1
|
Wilke AC, Gökbuget N. The role of blinatumomab in adult acute B lymphoblastic leukaemia. Br J Haematol 2025. [PMID: 40368871 DOI: 10.1111/bjh.20134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 04/25/2025] [Indexed: 05/16/2025]
Abstract
Blinatumomab is a CD19 × CD3-directed bispecific T-cell engager that has become an essential backbone of acute B lymphoblastic leukaemia (BCP-ALL) treatment. It is used in relapsed/refractory disease, minimal residual disease (MRD), as well as in first-line treatment. Blinatumomab is particularly effective in MRD-positive BCP-ALL, inducing a high rate of deep molecular response, with very good overall tolerability. Very promising data are reported for the addition of blinatumomab to first-line therapy. This review focusses on new aspects of blinatumomab treatment in Philadelphia-negative BCP-ALL treatment in adult patients, including MRD-positive and relapsed/refractory disease, as well as reviewing recent clinical trials implementing blinatumomab into first-line therapy of BCP-ALL treatment.
Collapse
Affiliation(s)
- Anne C Wilke
- Department of Medicine II, Department for Hematology/Oncology, Goethe University, Frankfurt am Main, Germany
| | - Nicola Gökbuget
- Department of Medicine II, Department for Hematology/Oncology, Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
2
|
Liu ZY, Zhang SJ, Yan ZY, Sun HM, Chen YB. [Clinical analysis of 16 cases of adult acute B-lymphoblastic leukemia treated with blinatumomab]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2025; 46:269-272. [PMID: 40355357 PMCID: PMC12038468 DOI: 10.3760/cma.j.cn121090-20240611-00219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Indexed: 05/14/2025]
Abstract
This study aimed to investigate the efficacy and safety of blinatumomab in adult patients with acute B-lymphoblastic leukemia (B-ALL) by conducting a retrospective analysis of the clinical data from 16 patients with B-ALL receiving blinatumomab at the Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, from June 2022 to April 2024. Among the 16 patients, 10 were classified as relapsed/refractory B-ALL and 6 were newly diagnosed Ph(-) B-ALL. Of the 10 patients with relapsed/refractory B-ALL, 8 achieved complete remission (CR) and minimal residual disease (MRD) negativity after one blinatumomab treatment cycle. In the 6 newly diagnosed patients, the bone marrow MRD was negative after one blinatumomab treatment cycle after initial induction chemotherapy followed by sequential blinatumomab treatment. Among them, four completed allogeneic hematopoietic stem cell transplantation and continuously maintained CR. This indicates that blinatumomab exhibits a high remission rate in both patients with relapsed/refractory and newly diagnosed B-ALL, thereby providing the possibility of bridging to transplantation and extending patient survival, with manageable adverse reactions.
Collapse
Affiliation(s)
- Z Y Liu
- Department of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201800, China
| | - S J Zhang
- Department of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201800, China
| | - Z Y Yan
- Department of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201800, China
| | - H M Sun
- Department of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201800, China
| | - Y B Chen
- Department of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201800, China
| |
Collapse
|
3
|
Guégan M, Bichon M, Chaput N, Houot R, Lemoine J. Cancer immunotherapy in elderly patients: The concept of immune senescence challenged by clinical experience. Eur J Cancer 2025; 214:115145. [PMID: 39615332 DOI: 10.1016/j.ejca.2024.115145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 12/20/2024]
Abstract
Cancer immunotherapy, including immune checkpoint inhibitors, chimeric antigen receptor T-cell therapy and bispecific antibodies, has led to major improvements in the treatment of a wide range of hematologic malignancies and solid tumors. However, age-mediated immune system modifications, known as immunosenescence, may preclude its efficacy in elderly patients. In this review, we assessed the efficacy of these different cancer immunotherapies in elderly patients compared to young patients to revisit the concept of immunosenescence from a therapeutic perspective.
Collapse
Affiliation(s)
- Mathilde Guégan
- Department of Hematology, CHU de Rennes, Université de Rennes, 2 rue Henri le Guilloux, 35033, Rennes Cedex 9, France
| | - Malvina Bichon
- Department of Hematology, CHU de Rennes, Université de Rennes, 2 rue Henri le Guilloux, 35033, Rennes Cedex 9, France
| | - Nathalie Chaput
- Laboratoire d'Immunomonitoring en Oncologie, INSERM US23, CNRS UMS 3655, Gustave Roussy, Université Paris-Saclay, Villejuif, Ile-de-France, France
| | - Roch Houot
- Department of Hematology, CHU de Rennes, Université de Rennes, 2 rue Henri le Guilloux, 35033, Rennes Cedex 9, France
| | - Jean Lemoine
- Department of Hematology, AP-HP, Université Paris Cité, Paris, France; Center for Cellular Immunotherapies and Division of Hematology-Oncology, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
4
|
Arvedson T, Bailis JM, Urbig T, Stevens JL. Considerations for design, manufacture, and delivery for effective and safe T-cell engager therapies. Curr Opin Biotechnol 2022; 78:102799. [PMID: 36179408 DOI: 10.1016/j.copbio.2022.102799] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 08/12/2022] [Accepted: 08/25/2022] [Indexed: 12/14/2022]
Abstract
T-cell engager (TCE) molecules provide a targeted immunotherapy approach to treat hematologic malignancies and solid tumors. Since the approval of the CD19-targeted BiTE® (bispecific T-cell engager) molecule blinatumomab, multiple TCE molecules against different targets have been developed in several tumor types, with the approval of three additional TCE molecules in 2022. Some of the initial challenges, such as the need for continuous intravenous administration and low productivity, have been addressed in subsequent iterations of the platform by advancing half-life extended, Fc-based molecules. As clinical data from these molecules emerge, additional optimization of formats and manufacturability will be necessary. Ongoing efforts are focused on further improving TCE efficacy, safety, and convenience of administration.
Collapse
Affiliation(s)
- Tara Arvedson
- Amgen Research, Amgen Inc., South San Francisco, CA, USA
| | - Julie M Bailis
- Amgen Research, Amgen Inc., South San Francisco, CA, USA
| | | | | |
Collapse
|
5
|
Mocquot P, Mossazadeh Y, Lapierre L, Pineau F, Despas F. The pharmacology of blinatumomab: state of the art on pharmacodynamics, pharmacokinetics, adverse drug reactions and evaluation in clinical trials. J Clin Pharm Ther 2022; 47:1337-1351. [PMID: 35906791 PMCID: PMC9796714 DOI: 10.1111/jcpt.13741] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 01/07/2023]
Abstract
WHAT IS KNOWN AND OBJECTIVE Bispecific drugs (BDs) belong to the family of immunotherapies along with checkpoint inhibitors and CAR-T cells. In the field of oncology, BDs are designed to simultaneously bind a tumour antigen on the one side and an antigen present on the surface of effector cells on the other. This review summarizes the information available to date on the first marketed BiTE-format bispecific antibody, blinatumomab BLINCYTO® in acute lymphoblastic leukaemia. METHODS A literature search was conducted in the PubMed database by including studies published in English using the term blinatumomab. Furthermore, bibliographies of selected references were also evaluated for relevant articles. Clinical trial (CT) data were retrieved from clinicaltrials.gov (ongoing trials, adverse events [AEs]) and global pharmacovigilance data were retrieved from VigiBase®. RESULTS AND DISCUSSION Blinatumomab is a fusion protein which consists of two single-chain variable fragments arranged in tandem: the first binds the CD19 surface antigen of all B cells and the second targets the CD3 antigen of T cells. Binding of blinatumomab to B and T cells induces apoptosis of B cells after secretion of granzymes and perforins by T cells. T-cell activation results in secretion of pro-inflammatory cytokines and upregulation of activation markers and adhesion molecules on the surface of T cells. The major CTs that led to an indication show increased overall survival with blinatumomab with better efficacy in patients in haematological remission with minimal residual disease ≥10-3 . The major AEs are cytokine release syndrome, neurotoxicity and hypogammaglobulinemia. The three most frequent system organ classes in CTs are haematological, gastrointestinal and general disorders. These results are also found in VigiBase® but neurological disorders and infections appear more frequently in real life. WHAT IS NEW AND CONCLUSION This review summarizes the current knowledge of blinatumomab in the literature. The subject of many CTs is to improve the route of administration and expand the indications for treatment.
