1
|
Messaoudi S, Wai K, Marple A, Baniahmad SF, Wylie RG, Pelletier M, Craig M, Durocher Y, Greschner AA, Gauthier MA. Rapid Systematic Screening of Bispecific Antibody Surrogate Geometries for T-Cell Engagement Using DNA Nanotechnology. J Am Chem Soc 2024; 146:29824-29835. [PMID: 39412838 PMCID: PMC11529601 DOI: 10.1021/jacs.4c11648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 11/01/2024]
Abstract
Bispecific antibodies (bsAbs) are emerging immune-therapeutics, and many formats exist that differ considerably in structure. However, little systematic data exist about how the spatial organization of their components influences activity, requiring innovative approaches combining empirical and quantitative frameworks. This study presents a modular DNA nanotechnology platform to generate numerous bsAbs with surrogate geometries that span the structural features of the BiTE, IgG-like, and IgG-conjugate platforms to screen for T-cell engagement. Results highlight interesting structure-activity relationships regarding bsAb potency and selectivity and raise questions regarding the molecular phenomena underlying activity. To elucidate some effects, the platform was paired with a simple mathematical model. This work is thus one of the first to systematically investigate and reveal the importance of the spatial organization of bsAb components on activity and equally provides an accessible and convenient tool for rapidly mapping out such trends for other combinations of target epitopes.
Collapse
Affiliation(s)
- Sabrine Messaoudi
- Institut
National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, Quebec J3X 1P7, Canada
| | - Kevin Wai
- Department
of Chemistry and Chemical Biology, McMaster
University, Hamilton, Ontario L8S 4M1, Canada
| | - April Marple
- Department
of Chemistry and Chemical Biology, McMaster
University, Hamilton, Ontario L8S 4M1, Canada
| | - Seyed Farzad Baniahmad
- Human
Health Therapeutics Research Centre, National
Research Council Canada, Montreal, Quebec H4P 2R2, Canada
| | - Ryan G. Wylie
- Department
of Chemistry and Chemical Biology, McMaster
University, Hamilton, Ontario L8S 4M1, Canada
| | - Marianne Pelletier
- Sainte-Justine
University Hospital Azrieli Research Centre, Montreal, Quebec H3T 1C5, Canada
- Département
de Mathématiques et de Statistique, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Morgan Craig
- Sainte-Justine
University Hospital Azrieli Research Centre, Montreal, Quebec H3T 1C5, Canada
- Département
de Mathématiques et de Statistique, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Yves Durocher
- Human
Health Therapeutics Research Centre, National
Research Council Canada, Montreal, Quebec H4P 2R2, Canada
| | - Andrea A. Greschner
- Institut
National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, Quebec J3X 1P7, Canada
| | - Marc A. Gauthier
- Institut
National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, Quebec J3X 1P7, Canada
| |
Collapse
|
2
|
Leotta S, Markovic U, Duminuco A, Mulè A, Porretto F, Federico V, Gentile M, Pastore D, Nigro LL, Selleri C, Serio B, Calafiore V, Patti C, Mauro E, Vetro C, Maugeri C, Parisi M, Fiumara P, Parrinello L, Marino S, Scuderi G, Garibaldi B, Musso M, Renzo ND, Vigna E, Martino EA, Raimondo FD, Milone G. Impact of minimal residual disease response and of status of disease on survival after blinatumomab in B-cell acute lymphoblastic leukemia: results from a real-life study. Ann Hematol 2024; 103:3701-3712. [PMID: 38609726 DOI: 10.1007/s00277-024-05725-9] [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: 02/25/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024]
Abstract
Blinatumomab is a bispecific T-cell engager approved for relapsed/refractory and minimal residual disease positive B-cell Acute Lymphoblastic Leukemia. We conducted a retrospective study evaluating the outcome of Blinatumomab. The impact of clinical and treatment-related variables on cumulative incidence of relapse/progression (CIRP), event-free (EFS) and overall survival (OS) was analyzed. From January 2016 to December 2022 50 Ph'- (37) and Ph+ (13) B-ALL patients received Blinatumomab. The median age was 37. Indications to blinatumomab were relapsed/refractory B-ALL in 29 and MRD-positive in 21 patients. Blinatumomab was the 2nd and 3rd line in 40 and in 10 patients, respectively. Twenty patients were treated pre-transplantation, ten were treated for relapse after transplant, twenty were not eligible for transplant. Out of 29 patients treated for relapsed/refractory disease, 16 (55%) achieved complete response and 12 achieved MRD-negativity. Out of 21 patients treated for MRD, 16 (76%) achieved MRD-negativity. At a median follow-up of 46 months the median EFS and OS were 11.5 and 16.2 months. The CIRP was 50%. In univariate analysis age, disease-status (overt vs. minimal disease) at blinatumomab, bridging to transplant after blinatumomab and MRD-response resulted significant for EFS and OS. In multivariate analysis only disease-status and MRD-response retained significance both for EFS and OS. Disease-status and MRD-response resulted significant for EFS and OS also after censoring at HSCT. This retrospective study on B-ALL patients treated with blinatumomab confirms a superior outcome for MRD-responsive over MRD non-responsive patients. Survival depends also on the disease-status prior treatment.
Collapse
Affiliation(s)
- Salvatore Leotta
- Hematology and BMT Unit, Azienda Ospedaliero Universitaria Policlinico "G.Rodolico-San Marco", Catania, Italy.
- Divisione di Ematologia - Azienda Ospedaliero, Universitaria Policlinico "G. Rodolico - San Marco", Via Santa Sofia, 78, Catania, 95123, Italy.
| | - Uros Markovic
- Hematology and BMT Unit, Azienda Ospedaliero Universitaria Policlinico "G.Rodolico-San Marco", Catania, Italy
| | - Andrea Duminuco
- Hematology and BMT Unit, Azienda Ospedaliero Universitaria Policlinico "G.Rodolico-San Marco", Catania, Italy
| | - Antonino Mulè
- Oncohematology Unit - AO Villa Sofia - Cervello, Palermo, Italy
| | | | - Vincenzo Federico
- Haematology and Stem Cell Transplant Unit, Presidio Ospedaliero "Vito Fazzi", Lecce, Italy
| | - Massimo Gentile
- Hematology Unit, Azienda Ospedaliera Annunziata, Cosenza, Italy
- Department of Pharmacy, Health and Nutritional Science, University of Calabria, Rende, Italy
| | | | - Luca Lo Nigro
- Pediatric Hematology-Oncology, Azienda Policlinico "G. Rodolico-San Marco", Catania, Italy
| | - Carmine Selleri
- Oncohematology Department and Transplant Center, University of Salerno - AOU San Giovanni di Dio e Ruggi d'Aragona, Salerno, Italy
| | - Bianca Serio
- Oncohematology Department and Transplant Center, University of Salerno - AOU San Giovanni di Dio e Ruggi d'Aragona, Salerno, Italy
| | | | - Caterina Patti
- Oncohematology Unit - AO Villa Sofia - Cervello, Palermo, Italy
| | - Elisa Mauro
- Hematology and BMT Unit, Azienda Ospedaliero Universitaria Policlinico "G.Rodolico-San Marco", Catania, Italy
| | - Calogero Vetro
- Hematology and BMT Unit, Azienda Ospedaliero Universitaria Policlinico "G.Rodolico-San Marco", Catania, Italy
| | - Cinzia Maugeri
- Hematology and BMT Unit, Azienda Ospedaliero Universitaria Policlinico "G.Rodolico-San Marco", Catania, Italy
| | - Marina Parisi
- Hematology and BMT Unit, Azienda Ospedaliero Universitaria Policlinico "G.Rodolico-San Marco", Catania, Italy
| | - Paolo Fiumara
- Hematology and BMT Unit, Azienda Ospedaliero Universitaria Policlinico "G.Rodolico-San Marco", Catania, Italy
| | - Laura Parrinello
- Cytometryc Lab, Division of Haematology, A.O.U. Policlinico "G.Rodolico - S. Marco", Catania, Italy
| | - Sara Marino
- Cytometryc Lab, Division of Haematology, A.O.U. Policlinico "G.Rodolico - S. Marco", Catania, Italy
| | - Grazia Scuderi
- Cytometryc Lab, Division of Haematology, A.O.U. Policlinico "G.Rodolico - S. Marco", Catania, Italy
| | - Bruno Garibaldi
- Hematology and BMT Unit, Azienda Ospedaliero Universitaria Policlinico "G.Rodolico-San Marco", Catania, Italy
| | - Maurizio Musso
- Hematology Unit, Ospedale "La Maddalena", Palermo, Italy
| | - Nicola Di Renzo
- Haematology and Stem Cell Transplant Unit, Presidio Ospedaliero "Vito Fazzi", Lecce, Italy
| | - Ernesto Vigna
- Hematology Unit, Azienda Ospedaliera Annunziata, Cosenza, Italy
| | | | - Francesco Di Raimondo
- Hematology and BMT Unit, Azienda Ospedaliero Universitaria Policlinico "G.Rodolico-San Marco", Catania, Italy
| | - Giuseppe Milone
- Hematology and BMT Unit, Azienda Ospedaliero Universitaria Policlinico "G.Rodolico-San Marco", Catania, Italy
| |
Collapse
|
3
|
Zaninelli S, Panna S, Tettamanti S, Melita G, Doni A, D’Autilia F, Valgardsdottir R, Gotti E, Rambaldi A, Golay J, Introna M. Functional Activity of Cytokine-Induced Killer Cells Enhanced by CAR-CD19 Modification or by Soluble Bispecific Antibody Blinatumomab. Antibodies (Basel) 2024; 13:71. [PMID: 39311376 PMCID: PMC11417890 DOI: 10.3390/antib13030071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/08/2024] [Accepted: 08/16/2024] [Indexed: 09/26/2024] Open
Abstract
Strategies to increase the anti-tumor efficacy of cytokine-induced killer cells (CIKs) include genetic modification with chimeric antigen receptors (CARs) or the addition of soluble T-cell engaging bispecific antibodies (BsAbs). Here, CIKs were modified using a transposon system integrating two distinct anti-CD19 CARs (CAR-MNZ and CAR-BG2) or combined with soluble CD3xCD19 BsAb blinatumomab (CIK + Blina). CAR-MNZ bearing the CD28-OX40-CD3ζ signaling modules, and CAR-BG2, designed on the Tisagenlecleucel CAR sequence (Kymriah®), carrying the 4-1BB and CD3ζ signaling elements, were employed. After transfection and CIK expansion, cells expressed CAR-CD19 to a similar extent (35.9% CAR-MNZ and 17.7% CAR-BG2). In vitro evaluations demonstrated robust proliferation and cytotoxicity (~50% cytotoxicity) of CARCIK-MNZ, CARCIK-BG2, and CIK + Blina against CD19+ target cells, suggesting similar efficacy. All effectors formed an increased number of synapses, activated NFAT and NFkB, and secreted IL-2 and IFN-ɣ upon encountering targets. CIK + Blina displayed strongest NFAT and IFN-ɣ induction, whereas CARCIK-BG2 demonstrated superior synapse formation. All the effectors have shown therapeutic activity in vivo against the CD19+ Daudi tumor model, with CARCIK cells showing a more durable response compared to CIK + Blina, likely due to the short half-life of Blina in this model.
