1
|
Ahmed AAM, Mekky AEM, Sanad SMH. Effective synthesis of new benzo-fused macrocyclic and heteromacrocyclic bis(Schiff bases). JOURNAL OF THE IRANIAN CHEMICAL SOCIETY 2022. [DOI: 10.1007/s13738-021-02409-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
2
|
Ahmed AAM, Mekky AEM, Sanad SMH. Effective synthesis of new benzo‐fused macrocyclic and thiamacrocyclic dilactams and related pyrazolo‐fused macrocycles. J Heterocycl Chem 2021. [DOI: 10.1002/jhet.4383] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Ahmed A. M. Ahmed
- Chemistry Department, Faculty of Science Cairo University Giza Egypt
- Common First Year Deanship Jouf University Sakaka Saudi Arabia
| | - Ahmed E. M. Mekky
- Chemistry Department, Faculty of Science Cairo University Giza Egypt
| | | |
Collapse
|
3
|
Jones HF, Molvi Z, Klatt MG, Dao T, Scheinberg DA. Empirical and Rational Design of T Cell Receptor-Based Immunotherapies. Front Immunol 2021; 11:585385. [PMID: 33569049 PMCID: PMC7868419 DOI: 10.3389/fimmu.2020.585385] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 12/04/2020] [Indexed: 01/04/2023] Open
Abstract
The use of T cells reactive with intracellular tumor-associated or tumor-specific antigens has been a promising strategy for cancer immunotherapies in the past three decades, but the approach has been constrained by a limited understanding of the T cell receptor's (TCR) complex functions and specificities. Newer TCR and T cell-based approaches are in development, including engineered adoptive T cells with enhanced TCR affinities, TCR mimic antibodies, and T cell-redirecting bispecific agents. These new therapeutic modalities are exciting opportunities by which TCR recognition can be further exploited for therapeutic benefit. In this review we summarize the development of TCR-based therapeutic strategies and focus on balancing efficacy and potency versus specificity, and hence, possible toxicity, of these powerful therapeutic modalities.
Collapse
Affiliation(s)
- Heather F. Jones
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Weill Cornell Medicine, New York, NY, United States
| | - Zaki Molvi
- Weill Cornell Medicine, New York, NY, United States
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Martin G. Klatt
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Tao Dao
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - David A. Scheinberg
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
4
|
Nanomaterials for Protein Delivery in Anticancer Applications. Pharmaceutics 2021; 13:pharmaceutics13020155. [PMID: 33503889 PMCID: PMC7910976 DOI: 10.3390/pharmaceutics13020155] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 01/22/2021] [Accepted: 01/22/2021] [Indexed: 12/16/2022] Open
Abstract
Nanotechnology platforms, such as nanoparticles, liposomes, dendrimers, and micelles have been studied extensively for various drug deliveries, to treat or prevent diseases by modulating physiological or pathological processes. The delivery drug molecules range from traditional small molecules to recently developed biologics, such as proteins, peptides, and nucleic acids. Among them, proteins have shown a series of advantages and potential in various therapeutic applications, such as introducing therapeutic proteins due to genetic defects, or used as nanocarriers for anticancer agents to decelerate tumor growth or control metastasis. This review discusses the existing nanoparticle delivery systems, introducing design strategies, advantages of using each system, and possible limitations. Moreover, we will examine the intracellular delivery of different protein therapeutics, such as antibodies, antigens, and gene editing proteins into the host cells to achieve anticancer effects and cancer vaccines. Finally, we explore the current applications of protein delivery in anticancer treatments.
