1
|
Feins S, Kong W, Williams EF, Milone MC, Fraietta JA. An introduction to chimeric antigen receptor (CAR) T-cell immunotherapy for human cancer. Am J Hematol 2019; 94:S3-S9. [PMID: 30680780 DOI: 10.1002/ajh.25418] [Citation(s) in RCA: 327] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 01/17/2019] [Accepted: 01/22/2019] [Indexed: 02/06/2023]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy represents a major advancement in personalized cancer treatment. In this strategy, a patient's own T cells are genetically engineered to express a synthetic receptor that binds a tumor antigen. CAR T cells are then expanded for clinical use and infused back into the patient's body to attack and destroy chemotherapy-resistant cancer. Dramatic clinical responses and high rates of complete remission have been observed in the setting of CAR T-cell therapy of B-cell malignancies. This resulted in two recent FDA approvals of CAR T cells directed against the CD19 protein for treatment of acute lymphoblastic leukemia and diffuse large B-cell lymphoma. Thus, CAR T cells are arguably one of the first successful examples of synthetic biology and personalized cellular cancer therapy to become commercially available. In this review, we introduce the concept of using CAR T cells to break immunological tolerance to tumors, highlight several challenges in the field, discuss the utility of biomarkers in the context of predicting clinical responses, and offer prospects for developing next-generation CAR T cell-based approaches that will improve outcome.
Collapse
Affiliation(s)
- Steven Feins
- Department of MicrobiologyPerelman School of Medicine, University of Pennsylvania Philadelphia Pennsylvania
- Department of Pathology and Laboratory MedicinePerelman School of Medicine, University of Pennsylvania Philadelphia Pennsylvania
- Center for Cellular ImmunotherapiesUniversity of Pennsylvania Philadelphia Pennsylvania
| | - Weimin Kong
- Department of MicrobiologyPerelman School of Medicine, University of Pennsylvania Philadelphia Pennsylvania
- Department of Pathology and Laboratory MedicinePerelman School of Medicine, University of Pennsylvania Philadelphia Pennsylvania
- Center for Cellular ImmunotherapiesUniversity of Pennsylvania Philadelphia Pennsylvania
| | - Erik F. Williams
- Department of MicrobiologyPerelman School of Medicine, University of Pennsylvania Philadelphia Pennsylvania
- Department of Pathology and Laboratory MedicinePerelman School of Medicine, University of Pennsylvania Philadelphia Pennsylvania
- Center for Cellular ImmunotherapiesUniversity of Pennsylvania Philadelphia Pennsylvania
| | - Michael C. Milone
- Department of Pathology and Laboratory MedicinePerelman School of Medicine, University of Pennsylvania Philadelphia Pennsylvania
- Center for Cellular ImmunotherapiesUniversity of Pennsylvania Philadelphia Pennsylvania
- Abramson Cancer CenterUniversity of Pennsylvania Philadelphia Pennsylvania
- Parker Institute for Cancer ImmunotherapyUniversity of Pennsylvania Philadelphia Pennsylvania
| | - Joseph A. Fraietta
- Department of MicrobiologyPerelman School of Medicine, University of Pennsylvania Philadelphia Pennsylvania
- Department of Pathology and Laboratory MedicinePerelman School of Medicine, University of Pennsylvania Philadelphia Pennsylvania
- Center for Cellular ImmunotherapiesUniversity of Pennsylvania Philadelphia Pennsylvania
- Abramson Cancer CenterUniversity of Pennsylvania Philadelphia Pennsylvania
- Parker Institute for Cancer ImmunotherapyUniversity of Pennsylvania Philadelphia Pennsylvania
| |
Collapse
|
2
|
Long KB, Young RM, Boesteanu AC, Davis MM, Melenhorst JJ, Lacey SF, DeGaramo DA, Levine BL, Fraietta JA. CAR T Cell Therapy of Non-hematopoietic Malignancies: Detours on the Road to Clinical Success. Front Immunol 2018; 9:2740. [PMID: 30559740 PMCID: PMC6287001 DOI: 10.3389/fimmu.2018.02740] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 11/07/2018] [Indexed: 12/12/2022] Open
Abstract
