austria
| | - Florian Stracke
- CD Laboratory for Innovative Immunotherapeutics, Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Muthgasse 18, 1190, Vienna, Austria
| | - Gerhard Stadlmayr
- CD Laboratory for Innovative Immunotherapeutics, Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Muthgasse 18, 1190, Vienna, Austria
| | - Katharina Stadlbauer
- CD Laboratory for Innovative Immunotherapeutics, Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Muthgasse 18, 1190, Vienna, Austria
| | - Florian Rüker
- CD Laboratory for Innovative Immunotherapeutics, Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Muthgasse 18, 1190, Vienna, Austria
| | - Gordana Wozniak-Knopp
- CD Laboratory for Innovative Immunotherapeutics, Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Muthgasse 18, 1190, Vienna, Austria
| |
Collapse
|
14
|
Mohammadi M, Jeddi-Tehrani M, Golsaz-Shirazi F, Arjmand M, Bahadori T, Judaki MA, Shiravi F, Zare HA, Haghighat FN, Mobini M, Amiri MM, Shokri F. A Novel Anti-HER2 Bispecific Antibody With Potent Tumor Inhibitory Effects In Vitro and In Vivo. Front Immunol 2021; 11:600883. [PMID: 33679691 PMCID: PMC7927792 DOI: 10.3389/fimmu.2020.600883] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 12/23/2020] [Indexed: 01/14/2023] Open
Abstract
Overexpression of HER2 has been reported in many types of cancer, making it a perfect candidate for targeted immunotherapy. The combination of two FDA approved monoclonal antibodies (mAbs), trastuzumab and pertuzumab, has more robust anti-tumor activity in patients with HER2-overexpressing breast cancer. We recently produced a new humanized anti-HER2 mAb, hersintuzumab, which recognizes a different epitope than trastuzumab and pertuzumab on HER2. This mAb, in combination with trastuzumab, exhibits more potent anti-tumor activity than each parental mAb alone. Here we have developed a novel bispecific anti-HER2 antibody (BsAb) designated as trasintuzumab, composed of trastuzumab and hersintuzumab, using dual variable domain immunoglobulin (DVD-Ig) technology. Both variable domains of trasintuzumab are fully functional and have similar affinities to the parental mAbs and are also able to bind to natural HER2 on the surface of several HER2-expressing cell lines. Trasintuzumab was found to inhibit the growth of different types of tumor cell lines through suppression of the AKT and ERK signaling pathways as efficiently as the combination of the parental mAbs. It also induced tumor regression as potently as the combination of the two mAbs in nude mice bearing ovarian and gastric cancer xenografts. Our data suggest that trasintuzumab may be a promising BsAb therapeutic candidate for the treatment of HER2-overexpressing cancers.
Collapse
Affiliation(s)
- Mehdi Mohammadi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmood Jeddi-Tehrani
- Monoclonal Antibody Research Center, Avicenna Research Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Forough Golsaz-Shirazi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Arjmand
- Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran
| | - Tannaz Bahadori
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Judaki
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Fariba Shiravi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Hengameh Ahmadi Zare
- Monoclonal Antibody Research Center, Avicenna Research Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Farzaneh Notash Haghighat
- Monoclonal Antibody Research Center, Avicenna Research Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Maryam Mobini
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mehdi Amiri
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Fazel Shokri
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Yamashita-Kashima Y, Shu S, Osada M, Fujimura T, Yoshiura S, Harada N, Yoshimura Y. Combination efficacy of pertuzumab and trastuzumab for trastuzumab emtansine-resistant cells exhibiting attenuated lysosomal trafficking or efflux pumps upregulation. Cancer Chemother Pharmacol 2020; 86:641-654. [PMID: 32997196 PMCID: PMC7561595 DOI: 10.1007/s00280-020-04138-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 09/04/2020] [Indexed: 11/28/2022]
Abstract
Purpose Trastuzumab emtansine (T-DM1) is the standard treatment in the current second-line therapy of human epidermal growth factor receptor 2 (HER2)-positive metastatic breast cancer. However, a useful therapy after T-DM1 resistance has not been established. In this study, we established two different HER2-positive T-DM1-resistant cancer cells and evaluated the antitumor effect of trastuzumab in combination with pertuzumab (TRAS + PER). Methods Single-cell-cloned OE19 and BT-474 cells were cultured with increasing concentrations of T-DM1 to generate T-DM1-resistant OE19bTDR and BT-474bTDR cells, respectively. HER2 expression was assessed by immunohistochemistry. Multidrug resistance proteins (MDR1 and MRP1) were evaluated by real-time polymerase chain reaction and western blotting. Intracellular trafficking of T-DM1 was examined by flow cytometry and immunofluorescence staining. Efficacy of TRAS + PER was evaluated by cell proliferation assay, HER3 and AKT phosphorylation, caspase 3/7 activity, and antitumor activity. Results HER2 expression of both resistant cells was equivalent to that of the parent cells. Overexpression of MDR1 and MRP1 was observed and affected the T-DM1 sensitivity in the OE19bTDR cells. Abnormal localization of T-DM1 into the lysosomes was observed in the BT-474bTDR cells. In BT-474bTDR cells, TRAS + PER inhibited the phosphorylation of AKT involved in HER2–HER3 signaling, and apoptosis induction and cell proliferation inhibition were significantly higher with TRAS + PER than with the individual drugs. TRAS + PER significantly suppressed tumor growth in the OE19bTDR xenograft model compared with each single agent. Conclusions The results suggest that the TRAS + PER combination may be effective in T-DM1-resistant cancer cells where HER2 overexpression is maintained. Electronic supplementary material The online version of this article (10.1007/s00280-020-04138-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yoriko Yamashita-Kashima
- Product Research Department, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa, 247-8530, Japan
| | - Sei Shu
- Product Research Department, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa, 247-8530, Japan
| | - Masahiro Osada
- Product Research Department, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa, 247-8530, Japan
| | - Takaaki Fujimura
- Product Research Department, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa, 247-8530, Japan
| | - Shigeki Yoshiura
- Product Research Department, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa, 247-8530, Japan
| | - Naoki Harada
- Product Research Department, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa, 247-8530, Japan.