Collapse
Affiliation(s)
- Pauline Mocquot
- Département de Pharmacologie Médicale, CHU de ToulouseUniversité Toulouse III ‐ Paul SabatierToulouseFrance
| | - Yasmine Mossazadeh
- Département de Pharmacologie Médicale, CHU de ToulouseUniversité Toulouse III ‐ Paul SabatierToulouseFrance
| | - Léopoldine Lapierre
- Département d'Hématologie et de Médecine InterneInstitut Universitaire du Cancer‐Oncopole, CHU de ToulouseToulouseFrance
| | - Fanny Pineau
- Département d'Hématologie et de Médecine InterneInstitut Universitaire du Cancer‐Oncopole, CHU de ToulouseToulouseFrance
| | - Fabien Despas
- Département de Pharmacologie Médicale, CHU de ToulouseUniversité Toulouse III ‐ Paul SabatierToulouseFrance,Université Toulouse III ‐ Paul SabatierToulouseFrance,INSERM CIC1436 CIC ToulouseFrance
| |
Collapse
|
6
|
IBI379, a novel B cell maturation antigen/CD3 bispecific T-cell engager, displays high antitumor efficacy in preclinical models of multiple myeloma. Cancer Lett 2022; 536:215663. [PMID: 35381307 DOI: 10.1016/j.canlet.2022.215663] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 02/21/2022] [Accepted: 03/21/2022] [Indexed: 11/24/2022]
Abstract
Multiple myeloma (MM) is a hematological malignancy that results from the malignant proliferation of plasma cells in the bone marrow. B cell maturation antigen (BCMA) is highly selectively expressed in malignant plasma cells and is a novel therapeutic target for MM. Here, we developed a bispecific T cell engager, IBI379, that targets BCMA and CD3, and investigated its antitumor efficacy against MM. IBI379 showed strong binding affinity with both BCMA and CD3, which triggered T cell activation, proliferation, and cytokine release. An in vitro study demonstrated that IBI379 induced the lysis of MM cells expressing differing levels of BCMA on the cell surface. Administration of IBI379 in H929 or Daudi-BCMA cell xenograft mouse models significantly inhibited tumor growth without inducing body weight loss. The mechanism of action study revealed the accumulation of CD4+CD8+ T cells and granzyme B-positive T cells in tumors that were treated with IBI379. Moreover, administration of low dose of IBI379 in cynomolgus monkeys was well-tolerated and induced the depletion of BCMA+ B cells and a mild transient increase of cytokines. Collectively, these results demonstrate that IBI379 is a highly potent therapeutic strategy for depleting BCMA-positive B cells and is a promising approach for the treatment of MM.
Collapse
|
7
|
Ahmed Z, Afridi SS, Shahid Z, Zamani Z, Rehman S, Aiman W, Khan M, Mir MA, Awan FT, Anwer F, Iftikhar R. Primary Mediastinal B-Cell Lymphoma: A 2021 Update on Genetics, Diagnosis, and Novel Therapeutics. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2021; 21:e865-e875. [PMID: 34330673 DOI: 10.1016/j.clml.2021.06.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/08/2021] [Accepted: 06/19/2021] [Indexed: 12/21/2022]
Abstract
Primary mediastinal large B-cell lymphoma (PMBCL) is an aggressive B-cell lymphoma arising from thymic B-cells having clinicopathologic features distinct from systemic diffuse large B-cell lymphoma (DLBCL). PMBCL comprises 2% to 4% of all non-Hodgkin lymphomas (NHL), 7% of DLBCL and seen predominantly in young females with a median age of 35 years at diagnosis. The annual incidence of PMBCL is 0.4 per million with a 5-year survival rate exceeding 70% with improving supportive care and genetic characterization of the disease. Pathogenesis involves dysregulation of Janus kinase-signal transducer and activator of transcription (JAK-STAT), nuclear factor-kB (NF-kB) pathways and amplification of the 9p24.1 region of chromosome 9. PMBCL patients have a prolonged life expectancy necessitating the need for treatment approaches that are based on maximizing cure with minimal long-term toxicity. Due to rarity and its recognition as a distinct entity, therapeutic decisions are guided by clinical presentation, clinician and center experience, and analysis of patients with PMBCL within DLBCL registries. Historically R-CHOP has been the usual first line treatment for PMBCL followed by involved site radiotherapy (ISRT), however clinical practice varies across centers with emerging consensus to avoid upfront RT by utilizing dose intense regimens (DA-EPOCH-R) in younger and fit patients. Prognosis of relapsed refractory PMBCL not responding to salvage chemotherapy is dismal, however there are many emerging options including Brentuximab Vedotin, immune check point inhibitors and chimeric antigen receptor T-cell therapy. In this article, we focus on the pathogenesis, current and evolving treatments, and provide recommendations for optimal management of patients with PMBCL.
Collapse
Affiliation(s)
- Zahoor Ahmed
- Department of Internal Medicine, King Edward Medical University, Lahore, Pakistan
| | - Safa Saadat Afridi
- Department of Internal Medicine, Khyber Medical College Peshawar, Peshawar, Pakistan
| | | | - Zarlakhta Zamani
- Department of Internal Medicine, King Edward Medical University, Lahore, Pakistan
| | - Sana Rehman
- Shaikh Khalifa Bin Zayyed al Nahyan Medical and Dental College, Lahore, Pakistan
| | - Wajeeha Aiman
- Department of Internal Medicine, Nishtar Medical College, Multan, Pakistan
| | - Maryam Khan
- Armed Forces Bone Marrow Transplant Centre, Rawalpindi, Pakistan
| | | | - Farrukh T Awan
- Department of Medicine, Division of Hematology and Oncology, UT Southwestern Medical Center, Dallas, TX
| | - Faiz Anwer
- Hematology, Oncology, Stem Cell Transplantation, Multiple Myeloma Program, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH
| | - Raheel Iftikhar
- Armed Forces Bone Marrow Transplant Centre, Rawalpindi, Pakistan.
| |
Collapse
|
8
|
Chen H, Pan T, He Y, Zeng R, Li Y, Yi L, Zang H, Chen S, Duan Q, Xiao L, Zhou H. Primary Mediastinal B-Cell Lymphoma: Novel Precision Therapies and Future Directions. Front Oncol 2021; 11:654854. [PMID: 33869061 PMCID: PMC8044947 DOI: 10.3389/fonc.2021.654854] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 03/01/2021] [Indexed: 11/13/2022] Open
Abstract
Primary mediastinal large B-cell lymphoma (PMBCL) is a distinct clinicopathologic disease from other types of diffuse large B-cell lymphoma (DLBCL) with unique prognostic features and limited availability of clinical data. The current standard treatment for newly diagnosed PMBCL has long been dependent on a dose-intensive, dose-adjusted multi-agent chemotherapy regimen of rituximab plus etoposide, prednisone, vincristine, cyclophosphamide, and doxorubicin (DA-R-EPOCH). Recent randomized trials have provided evidence that R-CHOP followed by consolidation radiotherapy (RT) is a valuable alternative option to first-line treatment. For recurrent/refractory PMBCL (rrPMBCL), new drugs such as pembrolizumab and CAR-T cell therapy have proven to be effective in a few studies. Positron emission tomography-computed tomography (PET-CT) is the preferred imaging modality of choice for the initial phase of lymphoma treatment and to assess response to treatment. In the future, baseline quantitative PET-CT can be used to predict prognosis in PMBCL. This review focuses on the pathology of PMBCL, underlying molecular basis, treatment options, radiotherapy, targeted therapies, and the potential role of PET-CT to guide treatment choices in this disease.
Collapse
Affiliation(s)
- Huan Chen
- Department of Lymphoma and Hematology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Tao Pan
- Department of Lymphoma and Hematology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yizi He
- Department of Lymphoma and Hematology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Ruolan Zeng
- Department of Lymphoma and Hematology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yajun Li
- Department of Lymphoma and Hematology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Liming Yi
- Department of Human Anatomy, Hunan University of Medicine, Huaihua, China
| | - Hui Zang
- Department of Basic Medicine, Yiyang Medical College, Yiyang, China
| | - Siwei Chen
- Department of Histology and Embryology of School of Basic Medical Science, Central South University, Changsha, China
| | - Qintong Duan
- Department of Histology and Embryology of School of Basic Medical Science, Central South University, Changsha, China
| | - Ling Xiao
- Department of Histology and Embryology of School of Basic Medical Science, Central South University, Changsha, China
| | - Hui Zhou
- Department of Lymphoma and Hematology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
9
|
Do P, Perdue LA, Chyong A, Hunter R, Dougan J, Henry CJ, Porter CC, Dreaden EC. Rapid Assembly and Screening of Multivalent Immune Cell-Redirecting Therapies for Leukemia. ACS COMBINATORIAL SCIENCE 2020; 22:533-541. [PMID: 32786324 PMCID: PMC8496977 DOI: 10.1021/acscombsci.0c00081] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Therapies that bind with immune cells and redirect their cytotoxic activity toward diseased cells represent a promising and versatile approach to immunotherapy with applications in cancer, lupus, and other diseases; traditional methods for discovering these therapies, however, are often time-intensive and lack the throughput of related target-based discovery approaches. Inspired by the observation that the cytokine, IL-12, can enhance antileukemic activity of the clinically approved T cell redirecting therapy, blinatumomab, here we describe the structure and assembly of a chimeric immune cell-redirecting agent which redirects the lytic activity of primary human T cells toward leukemic B cells and simultaneously cotargets the delivery of T cell-stimulating IL-12. We further describe a novel method for the parallel assembly of compositionally diverse libraries of these bispecific T cell engaging cytokines (BiTEokines) and their high-throughput phenotypic screening, requiring just days for hit identification and the analysis of composition-function relationships. Using this approach, we identified CD19 × CD3 × IL12 compounds that exhibit ex vivo lytic activity comparable to current FDA-approved therapies for leukemia and correlated drug treatment with specific cell–cell contact, cytokine delivery, and leukemia cell lysis. Given the modular nature of these multivalent compounds and their rapid assembly/screening, we anticipate facile extension of this therapeutic approach to a wide range of immune cells, diseased cells, and soluble protein combinations in the future.