Chimeric antigen receptor (CAR)-engineered T cells represent a breakthrough in personalized medicine. In this strategy, a patient's own T lymphocytes are genetically reprogrammed to encode a synthetic receptor that binds a tumor antigen, allowing T cells to recognize and kill antigen-expressing cancer cells. As a result of complete and durable responses in individuals who are refractory to standard of care therapy, CAR T cells directed against the CD19 protein have been granted United States Food and Drug Administration (FDA) approval as a therapy for treatment of pediatric and young adult acute lymphoblastic leukemia and diffuse large B cell lymphoma. Human trials of CAR T cells targeting CD19 or B cell maturation antigen in multiple myeloma have also reported early successes. However, a clear and consistently reproducible demonstration of the clinical efficacy of CAR T cells in the setting of solid tumors has not been reported to date. Here, we review the history and status of CAR T cell therapy for solid tumors, potential T cell-intrinsic determinants of response and resistance as well as extrinsic obstacles to the success of this approach for much more prevalent non-hematopoietic malignancies. In addition, we summarize recent strategies and innovations that aim to augment the potency of CAR T cells in the face of multiple immunosuppressive barriers operative within the solid tumor microenvironment. Advances in the field of CAR T cell biology over the coming years in the areas of safety, reliability and efficacy against non-hematopoietic cancers will ultimately determine how transformative adoptive T cell therapy will be in the broader battle against cancer.
Collapse
Affiliation(s)
- Kristen B Long
- Department of Biology, Mansfield University, Mansfield, PA, United States
| | - Regina M Young
- Center for Cellular Immunotherapies, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, United States.,Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, United States
| | - Alina C Boesteanu
- Center for Cellular Immunotherapies, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, United States
| | - Megan M Davis
- Center for Cellular Immunotherapies, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, United States.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - J Joseph Melenhorst
- Center for Cellular Immunotherapies, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, United States.,Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, United States.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Simon F Lacey
- Center for Cellular Immunotherapies, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, United States.,Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, United States.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - David A DeGaramo
- Department of Biology, Mansfield University, Mansfield, PA, United States
| | - Bruce L Levine
- Center for Cellular Immunotherapies, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, United States.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Joseph A Fraietta
- Center for Cellular Immunotherapies, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, United States.,Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, United States.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
3
|
Shank BR, Do B, Sevin A, Chen SE, Neelapu SS, Horowitz SB. Chimeric Antigen Receptor T Cells in Hematologic Malignancies. Pharmacotherapy 2017; 37:334-345. [PMID: 28079265 DOI: 10.1002/phar.1900] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Patients with B-cell hematologic malignancies who progress through first- or second-line chemotherapy have a poor prognosis. Early clinical trials with autologous anti-CD19 chimeric antigen receptor (CAR) T cells have demonstrated promising results for patients who have relapsed or refractory disease. Lymphodepleting conditioning regimens, including cyclophosphamide, fludarabine, pentostatin, bendamustine, interleukin-2, and total body irradiation, are often administered before the infusion of CAR T cells, allowing for greater T-cell expansion. The major toxicity associated with CAR T-cell infusions is cytokine release syndrome (CRS), a potentially life-threatening systemic inflammatory disorder. The quick onset and progression of CRS require rapid detection and intervention to reduce treatment-related mortality. Management with tocilizumab can help ameliorate the symptoms of severe CRS, allowing steroids, which diminish the expansion and persistence of CAR T cells, to be reserved for tocilizumab-refractory patients. Other toxicities of CAR T-cell therapy include neutropenia and/or febrile neutropenia, infection, tumor lysis syndrome, neurotoxicity and nausea/vomiting. A review of patients' medications is imperative to eliminate medications that may contribute to treatment-related toxicities. Studies are ongoing to help optimize patient selection, preparation, safety, and management of individuals receiving CAR T cells. Long-term follow-up will help establish the place of CAR T cells in therapy.