| | - Yasushi Yoshimura
- Product Research Department, Chugai Pharmaceutical Co., Ltd., 200 Kajiwara, Kamakura, Kanagawa, 247-8530, Japan
| |
Collapse
|
16
|
Skeie M, Nikolaysen F, Chitano Y, Stang E. Hsp90 inhibition and co-incubation with pertuzumab induce internalization and degradation of trastuzumab: Implications for use of T-DM1. J Cell Mol Med 2020; 24:10258-10262. [PMID: 32672902 PMCID: PMC7520337 DOI: 10.1111/jcmm.15643] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/11/2020] [Accepted: 06/28/2020] [Indexed: 12/15/2022] Open
Abstract
The receptor tyrosine kinase HER2 is associated with a number of human malignancies and is an important therapeutic target. The antibody-drug conjugate trastuzumab emtansine (T-DM1; Kadcyla® ) is recommended as a first-line treatment for patients with HER2-positive metastatic breast cancer. T-DM1 combines the antibody-induced effects of the anti-HER2 antibody trastuzumab (Herceptin® ) with the cytotoxic effect of the tubulin inhibitor mertansine (DM1). For DM1 to have effect, the T-DM1-HER2 complex has to be internalized and the trastuzumab part of T-DM1 has to be degraded. HER2 is, however, considered endocytosis-resistant. As a result of this, trastuzumab is only internalized to a highly limited extent, and if internalized, it is rapidly recycled. The exact reasons for the endocytosis resistance of HER2 are not clear, but it is stabilized by heat-shock protein 90 (Hsp90) and Hsp90 inhibitors induce internalization and degradation of HER2. HER2 can also be internalized upon activation of protein kinase C, and contrary to trastuzumab alone, the combination of two or more anti-HER2 antibodies can induce efficient internalization and degradation of HER2. With intention to find ways to improve the action of T-DM1, we investigated how different ways of inducing HER2 internalization leads to degradation of trastuzumab. The results show that although both Hsp90 inhibition and activation of protein kinase C induce internalization of trastuzumab, only Hsp90 inhibition induces degradation. Furthermore, we find that antibody internalization and degradation are increased when trastuzumab is combined with the clinically approved anti-HER2 antibody pertuzumab (Perjeta® ).
Collapse
Affiliation(s)
- Marianne Skeie
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Filip Nikolaysen
- Department of Pathology, Oslo University Hospital, Oslo, Norway.,Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| | - Ylenia Chitano
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Espen Stang
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
17
|
Leyton JV. Improving Receptor-Mediated Intracellular Access and Accumulation of Antibody Therapeutics-The Tale of HER2. Antibodies (Basel) 2020; 9:E32. [PMID: 32668710 PMCID: PMC7551051 DOI: 10.3390/antib9030032] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/03/2020] [Accepted: 07/08/2020] [Indexed: 12/14/2022] Open
Abstract
Therapeutic anti-HER2 antibodies and antibody-drug conjugates (ADCs) have undoubtedly benefitted patients. Nonetheless, patients ultimately relapse-some sooner than others. Currently approved anti-HER2 drugs are expensive and their cost-effectiveness is debated. There is increased awareness that internalization and lysosomal processing including subsequent payload intracellular accumulation and retention for ADCs are critical therapeutic attributes. Although HER2 preferential overexpression on the surface of tumor cells is attractive, its poor internalization and trafficking to lysosomes has been linked to poor therapeutic outcomes. To help address such issues, this review will comprehensively detail the most relevant findings on internalization and cellular accumulation for approved and investigational anti-HER2 antibodies and ADCs. The improved clarity of the HER2 system could improve antibody and ADC designs and approaches for next-generation anti-HER2 and other receptor targeting agents.
Collapse
Affiliation(s)
- Jeffrey V Leyton
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Centre Hospitalier Universitaire de Sherbrooke (CHUS), Université de Sherbrooke, Sherbrooke, QC J1H5N4, Canada
| |
Collapse
|
18
|
DaSilva JO, Yang K, Perez Bay AE, Andreev J, Ngoi P, Pyles E, Franklin MC, Dudgeon D, Rafique A, Dore A, Delfino FJ, Potocky TB, Babb R, Chen G, MacDonald D, Olson WC, Thurston G, Daly C. A Biparatopic Antibody That Modulates MET Trafficking Exhibits Enhanced Efficacy Compared with Parental Antibodies in MET-Driven Tumor Models. Clin Cancer Res 2019; 26:1408-1419. [PMID: 31848185 DOI: 10.1158/1078-0432.ccr-19-2428] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 11/01/2019] [Accepted: 12/11/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Recent clinical data demonstrate that tumors harboring MET genetic alterations (exon 14 skip mutations and/or gene amplification) respond to small-molecule tyrosine kinase inhibitors, validating MET as a therapeutic target. Although antibody-mediated blockade of the MET pathway has not been successful in the clinic, the failures are likely the result of inadequate patient selection strategies as well as suboptimal antibody design. Thus, our goal was to generate a novel MET blocking antibody with enhanced efficacy. EXPERIMENTAL DESIGN Here, we describe the activity of a biparatopic MET×MET antibody that recognizes two distinct epitopes in the MET Sema domain. We use a combination of in vitro assays and tumor models to characterize the effect of our antibody on MET signaling, MET intracellular trafficking, and the growth of MET-dependent cells/tumors. RESULTS In MET-driven tumor models, our biparatopic antibody exhibits significantly better activity than either of the parental antibodies or the mixture of the two parental antibodies and outperforms several clinical-stage MET antibodies. Mechanistically, the biparatopic antibody inhibits MET recycling, thereby promoting lysosomal trafficking and degradation of MET. In contrast to the parental antibodies, the biparatopic antibody fails to activate MET-dependent biological responses, consistent with the observation that it recycles inefficiently and induces very transient downstream signaling. CONCLUSIONS Our results provide strong support for the notion that biparatopic antibodies are a promising therapeutic modality, potentially having greater efficacy than that predicted from the properties of the parental antibodies.