Collapse
Affiliation(s)
- Priscilla Do
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30322, United States
| | - Lacey A Perdue
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30322, United States
| | - Andrew Chyong
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30322, United States
| | - Rae Hunter
- Department of Pediatrics, Emory School of Medicine, Atlanta, Georgia 30322, United States
- Winship Cancer Institute of Emory University, Atlanta, Georgia 30322, United States
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Emory School of Medicine, Atlanta, Georgia 30322, United States
| | - Jodi Dougan
- Department of Pediatrics, Emory School of Medicine, Atlanta, Georgia 30322, United States
- Winship Cancer Institute of Emory University, Atlanta, Georgia 30322, United States
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Emory School of Medicine, Atlanta, Georgia 30322, United States
| | - Curtis J Henry
- Department of Pediatrics, Emory School of Medicine, Atlanta, Georgia 30322, United States
- Winship Cancer Institute of Emory University, Atlanta, Georgia 30322, United States
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Emory School of Medicine, Atlanta, Georgia 30322, United States
| | - Christopher C Porter
- Department of Pediatrics, Emory School of Medicine, Atlanta, Georgia 30322, United States
- Winship Cancer Institute of Emory University, Atlanta, Georgia 30322, United States
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Emory School of Medicine, Atlanta, Georgia 30322, United States
| | - Erik C Dreaden
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30322, United States
- Department of Pediatrics, Emory School of Medicine, Atlanta, Georgia 30322, United States
- Winship Cancer Institute of Emory University, Atlanta, Georgia 30322, United States
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Emory School of Medicine, Atlanta, Georgia 30322, United States
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
10
|
Chitadze G, Laqua A, Lettau M, Baldus CD, Brüggemann M. Bispecific antibodies in acute lymphoblastic leukemia therapy. Expert Rev Hematol 2020; 13:1211-1233. [PMID: 33000968 DOI: 10.1080/17474086.2020.1831380] [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: 12/26/2022]
Abstract
INTRODUCTION Blinatumomab, first in a class of bispecific T-cell engagers, revolutionized treatment paradigm of B-cell precursor relapsed/refractory or minimal residual disease positive acute lymphoblastic leukemia (ALL) in adults and children, inducing deep remissions in a proportion of patients. However, significant numbers of patients do not respond or eventually relapse. Strategies for improvement of treatment outcomes are required. AREAS COVERED This review discusses the main structural and functional features of blinatumomab, and its place in the treatment of ALL. Furthermore, prospects to increase the efficacy of blinatumomab are addressed. The developments in the field of bispecific antibodies and their possible implications for treatment of ALL are reviewed. EXPERT OPINION Better understanding the mechanisms of response and resistance to blinatumomab might help us to identify the group of patients benefiting most from treatment and to spare potentially toxic subsequent treatment strategies. Data emerging from ongoing clinical trials might change the treatment landscape of ALL and beyond. Early use of blinatumomab in frontline protocols with more advantageous treatment sequences and in combination with other targeted therapies might reduce the failure rates. Exponentially increasing number of novel treatment options and their possible combinations might complicate treatment decision-making without data from randomized trials.
Collapse
Affiliation(s)
- Guranda Chitadze
- Department of Hematology, University Hospital Schleswig-Holstein , Campus Kiel, Kiel, Germany
| | - Anna Laqua
- Department of Hematology, University Hospital Schleswig-Holstein , Campus Kiel, Kiel, Germany
| | - Marcus Lettau
- Department of Hematology, University Hospital Schleswig-Holstein , Campus Kiel, Kiel, Germany.,Institute of Immunology, University Hospital Schleswig-Holstein , Campus Kiel, Kiel, Germany
| | - Claudia D Baldus
- Department of Hematology, University Hospital Schleswig-Holstein , Campus Kiel, Kiel, Germany
| | - Monika Brüggemann
- Department of Hematology, University Hospital Schleswig-Holstein , Campus Kiel, Kiel, Germany
| |
Collapse
|
11
|
Yoon JH, Min GJ, Park SS, Park S, Lee SE, Cho BS, Eom KS, Kim YJ, Kim HJ, Min CK, Cho SG, Kim DW, Lee JW, Lee S. Feasible outcome of blinatumomab followed by allogeneic hematopoietic cell transplantation for adults with Philadelphia chromosome-negative acute lymphoblastic leukemia in first salvage. Cancer Med 2019; 8:7650-7659. [PMID: 31691536 PMCID: PMC6912052 DOI: 10.1002/cam4.2680] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/24/2019] [Accepted: 10/21/2019] [Indexed: 01/15/2023] Open
Abstract
In adult patients with relapsed or refractory (R/R) Philadelphia chromosome‐negative (Ph‐negative) B‐cell presursor acute lymphoblastic leukemia (BCP‐ALL), complete remission (CR) and overall survival (OS) rates are poor. We analyzed treatment outcomes and prognostic factors for 32 adult patients with R/R Ph‐negative BCP‐ALL who received blinatumomab at first salvage. Patients who achieved CR proceeded to allogeneic hematopoietic cell transplantation (allo‐HCT). At the time of blinatumomab treatment, 11 patients (34.3%) were primary refractory, 10 (31.4%) had relapsed with first CR duration (CRD1) ≥12 months, and 11 (34.3%) had relapsed with CRD1 <12 months. After the first blinatumomab cycle, 22 (68.8%) achieved CR. At the end of the second cycle, 20 of the 22 patients remained in persistent CR, and 1 patient achieved new CR. The overall minimal residual disease negativity rate was 75% among evaluable patients with persistent CR. Patients with CRD1 <12 months were associated with poorer response to blinatumomab. Twenty (62.5%) of 32 patients underwent allo‐HCT in blinatumomab‐induced CR. After a median follow‐up of 15.2 months, the 1‐year OS rates for all patients and patients receiving allo‐HCT in CR were 55.5% (median OS, 18.2 months) and 70.7%, respectively. Patients with CRD1 <12 months, extramedullary disease (EMD), and high peripheral blood blasts were associated with poorer OS. Blinatumomab is effective for achieving good quality CR and bridging to allo‐HCT for adult patients with R/R Ph‐negative BCP‐ALL in first salvage. The role of blinatumomab in patients with CRD1 <12 months, EMD, or high tumor burden should be evaluated in future trials.
Collapse
Affiliation(s)
- Jae-Ho Yoon
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Gi June Min
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sung-Soo Park
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Silvia Park
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sung-Eun Lee
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Byung-Sik Cho
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ki-Seong Eom
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yoo-Jin Kim
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hee-Je Kim
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Chang-Ki Min
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seok-Goo Cho
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Dong-Wook Kim
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jong Wook Lee
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seok Lee
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
12
|
Panowski SH, Kuo TC, Zhang Y, Chen A, Geng T, Aschenbrenner L, Kamperschroer C, Pascua E, Chen W, Delaria K, Farias S, Bateman M, Dushin RG, Chin SM, Van Blarcom TJ, Yeung YA, Lindquist KC, Chunyk AG, Kuang B, Han B, Mirsky M, Pardo I, Buetow B, Martin TG, Wolf JL, Shelton D, Rajpal A, Strop P, Chaparro-Riggers J, Sasu BJ. Preclinical Efficacy and Safety Comparison of CD3 Bispecific and ADC Modalities Targeting BCMA for the Treatment of Multiple Myeloma. Mol Cancer Ther 2019; 18:2008-2020. [DOI: 10.1158/1535-7163.mct-19-0007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 06/03/2019] [Accepted: 08/12/2019] [Indexed: 11/16/2022]
|
13
|
Ma Y, Zhang Q, Kong P, Xiong J, Zhang X, Zhang C. Treatment Selection for Philadelphia Chromosome-Positive Acute Lymphoblastic Leukemia in the Era of Tyrosine Kinase Inhibitors. Chemotherapy 2019; 64:81-93. [PMID: 31390613 DOI: 10.1159/000501061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 05/18/2019] [Indexed: 12/20/2022]
Abstract
With the advent of tyrosine kinase inhibitors (TKIs), the treatment of Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL) has entered a new era. The efficacy of TKIs compared with other ALL treatment options is emphasized by a rapid increase in the number of TKI clinical trials. Subsequently, the use of traditional approaches, such as combined chemotherapy and even allogeneic hematopoietic stem cell transplantation (allo-HSCT), for the treatment of ALL is being challenged in the clinic. In light of the increased use of TKIs in the clinic, several questions have been raised. First, is it necessary to use intensive chemotherapy during the induction course of therapy to achieve a minimal residual disease (MRD)-negative status? Must a patient reach a complete molecular response/major molecular response before receiving allo-HSCT? Does MRD status affect long-term survival after allo-HSCT? Is auto-HSCT an appropriate alternative for allo-HSCT in those Ph+ ALL patients who lack suitable donors? Here, we review the recent literature in an attempt to summarize the current status of TKI usage in the clinic, including several new therapeutic approaches, provide answers for the above questions, and speculate on the future direction of TKI utilization for the treatment of Ph+ ALL patients.