Collapse
|
5
|
Chang J, Liu ZS, Song DF, Li M, Zhang S, Zhao K, Guan YT, Ren HL, Li YS, Zhou Y, Liu XL, Lu SY, Hu P. Cholecystokinin type 2 receptor in colorectal cancer: diagnostic and therapeutic target. J Cancer Res Clin Oncol 2020; 146:2205-2217. [PMID: 32488497 DOI: 10.1007/s00432-020-03273-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 05/23/2020] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Cholecystokinin type 2 receptor (CCK2R), which mediates the action of gastrin and cholecystokinin (CCK), has been demonstrated to promote the proliferation of colorectal cancer (CRC). A number of studies showed that CCK2R overexpressed in gastric cancer and pancreatic cancer but few in CRC. The correlation between CCK2R expression and clinicopathological characteristics is also not clear. METHODS This study investigated CCK2R expression in a wide range of cell lines and clinical CRC samples, and explored expression pattern and prognostic value of CCK2R in relation to clinicopathological parameters. The location and expression levels of CCK2R were measured by immunocytochemical (ICC), qRT-PCR and Western blot. The druggability and antineoplastic effects of CCK2R as a therapeutic target were investigated using an anti-CCK2R targeting recombinant toxin named rCCK8PE38 by CCK-8 assay. RESULTS Compared with paracarcinoma tissues, tumor samples showed overexpression of CCK2R (p = 0.028) including both CRC tissue and plasma samples, with plasma detection showing a significant indication for CCK2R evaluation. Aberrant expression correlated significantly with histological type (p = 0.032) and p53 status (p < 0.01), and patients with CCK2R overexpression had significantly lower disease-free survival. Application of rCCK8PE38 demonstrated the specificity and druggability of CCK2R as a therapeutic target, providing a strategy for clinical case screening of drugs targeting CCK2R. CONCLUSION This study highlighted the aberrant expression and clinical correlation of CCK2R and reveals its diagnostic, prognostic and treatment value in CRC. We hypothesize that CCK2R serve as a target for the diagnosis and treatment of this cancer.
Collapse
Affiliation(s)
- Jiang Chang
- Key Laboratory of Zoonosis Research, Ministry of Education/Institute of Zoonosis, College of Veterinary Medicine, Double-First Class Discipline of Human-Animal Medicine, Jilin University, Xi An Da Lu 5333, Changchun, 130062, China
| | - Zeng-Shan Liu
- Key Laboratory of Zoonosis Research, Ministry of Education/Institute of Zoonosis, College of Veterinary Medicine, Double-First Class Discipline of Human-Animal Medicine, Jilin University, Xi An Da Lu 5333, Changchun, 130062, China
| | - De-Feng Song
- China-Japan Union Hospital, Xian Tai Da Jie 126, Changchun, 130033, China
| | - Meng Li
- Key Laboratory of Zoonosis Research, Ministry of Education/Institute of Zoonosis, College of Veterinary Medicine, Double-First Class Discipline of Human-Animal Medicine, Jilin University, Xi An Da Lu 5333, Changchun, 130062, China
| | - Song Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education/Institute of Zoonosis, College of Veterinary Medicine, Double-First Class Discipline of Human-Animal Medicine, Jilin University, Xi An Da Lu 5333, Changchun, 130062, China
| | - Ke Zhao
- Key Laboratory of Zoonosis Research, Ministry of Education/Institute of Zoonosis, College of Veterinary Medicine, Double-First Class Discipline of Human-Animal Medicine, Jilin University, Xi An Da Lu 5333, Changchun, 130062, China
| | - Yu-Ting Guan
- Key Laboratory of Zoonosis Research, Ministry of Education/Institute of Zoonosis, College of Veterinary Medicine, Double-First Class Discipline of Human-Animal Medicine, Jilin University, Xi An Da Lu 5333, Changchun, 130062, China
| | - Hong-Lin Ren
- Key Laboratory of Zoonosis Research, Ministry of Education/Institute of Zoonosis, College of Veterinary Medicine, Double-First Class Discipline of Human-Animal Medicine, Jilin University, Xi An Da Lu 5333, Changchun, 130062, China
| | - Yan-Song Li
- Key Laboratory of Zoonosis Research, Ministry of Education/Institute of Zoonosis, College of Veterinary Medicine, Double-First Class Discipline of Human-Animal Medicine, Jilin University, Xi An Da Lu 5333, Changchun, 130062, China
| | - Yu Zhou
- Key Laboratory of Zoonosis Research, Ministry of Education/Institute of Zoonosis, College of Veterinary Medicine, Double-First Class Discipline of Human-Animal Medicine, Jilin University, Xi An Da Lu 5333, Changchun, 130062, China
| | - Xi-Lin Liu
- China-Japan Union Hospital, Xian Tai Da Jie 126, Changchun, 130033, China.
| | - Shi-Ying Lu
- Key Laboratory of Zoonosis Research, Ministry of Education/Institute of Zoonosis, College of Veterinary Medicine, Double-First Class Discipline of Human-Animal Medicine, Jilin University, Xi An Da Lu 5333, Changchun, 130062, China. .,Key Laboratory of Zoonosis Research, Ministry of Education/Institute of Zoonosis, Jilin University, Xi An Da Lu 5333, Changchun, 130062, China.