Collapse
Affiliation(s)
| | - Katie Yang
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | | | | | - Peter Ngoi
- UC Santa Cruz, Program for Biomedical Sciences and Engineering, Santa Cruz, California
| | - Erica Pyles
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | | | - Drew Dudgeon
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | | | - Anthony Dore
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | | | | | - Robert Babb
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | - Gang Chen
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | | | | | | | | |
Collapse
|
19
|
Bonelli P, Borrelli A, Tuccillo FM, Silvestro L, Palaia R, Buonaguro FM. Precision medicine in gastric cancer. World J Gastrointest Oncol 2019; 11:804-829. [PMID: 31662821 PMCID: PMC6815928 DOI: 10.4251/wjgo.v11.i10.804] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 07/11/2019] [Accepted: 09/05/2019] [Indexed: 02/05/2023] Open
Abstract
Gastric cancer (GC) is a complex disease linked to a series of environmental factors and unhealthy lifestyle habits, and especially to genetic alterations. GC represents the second leading cause of cancer-related deaths worldwide. Its onset is subtle, and the majority of patients are diagnosed once the cancer is already advanced. In recent years, there have been innovations in the management of advanced GC including the introduction of new classifications based on its molecular characteristics. Thanks to new technologies such as next-generation sequencing and microarray, the Cancer Genome Atlas and Asian Cancer Research Group classifications have also paved the way for precision medicine in GC, making it possible to integrate diagnostic and therapeutic methods. Among the objectives of the subdivision of GC into subtypes is to select patients in whom molecular targeted drugs can achieve the best results; many lines of research have been initiated to this end. After phase III clinical trials, trastuzumab, anti-Erb-B2 receptor tyrosine kinase 2 (commonly known as ERBB2) and ramucirumab, anti-vascular endothelial growth factor receptor 2 (commonly known as VEGFR2) monoclonal antibodies, were approved and introduced into first- and second-line therapies for patients with advanced/metastatic GC. However, the heterogeneity of this neoplasia makes the practical application of such approaches difficult. Unfortunately, scientific progress has not been matched by progress in clinical practice in terms of significant improvements in prognosis. Survival continues to be low in contrast to the reduction in deaths from many common cancers such as colorectal, lung, breast, and prostate cancers. Although several target molecules have been identified on which targeted drugs can act and novel products have been introduced into experimental therapeutic protocols, the overall approach to treating advanced stage GC has not substantially changed. Currently, surgical resection with adjuvant or neoadjuvant radiotherapy and chemotherapy are the most effective treatments for this disease. Future research should not underestimate the heterogeneity of GC when developing diagnostic and therapeutic strategies aimed toward improving patient survival.
Collapse
Affiliation(s)
- Patrizia Bonelli
- Molecular Biology and Viral Oncology, Istituto Nazionale Tumori - IRCCS - Fondazione G Pascale, Napoli 80131, Italy
| | - Antonella Borrelli
- Molecular Biology and Viral Oncology, Istituto Nazionale Tumori - IRCCS - Fondazione G Pascale, Napoli 80131, Italy
| | - Franca Maria Tuccillo
- Molecular Biology and Viral Oncology, Istituto Nazionale Tumori - IRCCS - Fondazione G Pascale, Napoli 80131, Italy
| | - Lucrezia Silvestro
- Abdominal Medical Oncology, Istituto Nazionale Tumori - IRCCS - Fondazione G Pascale, Napoli 80131, Italy
| | - Raffaele Palaia
- Gastro-pancreatic Surgery Division, Istituto Nazionale Tumori - IRCCS - Fondazione G Pascale, Napoli 80131, Italy
| | - Franco Maria Buonaguro
- Molecular Biology and Viral Oncology, Istituto Nazionale Tumori - IRCCS - Fondazione G Pascale, Napoli 80131, Italy
| |
Collapse
|
20
|
Sampera A, Sánchez-Martín FJ, Arpí O, Visa L, Iglesias M, Menéndez S, Gaye É, Dalmases A, Clavé S, Gelabert-Baldrich M, Poulsen TT, Kragh M, Bellosillo B, Albanell J, Rovira A, Montagut C. HER-Family Ligands Promote Acquired Resistance to Trastuzumab in Gastric Cancer. Mol Cancer Ther 2019; 18:2135-2145. [DOI: 10.1158/1535-7163.mct-19-0455] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/28/2019] [Accepted: 08/27/2019] [Indexed: 11/16/2022]
|
21
|
Mathematical modeling of drug-induced receptor internalization in the HER2-positive SKBR3 breast cancer cell-line. Sci Rep 2019; 9:12709. [PMID: 31481718 PMCID: PMC6722142 DOI: 10.1038/s41598-019-49019-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 08/19/2019] [Indexed: 12/22/2022] Open
Abstract
About 20% of breast cancer tumors over-express the HER2 receptor. Trastuzumab, an approved drug to treat this type of breast cancer, is a monoclonal antibody directly binding at the HER2 receptor and ultimately inhibiting cancer cell growth. The goal of our study was to understand the early impact of trastuzumab on HER2 internalization and recycling in the HER2-overexpressing breast cancer cell line SKBR3. To this end, fluorescence microscopy, monitoring the amount of HER2 expression in the plasma membrane, was combined with mathematical modeling to derive the flux of HER2 receptors from and to the membrane. We constructed a dynamic multi-compartment model based on ordinary differential equations. To account for cancer cell heterogeneity, a first, dynamic model was expanded to a second model including two distinct cell phenotypes, with implications for different conformational states of HER2, i.e. monomeric or homodimeric. Our mathematical model shows that the hypothesis of fast constitutive HER2 recycling back to the plasma membrane does not match the experimental data. It conclusively describes the experimental observation that trastuzumab induces sustained receptor internalization in cells with membrane ruffles. It is also concluded that for rare, non-ruffled (flat) cells, HER2 internalization occurs three orders of magnitude slower than for the bulk, ruffled cell population.
Collapse
|
22
|
Crosby EJ, Gwin W, Blackwell K, Marcom PK, Chang S, Maecker HT, Broadwater G, Hyslop T, Kim S, Rogatko A, Lubkov V, Snyder JC, Osada T, Hobeika AC, Morse MA, Lyerly HK, Hartman ZC. Vaccine-Induced Memory CD8 + T Cells Provide Clinical Benefit in HER2 Expressing Breast Cancer: A Mouse to Human Translational Study. Clin Cancer Res 2019; 25:2725-2736. [PMID: 30635338 PMCID: PMC6497539 DOI: 10.1158/1078-0432.ccr-18-3102] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/28/2018] [Accepted: 01/08/2019] [Indexed: 01/23/2023]
Abstract
PURPOSE Immune-based therapy for metastatic breast cancer has had limited success, particularly in molecular subtypes with low somatic mutations rates. Strategies to augment T-cell infiltration of tumors include vaccines targeting established oncogenic drivers such as the genomic amplification of HER2. We constructed a vaccine based on a novel alphaviral vector encoding a portion of HER2 (VRP-HER2). PATIENTS AND METHODS In preclinical studies, mice were immunized with VRP-HER2 before or after implantation of hHER2+ tumor cells and HER2-specific immune responses and antitumor function were evaluated. We tested VRP-HER2 in a phase I clinical trial where subjects with advanced HER2-overexpressing malignancies in cohort 1 received VRP-HER2 every 2 weeks for a total of 3 doses. In cohort 2, subjects received the same schedule concurrently with a HER2-targeted therapy. RESULTS Vaccination in preclinical models with VRP-HER2 induced HER2-specific T cells and antibodies while inhibiting tumor growth. VRP-HER2 was well tolerated in patients and vaccination induced HER2-specific T cells and antibodies. Although a phase I study, there was 1 partial response and 2 patients with continued stable disease. Median OS was 50.2 months in cohort 1 (n = 4) and 32.7 months in cohort 2 (n = 18). Perforin expression by memory CD8 T cells post-vaccination significantly correlated with improved PFS. CONCLUSIONS VRP-HER2 increased HER2-specific memory CD8 T cells and had antitumor effects in preclinical and clinical studies. The expansion of HER2-specific memory CD8 T cells in vaccinated patients was significantly correlated with increased PFS. Subsequent studies will seek to enhance T-cell activity by combining with anti-PD-1.