Collapse
Affiliation(s)
- Yingying Ma
- Hematology Department, State Key Laboratory of Trauma, Burns and Combined Injury, Xinqiao Hospital of Army Medical University, Chongqing, China
| | - Quanchao Zhang
- Shigatse Branch, Xinqiao Hospital, Army Medical University (Third Military Medical University), Shigatse, China
| | - Peiyan Kong
- Hematology Department, State Key Laboratory of Trauma, Burns and Combined Injury, Xinqiao Hospital of Army Medical University, Chongqing, China
| | - Jingkang Xiong
- Hematology Department, State Key Laboratory of Trauma, Burns and Combined Injury, Xinqiao Hospital of Army Medical University, Chongqing, China
| | - Xi Zhang
- Hematology Department, State Key Laboratory of Trauma, Burns and Combined Injury, Xinqiao Hospital of Army Medical University, Chongqing, China
| | - Cheng Zhang
- Hematology Department, State Key Laboratory of Trauma, Burns and Combined Injury, Xinqiao Hospital of Army Medical University, Chongqing, China,
| |
Collapse
|
14
|
Acheampong DO. Bispecific Antibody (bsAb) Construct Formats and their Application in Cancer Therapy. Protein Pept Lett 2019; 26:479-493. [DOI: 10.2174/0929866526666190311163820] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 02/28/2019] [Accepted: 03/03/2019] [Indexed: 12/15/2022]
Abstract
Development of cancers mostly involves more than one signal pathways, because of the complicated nature of cancer cells. As such, the most effective treatment option is the one that stops the cancer cells in their tracks by targeting these signal pathways simultaneously. This explains why therapeutic monoclonal antibodies targeted at cancers exert utmost activity when two or more are used as combination therapy. This notwithstanding, studies elsewhere have proven that when bispecific antibody (bsAb) is engineered from two conventional monoclonal antibodies or their chains, it produces better activity than when used as combination therapy. This therefore presents bispecific antibody (bsAb) as the appropriate and best therapeutic agent for the treatment of such cancers. This review therefore discusses the various engineering formats for bispecific antibodies (bsAbs) and their applications.
Collapse
Affiliation(s)
- Desmond O. Acheampong
- Department of Biomedical Sciences, School of Allied Health Sciences, College of Health and Allied Science, University of Cape Coast, Cape Coast, Ghana
| |
Collapse
|
15
|
Betts A, Haddish-Berhane N, Shah DK, van der Graaf PH, Barletta F, King L, Clark T, Kamperschroer C, Root A, Hooper A, Chen X. A Translational Quantitative Systems Pharmacology Model for CD3 Bispecific Molecules: Application to Quantify T Cell-Mediated Tumor Cell Killing by P-Cadherin LP DART ®. AAPS J 2019; 21:66. [PMID: 31119428 PMCID: PMC6531394 DOI: 10.1208/s12248-019-0332-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/08/2019] [Indexed: 01/12/2023] Open
Abstract
CD3 bispecific antibody constructs recruit cytolytic T cells to kill tumor cells, offering a potent approach to treat cancer. T cell activation is driven by the formation of a trimolecular complex (trimer) between drugs, T cells, and tumor cells, mimicking an immune synapse. A translational quantitative systems pharmacology (QSP) model is proposed for CD3 bispecific molecules capable of predicting trimer concentration and linking it to tumor cell killing. The model was used to quantify the pharmacokinetic (PK)/pharmacodynamic (PD) relationship of a CD3 bispecific targeting P-cadherin (PF-06671008). It describes the disposition of PF-06671008 in the central compartment and tumor in mouse xenograft models, including binding to target and T cells in the tumor to form the trimer. The model incorporates T cell distribution to the tumor, proliferation, and contraction. PK/PD parameters were estimated for PF-06671008 and a tumor stasis concentration (TSC) was calculated as an estimate of minimum efficacious trimer concentration. TSC values ranged from 0.0092 to 0.064 pM across mouse tumor models. The model was translated to the clinic and used to predict the disposition of PF-06671008 in patients, including the impact of binding to soluble P-cadherin. The predicted terminal half-life of PF-06671008 in the clinic was approximately 1 day, and P-cadherin expression and number of T cells in the tumor were shown to be sensitive parameters impacting clinical efficacy. A translational QSP model is presented for CD3 bispecific molecules, which integrates in silico, in vitro and in vivo data in a mechanistic framework, to quantify and predict efficacy across species.
Collapse
Affiliation(s)
- Alison Betts
- Department of Biomedicine Design, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts, 02139, USA.
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, 2300 RA, Leiden, The Netherlands.
| | | | - Dhaval K Shah
- Department of Pharmaceutical Sciences, 455 Kapoor Hall, University at Buffalo, The State University of New York, Buffalo, New York, 14214-8033, USA
| | - Piet H van der Graaf
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, 2300 RA, Leiden, The Netherlands
| | - Frank Barletta
- Oncology Research Unit, Pfizer Inc., 401 N Middletown Rd., Pearl River, New York, 10965, USA
| | - Lindsay King
- Department of Biomedicine Design, Pfizer Inc., 1 Burtt Road, Andover, Massachusetts, USA
| | - Tracey Clark
- Established Med Business, Pfizer Inc., Eastern Point Rd, Groton, Connecticut, 06340, USA
| | - Cris Kamperschroer
- Department of Immunotoxicology, Pfizer Inc., 558 Eastern Point Road, Groton, Connecticut, 06340, USA
| | - Adam Root
- Department of Biomedicine Design, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts, 02139, USA
| | - Andrea Hooper
- Oncology Research Unit, Pfizer Inc., 401 N Middletown Rd., Pearl River, New York, 10965, USA
| | - Xiaoying Chen
- Department of Clinical Pharmacology, Pfizer Inc., 10555 Science Center Dr., San Diego, California, 92121, USA
| |
Collapse
|
16
|
Abou Dalle I, Jabbour E, Short NJ, Ravandi F. Treatment of Philadelphia Chromosome-Positive Acute Lymphoblastic Leukemia. Curr Treat Options Oncol 2019; 20:4. [PMID: 30675645 DOI: 10.1007/s11864-019-0603-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OPINION STATEMENT With the introduction of tyrosine kinase inhibitors (TKIs) in the management of Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL), the prognosis of patients has improved dramatically. Currently, the standard of care in the frontline setting for fit patients is TKI in combination with chemotherapy. Age-adjusted chemotherapy or corticosteroids alone have been used with TKIs in elderly patients with comorbidities with modest long-term benefit. The primary goal of treatment is the achievement of early deep molecular remission as the achievement of complete molecular remission (CMR) at 3 months has been demonstrated to be predictive of higher long-term survival. The probability of attaining this goal by a more potent TKIs like dasatinib or ponatinib is higher, thus we recommend the use of second- or third-generation TKIs over imatinib. Clinicians should be aware of possible fatal cardiovascular events mainly related to ponatinib. Allogeneic hematopoietic stem cell transplantation (alloHSCT) should still be considered in first remission, especially for younger patients treated with imatinib combination therapy. A subset of patients achieving CMR at 3 months may be able to continue consolidation and maintenance with chemotherapy and TKI without the need for alloHSCT. Because of higher risk of relapses in the central nervous system, intrathecal chemoprophylaxis is mandatory for all patients. New strategies incorporating novel agents, such as antibody-drug conjugates, bispecific monoclonal antibodies, potent TKIs, and CAR T cells are under investigation.