Collapse
Affiliation(s)
- Silvia Zaninelli
- Center of Cellular Therapy “G. Lanzani”, Division of Hematology, ASST Papa Giovanni XXIII, 24122 Bergamo, Italy; (S.Z.)
| | - Silvia Panna
- Center of Cellular Therapy “G. Lanzani”, Division of Hematology, ASST Papa Giovanni XXIII, 24122 Bergamo, Italy; (S.Z.)
| | - Sarah Tettamanti
- M. Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Giusi Melita
- M. Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Andrea Doni
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital, 20089 Milano, Italy
| | - Francesca D’Autilia
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital, 20089 Milano, Italy
| | - Rut Valgardsdottir
- Center of Cellular Therapy “G. Lanzani”, Division of Hematology, ASST Papa Giovanni XXIII, 24122 Bergamo, Italy; (S.Z.)
| | - Elisa Gotti
- Center of Cellular Therapy “G. Lanzani”, Division of Hematology, ASST Papa Giovanni XXIII, 24122 Bergamo, Italy; (S.Z.)
| | - Alessandro Rambaldi
- Hematology and Bone Marrow Transplant Unit, ASST Papa Giovanni XXIII Hospital, 24127 Bergamo, Italy
- Department of Oncology and Hematology, Università degli Studi di Milano, 20122 Milan, Italy
| | - Josée Golay
- Center of Cellular Therapy “G. Lanzani”, Division of Hematology, ASST Papa Giovanni XXIII, 24122 Bergamo, Italy; (S.Z.)
| | - Martino Introna
- Center of Cellular Therapy “G. Lanzani”, Division of Hematology, ASST Papa Giovanni XXIII, 24122 Bergamo, Italy; (S.Z.)
| |
Collapse
|
4
|
Gambles MT, Li S, Kendell I, Li J, Sborov D, Shami P, Yang J, Kopeček J. Multiantigen T-Cell Hybridizers: A Two-Component T-Cell-Activating Therapy. ACS NANO 2024; 18:23341-23353. [PMID: 39149859 DOI: 10.1021/acsnano.4c06500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Multispecific T-cell-engaging scaffolds have emerged as effective anticancer therapies for the treatment of hematological malignancies. Approaches that modulate cancer cell targeting and provide personalized, multispecific immunotherapeutics are needed. Here, we report on a modular, split antibody-like approach consisting of Fab' fragments modified with complementary morpholino oligonucleotides (MORFs). We synthesized a library of B-cell-targeting Fab'-MORF1 conjugates that self-assemble, via a Watson-Crick base pairing hybridization, with a complementary T-cell-engaging Fab'-MORF2 conjugate. We aptly titled our technology multiantigen T-cell hybridizers (MATCH). Using MATCH, cancer-specific T-cell recruitment was achieved utilizing four B-cell antigen targets: CD20, CD38, BCMA, and SLAMF7. The antigen expression profiles of various malignant B-cell lines were produced, and using these distinct profiles, cell-specific T-cell activation was attained on lymphoma, leukemia, and multiple myeloma cell lines in vitro. T-cell rechallenge experiments demonstrated the modular approach of MATCH by sequentially activating the same T-cell cohort against three different cancers using cancer antigen-specific Fab'-MORF1 conjugates. Furthermore, MATCH's efficacy was demonstrated in vivo by treating xenograft mouse models of human non-Hodgkin's lymphoma with CD20-directed MATCH therapy. In the pilot study, a single dose of MATCH allowed for long-term survival of all treated mice compared to saline control. In a second in vivo model, insights regarding optimal T-cell-to-target cell ratio were gleaned when a ratio of 5:1 T-cell-to-target cell MATCH-treated mice significantly delayed the onset of disease compared to higher and lower ratios.
Collapse
Affiliation(s)
- M Tommy Gambles
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, Utah 84112, United States
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah 84112, United States
| | - Shannuo Li
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, Utah 84112, United States
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah 84112, United States
| | - Isaac Kendell
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah 84112, United States
| | - Jiahui Li
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, Utah 84112, United States
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah 84112, United States
| | - Douglas Sborov
- Huntsman Cancer Institute, University of Utah, Salt Lake City ,Utah 84112, United States
| | - Paul Shami
- Huntsman Cancer Institute, University of Utah, Salt Lake City ,Utah 84112, United States
| | - Jiyuan Yang
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, Utah 84112, United States
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah 84112, United States
| | - Jindřich Kopeček
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, Utah 84112, United States
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah 84112, United States
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah 84112, United States
| |
Collapse
|
5
|
Gambles MT, Yang J, Kopeček J. Multi-targeted immunotherapeutics to treat B cell malignancies. J Control Release 2023; 358:232-258. [PMID: 37121515 PMCID: PMC10330463 DOI: 10.1016/j.jconrel.2023.04.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 04/20/2023] [Accepted: 04/27/2023] [Indexed: 05/02/2023]
Abstract
The concept of multi-targeted immunotherapeutic systems has propelled the field of cancer immunotherapy into an exciting new era. Multi-effector molecules can be designed to engage with, and alter, the patient's immune system in a plethora of ways. The outcomes can vary from effector cell recruitment and activation upon recognition of a cancer cell, to a multipronged immune checkpoint blockade strategy disallowing evasion of the cancer cells by immune cells, or to direct cancer cell death upon engaging multiple cell surface receptors simultaneously. Here, we review the field of multi-specific immunotherapeutics implemented to treat B cell malignancies. The mechanistically diverse strategies are outlined and discussed; common B cell receptor antigen targeting strategies are outlined and summarized; and the challenges of the field are presented along with optimistic insights for the future.
Collapse
Affiliation(s)
- M Tommy Gambles
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
| | - Jiyuan Yang
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA.
| | - Jindřich Kopeček
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
6
|
Comparing the efficacy of salvage regimens for relapsed/refractory B-cell acute lymphoblastic leukaemia: a systematic review and network meta-analysis. Ann Hematol 2023; 102:155-165. [PMID: 36394582 DOI: 10.1007/s00277-022-05040-1] [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: 08/28/2022] [Accepted: 11/07/2022] [Indexed: 11/19/2022]
Abstract
The complete remission (CR) rate and overall survival (OS) of relapsed/refractory (R/R) B-cell acute lymphoblastic leukaemia (B-ALL) are not satisfactory. The available salvage regimens include standard chemotherapy, inotuzumab ozogamicin, blinatumomab and cluster of differentiation (CD)19 chimeric antigen receptor T cells (CAR T), and the NCCN guidelines recommend all of these therapies with no preference. Dual CD19/CD22 CAR T-cells have emerged as new treatments and have shown some efficacy, with high CR rates and preventing CD19-negative relapse. However, direct comparisons of the CR rate and long-term survival among the different salvage therapies are lacking. Databases including PubMed, Embase, Web of Science and Cochrane were searched from inception to January 31, 2022, for relevant studies. The outcomes of interest were complete remission/complete remission with incomplete haematologic recovery (CR/CRi) rates and 1-year overall survival (OS) rates. Odds ratios (ORs) were generated for binary outcomes, and the mean difference (MD) was generated for consecutive outcomes by network meta-analysis. CD19 CAR T-cells demonstrated a significantly better effect in improving the CR/CRi rate than blinatumomab (OR = 8.32, 95% CI: 1.18 to 58.44) and chemotherapy (OR = 16.4, 95% CI: 2.76 to 97.45). In terms of OS, CD19 CAR T-cells and dual CD19/CD22 CAR T-cells both had a higher 1-year OS rate than blinatumomab, inotuzumab ozogamicin and chemotherapy. There was no significant difference between CD19 CAR T-cells and dual CD19/CD22 CAR T-cells in terms of 1-year OS and CR/CRi rates. CD19 CAR T-cells are effective in inducing CR, and CD19 CAR T-cells and dual CD19/CD22 CAR T-cells show benefits for overall survival. More high-quality randomized controlled trials and longer follow-ups are needed to confirm and update the results of this analysis in the future.
Collapse
|
7
|
Glud EN, Rasmussen M, Zhang Y, Mandrup OA, Salachan PV, Borre M, Sørensen KD, Howard KA. Identification of a high-risk immunogenic prostate cancer patient subset as candidates for T-cell engager immunotherapy and the introduction of a novel albumin-fused anti-CD3 × anti-PSMA bispecific design. Br J Cancer 2022; 127:2186-2197. [PMID: 36243890 PMCID: PMC9727128 DOI: 10.1038/s41416-022-01994-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Cancer immunotherapies such as bispecific T-cell engagers have seen limited adoption in prostate cancer (PC), possibly due to differing levels of cancer receptor expression and effector T-cell infiltration between patients and inherent defects in T-cell engager design. METHODS CD8+ T-cell infiltration and PSMA expression were determined by RNA sequencing of primary PC tissue samples from 126 patients with localised PC and 17 patients with metastatic PC. Prognostic value was assessed through clinical parameters, including CAPRA-S risk score. A panel of albumin-fused anti-CD3 × anti-PSMA T-cell engagers with different neonatal Fc receptor (FcRn) affinity were characterised by flow cytometry, Bio-Layer Interferometry and functional cellular assays. RESULTS A subset of patients with localised (30/126 = 24%) and metastatic (10/17 = 59%) PC showed both high PSMA expression and high CD8+ T-cell enrichment. The High/High phenotype in localised PC associated with a clinically high-risk cancer subtype, confirmed in an external patient cohort (n = 550, PRAD/TCGA). The T-cell engagers exhibited tunable FcRn-driven cellular recycling, CD3 and PSMA cellular engagement, T-cell activation and PSMA level-dependent cellular cytotoxicity. CONCLUSION This work presents an albumin-fused bispecific T-cell engager with programmable FcRn engagement and identifies a high-risk PC patient subset as candidates for treatment with the T-cell engager class of immuno-oncology biologics.