Collapse
Affiliation(s)
- Brandon R Shank
- Division of Pharmacy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bryan Do
- Division of Pharmacy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Adrienne Sevin
- Division of Pharmacy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sheree E Chen
- Division of Pharmacy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sattva S Neelapu
- Division of Cancer Medicine, Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sandra B Horowitz
- Division of Pharmacy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
4
|
Fraietta JA, Schwab RD, Maus MV. Improving therapy of chronic lymphocytic leukemia with chimeric antigen receptor T cells. Semin Oncol 2016; 43:291-9. [PMID: 27040708 DOI: 10.1053/j.seminoncol.2016.02.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Adoptive cell immunotherapy for the treatment of chronic lymphocytic leukemia (CLL) has heralded a new era of synthetic biology. The infusion of genetically engineered, autologous chimeric antigen receptor (CAR) T cells directed against CD19 expressed by normal and malignant B cells represents a novel approach to cancer therapy. The results of recent clinical trials of CAR T cells in relapsed and refractory CLL have demonstrated long-term disease-free remissions, underscoring the power of harnessing and redirecting the immune system against cancer. This review will briefly summarize T-cell therapies in development for CLL disease. We discuss the role of T-cell function and phenotype, T-cell culture optimization, CAR design, and approaches to potentiate the survival and anti-tumor effects of infused lymphocytes. Future efforts will focus on improving the efficacy of CAR T cells for the treatment of CLL and incorporating adoptive cell immunotherapy into standard medical management of CLL.
Collapse
Affiliation(s)
- Joseph A Fraietta
- Center for Cellular Immunotherapy, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Robert D Schwab
- Center for Cellular Immunotherapy, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA.
| |
Collapse
|
5
|
Jin HE, Chung WJ, Lee SW. Phage Display for the Discovery of Hydroxyapatite-Associated Peptides. Methods Enzymol 2013; 532:305-23. [DOI: 10.1016/b978-0-12-416617-2.00014-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
6
|
Tóthová L, Bábíčková J, Celec P. Phage survival: The biodegradability of M13 phage display libraryin vitro. Biotechnol Appl Biochem 2012; 59:490-4. [DOI: 10.1002/bab.1050] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Accepted: 10/04/2012] [Indexed: 01/02/2023]
Affiliation(s)
- L'ubomíra Tóthová
- Institute of Molecular Biomedicine; Comenius University; Bratislava; Slovakia
| | - Janka Bábíčková
- Institute of Molecular Biomedicine; Comenius University; Bratislava; Slovakia
| | | |
Collapse
|
7
|
A phase I clinical trial of adoptive transfer of folate receptor-alpha redirected autologous T cells for recurrent ovarian cancer. J Transl Med 2012; 10:157. [PMID: 22863016 PMCID: PMC3439340 DOI: 10.1186/1479-5876-10-157] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Accepted: 07/18/2012] [Indexed: 11/10/2022] Open
Abstract
PURPOSE In spite of increased rates of complete response to initial chemotherapy, most patients with advanced ovarian cancer relapse and succumb to progressive disease. RATIONALE Genetically reprogrammed, patient-derived chimeric antigen receptor (CAR)-T lymphocytes with the ability to recognize predefined surface antigens with high specificity in a non-MHC restricted manner have shown increasing anti-tumor efficacy in preclinical and clinical studies. Folate receptor-α (FRα) is an ovarian cancer-specific tumor target; however, it is expressed at low levels in certain organs with risk for toxicity. DESIGN Here we propose a phase I study testing the feasibility, safety and preliminary activity of FRα-redirected CAR-T cells bearing the CD137 (4-1BB) costimulatory domain, administered after lymphodepletion for the treatment of recurrent ovarian cancer. A novel trial design is proposed that maximizes safety features. INNOVATION This design involves an initial accelerated dose escalation phase of FR-α CAR-T cells followed by a standard 3 + 3 escalation phase. A split-dose approach is proposed to mitigate acute adverse events. Furthermore, infusion of bulk untransduced autologous peripheral blood lymphocytes (PBL) is proposed two days after CAR-T cell infusion at the lower dose levels of CAR-T cells, to suppress excessive expansion of CAR-T cells in vivo and mitigate toxicity.