Collapse
Affiliation(s)
- Erika J Crosby
- Department of Surgery, Division of Surgical Sciences, Duke University Medical Center, Durham, North Carolina
| | - William Gwin
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, North Carolina
- Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, Washington
| | - Kimberly Blackwell
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, North Carolina
| | - Paul K Marcom
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, North Carolina
| | - Serena Chang
- Department of Microbiology and Immunology, Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, California
| | - Holden T Maecker
- Department of Microbiology and Immunology, Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, California
| | - Gloria Broadwater
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, North Carolina
| | - Terry Hyslop
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, North Carolina
| | - Sungjin Kim
- Department of Biomedical Sciences, Biostatistics and Bioinformatics Research Center, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Andre Rogatko
- Department of Biomedical Sciences, Biostatistics and Bioinformatics Research Center, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Veronica Lubkov
- Department of Surgery, Division of Surgical Sciences, Duke University Medical Center, Durham, North Carolina
| | - Joshua C Snyder
- Department of Surgery, Division of Surgical Sciences, Duke University Medical Center, Durham, North Carolina
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina
| | - Takuya Osada
- Department of Surgery, Division of Surgical Sciences, Duke University Medical Center, Durham, North Carolina
| | - Amy C Hobeika
- Department of Surgery, Division of Surgical Sciences, Duke University Medical Center, Durham, North Carolina
| | - Michael A Morse
- Department of Surgery, Division of Surgical Sciences, Duke University Medical Center, Durham, North Carolina
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, North Carolina
| | - H Kim Lyerly
- Department of Surgery, Division of Surgical Sciences, Duke University Medical Center, Durham, North Carolina.
| | - Zachary C Hartman
- Department of Surgery, Division of Surgical Sciences, Duke University Medical Center, Durham, North Carolina.
| |
Collapse
|
23
|
Wang H, Wang Y, Xiao Z, Li W, Dimitrov DS, Chen W. Human Domain Antibodies to Conserved Epitopes on HER2 Potently Inhibit Growth of HER2-Overexpressing Human Breast Cancer Cells In Vitro. Antibodies (Basel) 2019; 8:antib8010025. [PMID: 31544831 PMCID: PMC6640707 DOI: 10.3390/antib8010025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 03/05/2019] [Accepted: 03/12/2019] [Indexed: 12/18/2022] Open
Abstract
The FDA approval of two anti-HER2 monoclonal antibodies, trastuzumab and pertuzumab, and an antibody-drug conjugate, trastuzumab emtansine, has transformed clinical practice for HER2-positive cancers. However, not all patients respond to therapy, and the majority of responders eventually develop resistance. In addition, cardiotoxicity is a major safety concern for their clinical use. Thus, there remains a need for the discovery and development of novel classes of HER2-targeted therapeutics with high efficacy and specificity. In this study, we report the identification and characterization of fully human single-domain antibodies (dAbs) targeting HER2 epitopes that are highly conserved among various species and non-overlapping with those of trastuzumab and pertuzumab. An Fc-fusion protein of the best binder demonstrated much higher inhibitory activity against HER2-amplified human breast cancer cell lines than trastuzumab and pertuzumab. Unlike the latter, however, it did not have an effect on gastric and ovarian cancer cell lines with HER2 overexpression. The dAb-Fc fusion protein showed poor pharmacokinetics in mice, thus limiting its potential for therapeutic use. It could be useful as an agent for the exploration of functionally important conserved structures on HER2 with implications for the design of novel therapeutics and elucidation of mechanisms of HER2-mediated tumorigenesis.
Collapse
Affiliation(s)
- Hongqian Wang
- The Second Clinical Medicine School, Zhejiang Chinese Medical University, Hangzhou 310005, China.
- Zhejiang Shimai Pharmaceutical Co., Ltd., Hangzhou 311100, China.
| | - Yanping Wang
- CentryMed Pharmaceutical Inc., a subsidiary of Zhejiang Shimai Pharmaceutical Co., Ltd., Frederick, MD 21704, USA.
| | - Zuoxiang Xiao
- Zhejiang Shimai Pharmaceutical Co., Ltd., Hangzhou 311100, China.
- CentryMed Pharmaceutical Inc., a subsidiary of Zhejiang Shimai Pharmaceutical Co., Ltd., Frederick, MD 21704, USA.
| | - Wei Li
- Center for Antibody Therapeutics, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | - Dimiter S Dimitrov
- Center for Antibody Therapeutics, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | - Weizao Chen
- Zhejiang Shimai Pharmaceutical Co., Ltd., Hangzhou 311100, China.
- CentryMed Pharmaceutical Inc., a subsidiary of Zhejiang Shimai Pharmaceutical Co., Ltd., Frederick, MD 21704, USA.
| |
Collapse
|
24
|
Soltantoyeh T, Bahadori T, Hosseini-Ghatar R, Khoshnoodi J, Roohi A, Mobini M, Golsaz-Shirazi F, Jeddi-Tehrani M, Amiri MM, Shokri F. Differential Effects of Inhibitory and Stimulatory Anti-HER2 Monoclonal Antibodies on AKT/ERK Signaling Pathways. Asian Pac J Cancer Prev 2018; 19:2255-2262. [PMID: 30139234 PMCID: PMC6171393 DOI: 10.22034/apjcp.2018.19.8.2255] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Objective: Homo- and heterodimerization of the receptor tyrosine kinase HER2 hyperactivate several downstream signaling pathways, leading to uncontrolled growth and proliferation of tumor cells. Anti-HER2 monoclonal antibodies (mAbs) may induce different effects on HER2 dimerization and signaling. Methods: The effect of two inhibitory (2A8, 1T0) and one stimulatory (1H9) anti-HER2 mAbs either alone or in combination with trastuzumab was investigated on AKT and ERK signaling pathways and HER2 degradation in a human breast cancer cell line (BT-474) by Western blotting. Result: While 1H9 mAb had no significant effect on AKT and ERK signaling pathways, 1T0 and 2A8 mAbs inhibited phosphorylation of both pathways. Combination of 1T0 mAb with trastuzumab resulted in significant synergistic inhibition of both pathways and HER2 degradation, much more potently than the combination of trastuzumab and pertuzumab. Conclusion: Our data indicate that anti-HER2 mAbs may induce different signaling pathways depending on their effect on tumor cell growth and proliferation. The significant inhibition of ERK and AKT phosphorylation by 1T0 alone or particularly in combination with trastuzumab suggests its potential therapeutic application for targeted immunotherapy of HER2 overexpressing malignancies.