Collapse
Affiliation(s)
- Eske N. Glud
- grid.7048.b0000 0001 1956 2722Interdisciplinary Nanoscience Center (iNANO), Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| | - Martin Rasmussen
- grid.7048.b0000 0001 1956 2722Department of Molecular Medicine, Aarhus University Hospital & Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Yonghui Zhang
- grid.7048.b0000 0001 1956 2722Interdisciplinary Nanoscience Center (iNANO), Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| | - Ole A. Mandrup
- grid.7048.b0000 0001 1956 2722Interdisciplinary Nanoscience Center (iNANO), Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| | - Paul Vinu Salachan
- grid.7048.b0000 0001 1956 2722Department of Molecular Medicine, Aarhus University Hospital & Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Michael Borre
- grid.7048.b0000 0001 1956 2722Department of Urology, Aarhus University Hospital & Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Karina Dalsgaard Sørensen
- grid.7048.b0000 0001 1956 2722Department of Molecular Medicine, Aarhus University Hospital & Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Kenneth A. Howard
- grid.7048.b0000 0001 1956 2722Interdisciplinary Nanoscience Center (iNANO), Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| |
Collapse
|
8
|
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
|
9
|
IgG-Based Bispecific Anti-CD95 Antibodies for the Treatment of B Cell-Derived Malignancies and Autoimmune Diseases. Cancers (Basel) 2022; 14:cancers14163941. [PMID: 36010934 PMCID: PMC9405798 DOI: 10.3390/cancers14163941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/12/2022] [Accepted: 08/13/2022] [Indexed: 11/23/2022] Open
Abstract
Simple Summary Therapeutic antibodies have become a crucial cornerstone of the standard therapy for lymphoma and autoimmune diseases. However, the respective target antigens are also expressed on healthy B cells resulting in unspecific effects. In this article, we present a novel approach to selectively induce apoptosis in lymphoma cells and autoreactive B cells that express the CD95 death receptor. Therefore, we developed an improved IgG-based bispecific antibody format with favorable production properties and pharmacokinetics for CD20- and CD19-directed induction of apoptosis via CD95. We could show that our bispecific anti-CD95 antibodies are very efficient in the depletion of malignant and autoreactive B cells in vitro and in vivo. Therefore, our antibodies could help to provide a more selective therapy for patients with B cell-derived malignancies and autoimmune diseases. Abstract Antibodies against the B cell-specific antigens CD20 and CD19 have markedly improved the treatment of B cell-derived lymphoma and autoimmune diseases by depleting malignant and autoreactive B cells. However, since CD20 and CD19 are also expressed on healthy B cells, such antibodies lack disease specificity. Here, we optimize a previously developed concept that uses bispecific antibodies to induce apoptosis selectively in malignant and autoreactive B cells that express the death receptor CD95. We describe the development and characterization of bispecific antibodies with CD95xCD20 and CD95xCD19 specificity in a new IgG-based format. We could show that especially the CD95xCD20 antibody mediated a strong induction of apoptosis in malignant B cells in vitro. In vivo, the antibody was clearly superior to the previously used Fabsc format with identical specificities. In addition, both IgGsc antibodies depleted activated B cells in vitro, leading to a significant reduction in antibody production and cytokine secretion. The killing of resting B cells and hepatocytes that lack CD95 and CD20/CD19, respectively, was marginal. Thus, our results imply that bispecific anti-CD95 antibodies in the IgGsc format are an attractive tool for a more selective and efficient depletion of malignant as well as autoreactive B cells.
Collapse
|
10
|
Wo S, Levavi H, Mascarenhas J, Kremyanskaya M, Navada S, Bar-Natan M, Kim SS. Immunoglobulin repletion during blinatumomab therapy does not reduce the rate of secondary hypogammaglobulinemia and associated infectious risk. Blood Res 2022; 57:135-143. [PMID: 35551109 PMCID: PMC9242831 DOI: 10.5045/br.2022.2021163] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 01/09/2022] [Accepted: 04/18/2022] [Indexed: 11/17/2022] Open
Abstract
Background Blinatumomab has demonstrated efficacy in minimal residual disease (MRD) positive and relapsed/refractory B-cell acute lymphoblastic leukemia (B-ALL) by inciting rapid and sustained B-cell depletion. Methods Owing to its effect on B-cells, blinatumomab is associated with a higher rate of secondary hypogammaglobulinemia compared to chemotherapy. To mitigate blinatumomab-induced hypogammaglobulinemia, patients were pre-emptively repleted with intravenous immune globulin (IVIG) during blinatumomab therapy. In this retrospective study, we compared outcomes of 23 blinatumomab treated adults with ALL. Seventeen patients routinely received IVIG and 6 patients were in the control cohort. Results Our findings demonstrated no difference between the two cohorts in immunoglobulin G (IgG) nadir (338 mg/dL vs. 337 mg/dL, P=0.641), days to IgG nadir (120.5 vs. 85.5 days, P=0.13), infection rate (82.4% vs. 66.7%, P=0.58), infections requiring ICU admission (23.5% vs. 16.7%, P=1), and infection related mortality (17.6% vs. 16.7%, P=1). Conclusion Pre-emptive IVIG repletion during blinatumomab did not prevent hypogammaglobulinemia and associated infection risk.
Collapse
Affiliation(s)
- Stephanie Wo
- Department of Pharmacy, The Mount Sinai Hospital, New York, NY, USA
| | - Hannah Levavi
- Division of Hematology/Oncology, Icahn School of Medicine at The Mount Sinai Hospital, New York, NY, USA
| | - John Mascarenhas
- Division of Hematology/Oncology, Icahn School of Medicine at The Mount Sinai Hospital, New York, NY, USA
| | - Marina Kremyanskaya
- Division of Hematology/Oncology, Icahn School of Medicine at The Mount Sinai Hospital, New York, NY, USA
| | - Shyamala Navada
- Division of Hematology/Oncology, Icahn School of Medicine at The Mount Sinai Hospital, New York, NY, USA
| | - Michal Bar-Natan
- Division of Hematology/Oncology, Icahn School of Medicine at The Mount Sinai Hospital, New York, NY, USA
| | - Sara S Kim
- Department of Pharmacy, The Mount Sinai Hospital, New York, NY, USA
| |
Collapse
|
11
|
Tang J, Gong Y, Ma X. Bispecific Antibodies Progression in Malignant Melanoma. Front Pharmacol 2022; 13:837889. [PMID: 35401191 PMCID: PMC8984188 DOI: 10.3389/fphar.2022.837889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/28/2022] [Indexed: 02/05/2023] Open
Abstract
The discovery of oncogenes and immune checkpoints has revolutionized the treatment of melanoma in the past 10 years. However, the current PD-L1 checkpoints lack specificity for tumors and target normal cells expressing PD-L1, thus reducing the efficacy on malignant melanoma and increasing the side effects. In addition, the treatment options for primary or secondary drug-resistant melanoma are limited. Bispecific antibodies bind tumor cells and immune cells by simultaneously targeting two antigens, enhancing the anti-tumor targeting effect and cytotoxicity and reducing drug-resistance in malignant melanoma, thus representing an emerging strategy to improve the clinical efficacy. This review focused on the treatment of malignant melanoma by bispecific antibodies and summarized the effective results of the experiments that have been conducted, also discussing the different aspects of these therapies. The role of the melanoma epitopes, immune cell activation, cell death and cytotoxicity induced by bispecific antibodies were evaluated in the clinical or preclinical stage, as these therapies appear to be the most suitable in the treatment of malignant melanoma.
Collapse
Affiliation(s)
- Juan Tang
- Department of Oncology, West China Hospital of Sichuan University, Chengdu, China
| | - Youling Gong
- Department of Oncology, West China Hospital of Sichuan University, Chengdu, China
| | - Xuelei Ma
- Department of Oncology, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
12
|
Lee SC, Ma JSY, Kim MS, Laborda E, Choi SH, Hampton EN, Yun H, Nunez V, Muldong MT, Wu CN, Ma W, Kulidjian AA, Kane CJ, Klyushnichenko V, Woods AK, Joseph SB, Petrassi M, Wisler J, Li J, Jamieson CAM, Schultz PG, Kim CH, Young TS. A PSMA-targeted bispecific antibody for prostate cancer driven by a small-molecule targeting ligand. SCIENCE ADVANCES 2021; 7:7/33/eabi8193. [PMID: 34380625 PMCID: PMC8357232 DOI: 10.1126/sciadv.abi8193] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/23/2021] [Indexed: 06/13/2023]
Abstract
Despite the development of next-generation antiandrogens, metastatic castration-resistant prostate cancer (mCRPC) remains incurable. Here, we describe a unique semisynthetic bispecific antibody that uses site-specific unnatural amino acid conjugation to combine the potency of a T cell-recruiting anti-CD3 antibody with the specificity of an imaging ligand (DUPA) for prostate-specific membrane antigen. This format enabled optimization of structure and function to produce a candidate (CCW702) with specific, potent in vitro cytotoxicity and improved stability compared with a bispecific single-chain variable fragment format. In vivo, CCW702 eliminated C4-2 xenografts with as few as three weekly subcutaneous doses and prevented growth of PCSD1 patient-derived xenograft tumors in mice. In cynomolgus monkeys, CCW702 was well tolerated up to 34.1 mg/kg per dose, with near-complete subcutaneous bioavailability and a PK profile supporting testing of a weekly dosing regimen in patients. CCW702 is being evaluated in a first in-human clinical trial for men with mCRPC who had progressed on prior therapies (NCT04077021).
Collapse
Affiliation(s)
- Sung Chang Lee
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jennifer S Y Ma
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Min Soo Kim
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Eduardo Laborda
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Sei-Hyun Choi
- Department of Chemistry and Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Eric N Hampton
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Hwayoung Yun
- Department of Chemistry and Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Vanessa Nunez
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Michelle T Muldong
- Department of Urology, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Christina N Wu
- Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Wenxue Ma
- Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Anna A Kulidjian
- Department of Orthopedic Surgery, Scripps MD Anderson Cancer Center, La Jolla, CA 92093, USA
| | - Christopher J Kane
- Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Vadim Klyushnichenko
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ashley K Woods
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Sean B Joseph
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Mike Petrassi
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - John Wisler
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jing Li
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Christina A M Jamieson
- Department of Urology, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Peter G Schultz
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA.
- Department of Chemistry and Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Chan Hyuk Kim
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Travis S Young
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA.
- Department of Chemistry and Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
13
|
Bioassay Development for Bispecific Antibodies-Challenges and Opportunities. Int J Mol Sci 2021; 22:ijms22105350. [PMID: 34069573 PMCID: PMC8160952 DOI: 10.3390/ijms22105350] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/14/2021] [Accepted: 05/15/2021] [Indexed: 12/25/2022] Open
Abstract
Antibody therapeutics are expanding with promising clinical outcomes, and diverse formats of antibodies are further developed and available for patients of the most challenging disease areas. Bispecific antibodies (BsAbs) have several significant advantages over monospecific antibodies by engaging two antigen targets. Due to the complicated mechanism of action, diverse structural variations, and dual-target binding, developing bioassays and other types of assays to characterize BsAbs is challenging. Developing bioassays for BsAbs requires a good understanding of the mechanism of action of the molecule, principles and applications of different bioanalytical methods, and phase-appropriate considerations per regulatory guidelines. Here, we review recent advances and case studies to provide strategies and insights for bioassay development for different types of bispecific molecules.
