1
|
Kegyes D, Ghiaur G, Bancos A, Tomuleasa C, Gale RP. Immune therapies of B-cell acute lymphoblastic leukaemia in children and adults. Crit Rev Oncol Hematol 2024; 196:104317. [PMID: 38437908 DOI: 10.1016/j.critrevonc.2024.104317] [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: 10/14/2023] [Revised: 01/26/2024] [Accepted: 02/27/2024] [Indexed: 03/06/2024] Open
Abstract
B-cell acute lymphoblastic leukaemia (B-cell ALL) is a common haematologic cancer in children and adults. About 10 percent of children and 50 percent of adults fail to achieve a histological complete remission or subsequently relapse despite current anti-leukaemia drug therapies and/or haematopoietic cell transplants. Several new immune therapies including monoclonal antibodies and chimeric antigen receptor (CAR)-T-cells are proved safe and effective in this setting. We review data on US Food and Drug Administration (FDA)-approved immune therapies for B-cell ALL in children and adults including blinatumomab, inotuzumab ozogamicin, tisagenlecleucel, and brexucabtagene autoleucel. We also summarize pharmaco-dynamics, pharmaco-kinetics, and pharmaco-economics of these interventions.
Collapse
Affiliation(s)
- David Kegyes
- Department of Hematology-Medfuture Research Center for Advanced Medicine, Iuliu Hațieganu University of Medicine and Pharmacy Cluj Napoca, Romania; Department of Hematology, Ion Chiricuta Oncology Institute, Cluj Napoca, Romania; Academy of Romanian Scientists, Bucharest, Romania
| | - Gabriel Ghiaur
- Department of Hematology-Medfuture Research Center for Advanced Medicine, Iuliu Hațieganu University of Medicine and Pharmacy Cluj Napoca, Romania; Department of Leukemia, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University, Baltimore, MD, USA
| | - Anamaria Bancos
- Department of Hematology-Medfuture Research Center for Advanced Medicine, Iuliu Hațieganu University of Medicine and Pharmacy Cluj Napoca, Romania; Department of Hematology, Ion Chiricuta Oncology Institute, Cluj Napoca, Romania
| | - Ciprian Tomuleasa
- Department of Hematology-Medfuture Research Center for Advanced Medicine, Iuliu Hațieganu University of Medicine and Pharmacy Cluj Napoca, Romania; Department of Hematology, Ion Chiricuta Oncology Institute, Cluj Napoca, Romania; Academy of Romanian Scientists, Bucharest, Romania.
| | - Robert Peter Gale
- Centre for Haematology, Imperial College of Science, Technology and Medicine, London, UK; Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China; Department of Hematology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
2
|
Aureli A, Marziani B, Venditti A, Sconocchia T, Sconocchia G. Acute Lymphoblastic Leukemia Immunotherapy Treatment: Now, Next, and Beyond. Cancers (Basel) 2023; 15:3346. [PMID: 37444456 DOI: 10.3390/cancers15133346] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/14/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) is a blood cancer that primarily affects children but also adults. It is due to the malignant proliferation of lymphoid precursor cells that invade the bone marrow and can spread to extramedullary sites. ALL is divided into B cell (85%) and T cell lineages (10 to 15%); rare cases are associated with the natural killer (NK) cell lineage (<1%). To date, the survival rate in children with ALL is excellent while in adults continues to be poor. Despite the therapeutic progress, there are subsets of patients that still have high relapse rates after chemotherapy or hematopoietic stem cell transplantation (HSCT) and an unsatisfactory cure rate. Hence, the identification of more effective and safer therapy choices represents a primary issue. In this review, we will discuss novel therapeutic options including bispecific antibodies, antibody-drug conjugates, chimeric antigen receptor (CAR)-based therapies, and other promising treatments for both pediatric and adult patients.
Collapse
Affiliation(s)
- Anna Aureli
- CNR Institute of Translational Pharmacology, Via Carducci 32, 67100 L'Aquila, Italy
| | - Beatrice Marziani
- Emergency Medicine Department, Sant'Anna University Hospital, Via A. Moro, 8, Cona, 44124 Ferrara, Italy
| | - Adriano Venditti
- Department of Biomedicine and Prevention, The University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Tommaso Sconocchia
- Division of Hematology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Giuseppe Sconocchia
- CNR Institute of Translational Pharmacology, Via Carducci 32, 67100 L'Aquila, Italy
| |
Collapse
|
3
|
Kegyes D, Jitaru C, Ghiaur G, Ciurea S, Hoelzer D, Tomuleasa C, Gale RP. Switching from salvage chemotherapy to immunotherapy in adult B-cell acute lymphoblastic leukemia. Blood Rev 2023; 59:101042. [PMID: 36732205 DOI: 10.1016/j.blre.2023.101042] [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: 11/17/2022] [Revised: 12/27/2022] [Accepted: 01/18/2023] [Indexed: 01/21/2023]
Abstract
About one-half of adults with acute B-cell lymphoblastic leukemia (B-ALL) who do not achieve molecular complete remission or who subsequently relapse are not cured by current chemo- or targeted therapies. Previously, the sole therapeutic option for such persons was a hematopoietic stem cell transplant. Recently, several immune therapies including monoclonal antibodies, bispecific T-cell engagers (BiTEs), antibody-drug conjugates (ADCs), and chimeric antigen receptor T-cells (CARs) have been shown safe and effective in this setting. In this manuscript, we summarize data on US FDA-approved immune therapies of advanced adult B-ALL including rituximab, blinatumomab, inotuzumab ozogamicin, tisagenlecleucel and brexucabtagene autoleucel. We consider the results of clinical trials focusing on efficacy, safety, and quality of life (QoL). Real-world evidence is presented as well. We also briefly discuss pharmacodynamics, pharmacokinetics, and pharmacoeconomics followed by risk-benefit analyses. Lastly, we present future directions of immune therapies for advanced B-ALL in adults.
Collapse
Affiliation(s)
- David Kegyes
- Department of Hematology-Medfuture Research Center for Advanced Medicine, Iuliu Hațieganu University of Medicine and Pharmacy Cluj Napoca, Romania; Department of Hematology, Ion Chiricuta Oncology Institute, Cluj Napoca, Romania
| | - Ciprian Jitaru
- Department of Hematology-Medfuture Research Center for Advanced Medicine, Iuliu Hațieganu University of Medicine and Pharmacy Cluj Napoca, Romania; Department of Hematology, Ion Chiricuta Oncology Institute, Cluj Napoca, Romania
| | - Gabriel Ghiaur
- Department of Hematology-Medfuture Research Center for Advanced Medicine, Iuliu Hațieganu University of Medicine and Pharmacy Cluj Napoca, Romania; Department of Leukemia, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Stefan Ciurea
- Department of Stem Cell Transplant and Cellular Therapies, University of California, Irvine, CA, USA
| | - Dieter Hoelzer
- Department of Medicine, Goethe University, Frankfurt, Germany
| | - Ciprian Tomuleasa
- Department of Hematology-Medfuture Research Center for Advanced Medicine, Iuliu Hațieganu University of Medicine and Pharmacy Cluj Napoca, Romania; Department of Hematology, Ion Chiricuta Oncology Institute, Cluj Napoca, Romania.
| | - Robert Peter Gale
- Centre for Haematology, Imperial College of Science, Technology and Medicine, London, UK; Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
4
|
Liu D, Qi X, Wei X, Zhao L, Wang X, Li S, Wang Z, Shi L, Xu J, Hong M, Liu Z, Zhao L, Wang X, Zhang B, Zhang Y, Wang F, Cao YJ. A Novel Her2/VEGFR2/CD3 trispecific antibody with an optimal structural design showed improved T-cell-redirecting antitumor efficacy. Am J Cancer Res 2022; 12:7788-7803. [PMID: 36451856 PMCID: PMC9706591 DOI: 10.7150/thno.75037] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 11/02/2022] [Indexed: 12/03/2022] Open
Abstract
Rationale: T-cell-redirecting bispecific antibodies (bsAbs) and trispecific antibodies (tsAbs) designed to recognize different epitopes or antigens have emerged as promising cancer therapies. Current approaches are all designed to include another antibody specific to the site of the primary antibody, and the molecular structures are generally established. However, the dimensions of target molecule and epitope location play a key role in the efficiency of the immunological synapse (IS) formation and subsequent T-cell-redirecting activities, therefore the connection flexibility of these antibodies determines the geometries of different formats of these molecules and will have a major impact on the efficacy. Methods: We describe a novel recombination strategy using various linker designs to site-specifically fuse anti-Her2 (2Rs15) or anti-VEGFR2 (3VGR19) nanobodies to different positions of the anti-CD3 antibody fragment (Fab, SP34). Based on the comparison among the various antigen-specific bsAbs, we could determine the desired fusion site of each nanobody to SP34, and further ensure the optimal structure of tsAbs with synergistic dual-antigen enhanced T-cell-redirecting activities. Results: This approach allows precise control of the formation of IS between Her2- and/or VEGFR2-expressing cancer cells and T cells, to obtain the optimal structure of the Her2/VEGFR2/CD3 tsAb without the need to map antibody-binding epitopes. Optimization of Her2/VEGFR2/CD3 tsAb results in enhanced T-cell-redirecting in vitro and in vivo antitumor efficacy compared with the corresponding bsAbs alone or in combination, and the potency to overcome tumor relapse due to antigen escape or resistance to Herceptin and Cyramza therapy. Conclusion: The novel design strategy for developing tsAbs using a site-specific recombination approach represents a promising platform for immuno-oncology and in applications other than cancer therapy.