Collapse
Affiliation(s)
- Tahereh Soltantoyeh
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran. and
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Pollmann SE, Calvert VS, Rao S, Boca SM, Madhavan S, Horak ID, Kjaer A, Petricoin EF, Kragh M, Poulsen TT. Acquired Resistance to a MET Antibody In Vivo Can Be Overcome by the MET Antibody Mixture Sym015. Mol Cancer Ther 2018; 17:1259-1270. [DOI: 10.1158/1535-7163.mct-17-0787] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 01/08/2018] [Accepted: 03/07/2018] [Indexed: 11/16/2022]
|
26
|
Deci MB, Liu M, Dinh QT, Nguyen J. Precision engineering of targeted nanocarriers. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2018; 10:e1511. [PMID: 29436157 DOI: 10.1002/wnan.1511] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 12/11/2017] [Accepted: 01/16/2018] [Indexed: 12/15/2022]
Abstract
Since their introduction in 1980, the number of advanced targeted nanocarrier systems has grown considerably. Nanocarriers capable of targeting single receptors, multiple receptors, or multiple epitopes have all been used to enhance delivery efficiency and selectivity. Despite tremendous progress, preclinical studies and clinically translatable nanotechnology remain disconnected. The disconnect in targeting efficacy may stem from poorly-understood factors such as receptor clustering, spatial control of targeting ligands, ligand mobility, and ligand architecture. Further, the relationship between receptor distribution and ligand architecture remains elusive. Traditionally, targeted nanocarriers were engineered assuming a "static" target. However, it is becoming increasingly clear that receptor expression patterns change in response to external stimuli and disease progression. Here, we discuss how cutting-edge technologies will enable a better characterization of the spatiotemporal distribution of membrane receptors and their clustering. We further describe how this will enable the design of new nanocarriers that selectively target the site of disease. Ultimately, we explore how the precision engineering of targeted nanocarriers that adapt to receptor dynamics will have the potential to drive nanotechnology to the forefront of therapy and make targeted nanomedicine a clinical reality. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Biology-Inspired Nanomaterials > Lipid-Based Structures Biology-Inspired Nanomaterials > Protein and Virus-Based Structures.
Collapse
Affiliation(s)
- Michael B Deci
- Department of Pharmaceutical Sciences, School of Pharmacy, University at Buffalo, The State University of New York, Buffalo, New York
| | - Maixian Liu
- Department of Pharmaceutical Sciences, School of Pharmacy, University at Buffalo, The State University of New York, Buffalo, New York
| | - Quoc Thai Dinh
- Department of Experimental Pneumology and Allergology, Saarland University Faculty of Medicine, Homburg/Saar, Germany
| | - Juliane Nguyen
- Department of Pharmaceutical Sciences, School of Pharmacy, University at Buffalo, The State University of New York, Buffalo, New York
| |
Collapse
|
27
|
Milazzo FM, Anastasi AM, Chiapparino C, Rosi A, Leoni B, Vesci L, Petronzelli F, De Santis R. AvidinOX-anchored biotinylated trastuzumab and pertuzumab induce down-modulation of ErbB2 and tumor cell death at concentrations order of magnitude lower than not-anchored antibodies. Oncotarget 2017; 8:22590-22605. [PMID: 28186982 PMCID: PMC5410247 DOI: 10.18632/oncotarget.15145] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 01/25/2017] [Indexed: 01/22/2023] Open
Abstract
The oxidized version of Avidin, known as AvidinOX, was previously shown to link to tissue proteins upon injection or nebulization, thus becoming a stable receptor for biotinylated therapeutics. AvidinOX is currently under clinical investigation to target radioactive biotin to inoperable tumor lesions (ClinicalTrials.gov NCT02053324). Presently, we show that the anti-ErbB2 monoclonal antibodies Trastuzumab and Pertuzumab can be chemically biotinylated while maintaining their biochemical and biological properties. By using several and diverse experimental conditions, we show that when AvidinOX is conjugated to tumor cells, low antibody concentrations of biotinylated Trastuzumab (bTrast) or Pertuzumab (bPert) prevent internalization of ErbB2, induce endoplasmic reticulum stress, cell cycle arrest and apoptosis leading to inhibition of proliferation and ErbB2 signaling. Moreover, we found that the treatment is able to induce down-modulation of ErbB2 thus bypassing the known resistance of this receptor to degradation. Interestingly, we show that AvidinOX anchorage is a way to counteract agonistic activities of Trastuzumab and Pertuzumab. Present data are in agreement with previous observations from our group indicating that the engagement of the Epidermal Growth Factor Receptor (EGFR) by AvidinOX-bound biotinylated Cetuximab or Panitumumab, leads to potent tumor inhibition both in vitro and in animal models. All results taken together encourage further investigation of AvidinOX-based treatments with biotinylated antibodies directed to the members of the EGFR family.