Collapse
|
14
|
Woodchuck Hepatitis Virus Post-Transcriptional Regulation Element (WPRE) Promotes Anti-CD19 BiTE Expression in Expi293 Cells. IRANIAN BIOMEDICAL JOURNAL 2021; 25:275-83. [PMID: 34217158 PMCID: PMC8334396 DOI: 10.52547/ibj.25.4.275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Background: Bispecific antibodies represent an important class of mAbs, with great therapeutic potentials due to their ability to target simultaneously two distinct epitopes. The generation of functional bispecific antibodies with the highest possible yields is particularly critical for the production of these compounds on industrial scales. Anti- CD3 × CD19 bsAb is a bispecific T-cell engager (BiTE) currently used for treating ALL. Herein, we have tried to optimize the expression level of this antibody in mammalian hosts. Methods: WPRE sequence was incorporated at the 3’ end of the expression cassette. This modification resulted in a notable about two-fold increase in the expression of the bsAb in the Expi293 cell line. Results & Conclusion: Follow-up flow cytometry analysis demonstrated the binding properties of the produced antibody at acceptable levels, and in vitro bioactivity assays showed that this product is potent enough for targeting and destroying CD19-positive cells. Our findings show that WPRE enhances the expression of this type of bispecific mAbs in HEK-293 family cell lines. This approach can be used in biopharma industry for the mass production of anti-CD3 × CD19 bispecific antibody.
Collapse
|
15
|
Zhong L, Shi W, Gan L, Liu X, Huo Y, Wu P, Zhang Z, Wu T, Peng H, Huang Y, Zhao Y, Yuan Y, Deng Z, Tang H. Human endoglin-CD3 bispecific T cell engager antibody induces anti-tumor effect in vivo. Am J Cancer Res 2021; 11:6393-6406. [PMID: 33995664 PMCID: PMC8120215 DOI: 10.7150/thno.53121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 03/12/2021] [Indexed: 12/11/2022] Open
Abstract
Rationale: Endoglin, also known as CD105, is a homo-dimeric membrane glycoprotein required for angiogenesis and serves as a marker for cancer vasculature. In this study, we constructed a bispecific T-cell engager (BiTE) antibody that targets human endoglin and CD3 (hEND-CD3/BiTE). We examined BiTE binding to endoglin-expressing cells and its effects on the cytolytic activity of T cells and cancer development. Methods: The in vitro effects of hEND-CD3/BiTE, including binding to target cells, T-cell activation, proliferation, and cytotoxicity, were examined in endoglin-expressing 293T cells, human umbilical vascular endothelial cells, tumor-derived endothelial cells, and CD3+ T cells. An in vivo xenograft tumor model was established using A549 human lung cancer cells. The therapeutic efficacy of hEND-CD3/BiTE was assessed by monitoring tumor growth, angiogenesis, and mouse survival. Results: hEND-CD3/BiTE specifically bound to endoglin-expressing cells and CD3+ T cells in vitro and stimulated T-cell activation, proliferation, and Th1 cytokine secretion, and promoted T-cell-mediated cytolysis of endoglin-expressing cells. The hEND-CD3/BiTE in vivo caused minimal toxicity to major organs, reduced tumor neoangiogenesis, inhibited tumor growth, and significantly improved mouse survival. Conclusions: Our study demonstrated the therapeutic potential of hEND-CD3/BiTE and provided a novel approach to clinical cancer treatment.
Collapse
|
16
|
Lussana F, Gritti G, Rambaldi A. Immunotherapy of Acute Lymphoblastic Leukemia and Lymphoma With T Cell-Redirected Bispecific Antibodies. J Clin Oncol 2021; 39:444-455. [PMID: 33434063 PMCID: PMC8078487 DOI: 10.1200/jco.20.01564] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2020] [Indexed: 12/13/2022] Open
Affiliation(s)
- Federico Lussana
- Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Giuseppe Gritti
- Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Alessandro Rambaldi
- Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
- Department of Oncology-Hematology, University of Milan, Milan, Italy
| |
Collapse
|
17
|
Zekri L, Vogt F, Osburg L, Müller S, Kauer J, Manz T, Pflügler M, Maurer A, Heitmann JS, Hagelstein I, Märklin M, Hörner S, Todenhöfer T, Calaminus C, Stenzl A, Pichler B, la Fougère C, Schneider MA, Rammensee H, Zender L, Sipos B, Salih HR, Jung G. An IgG-based bispecific antibody for improved dual targeting in PSMA-positive cancer. EMBO Mol Med 2021; 13:e11902. [PMID: 33372710 PMCID: PMC7863392 DOI: 10.15252/emmm.201911902] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 11/26/2020] [Accepted: 12/01/2020] [Indexed: 12/14/2022] Open
Abstract
The prostate-specific membrane antigen (PSMA) has been demonstrated in numerous studies to be expressed specifically on prostate carcinoma cells and on the neovasculature of several other cancer entities. However, the simultaneous expression of PSMA on both, tumor cells as well as tumor vessels remains unclear, even if such "dual" expression would constitute an important asset to facilitate sufficient influx of effector cells to a given tumor site. We report here on the generation of a PSMA antibody, termed 10B3, which exerts superior dual reactivity on sections of prostate carcinoma and squamous cell carcinoma of the lung. 10B3 was used for the construction of T-cell recruiting bispecific PSMAxCD3 antibodies in Fab- and IgG-based formats, designated Fabsc and IgGsc, respectively. In vitro, both molecules exhibited comparable activity. In contrast, only the larger IgGsc molecule induced complete and durable elimination of established tumors in humanized mice due to favorable pharmacokinetic properties. Upon treatment of three patients with metastasized prostate carcinoma with the IgGsc reagent, marked activation of T cells and rapid reduction of elevated PSA levels were observed.
Collapse
|
18
|
Heitmann JS, Pfluegler M, Jung G, Salih HR. Bispecific Antibodies in Prostate Cancer Therapy: Current Status and Perspectives. Cancers (Basel) 2021; 13:549. [PMID: 33535627 PMCID: PMC7867165 DOI: 10.3390/cancers13030549] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/24/2021] [Accepted: 01/26/2021] [Indexed: 12/29/2022] Open
Abstract
Prostate carcinoma (PC) is the second most common cancer in men. When the disease becomes unresponsive to androgen deprivation therapy, the remaining treatment options are of limited benefit. Despite intense efforts, none of the T cell-based immunotherapeutic strategies that meanwhile have become a cornerstone for treatment of other malignancies is established in PC. This refers to immune checkpoint inhibition (CI), which generally reinforces T cell immunity as well as chimeric antigen receptor T (CAR-T) cells and bispecific antibodies (bsAbs) that stimulate the T cell receptor/CD3-complex and mobilize T cells in a targeted manner. In general, compared to CAR-T cells, bsAb would have the advantage of being an "off the shelf" reagent associated with less preparative effort, but at present, despite enormous efforts, neither CAR-T cells nor bsAbs are successful in solid tumors. Here, we focus on the various bispecific constructs that are presently in development for treatment of PC, and discuss underlying concepts and the state of clinical evaluation as well as future perspectives.
Collapse
Affiliation(s)
- Jonas S. Heitmann
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, 72076 Tübingen, Germany; (J.S.H.); (M.P.)
- DFG Cluster of Excellence 2180 “Image-Guided and Functional Instructed Tumor Therapy” (IFIT), University of Tübingen, 72076 Tübingen, Germany;
| | - Martin Pfluegler
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, 72076 Tübingen, Germany; (J.S.H.); (M.P.)
- DFG Cluster of Excellence 2180 “Image-Guided and Functional Instructed Tumor Therapy” (IFIT), University of Tübingen, 72076 Tübingen, Germany;
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany
| | - Gundram Jung
- DFG Cluster of Excellence 2180 “Image-Guided and Functional Instructed Tumor Therapy” (IFIT), University of Tübingen, 72076 Tübingen, Germany;
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany
- German Cancer Consortium (DKTK), DKFZ Partner Site Tübingen, 72076 Tübingen, Germany
| | - Helmut R. Salih
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, 72076 Tübingen, Germany; (J.S.H.); (M.P.)
- DFG Cluster of Excellence 2180 “Image-Guided and Functional Instructed Tumor Therapy” (IFIT), University of Tübingen, 72076 Tübingen, Germany;
| |
Collapse
|
19
|
Benmebarek MR, Cadilha BL, Herrmann M, Lesch S, Schmitt S, Stoiber S, Darwich A, Augsberger C, Brauchle B, Rohrbacher L, Oner A, Seifert M, Schwerdtfeger M, Gottschlich A, Rataj F, Fenn NC, Klein C, Subklewe M, Endres S, Hopfner KP, Kobold S. A modular and controllable T cell therapy platform for acute myeloid leukemia. Leukemia 2021; 35:2243-2257. [PMID: 33414484 PMCID: PMC7789085 DOI: 10.1038/s41375-020-01109-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 11/09/2020] [Accepted: 12/01/2020] [Indexed: 01/29/2023]
Abstract
Targeted T cell therapy is highly effective in disease settings where tumor antigens are uniformly expressed on malignant cells and where off-tumor on-target-associated toxicity is manageable. Although acute myeloid leukemia (AML) has in principle been shown to be a T cell-sensitive disease by the graft-versus-leukemia activity of allogeneic stem cell transplantation, T cell therapy has so far failed in this setting. This is largely due to the lack of target structures both sufficiently selective and uniformly expressed on AML, causing unacceptable myeloid cell toxicity. To address this, we developed a modular and controllable MHC-unrestricted adoptive T cell therapy platform tailored to AML. This platform combines synthetic agonistic receptor (SAR) -transduced T cells with AML-targeting tandem single chain variable fragment (scFv) constructs. Construct exchange allows SAR T cells to be redirected toward alternative targets, a process enabled by the short half-life and controllability of these antibody fragments. Combining SAR-transduced T cells with the scFv constructs resulted in selective killing of CD33+ and CD123+ AML cell lines, as well as of patient-derived AML blasts. Durable responses and persistence of SAR-transduced T cells could also be demonstrated in AML xenograft models. Together these results warrant further translation of this novel platform for AML treatment.