Collapse
Affiliation(s)
- Dong Liu
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, 518055, China
| | - Xuexiu Qi
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, 518055, China
| | - Xiaoyi Wei
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, 518055, China
| | - Lijun Zhao
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, 518055, China
| | - Xuechun Wang
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, 518055, China
| | - Shuhong Li
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, 518055, China
| | - Zhidong Wang
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, 518055, China
| | - Licai Shi
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, 518055, China
| | - Jiean Xu
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, 518055, China
| | - Mei Hong
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, 518055, China
| | - Zhong Liu
- Lunan Pharmaceutical Group Co., Ltd, Feixian County, Shandong, 273400, China
| | - Lili Zhao
- National Engineering Laboratory of High Level Expression in Mammalian Cells, Feixian County, Shandong, 273400, China.,✉ Corresponding authors: ;
| | - Xiankun Wang
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, 518055, China.,Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518132, China
| | - Bo Zhang
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, 518055, China.,Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518132, China
| | - Yuhan Zhang
- Key Laboratory of Protein and Peptide Pharmaceuticals, Beijing Translational Center for Biopharmaceuticals Institute of Biophysics, Chinese Academy of Sciences Beijing 100101, China
| | - Feng Wang
- Key Laboratory of Protein and Peptide Pharmaceuticals, Beijing Translational Center for Biopharmaceuticals Institute of Biophysics, Chinese Academy of Sciences Beijing 100101, China
| | - Yu J. Cao
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, 518055, China.,Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, 518132, China.,✉ Corresponding authors: ;
| |
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: 0.5] [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
|
Ordóñez-Reyes C, Garcia-Robledo JE, Chamorro DF, Mosquera A, Sussmann L, Ruiz-Patiño A, Arrieta O, Zatarain-Barrón L, Rojas L, Russo A, de Miguel-Perez D, Rolfo C, Cardona AF. Bispecific Antibodies in Cancer Immunotherapy: A Novel Response to an Old Question. Pharmaceutics 2022; 14:pharmaceutics14061243. [PMID: 35745815 PMCID: PMC9229626 DOI: 10.3390/pharmaceutics14061243] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 06/02/2022] [Accepted: 06/08/2022] [Indexed: 01/14/2023] Open
Abstract
Immunotherapy has redefined the treatment of cancer patients and it is constantly generating new advances and approaches. Among the multiple options of immunotherapy, bispecific antibodies (bsAbs) represent a novel thoughtful approach. These drugs integrate the action of the immune system in a strategy to redirect the activation of innate and adaptive immunity toward specific antigens and specific tumor locations. Here we discussed some basic aspects of the design and function of bsAbs, their main challenges and the state-of-the-art of these molecules in the treatment of hematological and solid malignancies and future perspectives.
Collapse
Affiliation(s)
- Camila Ordóñez-Reyes
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá 110111, Colombia; (C.O.-R.); (J.E.G.-R.); (D.F.C.); (A.M.); (A.R.-P.); (L.R.)
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad el Bosque, Bogotá 110121, Colombia
| | - Juan Esteban Garcia-Robledo
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá 110111, Colombia; (C.O.-R.); (J.E.G.-R.); (D.F.C.); (A.M.); (A.R.-P.); (L.R.)
- Division of Hematology/Oncology, Mayo Clinic, Phoenix, AZ 85054, USA
| | - Diego F. Chamorro
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá 110111, Colombia; (C.O.-R.); (J.E.G.-R.); (D.F.C.); (A.M.); (A.R.-P.); (L.R.)
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad el Bosque, Bogotá 110121, Colombia
| | - Andrés Mosquera
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá 110111, Colombia; (C.O.-R.); (J.E.G.-R.); (D.F.C.); (A.M.); (A.R.-P.); (L.R.)
| | - Liliana Sussmann
- Department of Neurology, Fundación Universitaria de Ciencias de la Salud, Bogotá 111221, Colombia;
| | - Alejandro Ruiz-Patiño
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá 110111, Colombia; (C.O.-R.); (J.E.G.-R.); (D.F.C.); (A.M.); (A.R.-P.); (L.R.)
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad el Bosque, Bogotá 110121, Colombia
| | - Oscar Arrieta
- Thoracic Oncology Unit and Personalized Oncology Laboratory, National Cancer Institute (INCan), Mexico City 14080, Mexico; (O.A.); (L.Z.-B.)
| | - Lucia Zatarain-Barrón
- Thoracic Oncology Unit and Personalized Oncology Laboratory, National Cancer Institute (INCan), Mexico City 14080, Mexico; (O.A.); (L.Z.-B.)
| | - Leonardo Rojas
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá 110111, Colombia; (C.O.-R.); (J.E.G.-R.); (D.F.C.); (A.M.); (A.R.-P.); (L.R.)
| | | | - Diego de Miguel-Perez
- Center for Thoracic Oncology, Tisch Cancer Institute and Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (D.d.M.-P.); (C.R.)
| | - Christian Rolfo
- Center for Thoracic Oncology, Tisch Cancer Institute and Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (D.d.M.-P.); (C.R.)
| | - Andrés F. Cardona
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá 110111, Colombia; (C.O.-R.); (J.E.G.-R.); (D.F.C.); (A.M.); (A.R.-P.); (L.R.)
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad el Bosque, Bogotá 110121, Colombia
- Direction of Research, Science and Education, Luis Carlos Sarmiento Angulo Cancer Treatment and Research Center (CTIC), Bogotá 110131, Colombia
- Correspondence: ; Tel.: +57-(1)-6190052; Fax: +57-(1)-6190053
| |
Collapse
|
7
|
Comparison of Anticancer Drug Toxicities: Paradigm Shift in Adverse Effect Profile. Life (Basel) 2021; 12:life12010048. [PMID: 35054441 PMCID: PMC8777973 DOI: 10.3390/life12010048] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/17/2021] [Accepted: 12/24/2021] [Indexed: 02/06/2023] Open
Abstract
The inception of cancer treatment with chemotherapeutics began in the 1940s with nitrogen mustards that were initially employed as weapons in World War II. Since then, treatment options for different malignancies have evolved over the period of last seventy years. Until the late 1990s, all the chemotherapeutic agents were small molecule chemicals with a highly nonspecific and severe toxicity spectrum. With the landmark approval of rituximab in 1997, a new horizon has opened up for numerous therapeutic antibodies in solid and hematological cancers. Although this transition to large molecules improved the survival and quality of life of cancer patients, this has also coincided with the change in adverse effect patterns. Typically, the anticancer agents are fraught with multifarious adverse effects that negatively impact different organs of cancer patients, which ultimately aggravate their sufferings. In contrast to the small molecules, anticancer antibodies are more targeted toward cancer signaling pathways and exhibit fewer side effects than traditional small molecule chemotherapy treatments. Nevertheless, the interference with the immune system triggers serious inflammation- and infection-related adverse effects. The differences in drug disposition and interaction with human basal pathways contribute to this paradigm shift in adverse effect profile. It is critical that healthcare team members gain a thorough insight of the adverse effect differences between the agents discovered during the last twenty-five years and before. In this review, we summarized the general mechanisms and adverse effects of small and large molecule anticancer drugs that would further our understanding on the toxicity patterns of chemotherapeutic regimens.
