1
|
Guo X, Xu C, Zhu X, Wang X, Hao Y, Wang X, Yao Y, Fang J, Wang K. In Silico Design and Evaluation of Novel Peptide-Based PET Probe for Noninvasive Imaging of HER2 Expression in Breast Cancer. J Med Chem 2025; 68:10461-10472. [PMID: 40322890 DOI: 10.1021/acs.jmedchem.5c00760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
The overexpression of HER2 has been strongly correlated with the pathogenesis and progression of breast cancer. Developing HER2-targeted PET tracers for noninvasive assessment of HER2 expression holds substantial clinical significance for screening patients suitable for HER2-targeted therapy, evaluating treatment efforts, and optimizing treatment. In this study, we employed computational simulation methods to develop peptide WC8 derived from trastuzumab, and designed HER2 targeting radiotracers [68Ga]Ga-DOTA-WC8 for detecting the HER2 expression. The radiotracer demonstrated nanomolar affinity, high specificity, and favorable pharmacokinetics. PET imaging revealed that [68Ga]Ga-DOTA-WC8 exhibited significant and specific uptake in HER2-positive breast cancer tissues, facilitating the rapid and accurate identification of HER2 expression. Notably, this radiotracer was successfully utilized to achieve direct visualization of tumor response to pyrotinib, a HER2 tyrosine kinase inhibitor. These findings suggest that the peptide-based radiotracer [68Ga]Ga-DOTA-WC8 represents a promising tool for accurately monitoring of HER2 expression after treatment with HER2-targeting drugs.
Collapse
Affiliation(s)
- Xu Guo
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China
- Department of Pharmacy, Affiliated Women's Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, Jiangsu 214002, China
| | - Chunwei Xu
- Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Xue Zhu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xiaojia Wang
- Department of Breast Medical Oncoloy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
| | - Yue Hao
- Department of Respiratory Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, China
| | - Xun Wang
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China
| | - Ying Yao
- Department of Pharmacy, Affiliated Women's Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, Jiangsu 214002, China
| | - Jing Fang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Ke Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
2
|
Zhang Y, Fan Y, Liu S, Guan Y, Wan J, Ren Q, Wang J, Zhong L, Hu Z, Shi W, Qian H. Development of Peptide Paratope Mimics Derived from the Anti-ROR1 Antibody and Long-Acting Peptide-Drug Conjugates for Targeted Cancer Therapy. J Med Chem 2024; 67:10967-10985. [PMID: 38943600 DOI: 10.1021/acs.jmedchem.4c00511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2024]
Abstract
Antibody-based targeted therapy in cancer faces a challenge due to uneven antibody distribution in solid tumors, hindering effective drug delivery. We addressed this by developing peptide mimetics with nanomolar-range affinity for Receptor Tyrosine Kinase-Like Orphan Receptor 1 (ROR1) using computational methods. These peptides showed both specific targeting and deep penetration in vitro and in vivo. Additionally, we created peptide-drug conjugates (PDCs) by linking targeting peptides to toxin drugs via various linkers and enhancing their in vivo half-life with fatty side chains for albumin binding. The antitumor candidate II-3 displayed exceptional affinity (KD = 1.72 × 10-9 M), internalization efficiency, anticancer potency (IC50 = 0.015 ± 0.002 μM), and pharmacokinetics (t1/2 = 2.6 h), showcasing a rational approach for designing PDCs with favorable tissue distribution and strong tumor penetration.
Collapse
Affiliation(s)
- Yang Zhang
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
- Department of Life Sciences, Changzhi University, Changzhi, Shanxi 046011, PR China
| | - Yiqing Fan
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Shuyu Liu
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Yonghui Guan
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Jiale Wan
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Qiang Ren
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Jialing Wang
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Li Zhong
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Zhipeng Hu
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Wei Shi
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Hai Qian
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing 210009, PR China
| |
Collapse
|
3
|
Murali R, Zhang H, Cai Z, Lam L, Greene M. Rational Design of Constrained Peptides as Protein Interface Inhibitors. Antibodies (Basel) 2021; 10:antib10030032. [PMID: 34449551 PMCID: PMC8395526 DOI: 10.3390/antib10030032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/06/2021] [Accepted: 08/10/2021] [Indexed: 11/26/2022] Open
Abstract
The lack of progress in developing targeted therapeutics directed at protein–protein complexes has been due to the absence of well-defined ligand-binding pockets and the extensive intermolecular contacts at the protein–protein interface. Our laboratory has developed approaches to dissect protein–protein complexes focusing on the superfamilies of erbB and tumor necrosis factor (TNF) receptors by the combined use of structural biology and computational biology to facilitate small molecule development. We present a perspective on the development and application of peptide inhibitors as well as immunoadhesins to cell surface receptors performed in our laboratory.