Collapse
Affiliation(s)
- Iman Abou Dalle
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd. Unit 0428, Houston, TX, 77030, USA
| | - Elias Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd. Unit 0428, Houston, TX, 77030, USA
| | - Nicholas J Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd. Unit 0428, Houston, TX, 77030, USA
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd. Unit 0428, Houston, TX, 77030, USA.
| |
Collapse
|
17
|
Fisher TS, Hooper AT, Lucas J, Clark TH, Rohner AK, Peano B, Elliott MW, Tsaparikos K, Wang H, Golas J, Gavriil M, Haddish-Berhane N, Tchistiakova L, Gerber HP, Root AR, May C. A CD3-bispecific molecule targeting P-cadherin demonstrates T cell-mediated regression of established solid tumors in mice. Cancer Immunol Immunother 2018; 67:247-259. [PMID: 29067496 PMCID: PMC11028296 DOI: 10.1007/s00262-017-2081-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 10/14/2017] [Indexed: 12/11/2022]
Abstract
Strong evidence exists supporting the important role T cells play in the immune response against tumors. Still, the ability to initiate tumor-specific immune responses remains a challenge. Recent clinical trials suggest that bispecific antibody-mediated retargeted T cells are a promising therapeutic approach to eliminate hematopoietic tumors. However, this approach has not been validated in solid tumors. PF-06671008 is a dual-affinity retargeting (DART®)-bispecific protein engineered with enhanced pharmacokinetic properties to extend in vivo half-life, and designed to engage and activate endogenous polyclonal T cell populations via the CD3 complex in the presence of solid tumors expressing P-cadherin. This bispecific molecule elicited potent P-cadherin expression-dependent cytotoxic T cell activity across a range of tumor indications in vitro, and in vivo in tumor-bearing mice. Regression of established tumors in vivo was observed in both cell line and patient-derived xenograft models engrafted with circulating human T lymphocytes. Measurement of in vivo pharmacodynamic markers demonstrates PF-06671008-mediated T cell activation, infiltration and killing as the mechanism of tumor inhibition.
Collapse
Affiliation(s)
- Timothy S Fisher
- Oncology Research and Development Pfizer Inc., La Jolla, CA, USA.
- Oncology Research and Development Pfizer Inc., Pearl River, NY, USA.
- Pfizer Inc., 10777 Science Center Drive, San Diego, CA, 92121, USA.
| | - Andrea T Hooper
- Oncology Research and Development Pfizer Inc., La Jolla, CA, USA
- Oncology Research and Development Pfizer Inc., Pearl River, NY, USA
| | - Justin Lucas
- Oncology Research and Development Pfizer Inc., La Jolla, CA, USA
- Oncology Research and Development Pfizer Inc., Pearl River, NY, USA
| | | | - Allison K Rohner
- Oncology Research and Development Pfizer Inc., La Jolla, CA, USA
- Oncology Research and Development Pfizer Inc., Pearl River, NY, USA
| | - Bryan Peano
- Oncology Research and Development Pfizer Inc., La Jolla, CA, USA
- Oncology Research and Development Pfizer Inc., Pearl River, NY, USA
| | - Mark W Elliott
- Oncology Research and Development Pfizer Inc., La Jolla, CA, USA
- Oncology Research and Development Pfizer Inc., Pearl River, NY, USA
| | - Konstantinos Tsaparikos
- Oncology Research and Development Pfizer Inc., La Jolla, CA, USA
- Oncology Research and Development Pfizer Inc., Pearl River, NY, USA
| | - Hui Wang
- Oncology Research and Development Pfizer Inc., La Jolla, CA, USA
- Oncology Research and Development Pfizer Inc., Pearl River, NY, USA
| | - Jonathan Golas
- Oncology Research and Development Pfizer Inc., La Jolla, CA, USA
- Oncology Research and Development Pfizer Inc., Pearl River, NY, USA
| | - Maria Gavriil
- Oncology Research and Development Pfizer Inc., La Jolla, CA, USA
- Oncology Research and Development Pfizer Inc., Pearl River, NY, USA
| | - Nahor Haddish-Berhane
- BioMedicine Design Pfizer Inc., Cambridge, MA, USA
- Johnson and Johnson Pharmaceutical Research and Development, Spring House, PA, USA
| | | | - Hans-Peter Gerber
- Oncology Research and Development Pfizer Inc., La Jolla, CA, USA
- Oncology Research and Development Pfizer Inc., Pearl River, NY, USA
- Maverick Therapeutics, Brisbane, CA, USA
| | - Adam R Root
- BioMedicine Design Pfizer Inc., Cambridge, MA, USA
| | - Chad May
- Oncology Research and Development Pfizer Inc., La Jolla, CA, USA
- Oncology Research and Development Pfizer Inc., Pearl River, NY, USA
- Maverick Therapeutics, Brisbane, CA, USA
| |
Collapse
|
18
|
Kuhlmann L, Cummins E, Samudio I, Kislinger T. Cell-surface proteomics for the identification of novel therapeutic targets in cancer. Expert Rev Proteomics 2018; 15:259-275. [DOI: 10.1080/14789450.2018.1429924] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Laura Kuhlmann
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Emma Cummins
- The Centre for Drug Research and Development, Division of Biologics, Vancouver, Canada
| | - Ismael Samudio
- The Centre for Drug Research and Development, Division of Biologics, Vancouver, Canada
| | - Thomas Kislinger
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| |
Collapse
|
19
|
A versatile pretargeting approach for tumour-selective delivery and activation of TNF superfamily members. Sci Rep 2017; 7:13301. [PMID: 29038485 PMCID: PMC5643434 DOI: 10.1038/s41598-017-13530-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/25/2017] [Indexed: 11/08/2022] Open
Abstract
TNFR superfamily (TNFRSF) members have important immunoregulatory functions and are of clear interest for cancer immunotherapy. Various TNFRSF agonists have been clinically evaluated, but have met with limited efficacy and/or toxicity. Recent insights indicate that 'first-generation' TNFRSF agonists lack efficacy as they do not effectively cross-link their corresponding receptor. Reversely, ubiquitous TNFRSF receptor(s) cross-linking by CD40 and Fas agonistic antibodies resulted in dose-limiting liver toxicity. To overcome these issues, we developed a novel pretargeting strategy exploiting recombinant fusion proteins in which a soluble form of TRAIL, FasL or CD40L is genetically fused to a high-affinity anti-fluorescein scFv antibody fragment (scFvFITC). Fusion proteins scFvFITC:sTRAIL and scFvFITC:sFasL induced potent target antigen-restricted apoptosis in a panel of cancer lines and in primary patient-derived cancer cells, but only when pretargeted with a relevant FITC-labelled antitumour antibody. In a similar pretargeting setting, fusion protein scFvFITC:sCD40L promoted tumour-directed maturation of immature monocyte-derived dendritic cells (iDCs). This novel tumour-selective pretargeting approach may be used to improve efficacy and/or reduce possible off-target toxicity of TNFSF ligands for cancer immunotherapy.
Collapse
|
20
|
Zhu M, Wu B, Brandl C, Johnson J, Wolf A, Chow A, Doshi S. Blinatumomab, a Bispecific T-cell Engager (BiTE(®)) for CD-19 Targeted Cancer Immunotherapy: Clinical Pharmacology and Its Implications. Clin Pharmacokinet 2017; 55:1271-1288. [PMID: 27209293 DOI: 10.1007/s40262-016-0405-4] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND AND OBJECTIVES Blinatumomab is a bispecific T-cell engager (BiTE(®)) antibody construct that transiently links CD19-positive B cells to CD3-positive T cells, resulting in induction of T-cell-mediated serial lysis of B cells and concomitant T-cell proliferation. Blinatumomab showed anti-leukemia activity in clinical trials and was approved by the US Food and Drug Administration for the treatment of Philadelphia chromosome-negative relapsed/refractory B-cell precursor acute lymphoblastic leukemia (r/r ALL). The objectives of this work were to characterize blinatumomab pharmacokinetics and pharmacodynamics and to evaluate dosing regimens. METHODS Data from six phase I and II trials in patients with r/r ALL, minimal residual disease-positive ALL, and non-Hodgkin's lymphoma (NHL) were analyzed. Blinatumomab pharmacokinetics was characterized by non-compartmental and population pharmacokinetic analyses and pharmacodynamics was described graphically. RESULTS Blinatumomab exhibited linear pharmacokinetics under continuous intravenous infusion for 4-8 weeks per cycle over a dose range of 5-90 µg/m(2)/day, without target-mediated disposition. Estimated mean (standard deviation) volume of distribution, clearance, and elimination half-life were 4.52 (2.89) L, 2.72 (2.71) L/h, and 2.11 (1.42) h, respectively. Pharmacokinetics was similar in patients with ALL and NHL and was not affected by patient demographics, supporting fixed dosing in adults. Although creatinine clearance was a significant covariate of drug clearance, no dose adjustment was required in patients with mild or moderate renal impairment. Incidence of neutralizing antidrug antibodies was <1 %. Blinatumomab pharmacodynamics featured T-cell redistribution and activation, B-cell depletion, and transient dose-dependent cytokine elevation. Blinatumomab did not affect cytochrome P450 enzymes directly; cytokines may trigger transient cytochrome P450 suppression with low potential for inducing drug interactions. CONCLUSIONS Blinatumomab has unique pharmacokinetic and immunological features that require indication-dependent dosing regimens. Stepped dosing is required to achieve adequate efficacy and minimize cytokine release in diseases with high tumor burden.