Collapse
|
8
|
Wei PS, Chen YJ, Lin SY, Chuang KH, Sheu MT, Ho HO. In situ subcutaneously injectable thermosensitive PEG-PLGA diblock and PLGA-PEG-PLGA triblock copolymer composite as sustained delivery of bispecific anti-CD3 scFv T-cell/anti-EGFR Fab Engager (BiTEE). Biomaterials 2021; 278:121166. [PMID: 34634663 DOI: 10.1016/j.biomaterials.2021.121166] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/12/2021] [Accepted: 09/28/2021] [Indexed: 12/12/2022]
Abstract
In this study, PEGylated poly (lactide-co-glycolide) (PLGA) thermosensitive composite hydrogels (DTgels) loaded with bispecific anti-cluster of differentiation 3 (CD3) scFv T-cell/anti-epidermal growth factor receptor (EGFR) Fab engager (BiTEE) were subcutaneously (s.c.) injected for the in situ formation of a drug deposit to resolve limitations of the clinical application of the BiTEE of a short half-life and potential side effects. Three kinds of DTgels prepared with different ratios of methoxy poly (ethylene glycol) (mPEG)-PLGA (diblock copolymer, DP) and PLGA-PEG-PLGA (triblock copolymer, TP) were designated DTgel-1, DTgel-2, and DTgel-2S. All three DTgel formulations showed thermosensitive properties with a sol-gel transition temperature at 28-34 °C, which is suitable for an injection. An in vitro release study showed that all DTgel formulations loaded with stabilized BiTEE extended the release of the BiTEE for up to 7 days. In an animal pharmacokinetics study, an s.c. injection of BiTEE/DTgel-1, BiTEE/DTgel-2, or BiTEE/DTgel-2S respectively prolonged the half-life of the BiTEE by 3.5-, 2.0-, and 2.2-fold compared to an intravenous injection of the BiTEE solution. Simultaneously, BiTEE/DTgel formulations showed almost no proinflammatory cytokine release in mice injected with T cells after s.c. administration. Results of an animal antitumor (MDA-MB-231) study indicated that an s.c. injection of the BiTEE/DTgel formulations significantly improved the antitumor efficacy compared to an intravenous (i.v.) or s.c. injection of the BiTEE solution. Moreover, BiTEE/DTgel formulations led to enhanced T-cell recruitment to solid-tumor sites. In conclusion, the in situ formation of injectable PEGylated PLGA thermosensitive hydrogels loaded with the BiTEE was successfully carried out to increase its half-life, maintain a constant blood level within therapeutic windows, and enhance T-cell recruitment to solid-tumor sites resulting in exceptional treatment efficacy.
Collapse
Affiliation(s)
- Pu-Sheng Wei
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Yi-Jou Chen
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan
| | - Shyr-Yi Lin
- Division of Gastroenterology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kuo-Hsiang Chuang
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan; Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Ming-Thau Sheu
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; Traditional Herbal Medicine Research Center of Taipei Medical University Hospital, Taipei, Taiwan.
| | - Hsiu-O Ho
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
9
|
Shenouda S, Kulkarni K, Abuetabh Y, Sergi C. Cancer Stem Cells and their Management in Cancer Therapy. Recent Pat Anticancer Drug Discov 2021; 15:212-227. [PMID: 32660407 DOI: 10.2174/1574892815666200713145931] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/16/2020] [Accepted: 06/20/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND In the last decade, the proposed Cancer Stem Cell (CSC) hypothesis has steadily changed the way cancer treatment is approached. CSCs may be the source of the heterogeneous non-tumorigenic cell population included in a neoplasm. Intratumor and intertumoral heterogeneity is a well-known phenomenon that massively entangles the diagnosis and treatment of cancer. The literature seems to suggest that heterogeneity develops progressively within tumor-initiating stem cells. CSCs harbor genetic and/or epigenetic alterations that allow them to differentiate into multiple tumor cell types sequentially. OBJECTIVE The CSC hypothesis, cellular therapy, and the most recent patents on CSCs were reviewed. METHODS PubMed, Scopus, and Google Scholar were screened for this information. Also, an analysis of the most recent data targeting CSCs in pediatric cancer developed at two Canadian institutions is provided. The genes involved with the activation of CSCs and the drugs used to antagonize them are also highlighted. RESULTS It is underlined that (1) CSCs possess stem cell-like properties, including the ability for self-renewal; (2) CSCs can start carcinogenesis and are responsible for tumor recurrence after treatment; (3) Although some limitations have been raised, which may oppose the CSC hypothesis, cancer progression and metastasis have been recognized to be caused by CSCs. CONCLUSION The significant roles of cell therapy may include an auto-transplant with high-dose treatment, an improvement of the immune function, creation of chimeric antigen receptor T cells, and the recruitment of NK cell-based immunotherapy.
Collapse
Affiliation(s)
- Suzan Shenouda
- Department of Lab. Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Ketan Kulkarni
- Department of Pediatrics, Pediatric Hematology/Oncology, Halifax, NS, Canada
| | - Yasser Abuetabh
- Department of Lab. Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Consolato Sergi
- Department of Lab. Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
10
|
Azadi A, Golchini A, Delazar S, Abarghooi Kahaki F, Dehnavi SM, Payandeh Z, Eyvazi S. Recent Advances on Immune Targeted Therapy of Colorectal Cancer Using bi-Specific Antibodies and Therapeutic Vaccines. Biol Proced Online 2021; 23:13. [PMID: 34193050 PMCID: PMC8245152 DOI: 10.1186/s12575-021-00147-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/12/2021] [Indexed: 12/22/2022] Open
Abstract
Colorectal cancer (CRC) is a universal heterogeneous disease that is characterized by genetic and epigenetic alterations. Immunotherapy using monoclonal antibodies (mAb) and cancer vaccines are substitute strategies for CRC treatment. When cancer immunotherapy is combined with chemotherapy, surgery, and radiotherapy, the CRC treatment would become excessively efficient. One of the compelling immunotherapy approaches to increase the efficiency of CRC therapy is the deployment of therapeutic mAbs, nanobodies, bi-specific antibodies and cancer vaccines, which improve clinical outcomes in patients. Also, among the possible therapeutic approaches for CRC patients, gene vaccines in combination with antibodies are recently introduced as a new perspective. Here, we aimed to present the current progress in CRC immunotherapy, especially using Bi-specific antibodies and dendritic cells mRNA vaccines. For this aim, all data were extracted from Google Scholar, PubMed, Scopus, and Elsevier, using keywords cancer vaccines; CRC immunotherapy and CRC mRNA vaccines. About 97 articles were selected and investigated completely based on the latest developments and novelties on bi-specific antibodies, mRNA vaccines, nanobodies, and MGD007.
Collapse
Affiliation(s)
- Ali Azadi
- Department of Medicine, De La Salle Health Sciences Institute, Dasmariñas, Philippines
| | - Alireza Golchini
- Cancer surgery Department; Shiraz Medical School, Shiraz University of medical Sciences, Shiraz, Iran
| | - Sina Delazar
- Department of Radiology, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Abarghooi Kahaki
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Mohsen Dehnavi
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Zahra Payandeh
- Immunology Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shirin Eyvazi
- Department of Biology, Tabriz Branch, Islamic Azad University, Tabriz, Iran
- Biotechnology Research Center, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| |
Collapse
|
11
|
Bogen JP, Carrara SC, Fiebig D, Grzeschik J, Hock B, Kolmar H. Design of a Trispecific Checkpoint Inhibitor and Natural Killer Cell Engager Based on a 2 + 1 Common Light Chain Antibody Architecture. Front Immunol 2021; 12:669496. [PMID: 34040611 PMCID: PMC8141644 DOI: 10.3389/fimmu.2021.669496] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/21/2021] [Indexed: 12/18/2022] Open
Abstract
Natural killer cell engagers gained enormous interest in recent years due to their potent anti-tumor activity and favorable safety profile. Simultaneously, chicken-derived antibodies entered clinical studies paving the way for avian-derived therapeutics. In this study, we describe the affinity maturation of a common light chain (cLC)-based, chicken-derived antibody targeting EGFR, followed by utilization of the same light chain for the isolation of CD16a- and PD-L1-specific monoclonal antibodies. The resulting binders target their respective antigen with single-digit nanomolar affinity while blocking the ligand binding of all three respective receptors. Following library-based humanization, bispecific and trispecific variants in a standard 1 + 1 or a 2 + 1 common light chain format were generated, simultaneously targeting EGFR, CD16a, and PD-L1. The trispecific antibody mediated an elevated antibody-dependent cellular cytotoxicity (ADCC) in comparison to the EGFR×CD16a bispecific variant by effectively bridging EGFR/PD-L1 double-positive cancer cells with CD16a-positive effector cells. These findings represent, to our knowledge, the first detailed report on the generation of a trispecific 2 + 1 antibodies exhibiting a common light chain and illustrate synergistic effects of trispecific antigen binding. Overall, this generic procedure paves the way for the engineering of tri- and oligospecific therapeutic antibodies derived from avian immunizations.