| | - Pan Hu
- Key Laboratory of Zoonosis Research, Ministry of Education/Institute of Zoonosis, College of Veterinary Medicine, Double-First Class Discipline of Human-Animal Medicine, Jilin University, Xi An Da Lu 5333, Changchun, 130062, China. .,Key Laboratory of Zoonosis Research, Ministry of Education/Institute of Zoonosis, Jilin University, Xi An Da Lu 5333, Changchun, 130062, China.
| |
Collapse
|
6
|
Conejo-Garcia JR, Biswas S, Chaurio R. Humoral immune responses: Unsung heroes of the war on cancer. Semin Immunol 2020; 49:101419. [PMID: 33183950 PMCID: PMC7738315 DOI: 10.1016/j.smim.2020.101419] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/23/2020] [Accepted: 10/30/2020] [Indexed: 02/07/2023]
Abstract
Solid cancers progress from primordial lesions through complex interactions between tumor-promoting and anti-tumor immune cell types, ultimately leading to the orchestration of humoral and T cell adaptive immune responses, albeit in an immunosuppressive environment. B cells infiltrating most established tumors have been associated with a dual role: Some studies have associated antibodies produced by tumor-associated B cells with the promotion of regulatory activities on myeloid cells, and also with direct immunosuppression through the production of IL-10, IL-35 or TGF-β. In contrast, recent studies in multiple human malignancies identify B cell responses with delayed malignant progression and coordinated T cell protective responses. This includes the elusive role of Tertiary Lymphoid Structures identified in many human tumors, where the function of B cells remains unknown. Here, we discuss emerging data on the dual role of B cell responses in the pathophysiology of human cancer, providing a perspective on future directions and possible novel interventions to restore the coordinated action of both branches of the adaptive immune response, with the goal of maximizing immunotherapeutic effectiveness.
Collapse
Affiliation(s)
- Jose R Conejo-Garcia
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
| | - Subir Biswas
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Ricardo Chaurio
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
7
|
Sabharwal P, Sushmitha C, Amritha CK, Natraj U, Murthy MRN, Savithri HS. Development of pepper vein banding virus chimeric virus-like particles for potential diagnostic and therapeutic applications. Arch Virol 2020; 165:1163-1176. [DOI: 10.1007/s00705-020-04581-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 02/05/2020] [Indexed: 12/18/2022]
|
8
|
Jauset T, Beaulieu ME. Bioactive cell penetrating peptides and proteins in cancer: a bright future ahead. Curr Opin Pharmacol 2019; 47:133-140. [PMID: 31048179 DOI: 10.1016/j.coph.2019.03.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/26/2019] [Accepted: 03/27/2019] [Indexed: 02/05/2023]
Abstract
Peptides and proteins bear an extraordinary therapeutic potential to effectively and selectively target many components of cells currently considered undruggable. However, their intracellular delivery remains a critical challenge. Cell penetrating peptides and protein domains (CPPs) can be employed to translocate therapeutic polypeptides through the cellular membrane. Here, we describe examples of linear peptides and proteins, byciclic macropeptides and nanobodies that target key players in cancer development, with intrinsic and engineered cell penetrating ability. We also describe current solutions to the main challenges to their clinical viability.
Collapse
Affiliation(s)
- Toni Jauset
- Peptomyc, Edifici Cellex, Hospital Vall d'Hebron, Barcelona, 08035, Spain
| | - Marie-Eve Beaulieu
- Peptomyc, Edifici Cellex, Hospital Vall d'Hebron, Barcelona, 08035, Spain.
| |
Collapse
|
9
|
Huang Y, Wiedmann MM, Suga H. RNA Display Methods for the Discovery of Bioactive Macrocycles. Chem Rev 2018; 119:10360-10391. [PMID: 30395448 DOI: 10.1021/acs.chemrev.8b00430] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The past two decades have witnessed the emergence of macrocycles, including macrocyclic peptides, as a promising yet underexploited class of de novo drug candidates. Both rational/computational design and in vitro display systems have contributed tremendously to the development of cyclic peptide binders of either traditional targets such as cell-surface receptors and enzymes or challenging targets such as protein-protein interaction surfaces. mRNA display, a key platform technology for the discovery of cyclic peptide ligands, has become one of the leading strategies that can generate natural-product-like macrocyclic peptide binders with antibody-like affinities. On the basis of the original cell-free transcription/translation system, mRNA display is highly evolvable to realize its full potential by applying genetic reprogramming and chemical/enzymatic modifications. In addition, mRNA display also allows the follow-up hit-to-lead development using high-throughput focused affinity maturation. Finally, mRNA-displayed peptides can be readily engineered to create chemical conjugates based on known small molecules or biologics. This review covers the birth and growth of mRNA display and discusses the above features of mRNA display with success stories and future perspectives and is up to date as of August 2018.