Collapse
Affiliation(s)
- Min Zhu
- Amgen Inc., Thousand Oaks, CA, 91320, USA.
| | | | | | | | | | | | | |
Collapse
|
21
|
|
22
|
Liu L, Lam CYK, Long V, Widjaja L, Yang Y, Li H, Jin L, Burke S, Gorlatov S, Brown J, Alderson R, Lewis MD, Nordstrom JL, Koenig S, Moore PA, Johnson S, Bonvini E. MGD011, A CD19 x CD3 Dual-Affinity Retargeting Bi-specific Molecule Incorporating Extended Circulating Half-life for the Treatment of B-Cell Malignancies. Clin Cancer Res 2016; 23:1506-1518. [PMID: 27663593 DOI: 10.1158/1078-0432.ccr-16-0666] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 08/08/2016] [Accepted: 09/01/2016] [Indexed: 11/16/2022]
Abstract
Purpose: CD19, a B-cell lineage-specific marker, is highly represented in B-cell malignancies and an attractive target for therapeutic interventions. MGD011 is a CD19 x CD3 DART bispecific protein designed to redirect T lymphocytes to eliminate CD19-expressing cells. MGD011 has been engineered with a modified human Fc domain for improved pharmacokinetic (PK) properties and designed to cross-react with the corresponding antigens in cynomolgus monkeys. Here, we report on the preclinical activity, safety and PK properties of MGD011.Experimental Design: The activity of MGD011 was evaluated in several in vitro and in vivo models. PK, safety and pharmacodynamic activity was also assessed in dose-escalation and repeat-dose studies of MGD011 administered once weekly in cynomolgus monkeys.Results: MGD011 mediated killing of human B-cell lymphoma lines by human or cynomolgus monkey PBMCs as well as autologous B-cell depletion in PBMCs from both species. MGD011-mediated killing was accompanied by target-dependent T-cell activation and expansion, cytokine release and upregulation of perforin and granzyme B. MGD011 demonstrated antitumor activity against localized and disseminated lymphoma xenografts reconstituted with human PBMCs. In cynomolgus monkeys, MGD011 displayed a terminal half-life of 6.7 days; once weekly intravenous infusion of MGD011 at doses up to 100 μg/kg, the highest dose tested, was well tolerated and resulted in dose-dependent, durable decreases in circulating B cells accompanied by profound reductions of B lymphocytes in lymphoid organs.Conclusions: The preclinical activity, safety and PK profile support clinical investigation of MGD011 as a therapeutic candidate for the treatment of B-cell malignancies. Clin Cancer Res; 23(6); 1506-18. ©2016 AACR.
Collapse
Affiliation(s)
- Liqin Liu
- Research, MacroGenics, Inc., Rockville, Maryland
| | | | - Vatana Long
- Research, MacroGenics, Inc., Rockville, Maryland
| | | | - Yinhua Yang
- Research, MacroGenics, Inc., Rockville, Maryland
| | - Hua Li
- Research, MacroGenics, Inc., Rockville, Maryland
| | - Linda Jin
- Research, MacroGenics, Inc., Rockville, Maryland
| | - Steve Burke
- Research, MacroGenics, Inc., Rockville, Maryland
| | | | | | | | | | | | - Scott Koenig
- Research, MacroGenics, Inc., Rockville, Maryland
| | - Paul A Moore
- Research, MacroGenics, Inc., Rockville, Maryland
| | - Syd Johnson
- Research, MacroGenics, Inc., Rockville, Maryland
| | - Ezio Bonvini
- Research, MacroGenics, Inc., Rockville, Maryland.
| |
Collapse
|
23
|
Baumann A, Fischmann S, Blaich G, Friedrich M. Leverage nonclinical development of bispecifics by translational science. DRUG DISCOVERY TODAY. TECHNOLOGIES 2016; 21-22:95-102. [PMID: 27978993 DOI: 10.1016/j.ddtec.2016.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 08/09/2016] [Accepted: 08/11/2016] [Indexed: 06/06/2023]
Abstract
Bispecific antibody constructs (Bispecifics, bsAbs) may have greater functionality compared to established monoclonal antibodies because they bind to 2 different targets or, potentially, to 2 epitopes on the same target (dual targeting). This may result in enhanced binding avidity with preferential binding to cells that express both targets or binding to targets on different cells. However, development of these next-generation biologics, including new formats, creates unique challenges due to their increased complexity. Here we review aspects of bsAbs preclinical development programs that may increase the success rates of bsAbs in clinical development.
Collapse
Affiliation(s)
| | | | - Guenter Blaich
- AbbVie Germany GmbH & Co. KG, Knollstrasse, 67061 Ludwigshafen, Germany
| | | |
Collapse
|
24
|
Chen X, Haddish-Berhane N, Moore P, Clark T, Yang Y, Li H, Xuan D, Barton HA, Betts AM, Barletta F. Mechanistic Projection of First-in-Human Dose for Bispecific Immunomodulatory P-Cadherin LP-DART: An Integrated PK/PD Modeling Approach. Clin Pharmacol Ther 2016; 100:232-41. [DOI: 10.1002/cpt.393] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 05/05/2016] [Accepted: 05/06/2016] [Indexed: 12/19/2022]
Affiliation(s)
- X Chen
- Pharmacokinetics, Dynamics and Metabolism; Pfizer; Cambridge Massachusetts USA
| | - N Haddish-Berhane
- Pharmacokinetics, Dynamics and Metabolism; Pfizer; Groton Connecticut USA
- Current Address: Clinical Pharmacology and Pharmacometrics; Quantitative Sciences Janssen Pharmaceuticals, Spring House, Pennsylvania USA
| | - P Moore
- MacroGenics; Rockville; Maryland USA
| | - T Clark
- Pharmacokinetics, Dynamics and Metabolism; Pfizer; Groton Connecticut USA
| | - Y Yang
- MacroGenics; Rockville; Maryland USA
| | - H Li
- MacroGenics; Rockville; Maryland USA
| | - D Xuan
- Clinical Pharmacology; Pfizer; San Diego California USA
| | - HA Barton
- Pharmacokinetics, Dynamics and Metabolism; Pfizer; Groton Connecticut USA
| | - AM Betts
- Pharmacokinetics, Dynamics and Metabolism; Pfizer; Groton Connecticut USA
| | - F Barletta
- Pharmacokinetics, Dynamics and Metabolism; Pfizer; Pearl River New York USA
| |
Collapse
|
25
|
Goebeler ME, Bargou R. Blinatumomab: a CD19/CD3 bispecific T cell engager (BiTE) with unique anti-tumor efficacy. Leuk Lymphoma 2016; 57:1021-32. [DOI: 10.3109/10428194.2016.1161185] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
26
|
Root AR, Cao W, Li B, LaPan P, Meade C, Sanford J, Jin M, O'Sullivan C, Cummins E, Lambert M, Sheehan AD, Ma W, Gatto S, Kerns K, Lam K, D'Antona AM, Zhu L, Brady WA, Benard S, King A, He T, Racie L, Arai M, Barrett D, Stochaj W, LaVallie ER, Apgar JR, Svenson K, Mosyak L, Yang Y, Chichili GR, Liu L, Li H, Burke S, Johnson S, Alderson R, Finlay WJJ, Lin L, Olland S, Somers W, Bonvini E, Gerber HP, May C, Moore PA, Tchistiakova L, Bloom L. Development of PF-06671008, a Highly Potent Anti-P-cadherin/Anti-CD3 Bispecific DART Molecule with Extended Half-Life for the Treatment of Cancer. Antibodies (Basel) 2016; 5:E6. [PMID: 31557987 PMCID: PMC6698862 DOI: 10.3390/antib5010006] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 02/14/2016] [Accepted: 02/17/2016] [Indexed: 01/13/2023] Open
Abstract
Bispecific antibodies offer a promising approach for the treatment of cancer but can be challenging to engineer and manufacture. Here we report the development of PF-06671008, an extended-half-life dual-affinity re-targeting (DART®) bispecific molecule against P-cadherin and CD3 that demonstrates antibody-like properties. Using phage display, we identified anti-P-cadherin single chain Fv (scFv) that were subsequently affinity-optimized to picomolar affinity using stringent phage selection strategies, resulting in low picomolar potency in cytotoxic T lymphocyte (CTL) killing assays in the DART format. The crystal structure of this disulfide-constrained diabody shows that it forms a novel compact structure with the two antigen binding sites separated from each other by approximately 30 Å and facing approximately 90° apart. We show here that introduction of the human Fc domain in PF-06671008 has produced a molecule with an extended half-life (-4.4 days in human FcRn knock-in mice), high stability (Tm1 > 68 °C), high expression (>1 g/L), and robust purification properties (highly pure heterodimer), all with minimal impact on potency. Finally, we demonstrate in vivo anti-tumor efficacy in a human colorectal/human peripheral blood mononuclear cell (PBMC) co-mix xenograft mouse model. These results suggest PF-06671008 is a promising new bispecific for the treatment of patients with solid tumors expressing P-cadherin.