Collapse
MESH Headings
- Animals
- Antibodies, Bispecific/immunology
- Antibodies, Bispecific/pharmacology
- Antibodies, Monoclonal, Humanized/immunology
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibody Specificity
- B7-H1 Antigen/antagonists & inhibitors
- B7-H1 Antigen/immunology
- B7-H1 Antigen/metabolism
- Cell Line, Tumor
- Chickens
- Cytotoxicity, Immunologic/drug effects
- Drug Design
- Epitopes
- ErbB Receptors/antagonists & inhibitors
- ErbB Receptors/immunology
- ErbB Receptors/metabolism
- Immune Checkpoint Inhibitors/immunology
- Immune Checkpoint Inhibitors/pharmacology
- Immunization
- Immunoglobulin Light Chains/immunology
- Immunoglobulin Light Chains/pharmacology
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Receptors, IgG/antagonists & inhibitors
- Receptors, IgG/immunology
- Receptors, IgG/metabolism
- Skin Neoplasms/drug therapy
- Skin Neoplasms/immunology
- Skin Neoplasms/metabolism
- Skin Neoplasms/pathology
Collapse
Affiliation(s)
- Jan P. Bogen
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
- Ferring Darmstadt Laboratory, Biologics Technology and Development, Darmstadt, Germany
| | - Stefania C. Carrara
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
- Ferring Darmstadt Laboratory, Biologics Technology and Development, Darmstadt, Germany
| | - David Fiebig
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
- Ferring Darmstadt Laboratory, Biologics Technology and Development, Darmstadt, Germany
| | - Julius Grzeschik
- Ferring Darmstadt Laboratory, Biologics Technology and Development, Darmstadt, Germany
| | - Björn Hock
- Global Pharmaceutical Research and Development, Ferring International Center S.A., Saint-Prex, Switzerland
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| |
Collapse
|
12
|
Concepts in immuno-oncology: tackling B cell malignancies with CD19-directed bispecific T cell engager therapies. Ann Hematol 2020; 99:2215-2229. [PMID: 32856140 PMCID: PMC7481145 DOI: 10.1007/s00277-020-04221-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 08/11/2020] [Indexed: 12/11/2022]
Abstract
The B cell surface antigen CD19 is a target for treating B cell malignancies, such as B cell precursor acute lymphoblastic leukemia and B cell non-Hodgkin lymphoma. The BiTE® immuno-oncology platform includes blinatumomab, which is approved for relapsed/refractory B cell precursor acute lymphoblastic leukemia and B cell precursor acute lymphoblastic leukemia with minimal residual disease. Blinatumomab is also being evaluated in combination with other agents (tyrosine kinase inhibitors, checkpoint inhibitors, and chemotherapy) in various treatment settings, including frontline protocols. An extended half-life BiTE molecule is also under investigation. Patients receiving blinatumomab may experience cytokine release syndrome and neurotoxicity; however, these events may be less frequent and severe than in patients receiving other CD19-targeted immunotherapies, such as chimeric antigen receptor T cell therapy. We review BiTE technology for treating malignancies that express CD19, analyzing the benefits and limitations of this bispecific T cell engager platform from clinical experience with blinatumomab.
Collapse
|
13
|
Godbersen-Palmer C, Coupet TA, Grada Z, Zhang SC, Sentman CL. Toxicity Induced by a Bispecific T Cell-Redirecting Protein Is Mediated by Both T Cells and Myeloid Cells in Immunocompetent Mice. THE JOURNAL OF IMMUNOLOGY 2020; 204:2973-2983. [PMID: 32295875 DOI: 10.4049/jimmunol.1901401] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 03/23/2020] [Indexed: 12/21/2022]
Abstract
Bispecific T cell engagers have demonstrated clinical efficacy; however, their use can be accompanied by severe toxicity. Mechanistic understanding of these toxicities is limited by a lack of suitable immunocompetent preclinical models. In this study, we describe an immunocompetent mouse tumor model that exhibits bispecific T cell engager-induced toxicity and recapitulates key features similar to those in human cytokine release syndrome. In this study, toxicity occurred between the second and fourth injections of an NK Group 2D bispecific T cell engager protein. Symptoms were transient, peaking 3-4 h after treatment and resolving by 8 h. Mice developed weight loss, elevated plasma cytokines, a significant reduction in spleen white pulp, and lymphocyte infiltration in the liver. Systemic cellular immune changes also occurred; notably, an increase in CD8+ T cell activation, an increase in myeloid cells in the blood, and a population of Ly-6Cint monocytes (CD11b+Ly-6G-F4/80-) emerged in the liver and spleens of bispecific protein-treated mice. IFN-γ was primarily produced by CD8+ T cells in the spleen and was required for the observed changes in both T cell and myeloid populations. Rag deficiency, IFN-γ deficiency, or depletion of either CD4+ or CD8+ T cells prevented toxicity, whereas perforin deficiency, GM-CSF deficiency, or modulation of the myeloid population through clodronate-mediated depletion showed a partial abrogation of toxicity. Together, these findings reveal that T cell activation by a bispecific T cell engager leads to changes in the host myeloid cell population, both of which contribute to treatment induced toxicity in immunocompetent mice.
Collapse
Affiliation(s)
- Claire Godbersen-Palmer
- Center for Synthetic Immunity, Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756; and
| | - Tiffany A Coupet
- Center for Synthetic Immunity, Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756; and
| | - Zakaria Grada
- Department of Pathology, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756
| | - Samuel C Zhang
- Center for Synthetic Immunity, Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756; and
| | - Charles L Sentman
- Center for Synthetic Immunity, Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756; and
| |
Collapse
|
14
|
Harada A, Shioda A, Ikuno T, Iwata Y, Shiraiwa H, Wakabayashi T, Sano Y, Mishima M. In vitro toxicological support to establish specification limit for anti-CD3 monospecific impurity in a bispecific T cell engager drug, ERY974. Toxicol In Vitro 2020; 66:104841. [PMID: 32247040 DOI: 10.1016/j.tiv.2020.104841] [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: 08/01/2019] [Revised: 02/25/2020] [Accepted: 03/27/2020] [Indexed: 11/25/2022]
Abstract
An emerging structure for anti-tumor antibody drugs utilizes a bispecific antibody (BiAb) that recognizes a tumor surface antigen and CD3 on T cells. An impurity that commonly contaminates these BiAb products is an anti-CD3 monoclonal antibody (mAb). The most plausible cause of toxic activity by an anti-CD3 mAb is the induction of cytokines via T cell activation. In this in vitro study, we compared cytokine induction and T cell activation after treatment with an anti-glypican-3/CD3 BiAb (ERY974), anti-CD3 mAb impurity (aCD3), or ERY974 spiked with 5% aCD3. We found that contamination with up to 5% aCD3 did not affect cytokine release by ERY974. Cytokine levels induced by ERY974 in the presence of target cells were significantly higher than those induced by aCD3, but were very similar to those by the spiked treatment. The results supported the specification of a 5% limit for aCD3. OKT-3 had much higher activity to induce cytokines from peripheral blood mononuclear cells in an in vitro assay than aCD3. This suggests that specification limit should be decided for each type of anti-CD3 impurity that affects T cell-activating BiAb drug products. In vitro cytokine assays can provide useful information for determining these specification limits.
Collapse
Affiliation(s)
- Asako Harada
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka 412-8513, Japan.
| | - Akifumi Shioda
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka 412-8513, Japan
| | - Tatsuya Ikuno
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka 412-8513, Japan
| | - Yoshika Iwata
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka 412-8513, Japan
| | - Hirotake Shiraiwa
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka 412-8513, Japan
| | - Tetsuya Wakabayashi
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka 412-8513, Japan
| | - Yuji Sano
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka 412-8513, Japan
| | - Masayuki Mishima
- Translational Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka 412-8513, Japan
| |
Collapse
|
15
|
Pharmacokinetic/pharmacodynamic relationship of therapeutic monoclonal antibodies used in oncology: Part 1, monoclonal antibodies, antibody-drug conjugates and bispecific T-cell engagers. Eur J Cancer 2020; 128:107-118. [DOI: 10.1016/j.ejca.2020.01.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 01/02/2020] [Accepted: 01/07/2020] [Indexed: 01/31/2023]
|
16
|
Witkowski MT, Kousteni S, Aifantis I. Mapping and targeting of the leukemic microenvironment. J Exp Med 2020; 217:e20190589. [PMID: 31873722 PMCID: PMC7041707 DOI: 10.1084/jem.20190589] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/04/2019] [Accepted: 11/13/2019] [Indexed: 12/13/2022] Open
Abstract
Numerous studies support a role of the microenvironment in maintenance of the leukemic clone, as well as in treatment resistance. It is clear that disruption of the normal bone marrow microenvironment is sufficient to promote leukemic transformation and survival in both a cell autonomous and non-cell autonomous manner. In this review, we provide a snapshot of the various cell types shown to contribute to the leukemic microenvironment as well as treatment resistance. Several of these studies suggest that leukemic blasts occupy specific cellular and biochemical "niches." Effective dissection of critical leukemic niche components using single-cell approaches has allowed a more precise and extensive characterization of complexity that underpins both the healthy and malignant bone marrow microenvironment. Knowledge gained from these observations can have an important impact in the development of microenvironment-directed targeted approaches aimed at mitigating disease relapse.