Collapse
Affiliation(s)
- Yichao Huang
- Department of Chemistry, Graduate School of Science , The University of Tokyo , 7-3-1 Hongo , Bunkyo-ku, Tokyo 113-0033 , Japan
| | - Mareike Margarete Wiedmann
- Department of Chemistry, Graduate School of Science , The University of Tokyo , 7-3-1 Hongo , Bunkyo-ku, Tokyo 113-0033 , Japan
| | - Hiroaki Suga
- Department of Chemistry, Graduate School of Science , The University of Tokyo , 7-3-1 Hongo , Bunkyo-ku, Tokyo 113-0033 , Japan
| |
Collapse
|
10
|
Zhang JF, Xiong HL, Cao JL, Wang SJ, Guo XR, Lin BY, Zhang Y, Zhao JH, Wang YB, Zhang TY, Yuan Q, Zhang J, Xia NS. A cell-penetrating whole molecule antibody targeting intracellular HBx suppresses hepatitis B virus via TRIM21-dependent pathway. Am J Cancer Res 2018; 8:549-562. [PMID: 29290826 PMCID: PMC5743566 DOI: 10.7150/thno.20047] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 09/27/2017] [Indexed: 12/11/2022] Open
Abstract
Rationale: Monoclonal antibodies (mAbs) mostly targeting extracellular or cell surface molecules have been widely used in the treatment of various diseases. However, mAbs cannot pass through the cell membrane as efficiently as small compounds, thus limiting their use against intracellular targets. Methods to shuttle antibodies into living cells may largely expand research and application in areas based on mAbs. Hepatitis B virus X protein (HBx) is an important intracellular multi-functional viral protein in the life cycle of hepatitis B virus (HBV). HBx plays essential roles in virus infection and replication and is strongly associated with HBV-related carcinogenesis. Methods: In this study, we developed a cell-penetrating whole molecule antibody targeting HBx (9D11-Tat) by the fusion of a cell penetrating peptide (CPP) on the C-terminus of the heavy chain of a potent mAb specific to HBx (9D11). The anti-HBV effect and mechanism of 9D11-Tat were investigated in cell and mouse models mimicking chronic HBV infection. Results: Our results demonstrated that the recombinant 9D11-Tat antibody could efficiently internalize into living cells and significantly suppress viral transcription, replication, and protein production both in vitro and in vivo. Further analyses suggested the internalized 9D11-Tat antibody could greatly reduce intracellular HBx via Fc binding receptor TRIM21-mediated protein degradation. This process simultaneously stimulated the activations of NF-κB, AP-1, and IFN-β, which promoted an antiviral state of the host cell. Conclusion: In summary, our study offers a new approach to target intracellular pathogenesis-related protein by engineered cell-penetrating mAb expanding their potential for therapeutic applications. Moreover, the 9D11-Tat antibody may provide a novel therapeutic agent against human chronic HBV infection.
Collapse
|
11
|
Davis AM, Plowright AT, Valeur E. Directing evolution: the next revolution in drug discovery? Nat Rev Drug Discov 2017; 16:681-698. [PMID: 28935911 DOI: 10.1038/nrd.2017.146] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The strong biological rationale to pursue challenging drug targets such as protein-protein interactions has stimulated the development of novel screening strategies, such as DNA-encoded libraries, to allow broader areas of chemical space to be searched. There has also been renewed interest in screening natural products, which are the result of evolutionary selection for a function, such as interference with a key signalling pathway of a competing organism. However, recent advances in several areas, such as understanding of the biosynthetic pathways for natural products, synthetic biology and the development of biosensors to detect target molecules, are now providing new opportunities to directly harness evolutionary pressure to identify and optimize compounds with desired bioactivities. Here, we describe innovations in the key components of such strategies and highlight pioneering examples that indicate the potential of the directed-evolution concept. We also discuss the scientific gaps and challenges that remain to be addressed to realize this potential more broadly in drug discovery.