Collapse
Affiliation(s)
- Mohamed-Reda Benmebarek
- grid.5252.00000 0004 1936 973XCenter of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU, Munich, Germany
| | - Bruno L. Cadilha
- grid.5252.00000 0004 1936 973XCenter of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU, Munich, Germany
| | - Monika Herrmann
- grid.5252.00000 0004 1936 973XDepartment of Medicine III, Klinikum der Universität München, LMU, Munich, Germany
| | - Stefanie Lesch
- grid.5252.00000 0004 1936 973XCenter of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU, Munich, Germany
| | - Saskia Schmitt
- grid.5252.00000 0004 1936 973XDepartment of Medicine III, Klinikum der Universität München, LMU, Munich, Germany
| | - Stefan Stoiber
- grid.5252.00000 0004 1936 973XCenter of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU, Munich, Germany
| | - Abbass Darwich
- grid.417728.f0000 0004 1756 8807Mucosal Immunology and Microbiota Lab, Humanitas Clinical and Research Center, Milan, Italy
| | - Christian Augsberger
- grid.5252.00000 0004 1936 973XDepartment of Medicine III, Klinikum der Universität München, LMU, Munich, Germany
| | - Bettina Brauchle
- grid.5252.00000 0004 1936 973XDepartment of Medicine III, Klinikum der Universität München, LMU, Munich, Germany ,grid.5252.00000 0004 1936 973XLaboratory for Translational Cancer Immunology, Gene Center, LMU Munich, Munich, Germany
| | - Lisa Rohrbacher
- grid.5252.00000 0004 1936 973XDepartment of Medicine III, Klinikum der Universität München, LMU, Munich, Germany ,grid.5252.00000 0004 1936 973XLaboratory for Translational Cancer Immunology, Gene Center, LMU Munich, Munich, Germany
| | - Arman Oner
- grid.5252.00000 0004 1936 973XCenter of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU, Munich, Germany
| | - Matthias Seifert
- grid.5252.00000 0004 1936 973XCenter of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU, Munich, Germany
| | - Melanie Schwerdtfeger
- grid.5252.00000 0004 1936 973XCenter of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU, Munich, Germany
| | - Adrian Gottschlich
- grid.5252.00000 0004 1936 973XCenter of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU, Munich, Germany
| | - Felicitas Rataj
- grid.5252.00000 0004 1936 973XCenter of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU, Munich, Germany
| | - Nadja C. Fenn
- grid.5252.00000 0004 1936 973XDepartment of Medicine III, Klinikum der Universität München, LMU, Munich, Germany
| | - Christian Klein
- grid.417570.00000 0004 0374 1269Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Marion Subklewe
- grid.5252.00000 0004 1936 973XDepartment of Medicine III, Klinikum der Universität München, LMU, Munich, Germany ,grid.5252.00000 0004 1936 973XLaboratory for Translational Cancer Immunology, Gene Center, LMU Munich, Munich, Germany ,German Center for Translational Cancer Research (DKTK), Partner Site Munich, Munich, Germany
| | - Stefan Endres
- grid.5252.00000 0004 1936 973XCenter of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU, Munich, Germany ,German Center for Translational Cancer Research (DKTK), Partner Site Munich, Munich, Germany ,grid.4567.00000 0004 0483 2525Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Neuherberg, Germany
| | | | - Sebastian Kobold
- grid.5252.00000 0004 1936 973XCenter of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU, Munich, Germany ,German Center for Translational Cancer Research (DKTK), Partner Site Munich, Munich, Germany ,grid.4567.00000 0004 0483 2525Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Neuherberg, Germany
| |
Collapse
|
20
|
Li L, Wang Y. Recent updates for antibody therapy for acute lymphoblastic leukemia. Exp Hematol Oncol 2020; 9:33. [PMID: 33292550 PMCID: PMC7697374 DOI: 10.1186/s40164-020-00189-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/19/2020] [Indexed: 02/07/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) is a hematologic malignancy arising from precursors of the lymphoid lineage. Conventional cytotoxic chemotherapies have resulted in high cure rates of up to 90% in pediatric ALL, but the outcomes for adult patients remain suboptimal with 5-year survival rates of only 30%-40%. Current immunotherapies exploit the performance of antibodies through several different mechanisms, including naked antibodies, antibodies linked to cytotoxic agents, and T-cell re-directing antibodies. Compared with chemotherapy, the application of an antibody-drug conjugates (ADC) called inotuzumab ozogamicin in relapsed or refractory (R/R) CD22+. ALL resulted in a complete remission (CR) rate of 81% and an overall median survival of 7.7 months with reduced toxicity. Similarly, blinatumomab, the first FDA-approved bispecific antibody (BsAb), produced a 44% complete response rate and an overall median survival of 7.7 months in a widely treated ALL population. In addition, approximately 80% of patients getting complete remission with evidence of minimal residual disease (MRD) achieved a complete MRD response with the use of blinatumomab. These results highlight the great promise of antibody-based therapy for ALL. How to reasonably determine the place of antibody drugs in the treatment of ALL remains a major problem to be solved for ongoing and future researches. Meanwhile the combination of antibody-based therapy with traditional standard of care (SOC) chemotherapy, chimeric antigen receptor (CAR) T-cell therapy and HSCT is also a challenge. Here, we will review some important milestones of antibody-based therapies, including combinational strategies, and antibodies under clinical development for ALL.
Collapse
Affiliation(s)
- Le Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Ying Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
| |
Collapse
|
21
|
Lejeune M, Köse MC, Duray E, Einsele H, Beguin Y, Caers J. Bispecific, T-Cell-Recruiting Antibodies in B-Cell Malignancies. Front Immunol 2020; 11:762. [PMID: 32457743 PMCID: PMC7221185 DOI: 10.3389/fimmu.2020.00762] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 04/03/2020] [Indexed: 12/12/2022] Open
Abstract
Bispecific antibodies (BsAbs) are designed to recognize and bind to two different antigens or epitopes. In the last few decades, BsAbs have been developed within the context of cancer therapies and in particular for the treatment of hematologic B-cell malignancies. To date, more than one hundred different BsAb formats exist, including bispecific T-cell engagers (BiTEs), and new constructs are constantly emerging. Advances in protein engineering have enabled the creation of BsAbs with specific mechanisms of action and clinical applications. Moreover, a better understanding of resistance and evasion mechanisms, as well as advances in the protein engineering and in immunology, will help generating a greater variety of BsAbs to treat various cancer types. This review focuses on T-cell-engaging BsAbs and more precisely on the various BsAb formats currently being studied in the context of B-cell malignancies, on ongoing clinical trials and on the clinical concerns to be taken into account in the development of new BsAbs.
Collapse
Affiliation(s)
- Margaux Lejeune
- Laboratory of Hematology, GIGA I3, University of Liège, Liège, Belgium
| | - Murat Cem Köse
- Laboratory of Hematology, GIGA I3, University of Liège, Liège, Belgium
| | - Elodie Duray
- Laboratory of Hematology, GIGA I3, University of Liège, Liège, Belgium
| | - Hermann Einsele
- Department of Internal Medicine II, University of Würzburg, Würzburg, Germany
| | - Yves Beguin
- Laboratory of Hematology, GIGA I3, University of Liège, Liège, Belgium.,Department of Hematology, CHU de Liège, Liège, Belgium
| | - Jo Caers
- Laboratory of Hematology, GIGA I3, University of Liège, Liège, Belgium.,Department of Hematology, CHU de Liège, Liège, Belgium
| |
Collapse
|
22
|
Lenk L, Alsadeq A, Schewe DM. Involvement of the central nervous system in acute lymphoblastic leukemia: opinions on molecular mechanisms and clinical implications based on recent data. Cancer Metastasis Rev 2020; 39:173-187. [PMID: 31970588 PMCID: PMC7098933 DOI: 10.1007/s10555-020-09848-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Acute lymphoblastic leukemia (ALL) is the most common childhood cancer. One of the major clinical challenges is adequate diagnosis and treatment of central nervous system (CNS) involvement in this disease. Intriguingly, there is little solid evidence on the mechanisms sustaining CNS disease in ALL. Here, we present and discuss recent data on this topic, which are mainly derived from preclinical model systems. We thereby highlight sites and routes of leukemic CNS infiltration, cellular features promoting infiltration and survival of leukemic cells in a presumably hostile niche, and dormancy as a potential mechanism of survival and relapse in CNS leukemia. We also focus on the impact of ALL cytogenetic subtypes on features associated with a particular CNS tropism. Finally, we speculate on new perspectives in the treatment of ALL in the CNS, including ideas on the impact of novel immunotherapies.
Collapse
Affiliation(s)
- Lennart Lenk
- Department of Pediatrics I, ALL-BFM Study Group, Christian-Albrechts University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Ameera Alsadeq
- Institute of Immunology, Ulm University Medical Center, Ulm, Germany
| | - Denis M Schewe
- Department of Pediatrics I, ALL-BFM Study Group, Christian-Albrechts University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany.
| |
Collapse
|
23
|
Deak D, Pop C, Zimta AA, Jurj A, Ghiaur A, Pasca S, Teodorescu P, Dascalescu A, Antohe I, Ionescu B, Constantinescu C, Onaciu A, Munteanu R, Berindan-Neagoe I, Petrushev B, Turcas C, Iluta S, Selicean C, Zdrenghea M, Tanase A, Danaila C, Colita A, Colita A, Dima D, Coriu D, Einsele H, Tomuleasa C. Let's Talk About BiTEs and Other Drugs in the Real-Life Setting for B-Cell Acute Lymphoblastic Leukemia. Front Immunol 2020; 10:2856. [PMID: 31921126 PMCID: PMC6934055 DOI: 10.3389/fimmu.2019.02856] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 11/20/2019] [Indexed: 01/07/2023] Open
Abstract
Background: Therapy for acute lymphoblastic leukemia (ALL) are currently initially efficient, but even if a high percentage of patients have an initial complete remission (CR), most of them relapse. Recent data shows that immunotherapy with either bispecific T-cell engagers (BiTEs) of chimeric antigen receptor (CAR) T cells can eliminate residual chemotherapy-resistant B-ALL cells. Objective: The objective of the manuscript is to present improvements in the clinical outcome for chemotherapy-resistant ALL in the real-life setting, by describing Romania's experience with bispecific antibodies for B-cell ALL. Methods: We present the role of novel therapies for relapsed B-cell ALL, including the drugs under investigation in phase I-III clinical trials, as a potential bridge to transplant. Blinatumomab is presented in a critical review, presenting both the advantages of this drug, as well as its limitations. Results: Bispecific antibodies are discussed, describing the clinical trials that resulted in its approval by the FDA and EMA. The real-life setting for relapsed B-cell ALL is described and we present the patients treated with blinatumomab in Romania. Conclusion: In the current manuscript, we present blinatumomab as a therapeutic alternative in the bridge-to-transplant setting for refractory or relapsed ALL, to gain a better understanding of the available therapies and evidence-based data for these patients in 2019.