Collapse
Affiliation(s)
| | - Anna Maria Anastasi
- Biotech Products, Research and Development, Sigma-Tau SpA, 00071 Pomezia (Rome), Italy
| | - Caterina Chiapparino
- Biotech Products, Research and Development, Sigma-Tau SpA, 00071 Pomezia (Rome), Italy
| | - Antonio Rosi
- Biotech Products, Research and Development, Sigma-Tau SpA, 00071 Pomezia (Rome), Italy
| | - Barbara Leoni
- Biotech Products, Research and Development, Sigma-Tau SpA, 00071 Pomezia (Rome), Italy
| | - Loredana Vesci
- Biotech Products, Research and Development, Sigma-Tau SpA, 00071 Pomezia (Rome), Italy
| | - Fiorella Petronzelli
- Biotech Products, Research and Development, Sigma-Tau SpA, 00071 Pomezia (Rome), Italy
| | - Rita De Santis
- Biotech Products, Research and Development, Sigma-Tau SpA, 00071 Pomezia (Rome), Italy
| |
Collapse
|
28
|
Grandal MM, Havrylov S, Poulsen TT, Koefoed K, Dahlman A, Galler GR, Conrotto P, Collins S, Eriksen KW, Kaufman D, Woude GF, Jacobsen HJ, Horak ID, Kragh M, Lantto J, Bouquin T, Park M, Pedersen MW. Simultaneous Targeting of Two Distinct Epitopes on MET Effectively Inhibits MET- and HGF-Driven Tumor Growth by Multiple Mechanisms. Mol Cancer Ther 2017; 16:2780-2791. [DOI: 10.1158/1535-7163.mct-17-0374] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/03/2017] [Accepted: 08/02/2017] [Indexed: 11/16/2022]
|
29
|
Poulsen TT, Grandal MM, Skartved NJØ, Hald R, Alifrangis L, Koefoed K, Lindsted T, Fröhlich C, Pollmann SE, Eriksen KW, Dahlman A, Jacobsen HJ, Bouquin T, Pedersen MW, Horak ID, Lantto J, Kragh M. Sym015: A Highly Efficacious Antibody Mixture against MET-Amplified Tumors. Clin Cancer Res 2017; 23:5923-5935. [PMID: 28679766 DOI: 10.1158/1078-0432.ccr-17-0782] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 06/01/2017] [Accepted: 06/29/2017] [Indexed: 02/03/2023]
Abstract
Purpose: Activation of the receptor tyrosine kinase MET is associated with poor clinical outcome in certain cancers. To target MET more effectively, we developed an antagonistic antibody mixture, Sym015, consisting of two humanized mAbs directed against nonoverlapping epitopes of MET.Experimental Design/Results: We screened a large panel of well-annotated human cancer cell lines and identified a subset with highly elevated MET expression. In particular, cell lines of lung cancer and gastric cancer origin demonstrated high MET expression and activation, and Sym015 triggered degradation of MET and significantly inhibited growth of these cell lines. Next, we tested Sym015 in patient- and cell line-derived xenograft models with high MET expression and/or MET exon 14 skipping alterations, and in models harboring MET amplification as a mechanism of resistance to EGFR-targeting agents. Sym015 effectively inhibited tumor growth in all these models and was superior to an analogue of emibetuzumab, a monoclonal IgG4 antibody against MET currently in clinical development. Sym015 also induced antibody-dependent cellular cytotoxicity (ADCC) in vitro, suggesting that secondary effector functions contribute to the efficacy of Sym015.Retrospectively, all responsive, high MET-expressing models were scored as highly MET-amplified by in situ hybridization, pointing to MET amplification as a predictive biomarker for efficacy. Preclinical toxicology studies in monkeys showed that Sym015 was well tolerated, with a pharmacokinetic profile supporting administration of Sym015 every second or third week in humans.Conclusions: The preclinical efficacy and safety data provide a clear rationale for the ongoing clinical studies of Sym015 in patients with MET-amplified tumors. Clin Cancer Res; 23(19); 5923-35. ©2017 AACR.
Collapse
|
30
|
Superior Suppression of ErbB2-positive Tumor Cells by a Novel Human Triparatopic Tribody. J Immunother 2017; 40:117-128. [DOI: 10.1097/cji.0000000000000152] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
31
|
Twomey JD, Brahme NN, Zhang B. Drug-biomarker co-development in oncology – 20 years and counting. Drug Resist Updat 2017; 30:48-62. [DOI: 10.1016/j.drup.2017.02.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 02/10/2017] [Accepted: 02/17/2017] [Indexed: 12/28/2022]
|
32
|
Sun X, Ackerstaff E, He F, Xing L, Hsiao HT, Koutcher JA, Ling CC, Li GC. Visualizing the antivascular effect of bortezomib on the hypoxic tumor microenvironment. Oncotarget 2016; 6:34732-44. [PMID: 26416246 PMCID: PMC4741486 DOI: 10.18632/oncotarget.5300] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 09/11/2015] [Indexed: 01/07/2023] Open
Abstract
Bortezomib, a novel proteasome inhibitor, has been approved for treating multiple myeloma and mantle cell lymphoma and studied pre-clinically and clinically for solid tumors. Preferential cytotoxicity of bortezomib was found toward hypoxic tumor cells and endothelial cells in vitro. The purpose of this study is to investigate the role of a pretreatment hypoxic tumor microenvironment on the effects of bortezomib in vitro and ex vivo, and explore the feasibility of dynamic contrast enhanced magnetic resonance imaging (DCE MRI) to noninvasively evaluate the biological effects of bortezomib. It was shown in vitro by Western blot, flow cytometry, and ELISA that bortezomib accumulated HIF-1α in non-functional forms and blocks its hypoxia response in human colorectal cancer cell lines. Ex vivo experiments were performed with fluorescent immunohistochemical staining techniques using multiple endogenous and exogenous markers to identify hypoxia (pimonidazole, HRE-TKeGFP), blood flow/permeability (Hoechst 33342), micro-vessels (CD31 and SMA), apoptosis (cleaved caspase 3) and hypoxia response (CA9). After bortezomib administration, overall apoptosis index was significantly increased and blood perfusion was dramatically decreased in tumor xenografts. More importantly, apoptosis signals were found preferentially located in moderate and severe pretreatment hypoxic regions in both tumor and endothelial cells. Meanwhile, DCE MRI examinations showed that the tumor blood flow and permeability decreased significantly after bortezomib administration. The present study revealed that bortezomib reduces tumor hypoxia response and blood perfusion, thus, presenting antivascular properties. It will be important to determine the hypoxic/perfusion status pre- and during treatment at further translational studies.
Collapse
Affiliation(s)
- Xiaorong Sun
- Department of Radiology, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, China.,Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ellen Ackerstaff
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fuqiu He
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ligang Xing
- Department of Radiation Oncology, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, China
| | - Hung Tsung Hsiao
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Current address: Department of Anesthesiology, E-Da Hospital, Yanchau District, Kaohsiung, Taiwan
| | - Jason A Koutcher
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - C Clifton Ling
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gloria C Li
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
33
|
Szymanska M, Fosdahl AM, Nikolaysen F, Pedersen MW, Grandal MM, Stang E, Bertelsen V. A combination of two antibodies recognizing non-overlapping epitopes of HER2 induces kinase activity-dependent internalization of HER2. J Cell Mol Med 2016; 20:1999-2011. [PMID: 27469139 PMCID: PMC5020627 DOI: 10.1111/jcmm.12899] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 05/06/2016] [Indexed: 12/14/2022] Open
Abstract
The human epidermal growth factor receptor 2 (HER2/ErbB2) is overexpressed in a number of human cancers. HER2 is the preferred heterodimerization partner for other epidermal growth factor receptor (EGFR) family members and is considered to be resistant to endocytic down-regulation, properties which both contribute to the high oncogenic potential of HER2. Antibodies targeting members of the EGFR family are powerful tools in cancer treatment and can function by blocking ligand binding, preventing receptor dimerization, inhibiting receptor activation and/or inducing receptor internalization and degradation. With respect to antibody-induced endocytosis of HER2, various results are reported, and the effect seems to depend on the HER2 expression level and whether antibodies are given as individual antibodies or as mixtures of two or more. In this study, the effect of a mixture of two monoclonal antibodies against non-overlapping epitopes of HER2 was investigated with respect to localization and stability of HER2. Individual antibodies had limited effect, but the combination of antibodies induced internalization and degradation of HER2 by multiple endocytic pathways. In addition, HER2 was phosphorylated and ubiquitinated upon incubation with the antibody combination, and the HER2 kinase activity was found to be instrumental in antibody-induced HER2 down-regulation.