Collapse
Affiliation(s)
- Matthew T. Witkowski
- Department of Pathology, New York University School of Medicine, New York, NY
- Laura & Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY
| | - Stavroula Kousteni
- Department of Physiology & Cellular Biophysics, Columbia University Irving Medical Center, New York, NY
| | - Iannis Aifantis
- Department of Pathology, New York University School of Medicine, New York, NY
- Laura & Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY
| |
Collapse
|
17
|
Dhakal P, Kaur J, Gundabolu K, Bhatt VR. Immunotherapeutic options for management of relapsed or refractory B-cell acute lymphoblastic leukemia: how to select newly approved agents? Leuk Lymphoma 2020; 61:7-17. [PMID: 31317803 PMCID: PMC7261514 DOI: 10.1080/10428194.2019.1641802] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 06/23/2019] [Accepted: 06/30/2019] [Indexed: 01/15/2023]
Abstract
Recently, immunotherapeutic agents such as inotuzumab ozogamicin (INO), blinatumomab (BLIN), and tisagenlecleucel (TISA) have been approved for treatment of relapsed or refractory (R/R) acute lymphoblastic leukemia (ALL). No head to head trials have compared these agents. Thus, various factors influence the decision to choose an appropriate treatment for R/R ALL. INO may be preferred in patients with high tumor burden; BLIN is preferred in patients with low tumor burden or to eradicate minimal residual disease (MRD). Both INO and BLIN, compared to standard chemotherapy, increase the probability of receiving subsequent hematopoietic stem cell transplant (HSCT). TISA, approved for patients ≤25 years of age, is effective regardless of tumor burden or prior receipt of HSCT and can be used as a definite treatment in some patients. Further studies comparing the efficacy, safety, and other outcomes related to different immunotherapeutic options in combination with other treatment modalities and among themselves are needed.
Collapse
Affiliation(s)
- Prajwal Dhakal
- Division of Oncology and Hematology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE
- Fred and Pamela Buffett Cancer Center, Omaha, NE
| | - Jasleen Kaur
- Department of Internal Medicine, Hurley Medical Center/ Michigan State University, Flint, Michigan, USA
| | - Krishna Gundabolu
- Division of Oncology and Hematology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE
- Fred and Pamela Buffett Cancer Center, Omaha, NE
| | - Vijaya Raj Bhatt
- Division of Oncology and Hematology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE
- Fred and Pamela Buffett Cancer Center, Omaha, NE
| |
Collapse
|
18
|
Ali S, Moreau A, Melchiorri D, Camarero J, Josephson F, Olimpier O, Bergh J, Karres D, Tzogani K, Gisselbrecht C, Pignatti F. Blinatumomab for Acute Lymphoblastic Leukemia: The First Bispecific T-Cell Engager Antibody to Be Approved by the EMA for Minimal Residual Disease. Oncologist 2019; 25:e709-e715. [PMID: 32297447 DOI: 10.1634/theoncologist.2019-0559] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 09/24/2019] [Indexed: 01/26/2023] Open
Abstract
On November 15, 2018, the Committee for Medicinal Products for Human Use (CHMP) recommended the extension of indication for blinatumomab to include the treatment of adults with minimal residual disease (MRD) positive B-cell precursor acute lymphoblastic leukemia (ALL). Blinatumomab was authorized to treat relapsed or refractory B-precursor ALL, and the change concerned an extension of use. On March 29, 2018, the U.S. Food and Drug Administration (FDA) granted accelerated approval to blinatumomab to treat both adults and children with B-cell precursor ALL who are in remission but still have MRD. On July 26, 2018, the CHMP had originally adopted a negative opinion on the extension. The reason for the initial refusal was that although blinatumomab helped to reduce the amount of residual cancer cells in many patients, there was no strong evidence that it led to improved survival. During the re-examination, the CHMP consulted the scientific advisory group. The CHMP agreed with the expert group's conclusion that, although there was no strong evidence of patients living longer, the available data from the main study (MT103-203) indicated a good durable response to blinatumomab, with an overall complete response rate for the primary endpoint full analysis set (defined as all subjects with an Ig or T-cell receptor polymerase chain reaction MRD assay with the minimum required sensitivity of 1 × 10-4 at central lab established at baseline [n = 113]) as 79.6% (90/113; 95% confidence interval, 71.0-86.6), with a median time to complete MRD response of 29.0 days (range, 5-71). Therefore, the CHMP concluded that the benefits of blinatumomab outweigh its risks and recommended granting the change to the marketing authorization. The Committee for Orphan Medicinal Products, following reassessment, considered that significant benefit continued to be met and recommended maintaining the orphan designation and thus 10 years market exclusivity (the Orphan Designation is a legal procedure that allows for the designation of a medicinal substance with therapeutic potential for a rare disease, before its first administration in humans or during its clinical development). The marketing authorization holder for this medicinal product is Amgen Europe B.V. IMPLICATIONS FOR PRACTICE: Immunotherapy with blinatumomab has excellent and sustainable results, offering new hope for patients with minimal residual disease-positive acute lymphoblastic leukemia, a disease with poor prognosis. New recommendations and change of practice for treatment of this patient group are detailed.
Collapse
Affiliation(s)
- Sahra Ali
- European Medicines Agency, Amsterdam, The Netherlands
| | - Alexandre Moreau
- French National Agency for Medicines and Health Products Safety, Saint-Denis Cedex, France
| | - Daniela Melchiorri
- Department of Physiology and Pharmacology, University of Rome, Sapienza, Rome, Italy
| | | | - Filip Josephson
- Medical Products Agency, Department of Efficacy and Safety 3, Uppsala, Sweden
| | | | - Jonas Bergh
- Department of Oncology-Pathology, Karolinska Institutet, BES, Cancer Theme, Karolinska University Hospital Bioclinicum, Solna, Sweden
| | | | | | | | | |
Collapse
|
19
|
Witkowski MT, Lasry A, Carroll WL, Aifantis I. Immune-Based Therapies in Acute Leukemia. Trends Cancer 2019; 5:604-618. [PMID: 31706508 PMCID: PMC6859901 DOI: 10.1016/j.trecan.2019.07.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 07/24/2019] [Accepted: 07/26/2019] [Indexed: 12/31/2022]
Abstract
Treatment resistance remains a leading cause of acute leukemia-related deaths. Thus, there is an unmet need to develop novel approaches to improve outcome. New immune-based therapies with chimeric antigen receptor (CAR) T cells, bi-specific T cell engagers (BiTEs), and immune checkpoint blockers (ICBs) have emerged as effective treatment options for chemoresistant B cell acute lymphoblastic leukemia (B-ALL) and acute myeloid leukemia (AML). However, many patients show resistance to these immune-based approaches. This review describes crucial lessons learned from immune-based approaches targeting high-risk B-ALL and AML, such as the leukemia-intrinsic (e.g., target antigen loss, tumor heterogeneity) and -extrinsic (e.g., immunosuppressive microenvironment) mechanisms that drive treatment resistance, and discusses alternative approaches to enhance the effectiveness of these immune-based treatment regimens.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- Disease Susceptibility
- Humans
- Immunity
- Immunotherapy/methods
- Immunotherapy, Adoptive
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/therapy
- Molecular Targeted Therapy
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Matthew T Witkowski
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA.