Collapse
Affiliation(s)
- Dalma Deak
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Cristina Pop
- Department of Pharmacology, Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Alina-Andreea Zimta
- Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ancuta Jurj
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Alexandra Ghiaur
- Department of Hematology, Fundeni Clinical Institute, Bucharest, Romania
| | - Sergiu Pasca
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Patric Teodorescu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Angela Dascalescu
- Department of Hematology, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania.,Department of Hematology, Regional Institute of Oncology, Iasi, Romania
| | - Ion Antohe
- Department of Hematology, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania.,Department of Hematology, Regional Institute of Oncology, Iasi, Romania
| | - Bogdan Ionescu
- Department of Hematology, Fundeni Clinical Institute, Bucharest, Romania
| | - Catalin Constantinescu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Anca Onaciu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Raluca Munteanu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Bobe Petrushev
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Cristina Turcas
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Sabina Iluta
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Cristina Selicean
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Mihnea Zdrenghea
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Alina Tanase
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Catalin Danaila
- Department of Hematology, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania.,Department of Hematology, Regional Institute of Oncology, Iasi, Romania
| | - Anca Colita
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania.,Department of Pediatrics, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Andrei Colita
- Department of Hematology, Coltea Hospital, Bucharest, Romania.,Department of Hematology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Delia Dima
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Daniel Coriu
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania.,Department of Hematology, Fundeni Clinical Institute, Bucharest, Romania.,Department of Hematology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital Wurzburg, Würzburg, Germany
| | - Ciprian Tomuleasa
- Department of Hematology/Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
24
|
Khalid A, Aslam S, Ahmed M, Hasnain S, Aslam A. Risk assessment of FLT3 and PAX5 variants in B-acute lymphoblastic leukemia: a case-control study in a Pakistani cohort. PeerJ 2019; 7:e7195. [PMID: 31565544 PMCID: PMC6743442 DOI: 10.7717/peerj.7195] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 05/28/2019] [Indexed: 11/20/2022] Open
Abstract
AIMS B-cell acute lymphoblastic leukemia (B-ALL) is amongst the most prevalent cancers of children in Pakistan. Genetic variations in FLT3 are associated with auto-phosphorylation of kinase domain that leads to increased proliferation of blast cells. Paired box family of transcription factor (PAX5) plays a critical role in commitment and differentiation of B-cells. Variations in PAX5 are associated with the risk of B-ALL. We aimed to analyze the association of FLT3 and PAX5 polymorphisms with B cell leukemia in Pakistani cohort. METHODS We collected 155 B-ALL subject and 155 control blood samples. For analysis, genotyping was done by tetra ARMS-PCR. SPSS was used to check the association of demographic factors of SNPs present in the population with the risk of B-ALL. RESULTS Risk allele frequency A at locus 13q12.2 (rs35958982, FLT3) was conspicuous and showed positive association (OR = 2.30, CI [1.20–4.50], P = 0.005) but genotype frequency (OR = 3.67, CI [0.75–18.10], P = 0.088) failed to show any association with the disease. At locus 9p13.2 (rs3780135, PAX5), the risk allele frequency was significantly higher in B-ALL subjects than ancestral allele frequency (OR = 2.17, CI [1.37–3.43], P = 0.000). Genotype frequency analysis of rs3780135 polymorphism exhibited the protective effect (OR = 0.55, CI [0.72–1.83], P = 0.029). At locus 13q12.2 (rs12430881, FLT3), the minor allele frequency G (OR = 1.15, CI [1.37–3.43], P = 0.043) and genotype frequency (OR = 2.52, P = 0.006) reached significance as showed p < 0.05. CONCLUSION In the present study, a strong risk of B-cell acute lymphoblastic leukemia was associated with rs35958982 and rs12430881 polymorphisms. However, rs3780135 polymorphism showed the protective effect. Additionally, other demographic factors like family history, smoking and consanguinity were also found to be important in risk assessment. We anticipate that the information from genetic variations in this study can aid in therapeutic approach in the future.
Collapse
Affiliation(s)
- Ammara Khalid
- Department of Microbiology & Molecular Genetics, Quaid-e-Azam Campus, University of the Punjab, Lahore, Pakistan
| | - Sara Aslam
- Department of Microbiology & Molecular Genetics, Quaid-e-Azam Campus, University of the Punjab, Lahore, Pakistan
| | - Mehboob Ahmed
- Department of Microbiology & Molecular Genetics, Quaid-e-Azam Campus, University of the Punjab, Lahore, Pakistan
| | - Shahida Hasnain
- Department of Microbiology & Molecular Genetics, Quaid-e-Azam Campus, University of the Punjab, Lahore, Pakistan
| | - Aimen Aslam
- Department of Statistics and Actuarial Science, Quaid-e-Azam Campus, University of the Punjab, Lahore, Pakistan
| |
Collapse
|
25
|
Wang Q, Chen Y, Park J, Liu X, Hu Y, Wang T, McFarland K, Betenbaugh MJ. Design and Production of Bispecific Antibodies. Antibodies (Basel) 2019; 8:antib8030043. [PMID: 31544849 PMCID: PMC6783844 DOI: 10.3390/antib8030043] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 07/18/2019] [Accepted: 07/31/2019] [Indexed: 02/07/2023] Open
Abstract
With the current biotherapeutic market dominated by antibody molecules, bispecific antibodies represent a key component of the next-generation of antibody therapy. Bispecific antibodies can target two different antigens at the same time, such as simultaneously binding tumor cell receptors and recruiting cytotoxic immune cells. Structural diversity has been fast-growing in the bispecific antibody field, creating a plethora of novel bispecific antibody scaffolds, which provide great functional variety. Two common formats of bispecific antibodies on the market are the single-chain variable fragment (scFv)-based (no Fc fragment) antibody and the full-length IgG-like asymmetric antibody. Unlike the conventional monoclonal antibodies, great production challenges with respect to the quantity, quality, and stability of bispecific antibodies have hampered their wider clinical application and acceptance. In this review, we focus on these two major bispecific types and describe recent advances in the design, production, and quality of these molecules, which will enable this important class of biologics to reach their therapeutic potential.
Collapse
Affiliation(s)
- Qiong Wang
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yiqun Chen
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jaeyoung Park
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Xiao Liu
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yifeng Hu
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Tiexin Wang
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Kevin McFarland
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Michael J Betenbaugh
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA.
| |
Collapse
|
26
|
Liu D, Zhao J, Song Y, Luo X, Yang T. Clinical trial update on bispecific antibodies, antibody-drug conjugates, and antibody-containing regimens for acute lymphoblastic leukemia. J Hematol Oncol 2019; 12:15. [PMID: 30736842 PMCID: PMC6368716 DOI: 10.1186/s13045-019-0703-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 02/01/2019] [Indexed: 02/07/2023] Open
Abstract
The relapse rate remains high after chemotherapy for adult patients with acute lymphoblastic leukemia (ALL). With better molecular diagnosis and classification as well as better assessment for minimal residual disease, major progress in the treatment for refractory and/or relapsed ALL is being made. In addition to the tyrosine kinase inhibitors (TKIs) for Philadelphia chromosome-positive ALL, immunotherapeutic agents, blinatumomab, inotuzumab ozogamicin (INO), and chimeric antigen receptor (CAR) T cells, are changing the treatment paradigm for ALL. Blinatumomab and INO are being incorporated into induction chemotherapy regimens and combined with TKIs for ALL therapy. A novel low-intensity regimen, miniHCVD-INO-blinatumomab, appears to be less toxic and more effective than conventional intensive chemotherapy regimens. This review summarized new therapeutic researches of ALL and updated latest progress in clinical trials on bispecific antibodies, antibody-drug conjugates, and new regimens incorporating these novel antibodies.
Collapse
Affiliation(s)
- Delong Liu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
| | - Juanjuan Zhao
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Yongping Song
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Xiaofeng Luo
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou, 350001 Fujian China
| | - Ting Yang
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou, 350001 Fujian China
| |
Collapse
|
27
|
James AR, Schroeder MP, Neumann M, Bastian L, Eckert C, Gökbuget N, Tanchez JO, Schlee C, Isaakidis K, Schwartz S, Burmeister T, von Stackelberg A, Rieger MA, Göllner S, Horstman M, Schrappe M, Kirschner-Schwabe R, Brüggemann M, Müller-Tidow C, Serve H, Akalin A, Baldus CD. Long non-coding RNAs defining major subtypes of B cell precursor acute lymphoblastic leukemia. J Hematol Oncol 2019; 12:8. [PMID: 30642353 PMCID: PMC6332539 DOI: 10.1186/s13045-018-0692-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 12/25/2018] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) have emerged as a novel class of RNA due to its diverse mechanism in cancer development and progression. However, the role and expression pattern of lncRNAs in molecular subtypes of B cell acute lymphoblastic leukemia (BCP-ALL) have not yet been investigated. Here, we assess to what extent lncRNA expression and DNA methylation is driving the progression of relapsed BCP-ALL subtypes and we determine if the expression and DNA methylation profile of lncRNAs correlates with established BCP-ALL subtypes. METHODS We performed RNA sequencing and DNA methylation (Illumina Infinium microarray) of 40 diagnosis and 42 relapse samples from 45 BCP-ALL patients in a German cohort and quantified lncRNA expression. Unsupervised clustering was applied to ascertain and confirm that the lncRNA-based classification of the BCP-ALL molecular subtypes is present in both our cohort and an independent validation cohort of 47 patients. A differential expression and differential methylation analysis was applied to determine the subtype-specific, relapse-specific, and differentially methylated lncRNAs. Potential functions of subtype-specific lncRNAs were determined by using co-expression-based analysis on nearby (cis) and distally (trans) located protein-coding genes. RESULTS Using an integrative Bioinformatics analysis, we developed a comprehensive catalog of 1235 aberrantly dysregulated BCP-ALL subtype-specific and 942 relapse-specific lncRNAs and the methylation profile of three subtypes of BCP-ALL. The 1235 subtype-specific lncRNA signature represented a similar classification of the molecular subtypes of BCP-ALL in the independent validation cohort. We identified a strong correlation between the DUX4-specific lncRNAs and genes involved in the activation of TGF-β and Hippo signaling pathways. Similarly, Ph-like-specific lncRNAs were correlated with genes involved in the activation of PI3K-AKT, mTOR, and JAK-STAT signaling pathways. Interestingly, the relapse-specific lncRNAs correlated with the activation of metabolic and signaling pathways. Finally, we found 23 promoter methylated lncRNAs epigenetically facilitating their expression levels. CONCLUSION Here, we describe a set of subtype-specific and relapse-specific lncRNAs from three major BCP-ALL subtypes and define their potential functions and epigenetic regulation. The subtype-specific lncRNAs are reproducible and can effectively stratify BCP-ALL subtypes. Our data uncover the diverse mechanism of action of lncRNAs in BCP-ALL subtypes defining which lncRNAs are involved in the pathogenesis of disease and are relevant for the stratification of BCP-ALL subtypes.