Collapse
Affiliation(s)
- Adam R Root
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Wei Cao
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Bilian Li
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Peter LaPan
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Caryl Meade
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Jocelyn Sanford
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Macy Jin
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Cliona O'Sullivan
- Global Biotherapeutics Technologies, Pfizer Inc., Grange Castle Business Park, Clondalkin, Dublin 22, Ireland.
| | - Emma Cummins
- Global Biotherapeutics Technologies, Pfizer Inc., Grange Castle Business Park, Clondalkin, Dublin 22, Ireland.
| | - Matthew Lambert
- Global Biotherapeutics Technologies, Pfizer Inc., Grange Castle Business Park, Clondalkin, Dublin 22, Ireland.
| | - Alfredo D Sheehan
- Global Biotherapeutics Technologies, Pfizer Inc., Grange Castle Business Park, Clondalkin, Dublin 22, Ireland.
| | - Weijun Ma
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Scott Gatto
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Kelvin Kerns
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Khetemenee Lam
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Aaron M D'Antona
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Lily Zhu
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - William A Brady
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Susan Benard
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Amy King
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Tao He
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Lisa Racie
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Maya Arai
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Dianah Barrett
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Wayne Stochaj
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Edward R LaVallie
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - James R Apgar
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Kristine Svenson
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Lidia Mosyak
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Yinhua Yang
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA.
| | | | - Liqin Liu
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA.
| | - Hua Li
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA.
| | - Steve Burke
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA.
| | - Syd Johnson
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA.
| | - Ralph Alderson
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA.
| | - William J J Finlay
- Global Biotherapeutics Technologies, Pfizer Inc., Grange Castle Business Park, Clondalkin, Dublin 22, Ireland.
| | - Laura Lin
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Stéphane Olland
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - William Somers
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Ezio Bonvini
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA.
| | - Hans-Peter Gerber
- Oncology Research Unit, Pfizer Inc., 401 N. Middletown Road, Pearl River, NY 10965, USA.
| | - Chad May
- Oncology Research Unit, Pfizer Inc., 401 N. Middletown Road, Pearl River, NY 10965, USA.
| | - Paul A Moore
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA.
| | - Lioudmila Tchistiakova
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Laird Bloom
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| |
Collapse
|
27
|
Emole JN, Locke FL, Pinilla-Ibarz J. An update on current and prospective immunotherapies for chronic lymphocytic leukemia. Immunotherapy 2016; 7:455-66. [PMID: 25917633 DOI: 10.2217/imt.15.14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) is the most common leukemia. Combined agent chemotherapy is the current standard front-line treatment for physically fit patients with CLL. Use of chemotherapy can be complicated by significant toxicity, especially in patients with advanced age or comorbid conditions. Moreover, patients may relapse and become refractory to further chemotherapy. Immunotherapy targets the aberrant immunological processes in CLL without the toxicity of chemotherapy. Immunotherapeutic strategies can also be combined with chemotherapy to improve response rates in this incurable disease. In this review, we evaluate current and future immune-based options in the treatment of CLL.
Collapse
Affiliation(s)
- Josephine N Emole
- Department of Malignant Hematology, H Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | | | | |
Collapse
|
28
|
Taking up Cancer Immunotherapy Challenges: Bispecific Antibodies, the Path Forward? Antibodies (Basel) 2015; 5:antib5010001. [PMID: 31557983 PMCID: PMC6698871 DOI: 10.3390/antib5010001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 12/15/2015] [Accepted: 12/18/2015] [Indexed: 02/07/2023] Open
Abstract
As evidenced by the recent approvals of Removab (EU, Trion Pharma) in 2009 and of Blincyto (US, Amgen) in 2014, the high potential of bispecific antibodies in the field of immuno-oncology is eliciting a renewed interest from pharmaceutical companies. Supported by rapid advances in antibody engineering and the development of several technological platforms such as Triomab or bispecific T cell engagers (BiTEs), the “bispecifics” market has increased significantly over the past decade and may occupy a pivotal space in the future. Over 30 bispecific molecules are currently in different stages of clinical trials and more than 70 in preclinical phase. This review focuses on the clinical potential of bispecific antibodies as immune effector cell engagers in the onco-immunotherapy field. We summarize current strategies targeting various immune cells and their clinical interests. Furthermore, perspectives of bispecific antibodies in future clinical developments are addressed.
Collapse
|
29
|
Osaki T, Fujisawa S, Kitaguchi M, Kitamura M, Nakanishi T. Development of a bispecific antibody tetramerized through hetero-associating peptides. FEBS J 2015; 282:4389-401. [PMID: 26337767 DOI: 10.1111/febs.13505] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 08/04/2015] [Accepted: 08/30/2015] [Indexed: 11/27/2022]
Abstract
The specific assembly of self-associating peptides can be useful in building a functional antibody complex from small antibody fragments. We have focused on the exceedingly specific heterotetrameric assembly of Lin-2 and Lin-7 (L27) domains, which work as protein-protein interaction modules in many scaffold proteins. Here, we describe a novel method for constructing a highly functional antibody based on the hetero-association of L27 domains. In this study, we used a bacterial expression system to produce a bispecific antibody that was heterotetramerized through L27 domains and that targeted both epidermal growth factor receptor (EGFR) and Fcγ receptor III (FcγRIII or CD16). Gel electrophoresis, mass spectrometry and gel filtration analyses revealed that the constructed recombinant antibody was a disulfide-linked heterotetramer. The tetramerized antibody bound to EGFR and CD16 simultaneously, according to results from flow cytometry and surface plasmon resonance spectroscopy, respectively. Furthermore, we demonstrated that the bispecific antibody showed cytotoxic activity against EGFR-expressing tumor cells by using CD16-positive lymphocytes as effectors, and its cytotoxicity was comparable to that of a commercial therapeutic antibody. Taken together, the results show that our method has high potential for the cost-efficient production of highly active therapeutic antibodies.
Collapse
Affiliation(s)
- Tomohiro Osaki
- Department of Applied Chemistry and Bioengineering, Graduate School of Engineering, Osaka City University, Japan
| | - Shingo Fujisawa
- Department of Applied Chemistry and Bioengineering, Graduate School of Engineering, Osaka City University, Japan
| | - Masahiro Kitaguchi
- Department of Applied Chemistry and Bioengineering, Graduate School of Engineering, Osaka City University, Japan
| | - Masaya Kitamura
- Department of Applied Chemistry and Bioengineering, Graduate School of Engineering, Osaka City University, Japan
| | - Takeshi Nakanishi
- Department of Applied Chemistry and Bioengineering, Graduate School of Engineering, Osaka City University, Japan
| |
Collapse
|
30
|
Kaplan JB, Grischenko M, Giles FJ. Blinatumomab for the treatment of acute lymphoblastic leukemia. Invest New Drugs 2015; 33:1271-9. [PMID: 26383529 DOI: 10.1007/s10637-015-0289-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 09/08/2015] [Indexed: 01/13/2023]
Abstract
BACKGROUND Acute lymphoblastic leukemia (ALL) is a potentially fatal disease that involves clonal expansion of early lymphoid progenitor cells. Much of drug development for ALL treatment involves targeting antigens of the clonal cell surface. Blinatumomab belongs to an emerging class of anti-cancer therapeutics referred to as bispecific T-cell engaging antibodies. The Food and Drug Administration approved its use in relapsed or refractory adult Philadelphia chromosome-negative B-cell precursor ALL in December of 2014. MECHANISM OF ACTION AND PHARMACODYNAMICS Blinatumomab contains both an anti-CD3 and anti-CD19 arm, allowing for the juxtaposition of CD3+ T-cells to malignant CD19+ B-cells, thereby resulting in granzyme- and perforin-mediated B-cell apoptosis. PRECLINICAL PHARMACOLOGY Preclinical studies suggest that blinatumomab's efficacy is related to the effector-to-target ratio and to the difference between its affinity for CD19 and CD3. PHARMACOKINETICS AND METABOLISM Preclinical and early phase clinical studies have allowed for the characterization of the pharmacokinetics of blinatumomab, including the determination of its short half-life. The metabolic pathway has not been fully characterized but is thought to be similar to that of other antibodies. CLINICAL STUDIES Phase I and II studies led to the identification of an ideal stepwise dose, involving long-term continuous intravenous infusion (CIVI), to optimize its efficacy and reduce the risk of certain toxicities. A high remission rate and duration were noted among a relapsed/refractory population of patients. SAFETY The results of clinical trials have identified cytokine release syndrome and neurotoxicity, among others, as serious drug-related toxicities, leading to the institution of a Risk Evaluation and Mitigation Strategy. DISCUSSION AND CONCLUSIONS Blinatumomab represents a significant addition to the treatment options for ALL, but it is not without its limitations, of which are its short-half life, necessitating long-term CIVI, and the eventual emergence of CD19-negative clones. Continual development of the agent involves assessing its role in the frontline setting and in combination with chemotherapy.