| | - Audrey Lasry
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA
| | - William L Carroll
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA; Department of Pediatrics, New York University School of Medicine, New York, NY 10016, USA
| | - Iannis Aifantis
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA
| |
Collapse
|
20
|
Strohl WR, Naso M. Bispecific T-Cell Redirection versus Chimeric Antigen Receptor (CAR)-T Cells as Approaches to Kill Cancer Cells. Antibodies (Basel) 2019; 8:E41. [PMID: 31544847 PMCID: PMC6784091 DOI: 10.3390/antib8030041] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/23/2019] [Accepted: 06/24/2019] [Indexed: 12/16/2022] Open
Abstract
The concepts for T-cell redirecting bispecific antibodies (TRBAs) and chimeric antigen receptor (CAR)-T cells are both at least 30 years old but both platforms are just now coming into age. Two TRBAs and two CAR-T cell products have been approved by major regulatory agencies within the last ten years for the treatment of hematological cancers and an additional 53 TRBAs and 246 CAR cell constructs are in clinical trials today. Two major groups of TRBAs include small, short-half-life bispecific antibodies that include bispecific T-cell engagers (BiTE®s) which require continuous dosing and larger, mostly IgG-like bispecific antibodies with extended pharmacokinetics that can be dosed infrequently. Most CAR-T cells today are autologous, although significant strides are being made to develop off-the-shelf, allogeneic CAR-based products. CAR-Ts form a cytolytic synapse with target cells that is very different from the classical immune synapse both physically and mechanistically, whereas the TRBA-induced synapse is similar to the classic immune synapse. Both TRBAs and CAR-T cells are highly efficacious in clinical trials but both also present safety concerns, particularly with cytokine release syndrome and neurotoxicity. New formats and dosing paradigms for TRBAs and CAR-T cells are being developed in efforts to maximize efficacy and minimize toxicity, as well as to optimize use with both solid and hematologic tumors, both of which present significant challenges such as target heterogeneity and the immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- William R Strohl
- BiStro Biotech Consulting, LLC, 1086 Tullo Farm Rd., Bridgewater, NJ 08807, USA.
| | - Michael Naso
- Century Therapeutics, 3675 Market St., Philadelphia, PA 19104, USA
| |
Collapse
|
21
|
Exposure-adjusted adverse events comparing blinatumomab with chemotherapy in advanced acute lymphoblastic leukemia. Blood Adv 2019; 2:1522-1531. [PMID: 29954814 DOI: 10.1182/bloodadvances.2018019034] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 05/25/2018] [Indexed: 11/20/2022] Open
Abstract
In the phase 3 TOWER study, blinatumomab demonstrated an overall survival benefit over standard-of-care chemotherapy (SOC) in adults with relapsed or refractory (r/r) Philadelphia chromosome-negative (Ph-) B-precursor acute lymphoblastic leukemia (ALL). Nearly all patients in both treatment arms experienced an adverse event (AE), and the incidence rate of serious AEs was higher for blinatumomab. However, as treatment exposure differed between the 2 arms, we conducted an exploratory safety analysis comparing exposure-adjusted event rates (EAERs) of blinatumomab vs SOC. Analyses were conducted for all patients who received therapy (safety population). Patients received a median (range) of 2 cycles (1-9) of blinatumomab (N = 267) vs 1 cycle (1-4) of SOC (N = 109). Grade ≥3 AE rates were generally higher in cycle 1 of blinatumomab than in cycle 2 (76% vs 37%). After adjusting for time on treatment, EAERs of grade ≥3 were significantly lower for blinatumomab vs SOC overall (10.73 vs 45.27 events per patient-year; P < .001) and for events of clinical interest, including infections (1.63 vs 6.49 events per patient-year; P < .001), cytopenias (3.64 vs 20.07 events per patient-year; P < .001), and neurologic events (0.38 vs 0.95 events per patient-year; P = .008). The EAER of grade ≥3 cytokine-release syndrome was higher for blinatumomab than for SOC (0.16 vs 0 events per patient-year; P = .038). These data further support the role of blinatumomab as an efficacious and well-tolerated treatment option for patients with r/r Ph- ALL. This trial was registered at www.clinicaltrials.gov as #NCT02013167.
Collapse
|
22
|
Schlake T, Thess A, Thran M, Jordan I. mRNA as novel technology for passive immunotherapy. Cell Mol Life Sci 2019; 76:301-328. [PMID: 30334070 PMCID: PMC6339677 DOI: 10.1007/s00018-018-2935-4] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 09/13/2018] [Accepted: 10/03/2018] [Indexed: 12/17/2022]
Abstract
While active immunization elicits a lasting immune response by the body, passive immunotherapy transiently equips the body with exogenously generated immunological effectors in the form of either target-specific antibodies or lymphocytes functionalized with target-specific receptors. In either case, administration or expression of recombinant proteins plays a fundamental role. mRNA prepared by in vitro transcription (IVT) is increasingly appreciated as a drug substance for delivery of recombinant proteins. With its biological role as transient carrier of genetic information translated into protein in the cytoplasm, therapeutic application of mRNA combines several advantages. For example, compared to transfected DNA, mRNA harbors inherent safety features. It is not associated with the risk of inducing genomic changes and potential adverse effects are only temporary due to its transient nature. Compared to the administration of recombinant proteins produced in bioreactors, mRNA allows supplying proteins that are difficult to manufacture and offers extended pharmacokinetics for short-lived proteins. Based on great progress in understanding and manipulating mRNA properties, efficacy data in various models have now demonstrated that IVT mRNA constitutes a potent and flexible platform technology. Starting with an introduction into passive immunotherapy, this review summarizes the current status of IVT mRNA technology and its application to such immunological interventions.
Collapse
Affiliation(s)
- Thomas Schlake
- CureVac AG, Paul-Ehrlich-Str. 15, 72076, Tübingen, Germany.
| | - Andreas Thess
- CureVac AG, Paul-Ehrlich-Str. 15, 72076, Tübingen, Germany
| | - Moritz Thran
- CureVac AG, Paul-Ehrlich-Str. 15, 72076, Tübingen, Germany
| | - Ingo Jordan
- CureVac AG, Paul-Ehrlich-Str. 15, 72076, Tübingen, Germany
| |
Collapse
|
23
|
Naddafi F, Mahboudi F, Tabarzad M, Aliabadi Farahani Z, Hosein Shirazi F, Davami F. The Epigenetic Regulation of Blinatumomab Gene Expression: Tumor Cell-dependent T cell Response against Lymphoma Cells and Cytotoxic Activity. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2019; 8:55-66. [PMID: 32195205 PMCID: PMC7073260 DOI: 10.22088/ijmcm.bums.8.1.55] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 06/03/2019] [Indexed: 11/24/2022]
Abstract
Conventional treatment for cancer such as surgical resection and chemotherapy can cause damage in cases with advanced cancers. Moreover, the identification of tumor-specific targets has great importance in T-cell therapies. For decades, T cell activity has been stimulated to improve anti-tumor activity. Bispecific antibodies have attracted strong interest from pharmaceutical companies, for their diagnostic and therapeutic use. Blinatumomab is a first-in-class bispecific T engager antibody for the treatment of relapsed or refractory precursor B- cell acute lymphoblastic leukemia. But, it can benefit several cases with CD19+ malignancies in the future. PhiC31 integrase-based vectors could selectively integrate therapeutic transgenes into pseudo-attP sites in CHO genome. In this study, production of Blinatumomab in CHO cells using this type of vectors was investigated. We evaluated the effects of histone deacetylases (HDACs) inhibitors such as sodium butyrate and valproic acid, on specific productivity and cell viability of antibody expressing cells. Although sodium butyrate increased specific productivity about 1.7-fold and valproic acid about 1.4-fold, valproic acid was found more efficient because of its less cytotoxic effect on cell growth. We examined the efficacy of expressed Blinatumomab at various effector to target (E/T) ratios. A dose-response analyses of calcein-acetoxymethyl release assay illustrated that the effective dose of expressed mAb required for antibody mediated cytotoxicity was 100 ng/ml and the expressed mAb was more effective at E/T ratios of 10:1 and 5:1. Results of this study indicated that the expressed blinatumomab can be useful for enhancing the cytotoxicity of CD3+ T-cells against CD19 + target cells in vitro.