Collapse
|
8
|
Marr A, Markert A, Altmann A, Askoxylakis V, Haberkorn U. Biotechnology techniques for the development of new tumor specific peptides. Methods 2011; 55:215-22. [PMID: 21640826 DOI: 10.1016/j.ymeth.2011.05.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Revised: 05/09/2011] [Accepted: 05/18/2011] [Indexed: 12/28/2022] Open
Abstract
Peptides, proteins and antibodies are promising candidates as carriers for radionuclides in endoradiotherapy. This novel class of pharmaceuticals offers a great potential for the targeted therapy of cancer. The fact that some receptors are overexpressed in several tumor types and can be targeted by small peptides, proteins or antibodies conjugated to radionuclides has been used in the past for the development of peptide endoradiotherapeutic agents such as (90)Y-DOTATOC or radioimmunotherapy of lymphomas with Zevalin. These procedures have been shown to be powerful options for the treatment of cancer patients. Design of new peptide libraries and scaffolds combined with biopanning techniques like phage and ribosome display may lead to the discovery of new specific ligands for target structures overexpressed in malignant tumors. Display methods are high throughput systems which select for high affinity binders. These methods allow the screening of a vast amount of potential binding motifs which may be exposed to either cells overexpressing the target structures or in a cell-free system to the protein itself. Labelling these binders with radionuclides creates new potential tracers for application in diagnosis and endoradiotherapy. This review highlights the advantages and problems of phage and ribosome display for the identification and evaluation of new tumor specific peptides.
Collapse
Affiliation(s)
- Annabell Marr
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center, INF 260, 69120 Heidelberg, Germany.
| | | | | | | | | |
Collapse
|
9
|
Lin X, Liu M, Chen XW. Assessing reliability of protein-protein interactions by integrative analysis of data in model organisms. BMC Bioinformatics 2009; 10 Suppl 4:S5. [PMID: 19426453 PMCID: PMC2681066 DOI: 10.1186/1471-2105-10-s4-s5] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Background Protein-protein interactions play vital roles in nearly all cellular processes and are involved in the construction of biological pathways such as metabolic and signal transduction pathways. Although large-scale experiments have enabled the discovery of thousands of previously unknown linkages among proteins in many organisms, the high-throughput interaction data is often associated with high error rates. Since protein interaction networks have been utilized in numerous biological inferences, the inclusive experimental errors inevitably affect the quality of such prediction. Thus, it is essential to assess the quality of the protein interaction data. Results In this paper, a novel Bayesian network-based integrative framework is proposed to assess the reliability of protein-protein interactions. We develop a cross-species in silico model that assigns likelihood scores to individual protein pairs based on the information entirely extracted from model organisms. Our proposed approach integrates multiple microarray datasets and novel features derived from gene ontology. Furthermore, the confidence scores for cross-species protein mappings are explicitly incorporated into our model. Applying our model to predict protein interactions in the human genome, we are able to achieve 80% in sensitivity and 70% in specificity. Finally, we assess the overall quality of the experimentally determined yeast protein-protein interaction dataset. We observe that the more high-throughput experiments confirming an interaction, the higher the likelihood score, which confirms the effectiveness of our approach. Conclusion This study demonstrates that model organisms certainly provide important information for protein-protein interaction inference and assessment. The proposed method is able to assess not only the overall quality of an interaction dataset, but also the quality of individual protein-protein interactions. We expect the method to continually improve as more high quality interaction data from more model organisms becomes available and is readily scalable to a genome-wide application.
Collapse
Affiliation(s)
- Xiaotong Lin
- Bioinformatics and Computational Life-Science Laboratory, ITTC, Department of Electrical Engineering and Computer Science, The University of Kansas, 1520 west 15th Street, Lawrence, KS 66045, USA.