Collapse
Affiliation(s)
- Monika Szymanska
- Department of Pathology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Anne M Fosdahl
- Department of Pathology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Filip Nikolaysen
- Department of Pathology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | | | - Espen Stang
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Vibeke Bertelsen
- Department of Pathology, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
34
|
Ellebaek S, Brix S, Grandal M, Lantto J, Horak ID, Kragh M, Poulsen TT. Pan-HER-An antibody mixture targeting EGFR, HER2 and HER3 abrogates preformed and ligand-induced EGFR homo- and heterodimers. Int J Cancer 2016; 139:2095-105. [PMID: 27342948 DOI: 10.1002/ijc.30242] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 06/01/2016] [Accepted: 06/06/2016] [Indexed: 12/16/2022]
Abstract
The human epidermal growth factor receptor (HER)-family is involved in development of many epithelial cancers. Therefore, HER-family members constitute important targets for anti-cancer therapeutics such as monoclonal antibodies (mAbs). A limitation to the success of single HER-targeting mAbs is development of acquired resistance through mechanisms such as alterted receptor dimerization patterns and dependencies. Pan-HER is a mixture of six mAbs simultaneously targeting epidermal growth factor receptor (EGFR), HER2 and HER3 with two mAbs against each receptor. Pan-HER has previously demonstrated broader efficacy than targeting single or dual receptor combinations also in resistant settings. In light of this broad efficacy, we decided to investigate the effect of Pan-HER compared with single HER-targeting with single and dual mAbs on HER-family cross-talk and dimerization focusing on EGFR. The effect of Pan-HER on cell proliferation and HER-family receptor degradation was superior to treatment with single mAbs targeting either single receptor, and similar to targeting a single receptor with two non-overlapping antibodies. Furthermore, changes in EGFR-dimerization patterns after treatment with Pan-HER were investigated by in situ proximity ligation assay and co-immunoprecipitation, demonstrating that Pan-HER and the EGFR-targeting mAb mixture efficiently down-regulate basal EGFR homo- and heterodimerization in two tested cell lines, whereas single mAbs had limited effects. Pan-HER and the EGFR-targeting mAb mixture also blocked EGF-binding and thereby ligand-induced changes in EGFR-dimerization levels. These results suggest that Pan-HER reduces the cellular capability to switch HER-dependency and dimerization pattern in response to treatment and thus hold promise for future clinical development of Pan-HER in resistant settings.
Collapse
Affiliation(s)
| | - Susanne Brix
- Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Kongens Lyngby, Denmark
| | | | | | | | | | | |
Collapse
|
35
|
Corti D, Kearns JD. Promises and pitfalls for recombinant oligoclonal antibodies-based therapeutics in cancer and infectious disease. Curr Opin Immunol 2016; 40:51-61. [PMID: 26995095 PMCID: PMC7127534 DOI: 10.1016/j.coi.2016.03.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Revised: 02/29/2016] [Accepted: 03/01/2016] [Indexed: 02/08/2023]
Abstract
Monoclonal antibodies (mAbs) have revolutionized the diagnosis and treatment of many human diseases and the application of combinations of mAbs has demonstrated improved therapeutic activity in both preclinical and clinical testing. Combinations of antibodies have several advantages such as the capacities to target multiple and mutating antigens in complex pathogens and to engage varied epitopes on multiple disease-related antigens (e.g. receptors) to overcome heterogeneity and plasticity. Oligoclonal antibodies are an emerging therapeutic format in which a novel antibody combination is developed as a single drug product. Here, we will provide historical context on the use of oligoclonal antibodies in oncology and infectious diseases and will highlight practical considerations related to their preclinical and clinical development programs.
Collapse
Affiliation(s)
| | - Jeffrey D Kearns
- Merrimack Pharmaceuticals, Inc., One Kendall Square, Suite B7201, Cambridge, MA 02139, USA.
| |
Collapse
|
36
|
Gaborit N, Lindzen M, Yarden Y. Emerging anti-cancer antibodies and combination therapies targeting HER3/ERBB3. Hum Vaccin Immunother 2016; 12:576-92. [PMID: 26529100 PMCID: PMC4964743 DOI: 10.1080/21645515.2015.1102809] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 09/11/2015] [Accepted: 09/26/2015] [Indexed: 12/22/2022] Open
Abstract
Cancer progression depends on stepwise accumulation of oncogenic mutations and a select group of growth factors essential for tumor growth, metastasis and angiogenesis. Agents blocking the epidermal growth factor receptor (EGFR, also called HER1 and ERBB1) and the co-receptor called HER2/ERBB2 have been approved over the last decade as anti-cancer drugs. Because the catalytically defective member of the family, HER3/ERBB3, plays critical roles in emergence of resistance of carcinomas to various drugs, current efforts focus on antibodies and other anti-HER3/ERBB3 agents, which we review herein with an emphasis on drug combinations and some unique biochemical features of HER3/ERBB3.
Collapse
Affiliation(s)
- Nadège Gaborit
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Moshit Lindzen
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Yosef Yarden
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
37
|
Dimasi N, Fleming R, Hay C, Woods R, Xu L, Wu H, Gao C. Development of a Trispecific Antibody Designed to Simultaneously and Efficiently Target Three Different Antigens on Tumor Cells. Mol Pharm 2015; 12:3490-501. [PMID: 26176328 DOI: 10.1021/acs.molpharmaceut.5b00268] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Targeting Eph (erythropoietin producing hepatoma) receptors with monoclonal antibodies is being explored as therapy for several types of cancer. To test whether simultaneous targeting of EphA2, EphA4, and EphB4 would be an effective approach to cancer therapy, we generated a recombinant trispecific antibody using the variable domain genes of anti-EphA2, anti-EphA4, and anti-EphB4 monoclonal antibodies. A multidisciplinary approach combining biochemical, biophysical, and cellular-based assays was used to characterize the trispecific antibody in vitro and in vivo. Here we demonstrate that the trispecific antibody is expressed at high levels by mammalian cells, monodispersed in solution, thermostable, capable of simultaneously binding the three receptors, and able to activate the three targets effectively as evidenced by receptor internalization and degradation both in vitro and in vivo. Furthermore, pharmacokinetic analysis using tumor-bearing nude mice showed that the trispecific antibody remains in the circulation similarly to its respective parental antibodies. These results indicate that simultaneous blockade of EphA2, EphA4, and EphB4 could be an attractive approach to cancer therapy.