Collapse
Affiliation(s)
- Fatemeh Naddafi
- Pharmaceutical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Maryam Tabarzad
- Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Farshad Hosein Shirazi
- Pharmaceutical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Davami
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
24
|
Ma J, Liu T, Jin J, Hu J, Liu Q, Wang J, Shen Z, Du X, Jiang B, Meng X. An observational study of Chinese adults with relapsed/refractory Philadelphia-negative acute lymphoblastic leukemia. Int J Hematol Oncol 2018; 7:IJH06. [PMID: 30405901 PMCID: PMC6219430 DOI: 10.2217/ijh-2018-0006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 06/27/2018] [Indexed: 02/05/2023] Open
Abstract
Aim: Chinese adults with relapsed/refractory Philadelphia chromosome-negative B-cell precursor acute lymphoblastic leukemia (Ph- ALL) have poor outcomes. Patients & methods: We conducted a nationwide, retrospective, observational study to assess outcomes in this patient population. Results: Of the 270 enrolled patients, 31% of patients at last salvage achieved complete remission (CR) or CR with partial hematologic recovery (CRh), with median time to CR/CRh of 30 days and median CR/CRh duration of 2.7 months. The CR/CRh rate was more favorable with earlier versus later lines of salvage (41, 24 and 17% at first, second and third or later salvages, respectively). Conclusion: This dataset serves as an important reference of real-world outcomes using currently available chemotherapy regimens for high-risk Chinese adults with relapsed/refractory Ph- ALL.
Collapse
Affiliation(s)
- Jun Ma
- Department of Hematology, Harbin Institute of Hematology & Oncology, Harbin, China.,Department of Hematology, Harbin Institute of Hematology & Oncology, Harbin, China
| | - Ting Liu
- Department of Hematology, West China Hospital of Sichuan University, Chengdu, China.,Department of Hematology, West China Hospital of Sichuan University, Chengdu, China
| | - Jie Jin
- Department of Hematology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China.,Department of Hematology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Jianda Hu
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, China.,Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Qifa Liu
- Department of Hematology, Southern Medical University Nanfang Hospital, Guangzhou, China.,Department of Hematology, Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Jianxiang Wang
- Department of Hematology, Institute of Hematology, Chinese Academy of Medical Sciences, Tianjin, China.,Department of Hematology, Institute of Hematology, Chinese Academy of Medical Sciences, Tianjin, China
| | - Zhixiang Shen
- Department of Hematology, Shanghai Ruijin Hospital, Shanghai, China.,Department of Hematology, Shanghai Ruijin Hospital, Shanghai, China
| | - Xin Du
- Department of Hematology, Guangdong General Hospital/Guangdong Academy of Medical Sciences, Guangzhou, China.,Department of Hematology, Guangdong General Hospital/Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Bin Jiang
- Department of Hematology, Peking University People's Hospital, Beijing, China.,Department of Hematology, Peking University People's Hospital, Beijing, China
| | - Xianhua Meng
- Department of Biostatistical Science, Amgen R&D Asia, Shanghai, China.,Boehringer Ingelheim (China) Investment Co., Ltd, Shanghai, China.,Department of Biostatistical Science, Amgen R&D Asia, Shanghai, China.,Boehringer Ingelheim (China) Investment Co., Ltd, Shanghai, China
| |
Collapse
|
25
|
Abstract
Bispecific antibodies have moved from being an academic curiosity with therapeutic promise to reality, with two molecules being currently commercialized (Hemlibra® and Blincyto®) and many more in clinical trials. The success of bispecific antibodies is mainly due to the continuously growing number of mechanisms of actions (MOA) they enable that are not accessible to monoclonal antibodies. One of the earliest MOA of bispecific antibodies and currently the one with the largest number of clinical trials is the redirecting of the cytotoxic activity of T-cells for oncology applications, now extending its use in infective diseases. The use of bispecific antibodies for crossing the blood-brain barrier is another important application because of its potential to advance the therapeutic options for neurological diseases. Another noteworthy application due to its growing trend is enabling a more tissue-specific delivery or activity of antibodies. The different molecular solutions to the initial hurdles that limited the development of bispecific antibodies have led to the current diverse set of bispecific or multispecific antibody formats that can be grouped into three main categories: IgG-like formats, antibody fragment-based formats, or appended IgG formats. The expanded applications of bispecific antibodies come at the price of additional challenges for clinical development. The rising complexity in their structure may increase the risk of immunogenicity and the multiple antigen specificity complicates the selection of relevant species for safety assessment.
Collapse
Affiliation(s)
- Bushra Husain
- Protein Chemistry Department, Genentech Inc., South San Francisco, CA, 94080, USA
| | - Diego Ellerman
- Protein Chemistry Department, Genentech Inc., South San Francisco, CA, 94080, USA.
| |
Collapse
|
26
|
Hickey JW, Kosmides AK, Schneck JP. Engineering Platforms for T Cell Modulation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 341:277-362. [PMID: 30262034 DOI: 10.1016/bs.ircmb.2018.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
T cells are crucial contributors to mounting an effective immune response and increasingly the focus of therapeutic interventions in cancer, infectious disease, and autoimmunity. Translation of current T cell immunotherapies has been hindered by off-target toxicities, limited efficacy, biological variability, and high costs. As T cell therapeutics continue to develop, the application of engineering concepts to control their delivery and presentation will be critical for their success. Here, we outline the engineer's toolbox and contextualize it with the biology of T cells. We focus on the design principles of T cell modulation platforms regarding size, shape, material, and ligand choice. Furthermore, we review how application of these design principles has already impacted T cell immunotherapies and our understanding of T cell biology. Recent, salient examples from protein engineering, synthetic particles, cellular and genetic engineering, and scaffolds and surfaces are provided to reinforce the importance of design considerations. Our aim is to provide a guide for immunologists, engineers, clinicians, and the pharmaceutical sector for the design of T cell-targeting platforms.
Collapse
Affiliation(s)
- John W Hickey
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Institute for NanoBiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Alyssa K Kosmides
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Institute for NanoBiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jonathan P Schneck
- Institute for NanoBiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
27
|
Early Fever after Haploidentical Bone Marrow Transplantation Correlates with Class II HLA-Mismatching and Myeloablation but Not Outcomes. Biol Blood Marrow Transplant 2018; 24:2056-2064. [PMID: 29909152 DOI: 10.1016/j.bbmt.2018.06.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 06/05/2018] [Indexed: 12/12/2022]
Abstract
Noninfectious fevers are common early after T cell-replete HLA haploidentical (haplo) peripheral blood transplants and have been associated with cytokine release syndrome and overall mortality. However, less is known regarding the incidence and associations of early fever after bone marrow transplantation (BMT) with post-transplant cyclophosphamide (PTCy). We hypothesized that early fever would be associated with myeloablative conditioning (MAC), because of its relative increase in tissue damage augmenting antigen presentation and class II HLA-mismatching because of recognition of antigen-presenting cells by CD4+ T cells. In 672 recipients of MAC HLA-matched related donor (MRD) (n = 183), MAC HLA-matched unrelated donor (MUD) (n = 115), MAC haplo (n = 79), or nonmyeloablative (NMA) haplo (n = 295) T cell-replete BMT with PTCy, we retrospectively analyzed early noninfectious fever defined as temperature of ≥38.3°C once or ≥38.0°C twice or more on days 1 to 6. Fever occurred in 13% after MAC MRD, 23% after MAC MUD, 44% after NMA haplo, and 84% after MAC haplo BMT (P < .0001). Survival outcomes did not differ between patients with and without early fever. In NMA haplo BMT, mismatch in the graft-versus-host direction at HLA-DRB1 or -DPB1 (but not HLA-A, -B, -Cw, or -DQB1) was associated with early fever compared with no mismatches at these loci (P < .0001 and P = .02, respectively). In multivariable modeling, -DRB1 or -DPB1 mismatch and higher CD3+ graft cell dose were significantly associated with early fever. Early fever is more common after haplo compared with HLA-matched BMT. Fever is associated with myeloablation, -DRB1 or -DPB1 mismatching, and higher CD3+ graft cell dose but not survival.
Collapse
|
28
|
Chen P, Liu Y, Zhao J, Pang X, Zhang P, Hou X, Chen P, He CY, Wang Z, Chen ZY. The synthesis of amphiphilic polyethyleneimine/calcium phosphate composites for bispecific T-cell engager based immunogene therapy. Biomater Sci 2018; 6:633-641. [DOI: 10.1039/c7bm01143a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Bispecific T-cell engagers (BiTEs) are single chain variable fragments, which could connect the surface antigen on cancer cells and CD3 ligands on T cells, and then engage the T cells for cancer immunotherapy.