| | | | | |
Collapse
|
10
|
B MP, J MD, M PG. Mimotopes and proteome analyses using human genomic and cDNA epitope phage display. Comp Funct Genomics 2008; 3:254-63. [PMID: 18628851 PMCID: PMC2447272 DOI: 10.1002/cfg.174] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2001] [Accepted: 04/22/2002] [Indexed: 11/29/2022] Open
Abstract
In the post-genomic era, validation of candidate gene targets frequently requires proteinbased
strategies. Phage display is a powerful tool to define protein-protein interactions by
generating peptide binders against target antigens. Epitope phage display libraries have the
potential to enrich coding exon sequences from human genomic loci. We evaluated genomic
and cDNA phage display strategies to identify genes in the 5q31 Interleukin gene cluster
and to enrich cell surface receptor tyrosine kinase genes from a breast cancer cDNA
library. A genomic display library containing 2 × 106 clones with exon-sized inserts was
selected with antibodies specific for human Interleukin-4 (IL-4) and Interleukin-13. The
library was enriched significantly after two selection rounds and DNA sequencing revealed
unique clones. One clone matched a cognate IL-4 epitope; however, the majority of clone
insert sequences corresponded to E. coli genomic DNA. These bacterial sequences act as
‘mimotopes’ (mimetic sequences of the true epitope), correspond to open reading frames,
generate displayed peptides, and compete for binding during phage selection. The specificity
of these mimotopes for IL-4 was confirmed by competition ELISA. Other E. coli
mimotopes were generated using additional antibodies. Mimotopes for a receptor tyrosine
kinase gene were also selected using a breast cancer SKBR-3 cDNA phage display library,
screened against an anti-erbB2 monoclonal antibody. Identification of mimotopes in
genomic and cDNA phage libraries is essential for phage display-based protein validation
assays and two-hybrid phage approaches that examine protein-protein interactions. The
predominance of E. coli mimotopes suggests that the E. coli genome may be useful to
generate peptide diversity biased towards protein coding sequences. Abbreviations used: IL, interleukin; ELISA, enzyme linked immunoabsorbant assay;
PBS, phospho-buffered saline; cfu, colony forming units.
Collapse
Affiliation(s)
- Mullaney P B
- Departmentsof Laboratory Medicine and Cancer Center, University of California at San Francisco, San Francisco, CA 94143, USA
| | | | | |
Collapse
|
11
|
Tan FL, Ginsburg D. What a polyclonal antibody sees in von Willebrand factor. Thromb Res 2007; 121:519-26. [PMID: 17614124 PMCID: PMC2258224 DOI: 10.1016/j.thromres.2007.05.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2007] [Revised: 05/21/2007] [Accepted: 05/29/2007] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Von Willebrand factor (VWF) plays a critical role in hemostasis by carrying factor VIII (FVIII) and binding to specific ligands on the surface of blood platelets and within the blood vessel wall. MATERIALS AND METHODS We constructed a gene-specific phage display library containing small, random VWF fragments. Using this library, we mapped the repertoire of immune epitopes recognized by a commonly used commercial rabbit antihuman VWF polyclonal antibody. RESULTS A total of eight discrete epitopes within the VWF protein were identified, including two dominant epitopes that account for 74% of immuno selected VWF fragments. Comparison with previously mapped epitopes for mouse monoclonal antibodies reveals four overlapping regions that may identify common antigenic determinants. The distribution of these epitopes was not readily predicted from primary amino acid sequence divergence among these mammalian species or standard algorithms for the prediction of antigenicity, hydrophobicity, or surface probability. CONCLUSION Taken together with previous monoclonal antibody epitope mapping studies, our results suggest that a limited number of exposed domains on the surface of the human VWF protein may be the primary determinants of immunogenicity.