Collapse
Affiliation(s)
- Ramachandran Murali
- Cedars-Sinai Medical Center, Department of Biomedical Science, Research Division of Immunology, Los Angeles, CA 90211, USA
- Correspondence: (R.M.); (M.G.)
| | - Hongtao Zhang
- Department of Pathology and Laboratory of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (H.Z.); (Z.C.); (L.L.)
| | - Zheng Cai
- Department of Pathology and Laboratory of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (H.Z.); (Z.C.); (L.L.)
| | - Lian Lam
- Department of Pathology and Laboratory of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (H.Z.); (Z.C.); (L.L.)
| | - Mark Greene
- Department of Pathology and Laboratory of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (H.Z.); (Z.C.); (L.L.)
- Correspondence: (R.M.); (M.G.)
| |
Collapse
|
4
|
Kieber-Emmons T. Antibodies and Structure. Monoclon Antib Immunodiagn Immunother 2020; 39:193-194. [DOI: 10.1089/mab.2020.29003.tke] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
5
|
Murali R. Perspective on Crystallographic Studies of Antibody Structures. Monoclon Antib Immunodiagn Immunother 2020; 39:195-198. [PMID: 33156727 DOI: 10.1089/mab.2020.0037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
In the past 50 years, there has been a great progress made in understanding and deploying antibodies in biology, medicine, and therapy. In this study, a brief overview is presented on how the crystal structures of antibody fragments guided therapeutic strategies emanating from our laboratories along with some historical perspective.
Collapse
Affiliation(s)
- Ramachandran Murali
- Research Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
6
|
Murali R, Greene MI. Structure based antibody-like peptidomimetics. Pharmaceuticals (Basel) 2012; 5:209-35. [PMID: 24288089 PMCID: PMC3763629 DOI: 10.3390/ph5020209] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Revised: 01/17/2012] [Accepted: 01/19/2012] [Indexed: 12/22/2022] Open
Abstract
Biologics such as monoclonal antibodies (mAb) and soluble receptors represent new classes of therapeutic agents for treatment of several diseases. High affinity and high specificity biologics can be utilized for variety of clinical purposes. Monoclonal antibodies have been used as diagnostic agents when coupled with radionuclide, immune modulatory agents or in the treatment of cancers. Among other limitations of using large molecules for therapy the actual cost of biologics has become an issue. There is an effort among chemists and biologists to reduce the size of biologics which includes monoclonal antibodies and receptors without a reduction of biological efficacy. Single chain antibody, camel antibodies, Fv fragments are examples of this type of deconstructive process. Small high-affinity peptides have been identified using phage screening. Our laboratory used a structure-based approach to develop small-size peptidomimetics from the three-dimensional structure of proteins with immunoglobulin folds as exemplified by CD4 and antibodies. Peptides derived either from the receptor or their cognate ligand mimics the functions of the parental macromolecule. These constrained peptides not only provide a platform for developing small molecule drugs, but also provide insight into the atomic features of protein-protein interactions. A general overview of the reduction of monoclonal antibodies to small exocyclic peptide and its prospects as a useful diagnostic and as a drug in the treatment of cancer are discussed.
Collapse
Affiliation(s)
- Ramachandran Murali
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, D5091 Davis Building, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
- Department of Pathology and Laboratory of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mark I. Greene
- Department of Pathology and Laboratory of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
7
|
Cai Z, Zhang H, Liu J, Berezov A, Murali R, Wang Q, Greene MI. Targeting erbB receptors. Semin Cell Dev Biol 2010; 21:961-6. [PMID: 20850557 PMCID: PMC5940346 DOI: 10.1016/j.semcdb.2010.09.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Accepted: 09/09/2010] [Indexed: 10/19/2022]
Abstract
Our work is concerned with the origins and therapy of human cancers. Members of the epidermal growth factor receptor (EGFR) family of tyrosine kinases, also known as erbB or HER receptors, are over expressed and/or activated in many types of human tumors and represent important therapeutic targets in cancer therapy. Studies from our laboratory identified targeted therapy as a way to treat cancer. Rational therapeutics targeting and disabling erbB receptors have been developed to reverse the malignant properties of tumors. Reversal of the malignant phenotype, best seen with disabling the HER2 receptors using monoclonal antibodies is a distinct process from that seen with blocking of ligand binding to cognate receptors as has been done for EGFr receptors. Here we review the mechanisms of action deduced from a number of approaches developed in our laboratory and elsewhere, including monoclonal antibodies, peptide mimetics, recombinant proteins and small molecules. The biochemical and biological principles which have been uncovered during these studies of disabling HER2 homomeric or HER2-EGFr heteromeric receptors will help the development of novel and more efficient therapeutics targeting erbB family receptors.