Collapse
Affiliation(s)
- Pingzhang Chen
- Sun Yat-sen University
- Guangzhou 510275
- P. R. China
- Shenzhen College of Advanced Technology
- University of Chinese Academy of Sciences
| | - Yunhong Liu
- Department of Clinical Laboratory
- The People's Hospital of Longhua
- Shenzhen
- China
| | - Jing Zhao
- Sun Yat-sen University
- Guangzhou 510275
- P. R. China
| | | | - Peifa Zhang
- Sun Yat-sen University
- Guangzhou 510275
- P. R. China
| | - Xiaohu Hou
- Sun Yat-sen University
- Guangzhou 510275
- P. R. China
| | - Ping Chen
- Sun Yat-sen University
- Guangzhou 510275
- P. R. China
| | - Cheng-yi He
- Sun Yat-sen University
- Guangzhou 510275
- P. R. China
| | - Zhiyong Wang
- Sun Yat-sen University
- Guangzhou 510275
- P. R. China
- School of Materials Science and Engineering
- Sun Yat-sen University
| | - Zhi-ying Chen
- Sun Yat-sen University
- Guangzhou 510275
- P. R. China
- Shenzhen College of Advanced Technology
- University of Chinese Academy of Sciences
| |
Collapse
|
29
|
Abstract
As of May 1, 2017, 74 antibody-based molecules have been approved by a regulatory authority in a major market. Additionally, there are 70 and 575 antibody-based molecules in phase III and phase I/II clinical trials, respectively. These total 719 antibody-based clinical stage molecules include 493 naked IgGs, 87 antibody-drug conjugates, 61 bispecific antibodies, 37 total Fc fusion proteins, 17 radioimmunoglobulins, 13 antibody fragments, and 11 immunocytokines. New uses for these antibodies are being discovered each year. For oncology, many of the exciting new approaches involve antibody modulation of T-cells. There are over 80 antibodies in clinical trials targeting T cell checkpoints, 26 T-cell-redirected bispecific antibodies, and 145 chimeric antigen receptor (CAR) cell-based candidates (all currently in phase I or II clinical trials), totaling more than 250 T cell interacting clinical stage antibody-based candidates. Finally, significant progress has been made recently on routes of delivery, including delivery of proteins across the blood-brain barrier, oral delivery to the gut, delivery to the cellular cytosol, and gene- and viral-based delivery of antibodies. Thus, there are currently at least 864 antibody-based clinical stage molecules or cells, with incredible diversity in how they are constructed and what activities they impart. These are followed by a next wave of novel molecules, approaches, and new methods and routes of delivery, demonstrating that the field of antibody-based biologics is very innovative and diverse in its approaches to fulfill their promise to treat unmet medical needs.
Collapse
|
30
|
Goulet DR, Orcutt SJ, Zwolak A, Rispens T, Labrijn AF, de Jong RN, Atkins WM, Chiu ML. Kinetic mechanism of controlled Fab-arm exchange for the formation of bispecific immunoglobulin G1 antibodies. J Biol Chem 2017; 293:651-661. [PMID: 29150443 PMCID: PMC5767869 DOI: 10.1074/jbc.ra117.000303] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Indexed: 01/04/2023] Open
Abstract
Bispecific antibodies (bsAbs) combine the antigen specificities of two distinct Abs and demonstrate therapeutic promise based on novel mechanisms of action. Among the many platforms for creating bsAbs, controlled Fab-arm exchange (cFAE) has proven useful based on minimal changes to native Ab structure and the simplicity with which bsAbs can be formed from two parental Abs. Despite a published protocol for cFAE and its widespread use in the pharmaceutical industry, the reaction mechanism has not been determined. Knowledge of the mechanism could lead to improved yields of bsAb at faster rates as well as foster adoption of process control. In this work, a combination of Förster resonance energy transfer (FRET), nonreducing SDS-PAGE, and strategic mutation of the Ab hinge region was employed to identify and characterize the individual steps of cFAE. Fluorescence correlation spectroscopy (FCS) was used to determine the affinity of parental (homodimer) and bispecific (heterodimer) interactions within the CH3 domain, further clarifying the thermodynamic basis for bsAb formation. The result is a clear sequence of events with rate constants that vary with experimental conditions, where dissociation of the K409R parental Ab into half-Ab controls the rate of the reaction.
Collapse
Affiliation(s)
- Dennis R Goulet
- From the Department of Medicinal Chemistry, University of Washington, Seattle, Washington 98195
| | - Steven J Orcutt
- Biologics Discovery, Janssen Research & Development, LLC, Spring House, Pennsylvania 19477
| | - Adam Zwolak
- Biologics Discovery, Janssen Research & Development, LLC, Spring House, Pennsylvania 19477
| | - Theo Rispens
- the Sanquin Research and Landsteiner Laboratory, Department of Immunopathology, Academic Medical Centre, University of Amsterdam, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands, and
| | | | - Rob N de Jong
- Genmab, Yalelaan 60, 3584 CM Utrecht, The Netherlands
| | - William M Atkins
- From the Department of Medicinal Chemistry, University of Washington, Seattle, Washington 98195
| | - Mark L Chiu
- Biologics Discovery, Janssen Research & Development, LLC, Spring House, Pennsylvania 19477,
| |
Collapse
|
31
|
Ribera JM. Efficacy and safety of bispecific T-cell engager blinatumomab and the potential to improve leukemia-free survival in B-cell acute lymphoblastic leukemia. Expert Rev Hematol 2017; 10:1057-1067. [PMID: 29082835 DOI: 10.1080/17474086.2017.1396890] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Immunotherapy is a promising modality of treatment of neoplastic diseases, including acute lymphoblastic leukemia (ALL). The CD19/CD3-bispecific T cell-engaging (BiTE®) monoclonal antibody blinatumomab can transiently bind cytotoxic T cells to CD19+ target B cells of ALL inducing their serial lysis. Areas covered: This review focuses on the efficacy and safety of blinatumomab used for the treatment of relapsed/refractory (R/R) ALL and minimal residual disease (MRD)-positive B-cell precursor (BCP) ALL in adults and children, as well as the future prospects of this drug in the treatment of ALL. Expert commentary: Blinatumomab has demonstrated encouraging response rates in MRD-positive and R/R in adults with Philadelphia chromosome-positive and -negative ALL, as well as in children with R/R ALL. Blinatumomab has a favorable safety profile, although reversible CNS events and cytokine release syndrome can occur. Ongoing trials in ALL incorporate blinatumomab in the first line therapy of BCP ALL in combination with chemotherapy, targeted therapies or other immunotherapies with the aim of increasing the depth of the remission and decreasing the probability of relapse.
Collapse
Affiliation(s)
- Josep-Maria Ribera
- a Clinical Hematology Department, ICO-Hospital Germans Trias i Pujol, Jose Carreras Research Institute, Badalona , Universitat Autonoma de Barcelona , Spain
| |
Collapse
|
32
|
Horn LA, Ciavattone NG, Atkinson R, Woldergerima N, Wolf J, Clements VK, Sinha P, Poudel M, Ostrand-Rosenberg S. CD3xPDL1 bi-specific T cell engager (BiTE) simultaneously activates T cells and NKT cells, kills PDL1 + tumor cells, and extends the survival of tumor-bearing humanized mice. Oncotarget 2017; 8:57964-57980. [PMID: 28938530 PMCID: PMC5601626 DOI: 10.18632/oncotarget.19865] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 07/26/2017] [Indexed: 02/06/2023] Open
Abstract
Bi-specific T cell engagers (BiTEs) activate T cells through CD3 and target activated T cells to tumor-expressed antigens. BiTEs have shown therapeutic efficacy in patients with liquid tumors; however, they do not benefit all patients. Anti-tumor immunity is limited by Programmed Death 1 (PD1) pathway-mediated immune suppression, and patients who do not benefit from existing BiTES may be non-responders because their T cells are anergized via the PD1 pathway. We have designed a BiTE that activates and targets both T cells and NKT cells to PDL1+ cells. In vitro studies demonstrate that the CD3xPDL1 BiTE simultaneously binds to both CD3 and PDL1, and activates healthy donor CD4+ and CD8+ T cells and NKT cells that are specifically cytotoxic for PDL1+ tumor cells. Cancer patients’ PBMC are also activated and cytotoxic, despite the presence of myeloid-derived suppressor cells. The CD3xPDL1 BiTE significantly extends the survival time and maintains activated immune cell levels in humanized NSG mice reconstituted with human PBMC and carrying established human melanoma tumors. These studies suggest that the CD3xPDL1 BiTE may be efficacious for patients with PDL1+ solid tumors, in combination with other immunotherapies that do not specifically neutralize PD1 pathway-mediated immune suppression.
Collapse
Affiliation(s)
- Lucas A Horn
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD, USA
| | - Nicholas G Ciavattone
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, USA
| | - Ryan Atkinson
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD, USA
| | - Netsanet Woldergerima
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD, USA
| | - Julia Wolf
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD, USA
| | - Virginia K Clements
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD, USA
| | - Pratima Sinha
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD, USA
| | - Munanchu Poudel
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD, USA
| | | |
Collapse
|