Collapse
Affiliation(s)
- Alva Rani James
- Department of Hematology and Oncology, Charité, University Hospital Berlin, Campus Benjamin Franklin, 12203, Berlin, Germany
- German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
- German Cancer Consortium (DKTK), 69120, Heidelberg, Germany
| | - Michael P Schroeder
- Department of Hematology and Oncology, Charité, University Hospital Berlin, Campus Benjamin Franklin, 12203, Berlin, Germany
| | - Martin Neumann
- Department of Hematology and Oncology, Charité, University Hospital Berlin, Campus Benjamin Franklin, 12203, Berlin, Germany
- German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
- German Cancer Consortium (DKTK), 69120, Heidelberg, Germany
| | - Lorenz Bastian
- Department of Hematology and Oncology, Charité, University Hospital Berlin, Campus Benjamin Franklin, 12203, Berlin, Germany
- German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
- German Cancer Consortium (DKTK), 69120, Heidelberg, Germany
| | - Cornelia Eckert
- German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
- German Cancer Consortium (DKTK), 69120, Heidelberg, Germany
- Department of Pediatric Hematology/Oncology, Charité, University Hospital Berlin, Campus Rudolf Virchow, 13353, Berlin, Germany
| | - Nicola Gökbuget
- German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
- German Cancer Consortium (DKTK), 69120, Heidelberg, Germany
- Department of Medicine II, Department of Hematology/Oncology, Goethe University Hospital, 60590, Frankfurt/M, Germany
| | - Jutta Ortiz Tanchez
- Department of Hematology and Oncology, Charité, University Hospital Berlin, Campus Benjamin Franklin, 12203, Berlin, Germany
| | - Cornelia Schlee
- Department of Hematology and Oncology, Charité, University Hospital Berlin, Campus Benjamin Franklin, 12203, Berlin, Germany
| | - Konstandina Isaakidis
- Department of Hematology and Oncology, Charité, University Hospital Berlin, Campus Benjamin Franklin, 12203, Berlin, Germany
| | - Stefan Schwartz
- Department of Hematology and Oncology, Charité, University Hospital Berlin, Campus Benjamin Franklin, 12203, Berlin, Germany
| | - Thomas Burmeister
- Department of Hematology, Oncology and Tumor Immunology, Charité University Hospital Berlin, Campus Virchow-Klinikum, 13353, Berlin, Germany
| | - Arend von Stackelberg
- German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
- German Cancer Consortium (DKTK), 69120, Heidelberg, Germany
- Department of Pediatric Hematology/Oncology, Charité, University Hospital Berlin, Campus Rudolf Virchow, 13353, Berlin, Germany
| | - Michael A Rieger
- German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
- German Cancer Consortium (DKTK), 69120, Heidelberg, Germany
- Department of Medicine II, Department of Hematology/Oncology, Goethe University Hospital, 60590, Frankfurt/M, Germany
| | - Stefanie Göllner
- Department of Hematology, Oncology & Rheumatology, University Clinic Heidelberg, 69120, Heidelberg, Germany
| | - Martin Horstman
- Department of Pediatric Hematology and Oncology, Research Institute Children's Cancer Center, University Medical Center Hamburg, 20251, Hamburg, Germany
| | - Martin Schrappe
- Department of Pediatrics, University Hospital Schleswig-Holstein, Campus Kiel, 24105, Kiel, Germany
| | - Renate Kirschner-Schwabe
- German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
- German Cancer Consortium (DKTK), 69120, Heidelberg, Germany
- Department of Pediatric Hematology/Oncology, Charité, University Hospital Berlin, Campus Rudolf Virchow, 13353, Berlin, Germany
| | - Monika Brüggemann
- Department of Hematology and Oncology, University Hospital Schleswig-Holstein, Campus Kiel, 24105, Kiel, Germany
| | - Carsten Müller-Tidow
- Department of Hematology, Oncology & Rheumatology, University Clinic Heidelberg, 69120, Heidelberg, Germany
| | - Hubert Serve
- German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
- German Cancer Consortium (DKTK), 69120, Heidelberg, Germany
- Department of Medicine II, Department of Hematology/Oncology, Goethe University Hospital, 60590, Frankfurt/M, Germany
| | - Altuna Akalin
- Bioinformatics Platform, Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center (MDC), 13125, Berlin, Germany
| | - Claudia D Baldus
- Department of Hematology and Oncology, Charité, University Hospital Berlin, Campus Benjamin Franklin, 12203, Berlin, Germany.
- German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.
- German Cancer Consortium (DKTK), 69120, Heidelberg, Germany.
- Department of Hematology and Oncology, University Hospital Schleswig-Holstein, Campus Kiel, 24105, Kiel, Germany.
| |
Collapse
|
28
|
Ghelli Luserna Di Rorà A, Beeharry N, Imbrogno E, Ferrari A, Robustelli V, Righi S, Sabattini E, Verga Falzacappa MV, Ronchini C, Testoni N, Baldazzi C, Papayannidis C, Abbenante MC, Marconi G, Paolini S, Parisi S, Sartor C, Fontana MC, De Matteis S, Iacobucci I, Pelicci PG, Cavo M, Yen TJ, Martinelli G. Targeting WEE1 to enhance conventional therapies for acute lymphoblastic leukemia. J Hematol Oncol 2018; 11:99. [PMID: 30068368 PMCID: PMC6090987 DOI: 10.1186/s13045-018-0641-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/12/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Despite the recent progress that has been made in the understanding and treatment of acute lymphoblastic leukemia (ALL), the outcome is still dismal in adult ALL cases. Several studies in solid tumors identified high expression of WEE1 kinase as a poor prognostic factor and reported its role as a cancer-conserving oncogene that protects cancer cells from DNA damage. Therefore, the targeted inhibition of WEE1 kinase has emerged as a rational strategy to sensitize cancer cells to antineoplastic compounds, which we evaluate in this study. METHODS The effectiveness of the selective WEE1 inhibitor AZD-1775 as a single agent and in combination with different antineoplastic agents in B and T cell precursor ALL (B/T-ALL) was evaluated in vitro and ex vivo studies. The efficacy of the compound in terms of cytotoxicity, induction of apoptosis, and changes in gene and protein expression was assessed using different B/T-ALL cell lines and confirmed in primary ALL blasts. RESULTS We showed that WEE1 was highly expressed in adult primary ALL bone marrow and peripheral blood blasts (n = 58) compared to normal mononuclear cells isolated from the peripheral blood of healthy donors (p = 0.004). Thus, we hypothesized that WEE1 could be a rational target in ALL, and its inhibition could enhance the cytotoxicity of conventional therapies used for ALL. We evaluated the efficacy of AZD-1775 as a single agent and in combination with several antineoplastic agents, and we elucidated its mechanisms of action. AZD-1775 reduced cell viability in B/T-ALL cell lines by disrupting the G2/M checkpoint and inducing apoptosis. These findings were confirmed in human primary ALL bone marrow and peripheral blood blasts (n = 15). In both cell lines and primary leukemic cells, AZD-1775 significantly enhanced the efficacy of several tyrosine kinase inhibitors (TKIs) such as bosutinib, imatinib, and ponatinib, and of chemotherapeutic agents (clofarabine and doxorubicin) in terms of the reduction of cell viability, apoptosis induction, and inhibition of proliferation. CONCLUSIONS Our data suggest that WEE1 plays a role in ALL blast's survival and is a bona fide target for therapeutic intervention. These data support the evaluation of the therapeutic potential of AZD-1775 as chemo-sensitizer agent for the treatment of B/T-ALL.
Collapse
Affiliation(s)
- Andrea Ghelli Luserna Di Rorà
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology “L. e A. Seràgnoli”, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Neil Beeharry
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA USA
- LAM Therapeutics, Guilford, CT USA
| | - Enrica Imbrogno
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology “L. e A. Seràgnoli”, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Anna Ferrari
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology “L. e A. Seràgnoli”, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Valentina Robustelli
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology “L. e A. Seràgnoli”, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Simona Righi
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology “L. e A. Seràgnoli”, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Elena Sabattini
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology “L. e A. Seràgnoli”, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | | | - Chiara Ronchini
- Laboratory of Clinical Genomics, European Institute of Oncology, Milan, Italy
| | - Nicoletta Testoni
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology “L. e A. Seràgnoli”, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Carmen Baldazzi
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology “L. e A. Seràgnoli”, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Cristina Papayannidis
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology “L. e A. Seràgnoli”, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Maria Chiara Abbenante
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology “L. e A. Seràgnoli”, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Giovanni Marconi
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology “L. e A. Seràgnoli”, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Stefania Paolini
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology “L. e A. Seràgnoli”, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Sarah Parisi
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology “L. e A. Seràgnoli”, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Chiara Sartor
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology “L. e A. Seràgnoli”, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Maria Chiara Fontana
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology “L. e A. Seràgnoli”, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Serena De Matteis
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Ilaria Iacobucci
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology “L. e A. Seràgnoli”, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN USA
| | | | - Michele Cavo
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology “L. e A. Seràgnoli”, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Timothy J. Yen
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA USA
| | - Giovanni Martinelli
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| |
Collapse
|
29
|
Buatois V, Johnson Z, Salgado-Pires S, Papaioannou A, Hatterer E, Chauchet X, Richard F, Barba L, Daubeuf B, Cons L, Broyer L, D'Asaro M, Matthes T, LeGallou S, Fest T, Tarte K, Clarke Hinojosa RK, Genescà Ferrer E, Ribera JM, Dey A, Bailey K, Fielding AK, Eissenberg L, Ritchey J, Rettig M, DiPersio JF, Kosco-Vilbois MH, Masternak K, Fischer N, Shang L, Ferlin WG. Preclinical Development of a Bispecific Antibody that Safely and Effectively Targets CD19 and CD47 for the Treatment of B-Cell Lymphoma and Leukemia. Mol Cancer Ther 2018; 17:1739-1751. [PMID: 29743205 DOI: 10.1158/1535-7163.mct-17-1095] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 02/15/2018] [Accepted: 05/04/2018] [Indexed: 12/14/2022]
Abstract
CD47, an ubiquitously expressed innate immune checkpoint receptor that serves as a universal "don't eat me" signal of phagocytosis, is often upregulated by hematologic and solid cancers to evade immune surveillance. Development of CD47-targeted modalities is hindered by the ubiquitous expression of the target, often leading to rapid drug elimination and hemotoxicity including anemia. To overcome such liabilities, we have developed a fully human bispecific antibody, NI-1701, designed to coengage CD47 and CD19 selectively on B cells. NI-1701 demonstrates favorable elimination kinetics with no deleterious effects seen on hematologic parameters following single or multiple administrations to nonhuman primates. Potent in vitro and in vivo activity is induced by NI-1701 to kill cancer cells across a plethora of B-cell malignancies and control tumor growth in xenograft mouse models. The mechanism affording maximal tumor growth inhibition by NI-1701 is dependent on the coengagement of CD47/CD19 on B cells inducing potent antibody-dependent cellular phagocytosis of the targeted cells. NI-1701-induced control of tumor growth in immunodeficient NOD/SCID mice was more effective than that achieved with the anti-CD20 targeted antibody, rituximab. Interestingly, a synergistic effect was seen when tumor-implanted mice were coadministered NI-1701 and rituximab leading to significantly improved tumor growth inhibition and regression in some animals. We describe herein, a novel bispecific antibody approach aimed at sensitizing B cells to become more readily phagocytosed and eliminated thus offering an alternative or adjunct therapeutic option to patients with B-cell malignancies refractory/resistant to anti-CD20-targeted therapy. Mol Cancer Ther; 17(8); 1739-51. ©2018 AACR.