Collapse
Affiliation(s)
- Zheng Cai
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104-6082, USA
| | - Hongtao Zhang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104-6082, USA
| | - Jing Liu
- School of Life Sciences, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230027, China
| | - Alan Berezov
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, S122A Steven Spielberg Building, 8725 Alden Dr., Los Angeles, CA 90048, USA
| | - Ramachandran Murali
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Davis Building, # 4092, Los Angeles, CA 90048, USA
| | - Qiang Wang
- Women’s Cancer Research Institute at the Samuel Oschin Comprehensive Cancer Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Mark I. Greene
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104-6082, USA
| |
Collapse
|
8
|
Masuda K, Richter M, Song X, Berezov A, Masuda K, Murali R, Greene MI, Zhang H. AHNP-streptavidin: a tetrameric bacterially produced antibody surrogate fusion protein against p185her2/neu. Oncogene 2006; 25:7740-6. [PMID: 16785990 DOI: 10.1038/sj.onc.1209745] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The anti-p185(her2/neu) peptidomimetic (AHNP) is a small exo-cyclic peptide derived from the anti-p185(her2/neu) rhumAb 4D5 (h4D5). AHNP mimics many but not all of the antitumor characteristics exhibited by h4D5. However, the pharmacokinetic profiles of AHNP are less than optimal for therapeutic or diagnostic purposes. To improve the binding affinity to p185(her2/neu) and the antitumor efficacy, we have engineered a fusion protein containing AHNP and a nonimmunoglobulin protein scaffold, streptavidin (SA). The recombinant protein, AHNP-SA (ASA) bound to p185(her2/neu) with high affinity, inhibited the proliferation of p185(her2/neu)-overexpressing cells, and reduced tumor growth induced by p185(her2/neu)-transformed cells. These data suggest that the bacterially produced tetrameric ASA can be used as an antibody-surrogate molecule. This class of molecule will play a role in the diagnosis and treatment of p185(her2/neu)-related tumors. Our studies establish a general principle by which a small biologically active synthetic exo-cyclic peptide can be engineered to enhance functional aspects by structured oligomerization and can be produced recombinantly using bacterial expression.
Collapse
Affiliation(s)
- K Masuda
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-6082, USA
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Guillemard V, Nedev HN, Berezov A, Murali R, Saragovi HU. HER2-mediated internalization of a targeted prodrug cytotoxic conjugate is dependent on the valency of the targeting ligand. DNA Cell Biol 2005; 24:350-8. [PMID: 15941387 DOI: 10.1089/dna.2005.24.351] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
HER2 is a validated therapeutic target for cancer. There are no natural ligands, but monoclonal antibodies and peptides that bind HER2 act as artificial ligands, selectively affecting HER2-overexpressing tumors. One reported mechanism for this effect is receptor downregulation, but the expected correlation of ligand-dependent HER2 internalization and tumor inhibition remain poorly characterized. Moreover, HER2 ligands have limited therapeutic efficacy and often they require adjuvant treatment with the chemotherapeutic Taxol. Here, we generated a series of HER2 ligands (Anti-HER2/neu peptide ligands, AHNPmonovalent and AHNPbivalent) with different valency and correlated their internalization-promoting ability to biological potency. Since AHNPbivalent (but not AHNPmonovalent) induces rapid receptor internalization, we exploited this feature to deliver cytotoxic conjugates coupling AHNPbivalent and Taxol (Taxol . AHNPbivalent). The prodrug conjugate releases Taxol after receptor-mediated internalization, and cytotoxicity can be used as a marker of internalization. Taxol . AHNPbivalent is significantly more cytotoxic than free Taxol + free AHNPbivalent. Hence, the Taxol x AHNP(bivalent) prodrug binds to HER2, induces receptor internalization and downregulation, and the subsequent release of free Taxol inside the targeted cell results in synergistic toxicity, The effect is selective towards HER2- expressing cells. This work links HER2 receptor internalization and growth arrest, and the chemical conjugation strategy may yield improved and HER2 selective therapeutics.