Collapse
Affiliation(s)
- Nazzareno Dimasi
- Antibody Discovery and Protein Engineering and ‡Oncology Research, MedImmune , Gaithersburg, Maryland 20878, United States
| | - Ryan Fleming
- Antibody Discovery and Protein Engineering and ‡Oncology Research, MedImmune , Gaithersburg, Maryland 20878, United States
| | - Carl Hay
- Antibody Discovery and Protein Engineering and ‡Oncology Research, MedImmune , Gaithersburg, Maryland 20878, United States
| | - Rob Woods
- Antibody Discovery and Protein Engineering and ‡Oncology Research, MedImmune , Gaithersburg, Maryland 20878, United States
| | - Linda Xu
- Antibody Discovery and Protein Engineering and ‡Oncology Research, MedImmune , Gaithersburg, Maryland 20878, United States
| | - Herren Wu
- Antibody Discovery and Protein Engineering and ‡Oncology Research, MedImmune , Gaithersburg, Maryland 20878, United States
| | - Changshou Gao
- Antibody Discovery and Protein Engineering and ‡Oncology Research, MedImmune , Gaithersburg, Maryland 20878, United States
| |
Collapse
|
38
|
Kearns JD, Bukhalid R, Sevecka M, Tan G, Gerami-Moayed N, Werner SL, Kohli N, Burenkova O, Sloss CM, King AM, Fitzgerald JB, Nielsen UB, Wolf BB. Enhanced Targeting of the EGFR Network with MM-151, an Oligoclonal Anti-EGFR Antibody Therapeutic. Mol Cancer Ther 2015; 14:1625-36. [PMID: 25911688 DOI: 10.1158/1535-7163.mct-14-0772] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 04/17/2015] [Indexed: 12/16/2022]
Abstract
Although EGFR is a validated therapeutic target across multiple cancer indications, the often modest clinical responses to current anti-EGFR agents suggest the need for improved therapeutics. Here, we demonstrate that signal amplification driven by high-affinity EGFR ligands limits the capacity of monoclonal anti-EGFR antibodies to block pathway signaling and cell proliferation and that these ligands are commonly coexpressed with low-affinity EGFR ligands in epithelial tumors. To develop an improved antibody therapeutic capable of overcoming high-affinity ligand-mediated signal amplification, we used a network biology approach comprised of signaling studies and computational modeling of receptor-antagonist interactions. Model simulations suggested that an oligoclonal antibody combination may overcome signal amplification within the EGFR:ERK pathway driven by all EGFR ligands. Based on this, we designed MM-151, a combination of three fully human IgG1 monoclonal antibodies that can simultaneously engage distinct, nonoverlapping epitopes on EGFR with subnanomolar affinities. In signaling studies, MM-151 antagonized high-affinity EGFR ligands more effectively than cetuximab, leading to an approximately 65-fold greater decrease in signal amplification to ERK. In cell viability studies, MM-151 demonstrated antiproliferative activity against high-affinity EGFR ligands, either singly or in combination, while cetuximab activity was largely abrogated under these conditions. We confirmed this finding both in vitro and in vivo in a cell line model of autocrine high-affinity ligand expression. Together, these preclinical studies provide rationale for the clinical study of MM-151 and suggest that high-affinity EGFR ligand expression may be a predictive response marker that distinguishes MM-151 from other anti-EGFR therapeutics.
Collapse
Affiliation(s)
| | | | - Mark Sevecka
- Merrimack Pharmaceuticals, Cambridge, Massachusetts
| | - Gege Tan
- Merrimack Pharmaceuticals, Cambridge, Massachusetts
| | | | | | - Neeraj Kohli
- Merrimack Pharmaceuticals, Cambridge, Massachusetts
| | | | | | - Anne M King
- Merrimack Pharmaceuticals, Cambridge, Massachusetts
| | | | | | - Beni B Wolf
- Merrimack Pharmaceuticals, Cambridge, Massachusetts
| |
Collapse
|
39
|
Jacobsen HJ, Poulsen TT, Dahlman A, Kjær I, Koefoed K, Sen JW, Weilguny D, Bjerregaard B, Andersen CR, Horak ID, Pedersen MW, Kragh M, Lantto J. Pan-HER, an Antibody Mixture Simultaneously Targeting EGFR, HER2, and HER3, Effectively Overcomes Tumor Heterogeneity and Plasticity. Clin Cancer Res 2015; 21:4110-22. [PMID: 25908781 DOI: 10.1158/1078-0432.ccr-14-3312] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 04/01/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Accumulating evidence indicates a high degree of plasticity and compensatory signaling within the human epidermal growth factor receptor (HER) family, leading to resistance upon therapeutic intervention with HER family members. EXPERIMENTAL DESIGN/RESULTS We have generated Pan-HER, a mixture of six antibodies targeting each of the HER family members EGFR, HER2, and HER3 with synergistic pairs of antibodies, which simultaneously remove all three targets, thereby preventing compensatory tumor promoting mechanisms within the HER family. Pan-HER induces potent growth inhibition in a range of cancer cell lines and xenograft models, including cell lines with acquired resistance to therapeutic antibodies. Pan-HER is also highly efficacious in the presence of HER family ligands, indicating that it is capable of overcoming acquired resistance due to increased ligand production. All three target specificities contribute to the enhanced efficacy, demonstrating a distinct benefit of combined HER family targeting when compared with single-receptor targeting. CONCLUSIONS Our data show that simultaneous targeting of three receptors provides broader efficacy than targeting a single receptor or any combination of two receptors in the HER family, especially in the presence of HER family ligands. Pan-HER represents a novel strategy to deal with primary and acquired resistance due to tumor heterogeneity and plasticity in terms of HER family dependency and as such may be a viable alternative in the clinic.
Collapse
Affiliation(s)
| | | | | | - Ida Kjær
- Symphogen A/S, Ballerup, Denmark
| | | | | | | | | | | | | | | | | | | |
Collapse
|