Collapse
Affiliation(s)
- Jason B Kaplan
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, 233 E. Superior St., First floor, Chicago, IL, 60611, USA. .,Division of Hematology and Oncology and Northwestern Medicine Developmental Therapeutics Institute (NMDTI), Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| | - Marina Grischenko
- Division of Hematology and Oncology and Northwestern Medicine Developmental Therapeutics Institute (NMDTI), Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Francis J Giles
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, 233 E. Superior St., First floor, Chicago, IL, 60611, USA.,Division of Hematology and Oncology and Northwestern Medicine Developmental Therapeutics Institute (NMDTI), Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
31
|
Targeted Therapies in Adult B-Cell Malignancies. BIOMED RESEARCH INTERNATIONAL 2015; 2015:217593. [PMID: 26425544 PMCID: PMC4575712 DOI: 10.1155/2015/217593] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 05/03/2015] [Accepted: 05/05/2015] [Indexed: 12/17/2022]
Abstract
B-lymphocytes are programmed for the production of immunoglobulin (Ig) after antigen presentation, in the context of T-lymphocyte control within lymphoid organs. During this differentiation/activation process, B-lymphocytes exhibit different restricted or common surface markers, activation of cellular pathways that regulate cell cycle, metabolism, proteasome activity, and protein synthesis. All molecules involved in these different cellular mechanisms are potent therapeutic targets. Nowadays, due to the progress of the biology, more and more targeted drugs are identified, a situation that is correlated with an extended field of the targeted therapy. The full knowledge of the cellular machinery and cell-cell communication allows making the best choice to treat patients, in the context of personalized medicine. Also, focus should not be restricted to the immediate effects observed as clinical endpoints, that is, response rate, survival markers with conventional statistical methods, but it should consider the prediction of different clinical consequences due to other collateral drug targets, based on new methodologies. This means that new reflection and new bioclinical follow-up have to be monitored, particularly with the new drugs used with success in B-cell malignancies. This review discussed the principal aspects of such evident bioclinical progress.
Collapse
|
32
|
Wu J, Fu J, Zhang M, Liu D. Blinatumomab: a bispecific T cell engager (BiTE) antibody against CD19/CD3 for refractory acute lymphoid leukemia. J Hematol Oncol 2015; 8:104. [PMID: 26337639 PMCID: PMC4558758 DOI: 10.1186/s13045-015-0195-4] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 08/04/2015] [Indexed: 11/11/2022] Open
Abstract
Targeted therapy has been the forefront of cancer treatment. Cancer immunotherapy is the most recent focus. In addition, novel immunotherapeutics targeting B cell receptor signaling (e.g., ibrutinib), T cell receptor ( e.g., CART19), and NK cells (e.g., AFM13) are being developed. This review summarized the new development in blinatumomab (MT103/MEDI-538), a first-in-class bispecific T engager (BiTE) antibody against CD19/CD3 in patients with relapsed/refractory precursor B cell acute lymphoid leukemia.
Collapse
Affiliation(s)
- Jingjing Wu
- Department of Oncology, The first Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jiaping Fu
- Department of Hematology, Shaoxing People's Hospital, Shaoxing, Zhejiang Province, China
| | - Mingzhi Zhang
- Department of Oncology, The first Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Delong Liu
- Department of Oncology, The first Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China. .,Division of Hematology & Oncology, New York Medical College, Valhalla, NY, 10595, USA.
| |
Collapse
|
33
|
Ribera JM, Ferrer A, Ribera J, Genescà E. Profile of blinatumomab and its potential in the treatment of relapsed/refractory acute lymphoblastic leukemia. Onco Targets Ther 2015; 8:1567-74. [PMID: 26170691 PMCID: PMC4485855 DOI: 10.2147/ott.s70524] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The CD19 marker is expressed on the surface of normal and malignant immature or mature B-cells. On the other hand, immunotherapy involving T-cells is a promising modality of treatment for many neoplastic diseases including leukemias and lymphomas. The CD19/CD3-bispecific T-cell-engaging (BiTE®) monoclonal antibody blinatumomab can transiently engage cytotoxic T-cells to CD19+ target B-cells inducing serial perforin-mediated lysis. In the first clinical trial, blinatumomab showed efficacy in non-Hodgkin’s lymphomas, but the most important trials have been conducted in relapsed/refractory (R/R) acute lymphoblastic leukemia (ALL) and in ALL with minimal residual disease. Encouraging reports on the activity of blinatumomab in R/R Philadelphia chromosome-negative B-cell precursor ALL led to its approval by the US Food and Drug Administration on December 3, 2014 after an accelerated review process. This review focuses on the profile of blinatumomab and its activity in R/R ALL.
Collapse
Affiliation(s)
- Josep-Maria Ribera
- Clinical Hematology Department, ICO-Hospital Germans Trias i Pujol, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Albert Ferrer
- Clinical Hematology Department, ICO-Hospital Germans Trias i Pujol, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Jordi Ribera
- Clinical Hematology Department, ICO-Hospital Germans Trias i Pujol, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Eulàlia Genescà
- Clinical Hematology Department, ICO-Hospital Germans Trias i Pujol, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona, Spain
| |
Collapse
|
34
|
Stieglmaier J, Benjamin J, Nagorsen D. Utilizing the BiTE (bispecific T-cell engager) platform for immunotherapy of cancer. Expert Opin Biol Ther 2015; 15:1093-9. [PMID: 25971805 DOI: 10.1517/14712598.2015.1041373] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Various approaches of T-cell-based cancer immunotherapy are currently under investigation, among these are BiTE® (bispecific T-cell engager) antibody constructs, which have a unique design and mechanism of action. They are constructed by genetically linking onto a single polypeptide chain the minimal binding domains of monoclonal antibodies for tumor-associated surface antigens and for the T-cell receptor-associated molecule CD3. Concurrent engagement of the target cell antigen and CD3 leads to activation of polyclonal cytotoxic T-cells, resulting in target cell lysis. Blinatumomab, a BiTE targeting CD19, is being investigated in a broad range of B-cell malignancies and has recently been approved in the USA by the US FDA for Philadelphia chromosome-negative relapsed/refractory B-acute lymphoblastic leukemia under the trade name BLINCYTO™. The BiTE platform is one of the clinically most advanced T-cell immunotherapy options.
Collapse
Affiliation(s)
- Julia Stieglmaier
- Amgen Research (Munich), Global Clinical Development, Therapeutic Area Oncology , Staffelseestraße 2, Munich 81477 , Germany
| | | | | |
Collapse
|
35
|
Ribera JM, Vives S, Motlló C, Jiménez-Lorenzo MJ. Blinatumomab for treating acute lymphoblastic leukemia. Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.1024223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
36
|
Deonarain MP, Yahioglu G, Stamati I, Marklew J. Emerging formats for next-generation antibody drug conjugates. Expert Opin Drug Discov 2015; 10:463-81. [PMID: 25797303 DOI: 10.1517/17460441.2015.1025049] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Antibody drug conjugates now make up a significant fraction of biopharma's oncology pipeline due to great advances in the understanding of the three key components and how they should be optimised together. With this clinical success comes innovation to produce new enabling technologies that can deliver more effective antibody-drug conjugates (ADCs) with a larger therapeutic index. AREAS COVERED There are many reviews that discuss the various strategies for ADCs design but the last 5 years or so have witnessed the emergence of a number of different antibody formats compete with the standard whole immunoglobulin. Using published research, patent applications and conference disclosures, the authors review the many antibody and antibody-like formats, discussing innovations in protein engineering and how these new formats impact on the conjugation strategy and ultimately the performance. The alternative chemistries that are now available offer new linkages, stability profiles, drug:antibody ratio, pharmacokinetics and efficacy. The different sizes being considered promise to address issues, such as tumour penetration, circulatory half-life and side-effects. EXPERT OPINION ADCs are at the beginning of the next stage in their evolution and as these newer formats are developed and examined in the clinic, we will discover if the predicted features have a clinical benefit. From the commercial activity, it is envisaged that smaller or fragment-based ADCs will expand oncological applications.
Collapse
Affiliation(s)
- Mahendra P Deonarain
- Antikor Biopharma Ltd, Stevenage Bioscience Catalyst , Gunnels Wood Road, Stevenage, Herts, SG1 2FX , UK
| | | | | | | |
Collapse
|
37
|
Immunotherapy: An Evolving Paradigm in the Treatment of Advanced Cervical Cancer. Clin Ther 2015; 37:20-38. [DOI: 10.1016/j.clinthera.2014.11.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 11/14/2014] [Accepted: 11/14/2014] [Indexed: 11/23/2022]
|