Collapse
Affiliation(s)
- Veronique Guillemard
- McGill University, Pharmacology and Therapeutics and Oncology/Cancer Center, Lady Davis Research Institute-Jewish General Hospital, Montreal, Quebec, Canada
| | | | | | | | | |
Collapse
|
10
|
Peterson NC. Advances in monoclonal antibody technology: genetic engineering of mice, cells, and immunoglobulins. ILAR J 2005; 46:314-9. [PMID: 15953839 DOI: 10.1093/ilar.46.3.314] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The ability to produce antibodies that are directed against specific antigens has played a crucial role in advancing scientific discoveries. Recombinant technologies have extended the application of antibodies beyond the research laboratory and into the clinic for the treatment of cancer and other diseases. Creative approaches using these technologies have been used to reduce the antibody to its minimal functional size, and/or make them bifunctional (immunotoxins), bispecific, or less immunoreactive (humanized). Additionally, mice that are engineered to generate antibodies of human genomic origin have been used to produce therapeutic antibodies and are being further developed. As the research and clinical demands for antibodies continue to increase, the development of improved resources (cell lines and animals) to improve production efficiency, generate larger repertoires, and deliver greater yields of antibodies is being explored, and advances in this area are discussed further in this review.
Collapse
Affiliation(s)
- Norman C Peterson
- Department of Comparative Medicine, The Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
11
|
Cheng X, Kinosaki M, Murali R, Greene MI. The TNF receptor superfamily: role in immune inflammation and bone formation. Immunol Res 2004; 27:287-94. [PMID: 12857975 DOI: 10.1385/ir:27:2-3:287] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Tumor necrosis factor (TNF) and TNF receptor (TNFR) family proteins play important roles in many biological processes. Recently, the TNF-family molecule, RANKL (also called TRANCE, ODF, and OPGL), and its receptors, RANK and OPG, were found to be regulators of the development and activation of osteoclasts in bone remodeling. TNFalphaalso activates osteoclasts both by themselves and in synergy with RANKL. We used structure-based design to create peptidomimetics and organic therapeutics that inhibit osteoclastogenesis by inhibiting the interaction of ligands and receptors. Here we show for the first time that blocking TNFalpha by these small molecules effectively inhibited osteoclast formation in vitro. These mimetics can be used as a probe to understand the molecular basis of osteoclastogenesis and also as a platform to create useful therapeutic agent.
Collapse
Affiliation(s)
- Xin Cheng
- Department of Pathology, Abramson Institute for Cancer Research, University of Pennsylvania, Philadelphia, PA 19104-6082, USA
| | | | | | | |
Collapse
|
12
|
Cheng X, Kinosaki M, Takami M, Choi Y, Zhang H, Murali R. Disabling of Receptor Activator of Nuclear Factor-κB (RANK) Receptor Complex by Novel Osteoprotegerin-like Peptidomimetics Restores Bone Loss in Vivo. J Biol Chem 2004; 279:8269-77. [PMID: 14679212 DOI: 10.1074/jbc.m309690200] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The tumor necrosis factor family ligand, tumor necrosis factor-related activation-induced cytokine (TRANCE), and its receptors, receptor activator of nuclear factor-kappaB (RANK) and osteoprotegerin (OPG), are known to be regulators of development and activation of osteoclasts in bone remodeling. Sustained osteoclast activation that occurs through TRANCE-RANK causes osteopenic disorders such as osteoporosis and contributes to osteolytic metastases. Here, we report a rationally designed small molecule mimic of osteoprotegerin to inhibit osteoclast formation in vitro and limit bone loss in an animal model of osteoporosis. One of the mimetics, OP3-4, significantly inhibited osteoclast formation in vitro (IC(50) = 10 microm) and effectively inhibited total bone loss in ovariectomized mice at a dosage of 2 mg/kg/day. Unlike soluble OPG receptors, which preclude TRANCE binding to RANK, OP3-4 shows the ability to modulate RANK-TRANCE signaling pathways and alters the biological functions of the RANK-TRANCE receptor complex by facilitating a defective receptor complex. These features suggest that OPG-derived small molecules can be used as a probe to understand complex biological functions of RANK-TRANCE-OPG receptors and also can be used as a platform to develop more useful therapeutic agents for inflammation and bone disease.