Collapse
Affiliation(s)
- Fen-Lai Tan
- Department of Internal Medicine and Department of Human Genetics, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
- Life Science Institute, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - David Ginsburg
- Department of Internal Medicine and Department of Human Genetics, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
- Life Science Institute, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
- Howard Hughes Medical Institute, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| |
Collapse
|
12
|
Structure Prediction of Protein Complexes. COMPUTATIONAL METHODS FOR PROTEIN STRUCTURE PREDICTION AND MODELING 2007. [DOI: 10.1007/978-0-387-68825-1_4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
13
|
Sergeeva A, Kolonin MG, Molldrem JJ, Pasqualini R, Arap W. Display technologies: application for the discovery of drug and gene delivery agents. Adv Drug Deliv Rev 2006; 58:1622-54. [PMID: 17123658 PMCID: PMC1847402 DOI: 10.1016/j.addr.2006.09.018] [Citation(s) in RCA: 170] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2006] [Accepted: 09/29/2006] [Indexed: 01/17/2023]
Abstract
Recognition of molecular diversity of cell surface proteomes in disease is essential for the development of targeted therapies. Progress in targeted therapeutics requires establishing effective approaches for high-throughput identification of agents specific for clinically relevant cell surface markers. Over the past decade, a number of platform strategies have been developed to screen polypeptide libraries for ligands targeting receptors selectively expressed in the context of various cell surface proteomes. Streamlined procedures for identification of ligand-receptor pairs that could serve as targets in disease diagnosis, profiling, imaging and therapy have relied on the display technologies, in which polypeptides with desired binding profiles can be serially selected, in a process called biopanning, based on their physical linkage with the encoding nucleic acid. These technologies include virus/phage display, cell display, ribosomal display, mRNA display and covalent DNA display (CDT), with phage display being by far the most utilized. The scope of this review is the recent advancements in the display technologies with a particular emphasis on molecular mapping of cell surface proteomes with peptide phage display. Prospective applications of targeted compounds derived from display libraries in the discovery of targeted drugs and gene therapy vectors are discussed.
Collapse
Affiliation(s)
- Anna Sergeeva
- Department of Blood and Marrow Transplantation, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Mikhail G. Kolonin
- Department of Genitourinary Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Jeffrey J. Molldrem
- Department of Blood and Marrow Transplantation, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Renata Pasqualini
- Department of Genitourinary Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, 77030, USA
- Department of Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Wadih Arap
- Department of Genitourinary Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, 77030, USA
- Department of Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, 77030, USA
| |
Collapse
|
14
|
Chen Y, Xu D. Bioinformatics analysis for interactive proteomics. CURRENT PROTOCOLS IN PROTEIN SCIENCE 2005; Chapter 25:25.1.1-25.1.14. [PMID: 18429286 DOI: 10.1002/0471140864.ps2501s42] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
High-throughput protein-protein interaction data are becoming a foundation for new biological discoveries. A major challenge is to manage, analyze, and model these data. In this unit several databases are described that are used to store, query, and visualize protein-protein interaction data. Comparison between experimental techniques reveals that each high-throughput technique has its limitations in detecting certain types of interactions; however, the techniques are generally complementary. In silico prediction methods for protein-protein interactions can expand the scope of experimental data and increase the confidence of certain interactions. Use of protein-protein interaction networks, preferably integrating them with other types of data, allows assignment of cellular functions to novel proteins and derivation of new biological pathways. As demonstrated in this unit, bioinformatics can be used to transform protein-protein interaction data from noisy information into knowledge of cellular mechanisms.
Collapse
Affiliation(s)
- Yu Chen
- Monsanto Company, St. Louis, Missouri
| | - Dong Xu
- University of Missouri-Columbia, Columbia, Missouri
| |
Collapse
|
15
|
Nowakowski GS, Dooner MS, Valinski HM, Mihaliak AM, Quesenberry PJ, Becker PS. A specific heptapeptide from a phage display peptide library homes to bone marrow and binds to primitive hematopoietic stem cells. Stem Cells 2005; 22:1030-8. [PMID: 15536193 DOI: 10.1634/stemcells.22-6-1030] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Phage display peptide libraries have enabled the discovery of peptides that selectively target specific organs. Selection of organ-specific peptides is mediated through binding of peptides displayed on phage coat protein to adhesion molecules expressed within targeted organs. Hematopoietic stem cells selectively home to bone marrow, and certain adhesion receptors critical to this function have been demonstrated. Using a phage display library, we identified a specific peptide that trafficked to murine bone marrow in vivo. We independently isolated exactly the same heptapeptide from the entire library by in vitro biopanning on primitive lineage-depleted, Hoechst 33342(dull)/rhodamine 123(dull) murine bone marrow stem cells and confirmed peptide binding to these cells by immunofluorescence studies. We demonstrated bone marrow-specific homing of the peptide by an in vivo assay in which the animals were injected with the phage displaying peptide sequence, and immunofluorescence analysis of multiple organs was performed. We also showed that the peptide significantly decreased the homing of stem cells to the bone marrow but not to the spleen 3 hours after transplantation using fluorescently labeled Lin(-)Sca(+) hematopoietic cells in an in vivo homing assay. The peptide sequence has a partial (5/7) amino acid sequence homology with a region of CD84. This discovery represents the first application of the phage display methodology to the bone marrow and stem cells and led to the identification of a specific heptapeptide that homes to bone marrow, binds to primitive stem cells, and plays a role in stem cell homing.