Collapse
Affiliation(s)
- Andrew M Davis
- AstraZeneca R&D Gothenburg, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Alleyn T Plowright
- Integrated Drug Discovery, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Eric Valeur
- AstraZeneca R&D Gothenburg, Pepparedsleden 1, Mölndal, 43150, Sweden
| |
Collapse
|
12
|
Gabri MR, Cacciavillano W, Chantada GL, Alonso DF. Racotumomab for treating lung cancer and pediatric refractory malignancies. Expert Opin Biol Ther 2016; 16:573-8. [DOI: 10.1517/14712598.2016.1157579] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
13
|
Veomett N, Dao T, Liu H, Xiang J, Pankov D, Dubrovsky L, Whitten JA, Park SM, Korontsvit T, Zakhaleva V, Casey E, Curcio M, Kharas MG, O'Reilly RJ, Liu C, Scheinberg DA. Therapeutic efficacy of an Fc-enhanced TCR-like antibody to the intracellular WT1 oncoprotein. Clin Cancer Res 2014; 20:4036-46. [PMID: 24850840 DOI: 10.1158/1078-0432.ccr-13-2756] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE RMFPNAPYL (RMF), a Wilms' tumor gene 1 (WT1)-derived CD8 T-cell epitope presented by HLA-A*02:01, is a validated target for T-cell-based immunotherapy. We previously reported ESK1, a high avidity (Kd < 0.2 nmol/L), fully-human monoclonal antibody (mAb) specific for the WT1 RMF peptide/HLA-A*02:01 complex, which selectively bound and killed WT1(+) and HLA-A*02:01(+) leukemia and solid tumor cell lines. EXPERIMENTAL DESIGN We engineered a second-generation mAb, ESKM, to have enhanced antibody-dependent cell-mediated cytotoxicity (ADCC) function due to altered Fc glycosylation. ESKM was compared with native ESK1 in binding assays, in vitro ADCC assays, and mesothelioma and leukemia therapeutic models and pharmacokinetic studies in mice. ESKM toxicity was assessed in HLA-A*02:01(+) transgenic mice. RESULTS ESK antibodies mediated ADCC against hematopoietic and solid tumor cells at concentrations below 1 μg/mL, but ESKM was about 5- to 10-fold more potent in vitro against multiple cancer cell lines. ESKM was more potent in vivo against JMN mesothelioma, and effective against SET2 AML and fresh ALL xenografts. ESKM had a shortened half-life (4.9 days vs. 6.5 days), but an identical biodistribution pattern in C57BL/6J mice. At therapeutic doses of ESKM, there was no difference in half-life or biodistribution in HLA-A*02:01(+) transgenic mice compared with the parent strain. Importantly, therapeutic doses of ESKM in these mice caused no depletion of total WBCs or hematopoetic stem cells, or pathologic tissue damage. CONCLUSIONS The data provide proof of concept that an Fc-enhanced mAb can improve efficacy against a low-density, tumor-specific, peptide/MHC target, and support further development of this mAb against an important intracellular oncogenic protein.
Collapse
Affiliation(s)
- Nicholas Veomett
- Sloan Kettering Institute; Weill Cornell Medical College, New York, New York
| | | | - Hong Liu
- Eureka Therapeutics Inc., Emeryville, California; and
| | - Jingyi Xiang
- Eureka Therapeutics Inc., Emeryville, California; and
| | | | | | | | | | | | | | | | | | | | | | - Cheng Liu
- Eureka Therapeutics Inc., Emeryville, California; and
| | - David A Scheinberg
- Sloan Kettering Institute; Weill Cornell Medical College, New York, New York;
| |
Collapse
|
14
|
A TCR-mimic antibody to WT1 bypasses tyrosine kinase inhibitor resistance in human BCR-ABL+ leukemias. Blood 2014; 123:3296-304. [PMID: 24723681 DOI: 10.1182/blood-2014-01-549022] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Acute and chronic leukemias, including CD34(+) CML cells, demonstrate increased expression of the Wilms tumor gene 1 product (WT1), making WT1 an attractive therapeutic target. However, WT1 is a currently undruggable, intracellular protein. ESKM is a human IgG1 T-cell receptor mimic monoclonal antibody directed to a 9-amino acid sequence of WT1 in the context of cell surface HLA-A*02. ESKM was therapeutically effective, alone and in combination with tyrosine kinase inhibitors (TKIs), against Philadelphia chromosome-positive acute leukemia in murine models, including a leukemia with the most common, pan-TKI, gatekeeper resistance mutation, T315I. ESKM was superior to the first-generation TKI, imatinib. Combination therapy with ESKM and TKIs was superior to either drug alone, capable of curing mice. ESKM showed no toxicity to human HLA-A*02:01(+) stem cells under the conditions of this murine model. These features of ESKM make it a promising nontoxic therapeutic agent for sensitive and resistant Ph(+) leukemias.
Collapse
|