Collapse
Affiliation(s)
- Xin Cheng
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute for Cancer Research, University of Pennsylvania, School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | |
Collapse
|
13
|
Sillerud LO, Burks EJ, Wester MJ, Brown DC, Vijayan S, Larson RS. NMR-derived model of interconverting conformations of an ICAM-1 inhibitory cyclic nonapeptide. THE JOURNAL OF PEPTIDE RESEARCH : OFFICIAL JOURNAL OF THE AMERICAN PEPTIDE SOCIETY 2003; 62:97-116. [PMID: 12895272 DOI: 10.1034/j.1399-3011.2003.00070.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
We have produced by phage-display a disulfide-linked cyclic nonapeptide (inhibitory peptide-01, IP01), CLLRMRSIC, that binds to intracellular adhesion molecule-1 (ICAM-1) and blocks binding to its counter-structure, leukocyte functional antigen-1 (LFA-1). As a first step towards improving its pharmacologic properties, we have performed a structural and functional analysis of this peptide inhibitor to determine the features relevant to ICAM-1 binding. We report here the solution model of our initial product, IP01, as derived from two-dimensional nuclear magnetic resonance (NMR) restraints and molecular modeling. Distance and dihedral angle restraints, generated from nuclear Overhauser effect spectroscopy (NOESY) and one-dimensional-NMR experiments respectively, were used to generate an ensemble of structures using distance geometry and simulated annealing. Molecular dynamic simulations produced three interconverting conformational families consistent with the NMR-derived constraints. We describe these conformations and their mechanism of interconversion. Furthermore, we have measured the IC50 s of a series of inhibitors generated from IP01 through alanine substitution of each residue. These results show that the L2-L3-R4-M5-R6 segment is functionally active, conformationally flexible, and contains a beta-turn involving residues R4-S7, while the C1-C9-I8-S7 segment is less functionally-active but adopts a more defined solution conformation, consistent with a scaffolding function. This model will be useful for designing nonpeptide-based organic inhibitors with improved pharmacologic properties.
Collapse
Affiliation(s)
- L O Sillerud
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | | | | | | | | | | |
Collapse
|
14
|
Kumagai T, Katsumata M, Hasegawa A, Furuuchi K, Funakoshi T, Kawase I, Greene MI. Role of extracellular subdomains of p185c-neu and the epidermal growth factor receptor in ligand-independent association and transactivation. Proc Natl Acad Sci U S A 2003; 100:9220-5. [PMID: 12867596 PMCID: PMC170899 DOI: 10.1073/pnas.1633546100] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We investigated the assembly and activation of the epidermal growth factor receptor (EGFR)-p185c-neu heterodimer by using a sequential immunoprecipitation methodology. Using this approach we detected heterodimers and also higher-ordered oligomeric complexes. Phosphorylated EGFR-p185c-neu heterodimeric forms were detected in the absence of EGF, but the species became highly phosphorylated after EGF stimulation. To evaluate heterodimer formation and additional transactivation by EGF, we investigated the roles of the four extracellular subdomains of p185c-neu and the EGFR. Subdomains I-IV of the EGFR dimerized with subdomains I-IV of p185c-neu, respectively, in a parallel manner. In addition, subdomains I-IV of the EGFR also associated with p185c-neu subdomains III, IV, I, and II, respectively. A lack of one of the p185c-neu cysteine-rich domains (subdomains II or IV) resulted in a loss of EGF-induced transactivation. These data suggest that two cysteine-rich domains play defining roles in ligand-dependent transactivation and that both of these cysteine-rich extracellular subdomains as well as non-cysteine-rich extracellular subdomains are involved in ligand-independent interactions with the EGFR. Our studies provide biochemical evidence of the role of the cysteine-rich domains of p185c-neu in assembly and transactivation of erbB complexes and also indicate that these subdomains might be useful clinical targets.
Collapse
Affiliation(s)
- Toru Kumagai
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Horie T, Shen Y, Kajino K, Gaubin M, Bonomi G, Mani JC, Berezov A, Piatier-Tonneau D, Guardiola J, Hillard B, Rostami A, Greene M, Murali R. Study of disabling T-cell activation and inhibiting T-cell-mediated immunopathology reveals a possible inverse agonist activity of CD4 peptidomimetics. Exp Mol Pathol 2002; 73:93-103. [PMID: 12231211 DOI: 10.1006/exmp.2002.2444] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We designed a new class of aromatically modified exocyclic peptides based on the structure of CD4 by engineering one of the cysteine residues in a peptidomimetic derived from the CDR3 region of the CD4 molecule. All three species mediate inhibition of T-cell proliferation at concentrations ranging from 10 to 100 microM. The mimetics CD4-Cys and CD4-Met bind to sCD4 with affinities ranging from 1 to 2 microM, while CD4-Ser shows poor binding in radioisotope assay. Though these mimetics have similar structures, they exhibit different biochemical and biological functions. Activation of T-cells as measured by thymidine incorporation or IL-2 production revealed that CD4-Cys and CD4-Ser mimetics behave as classical antagonists. On the other hand, the CD4-Met species inhibited T-cell proliferation with an IC(50) of 30 microM but unexpectedly increased IL-2 secretion modestly at a less than 3 microM concentration. In experimental autoimmune encephalitis (EAE), CD4-Ser and CD4-Cys mimetics reduced the severity of EAE symptoms while the CD4-Met mimetic exacerbated the conditions. We propose that CD4-Cys and CD4-Ser are classical antagonists, but CD4-Met may possess properties of an inverse agonist. The structure-activity relationship of mimetics reveals that a minor change in the net hydropathic value is enough to alter the dynamic nature of the receptor-ligand complex.