Collapse
Affiliation(s)
- Grzegorz S Nowakowski
- Division of Gene Therapy, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | | | | | | | | | | |
Collapse
|
16
|
Quesenberry PJ, Colvin G, Abedi M. Perspective: fundamental and clinical concepts on stem cell homing and engraftment: a journey to niches and beyond. Exp Hematol 2005; 33:9-19. [PMID: 15661393 DOI: 10.1016/j.exphem.2004.10.012] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2004] [Revised: 09/29/2004] [Accepted: 10/12/2004] [Indexed: 11/16/2022]
Abstract
In many ways, the homing of hematopoietic stem cells to bone marrow and other tissues defines these cells and their immediate and long-term fates Once homed, an inevitable series of proliferative and differentiative events presumptively follows. These comments, of course, hold for both homing to marrow, or alternatively, to other nonmarrow tissues. In this review, we will specifically focus on homing and engraftment to bone marrow because this is the best-studied and clinically applicable system.
Collapse
Affiliation(s)
- Peter J Quesenberry
- Department of Research, Roger Williams Medical Center, Providence, RI 02908-4735, USA.
| | | | | |
Collapse
|
17
|
Gasparetto M, Gentry T, Sebti S, O'Bryan E, Nimmanapalli R, Blaskovich MA, Bhalla K, Rizzieri D, Haaland P, Dunne J, Smith C. Identification of compounds that enhance the anti-lymphoma activity of rituximab using flow cytometric high-content screening. J Immunol Methods 2004; 292:59-71. [PMID: 15350512 DOI: 10.1016/j.jim.2004.06.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2004] [Revised: 05/27/2004] [Accepted: 06/02/2004] [Indexed: 11/22/2022]
Abstract
In this report, we describe a new flow cytometry technique termed flow cytometric high-content screening (FC-HCS) which involves semi-automated processing and analysis of multiparameter flow cytometry samples. As a first test of the FC-HCS technique, we used it to screen a 2000-compound library, called the National Cancer Institute (NCI) Diversity Set, to identify agents that would enhance the anti-lymphoma activity of the therapeutic monoclonal antibody rituximab. FC-HCS identified 15 compounds from the Diversity Set that significantly enhanced the ability of rituximab to inhibit cell cycle progression and induce apoptosis in lymphoma cells. The validity of the screening results was confirmed for several compounds using additional assays of cell proliferation, apoptosis and cell growth. The FC-HCS technique was relatively simple and reliable and could process up to 1000 samples/day on a single flow cytometer. The FC-HCS technique may be useful for a variety of applications including drug discovery, immunologic monitoring of patients, functional genomics studies and tissue engineering efforts.
Collapse
Affiliation(s)
- Maura Gasparetto
- Duke University Medical Center, Duke University, Durham, NC, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
The field of combinatorial peptide chemistry has emerged as a powerful tool in the study of many biological systems. This review focuses on combinatorial peptide library methodology, which includes biological library methods, spatially addressable parallel library methods, library methods requiring deconvolution, the "one-bead one-compound" library method, and affinity chromatography selection method. These peptide libraries have successfully been employed to study a vast array of cell surface receptors, as well as have been useful in identifying protein kinase substrates and inhibitors. In recent immunobiological applications, peptide libraries have proven monumental in the definition of MHC anchor residues, in lymphocyte epitope mapping, and in the development of peptide vaccines. Peptides identified from such libraries, when presented in a chemical microarray format, may prove useful in immunodiagnostics. Combinatorial peptide libraries offer a high-throughput approach to study limitless biological targets. Peptides discovered from such studies may be therapeutically and diagnostically useful agents.
Collapse
Affiliation(s)
- Ruiwu Liu
- UC Davis Cancer Center, Division of Hematology/Oncology, and Department of Internal Medicine, University of California Davis, Sacramento, CA, USA
| | | | | |
Collapse
|