Collapse
MESH Headings
- Animals
- Biotechnology
- CD4 Antigens/chemistry
- CD4 Antigens/metabolism
- CD4 Antigens/pharmacology
- Complementarity Determining Regions/chemistry
- Complementarity Determining Regions/metabolism
- Computer Simulation
- Dimerization
- Dose-Response Relationship, Drug
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Guinea Pigs
- Humans
- In Vitro Techniques
- Interleukin-2/biosynthesis
- Interleukin-2/immunology
- Lymphocyte Activation/drug effects
- Major Histocompatibility Complex
- Mice
- Mice, Inbred C3H
- Mice, Inbred Strains
- Models, Biological
- Models, Molecular
- Peptide Fragments/chemistry
- Peptide Fragments/metabolism
- Peptide Fragments/pharmacology
- Protein Conformation
- Receptors, Antigen, T-Cell/metabolism
- Structure-Activity Relationship
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Takeo Horie
- Department of Pathology and School of Medicine, University of Pensylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Berezov A, Chen J, Liu Q, Zhang HT, Greene MI, Murali R. Disabling receptor ensembles with rationally designed interface peptidomimetics. J Biol Chem 2002; 277:28330-9. [PMID: 12011054 DOI: 10.1074/jbc.m202880200] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Members of the erbB family receptor tyrosine kinases (erbB1, erbB2, erbB3, and erbB4) are overexpressed in a variety of human cancers and represent important targets for the structure-based drug design. Homo- and heterodimerization (oligomerization) of the erbB receptors are known to be critical events for receptor signaling. To block receptor self-associations, we have designed a series of peptides derived from potential dimerization surfaces in the extracellular subdomain IV of the erbB receptors (erbB peptides). In surface plasmon resonance (BIAcore) studies, the designed peptides have been shown to selectively bind to the erbB receptor ectodomains and isolated subdomain IV of erbB2 with submicromolar affinities and to inhibit heregulin-induced interactions of erbB3 with different erbB receptors. A dose-dependent inhibition of native erbB receptor dimerization by the erbB peptides has been observed in 32D cell lines transfected with different combinations of erbB receptors. The peptides effectively inhibited growth of two types of transformed cells overexpressing different erbB receptors, T6-17 and 32D, in standard MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) and cell viability assays. The study identifies distinct loops within the membrane-proximal part of the subdomain IV as potential receptor-receptor interaction sites for the erbB receptors and demonstrates the possibility of disabling receptor activity by structure-based targeting of the dimerization interfaces. Molecular models for possible arrangement of the erbB1.EGF complex, consistent with the involvement of subdomain IV in inter-receptor interactions, are proposed. Small dimerization inhibitors described herein can be useful as probes to elucidate different erbB signaling pathways and may be developed as therapeutic agents.
Collapse
Affiliation(s)
- Alan Berezov
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine and the Abramson Family Cancer Research Institute, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | |
Collapse
|
17
|
Bès C, Briant-Longuet L, Cerruti M, De Berardinis P, Devauchelle G, Devaux C, Granier C, Chardès T, DeBerardinis P. Efficient CD4 binding and immunosuppressive properties of the 13B8.2 monoclonal antibody are displayed by its CDR-H1-derived peptide CB1. FEBS Lett 2001; 508:67-74. [PMID: 11707270 DOI: 10.1016/s0014-5793(01)03036-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
A systematic exploration of the V(H)2/V(kappa)12-13 variable domains of the anti-CD4 monoclonal antibody (mAb) 13B8.2 was performed by the Spot method to screen for paratope-derived peptides (PDPs) demonstrating CD4 binding ability. Nine peptides, named CB1 to CB9, were identified, synthesized in a cyclic and soluble form and tested for binding to recombinant soluble CD4. Among them, CB1, CB2 and CB8 showed high anti-CD4 activity. Competition studies for CD4 binding indicated that PDPs CB1, CB8, and the parental mAb 13B8.2 recognized the same complementarity determining region (CDR)3-like loop region. PDP CB1 was shown to mimic the biological properties of 13B8.2 mAb in two independent cellular assays, demonstrating inhibitory activities in the micromolar range on antigen presentation and human immunodeficiency virus promoter activation. Our results indicate that the bioactive CDR-H1 PDP CB1 has retained a significant part of the parental 13B8.2 mAb properties and might be a lead for the design of anti-CD4 peptidomimetics of clinical interest.
Collapse
Affiliation(s)
- C Bès
- CNRS-UMR 5094, Faculté de Pharmacie, Institut de Biotechnologie et Pharmacologie, 15 avenue Charles Flahault, 34060 Montpellier Cedex 2, France
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Preissner R, Goede A, Rother K, Osterkamp F, Koert U, Froemmel C. Matching organic libraries with protein-substructures. J Comput Aided Mol Des 2001; 15:811-7. [PMID: 11776292 DOI: 10.1023/a:1013158818807] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
We present a general approach which allows automatic identification of sub-structures in proteins that resemble given three-dimensional templates. This paper documents its success with non-peptide templates such as beta-turn mimetics. We considered well-tested turn-mimetics such as the bicyclic turned dipeptide (BTD), spiro lactam (Spiro) and the 2,5-disubstituded tetrahydrofuran (THF), a new furan-derivative which was recently developed and characterized. The detected geometric similarity between the templates and the protein patches corresponds to r.m.s.-values of 0.3 A for more than 80% of the constituting atoms, which is typical for active site comparisons of homologous proteins. This fast automatic procedure might be of biomedical value for finding special mimicking leads for particular protein sub-structures as well as for template-assembled synthetic protein (TASP) design.
Collapse
Affiliation(s)
- R Preissner
- Institute of Biochemistry, Charité, Medical Faculty of the Hunboldt-University, Berlin, Germany
| | | | | | | | | | | |
Collapse
|
19
|
Berezov A, Zhang HT, Greene MI, Murali R. Disabling erbB receptors with rationally designed exocyclic mimetics of antibodies: structure-function analysis. J Med Chem 2001; 44:2565-74. [PMID: 11472210 DOI: 10.1021/jm000527m] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Overexpression of the HER2 receptor is observed in about 30% of breast and ovarian cancers and is often associated with an unfavorable prognosis. We have recently designed an anti-HER2 peptide (AHNP) based on the structure of the CDR-H3 loop of the anti-HER2 rhumAb 4D5 and showed that this peptide can mimic some functions of rhumAb 4D5. The peptide disabled HER2 tyrosine kinases in vitro and in vivo similar to the monoclonal antibody (Park, B.-W. et al. Nat. Biotechnol. 2000, 18, 194--198). AHNP has been shown to selectively bind to the extracellular domain of the HER2 receptor with a submicromolar affinity in Biacore assays. In the present paper, we demonstrate that in addition to being a structural and functional mimic of rhumAb 4D5, AHNP can also effectively compete with the antibody for binding to the HER2 receptor indicating a similar binding site for the peptide and the parental antibody. To further develop AHNP as an antitumor agent useful for preclinical trials and as a radiopharmaceutical to be used for tumor imaging, a number of derivatives of AHNP have been designed. Structure--function relationships have been studied using surface plasmon resonance technology. Some of the AHNP analogues have improved binding properties, solubility, and cytotoxic activity relative to AHNP. Residues in the exocyclic region of AHNP appear to be essential for high-affinity binding. Kinetic and equilibrium analysis of peptide-receptor binding for various AHNP analogues revealed a strong correlation between peptide binding characteristics and their biological activity. For AHNP analogues, dissociation rate constants have been shown to be better indicators of peptide biological activity than receptor-binding affinities. This study demonstrates a possibility of mimicking the well-documented antibody effects and its applications in tumor therapy by much smaller antibody-based cyclic peptides with potentially significant therapeutic advantages. Strategies used to improve binding properties of rationally designed AHNP analogues are discussed.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/chemistry
- Antineoplastic Agents/chemical synthesis
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/metabolism
- Antineoplastic Agents/pharmacology
- Binding, Competitive
- Cell Division/drug effects
- Drug Design
- Kinetics
- Mice
- Models, Molecular
- Molecular Mimicry
- Oligopeptides/chemical synthesis
- Oligopeptides/chemistry
- Oligopeptides/metabolism
- Oligopeptides/pharmacology
- Peptides, Cyclic/chemical synthesis
- Peptides, Cyclic/chemistry
- Peptides, Cyclic/metabolism
- Peptides, Cyclic/pharmacology
- Radiopharmaceuticals/chemical synthesis
- Radiopharmaceuticals/chemistry
- Receptor, ErbB-2/drug effects
- Receptor, ErbB-2/metabolism
- Solubility
- Structure-Activity Relationship
- Surface Plasmon Resonance
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- A Berezov
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, 36th and Hamilton Walk, Philadelphia, Pennsylvania 19104, USA
| | | | | | | |
Collapse
|
20
|
Kumagai T, Davis JG, Horie T, O'Rourke DM, Greene MI. The role of distinct p185neu extracellular subdomains for dimerization with the epidermal growth factor (EGF) receptor and EGF-mediated signaling. Proc Natl Acad Sci U S A 2001; 98:5526-31. [PMID: 11320205 PMCID: PMC33246 DOI: 10.1073/pnas.071060598] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The extracellular domain of p185(c-neu) can be viewed as a complex structure of four subdomains, two of which are cysteine-rich subdomains. We have investigated the contribution of these distinct p185(c-neu) extracellular subdomains to p185/epidermal growth factor receptor (EGFR) heteromer formation and EGF-induced heteromeric signaling. Our studies indicate that at least two separate p185 subdomains, a region spanning subdomains I and II and subdomain IV are involved in association of p185 with the EGFR. We also demonstrated that subdomain IV reduced the heteromeric signaling and transforming activities induced by EGF after associating with EGFR. When 126 aa were deleted from subdomain IV, this small subdomain IV-derived fragment could still lead to heterodimers with EGFR and suppress EGF-induced mitogen-activated protein kinase activation and subsequent transformation abilities. These data provide information about trans-inhibitory mechanisms of mutant p185 species and also indicate that both the entire and a part of subdomain IV may represent a therapeutic target for erbB-overexpressing tumors. Finally, these studies define a basic feature of receptor-receptor associations that are determined by cystine-knot containing subdomains.
Collapse
Affiliation(s)
- T Kumagai
- Department of Pathology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|
21
|
Briant L, Devaux C. Bioactive CD4 ligands as pre- and/or postbinding inhibitors of HIV-1. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2001; 48:373-407. [PMID: 10987097 DOI: 10.1016/s1054-3589(00)48012-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- L Briant
- Laboratoire Infections Rétrovirales et Signalisation Cellulaire, CNRS EP 2104, Montpellier, France
| | | |
Collapse
|
22
|
Park BW, Zhang HT, Wu C, Berezov A, Zhang X, Dua R, Wang Q, Kao G, O'Rourke DM, Greene MI, Murali R. Rationally designed anti-HER2/neu peptide mimetic disables P185HER2/neu tyrosine kinases in vitro and in vivo. Nat Biotechnol 2000; 18:194-8. [PMID: 10657127 DOI: 10.1038/72651] [Citation(s) in RCA: 157] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Monoclonal antibodies specific for the p185HER2/neu growth factor receptor represent a significant advance in receptor-based therapy for p185HER2/neu-expressing human cancers. We have used a structure-based approach to develop a small (1.5 kDa) exocyclic anti-HER2/neu peptide mimic (AHNP) functionally similar to an anti-p185HER2/neu monoclonal antibody, 4D5 (Herceptin). The AHNP mimetic specifically binds to p185HER2/neu with high affinity (KD=300 nM). This results in inhibition of proliferation of p185HER2/neu-overexpressing tumor cells, and inhibition of colony formation in vitro and growth of p185HER2/neu-expressing tumors in athymic mice. In addition, the mimetic sensitizes the tumor cells to apoptosis when used in conjunction with ionizing radiation or chemotherapeutic agents. A comparison of the molar quantities of the Herceptin antibody and the AHNP mimetic required for inhibiting cell growth and anchorage-independent growth showed generally similar activities. The structure-based derivation of the AHNP represents a novel strategy for the design of receptor-specific tumor therapies.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Antineoplastic Agents/chemistry
- Apoptosis
- Astrocytoma
- Biosensing Techniques
- Doxorubicin/therapeutic use
- Drug Design
- Drug Therapy, Combination
- Gamma Rays
- Mice
- Mice, Nude
- Molecular Mimicry
- Protein Binding
- Radiotherapy, Adjuvant
- Receptor, ErbB-2/antagonists & inhibitors
- Trastuzumab
- Tumor Cells, Cultured/drug effects
- Tumor Cells, Cultured/radiation effects
Collapse
Affiliation(s)
- B W Park
- Department of Pathology and Laboratory Medicine, Center for Receptor Biology and Cell Growth, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|