Collapse
Affiliation(s)
| | - Zoë Johnson
- Novimmune S.A., Plan-les-Ouates, Switzerland
| | | | | | | | | | | | | | | | - Laura Cons
- Novimmune S.A., Plan-les-Ouates, Switzerland
| | | | | | - Thomas Matthes
- Medical Faculty, University of Geneva, Genève, Switzerland
| | | | - Thierry Fest
- Rennes 1 University, Inserm U1236, Rennes, France
| | - Karin Tarte
- Rennes 1 University, Inserm U1236, Rennes, France
| | - Robert K Clarke Hinojosa
- Josep Carreras Leukaemia Research Institute (IJC), Campus ICO-Germans Trias Pujol, Badalona, Barcelona, Spain
| | - Eulàlia Genescà Ferrer
- Josep Carreras Leukaemia Research Institute (IJC), Campus ICO-Germans Trias Pujol, Badalona, Barcelona, Spain
| | - José María Ribera
- Josep Carreras Leukaemia Research Institute (IJC), Campus ICO-Germans Trias Pujol, Badalona, Barcelona, Spain
| | - Aditi Dey
- Paul O'Gorman Building, University College London (UCL) Cancer Institute, London, United Kingdom
| | - Katharine Bailey
- Paul O'Gorman Building, University College London (UCL) Cancer Institute, London, United Kingdom
| | - Adele K Fielding
- Paul O'Gorman Building, University College London (UCL) Cancer Institute, London, United Kingdom
| | - Linda Eissenberg
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Julie Ritchey
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Michael Rettig
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - John F DiPersio
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | | | | | | | - Limin Shang
- Novimmune S.A., Plan-les-Ouates, Switzerland
| | | |
Collapse
|
30
|
McCusker MG, El Chaer F, Duffy A, Emadi A, Duong VH. Combination of Blinatumomab and Vincristine Sulfate Liposome Injection for Treatment of Relapsed Philadelphia Chromosome Positive B-cell Acute Lymphoblastic Leukemia. AMERICAN JOURNAL OF LEUKEMIA RESEARCH 2018; 2:1010. [PMID: 29953129 PMCID: PMC6018018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Relapsed Philadelphia chromosome (Ph) positive Acute Lymphoblastic Leukemia (ALL) is an aggressive lymphoid malignancy with a poor prognosis and no randomized studies demonstrating superiority of any single salvage regimen. We present the case of a 33-year-old woman with relapsed Ph positive precursor (pre) B-cell ALL with rapidly rising peripheral blasts while on blinatumomab monotherapy initially, but ultimately responded with the addition of Vincristine Sulfate Liposome Injection (VSLI). Ponatinib was added later when it became available for the patient, and she ultimately achieved a complete remission. Further study is warranted to explore mechanisms of potential synergy, and the safety and efficacy of the combination of blinatumomab and VSLI.
Collapse
Affiliation(s)
| | | | - Alison Duffy
- University of Maryland School of Medicine, USA
- University of Maryland School of Pharmacy, USA
| | | | - Vu H. Duong
- University of Maryland School of Medicine, USA
| |
Collapse
|
31
|
Schütz C, Varela JC, Perica K, Haupt C, Oelke M, Schneck JP. Antigen-specific T cell Redirectors: a nanoparticle based approach for redirecting T cells. Oncotarget 2018; 7:68503-68512. [PMID: 27602488 PMCID: PMC5356569 DOI: 10.18632/oncotarget.11785] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 07/27/2016] [Indexed: 12/20/2022] Open
Abstract
Redirection of T cells to target and destroy tumors has become an important clinical tool and major area of research in tumor immunology. Here we present a novel, nanoparticle-based approach to selectively bind antigen-specific cytotoxic T cells (CTL) and redirect them to kill tumors, termed ATR (Antigen-specific T cell Redirectors). ATR were generated by decorating nanoparticles with both an antigen-specific T cell binding moiety, either peptide loaded MHC-Ig dimer or clonotypic anti-TCR antibody, and a model tumor cell binding moiety, anti-CD19 antibody to engage CD19+ tumor cells. ATR stably bind tumor cells and CTL in a dose dependent fashion and stimulate antigen-specific conjugate formation between those cells. ATR induced redirected lysis of tumor cells in vitro, as demonstrated by 51Cr-release killing. In vivo ATR administration led to reduced tumor growth in a SCID/beige human lymphoma treatment model. In summary, ATR represent a novel, nanoparticle based approach for redirecting antigen-specific CTL to kill tumors.
Collapse
Affiliation(s)
- Christian Schütz
- Institute of Cell Engineering and Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA.,Current address: Division of Immunology, Paul-Ehrlich-Institut, Langen, Germany
| | - Juan Carlos Varela
- Division of Hematology, Department of Medicine, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Karlo Perica
- Institute of Cell Engineering and Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Carl Haupt
- Institute of Cell Engineering and Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Mathias Oelke
- Institute of Cell Engineering and Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA.,NexImmune Inc., Gaithersburg, Maryland, USA
| | - Jonathan P Schneck
- Institute of Cell Engineering and Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
32
|
Postel-Vinay S, Soria JC. Immune Therapies in Phase 1 Trials. Oncoimmunology 2018. [DOI: 10.1007/978-3-319-62431-0_32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
33
|
Fleisher B, Ait-Oudhia S. A retrospective examination of the US Food and Drug Administration's clinical pharmacology reviews of oncology biologics for potential use of therapeutic drug monitoring. Onco Targets Ther 2017; 11:113-121. [PMID: 29343970 PMCID: PMC5749565 DOI: 10.2147/ott.s153056] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background Biologics have gained traction for use in oncology, but have demonstrate clinical variability for efficacy and safety. Therapeutic drug monitoring (TDM) can benefit patients’ outcomes from a biologic therapy when the latter has a defined therapeutic window. A clinically relevant therapeutic window may exist for biologics with established exposure-response (E–R) relationships for efficacy and/or safety and a documented maximum tolerated dose (MTD). Additionally, the inter-individual variability (IIV) on the clearance (CL) parameter could determine risks for patients falling outside the proposed therapeutic window. Materials and methods The US Food and Drug Administration (FDA)-approved oncology biologics between 2005–2016 were reviewed via FDA “Purple Book” (FDA-repository for licensed biologics). Data were extracted from biologics’ pharmacokinetic models available on the clinical pharmacology reviews published on the FDA-Approved Drug Products website. Evaluated features for biologics with established E–R relationships for efficacy and/or safety and MTD include an IIV for the CL and various other covariates including demographic factors, disease factors, blood chemistry, or immunogenicity. Results Five therapies were identified with documented E–R relationships for both efficacy and safety including, Yervoy®(ipilimumab), Zaltrap® (ziv-aflibercept), Portrazza® (necitumumab), Adcetris® (brentuximab-vedotin), and Blincyto® (blinatumomab). The corresponding IIV on CL were: 34%, 33%, 29%, 47%, and 97%, respectively. Among the five therapies, only three had defined MTD including, brentuximab-vedotin, necitumumab, and blinatumomab. Conclusion Of the medications examined, blinatumomab was identified as the anticancer drug with the most available information for the establishment of TDM, and hence, may benefit through the use of TDM to optimize effectiveness and minimize patients’ toxicity. The approach used here may provide a generalizable framework to retrospectively identify anticancer biologics with high IIV that may benefit from TDM to improve patients’ clinical outcome.
Collapse
Affiliation(s)
- Brett Fleisher
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, USA
| | - Sihem Ait-Oudhia
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, USA
| |
Collapse
|
34
|
Du J, Cao Y, Liu Y, Wang Y, Zhang Y, Fu G, Zhang Y, Lu L, Luo X, Kim CH, Schultz PG, Wang F. Engineering Bifunctional Antibodies with Constant Region Fusion Architectures. J Am Chem Soc 2017; 139:18607-18615. [PMID: 29186655 DOI: 10.1021/jacs.7b09641] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We report a method to generate bifunctional antibodies by grafting full-length proteins into constant region loops of a full-length antibody or an antigen-binding fragment (Fab). The fusion proteins retain the antigen binding activity of the parent antibody but have an additional activity associated with the protein insert. The engineered antibodies have excellent in vitro activity, physiochemical properties, and stability. Among these, a Her2 × CD3 bispecific antibody (BsAb) was constructed by inserting an anti-Her2 single-chain variable fragment (ScFv) into an anti-CD3 Fab. This bispecific antibody efficiently induces targeted cell lysis in the presence of effector cells at as low as sub-picomolar concentrations in vitro. Moreover, the Her2 × CD3 BsAb shows potent in vivo antitumor activity in mouse Her22+ and Her21+ xenograft models. These results demonstrate that insertion of a full-length protein into non-CDR loops of antibodies provides a feasible approach to generate multifunctional antibodies for therapeutic applications.
Collapse
Affiliation(s)
- Juanjuan Du
- California Institute for Biomedical Research , 11119 N. Torrey Pines Road, La Jolla, California 92037, United States.,Department of Chemistry, The Scripps Research Institute , 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Yu Cao
- Department of Chemistry, The Scripps Research Institute , 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Yan Liu
- California Institute for Biomedical Research , 11119 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Ying Wang
- California Institute for Biomedical Research , 11119 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Yong Zhang
- California Institute for Biomedical Research , 11119 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Guangsen Fu
- California Institute for Biomedical Research , 11119 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Yuhan Zhang
- California Institute for Biomedical Research , 11119 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Lucy Lu
- California Institute for Biomedical Research , 11119 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Xiaozhou Luo
- Department of Chemistry, The Scripps Research Institute , 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Chan Hyuk Kim
- California Institute for Biomedical Research , 11119 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Peter G Schultz
- California Institute for Biomedical Research , 11119 N. Torrey Pines Road, La Jolla, California 92037, United States.,Department of Chemistry, The Scripps Research Institute , 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Feng Wang
- California Institute for Biomedical Research , 11119 N. Torrey Pines Road, La Jolla, California 92037, United States
| |
Collapse
|
35
|
Fedorov VD, Upadhyay VA, Fathi AT. The Approach to Acute Lymphoblastic Leukemia in Older Patients: Conventional Treatments and Emerging Therapies. Curr Hematol Malig Rep 2017; 11:165-74. [PMID: 26939921 DOI: 10.1007/s11899-016-0316-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Acute lymphoblastic leukemia (ALL) among older adult patients presents significant clinical challenges. As opposed to pediatric populations, in whom long-term outcomes are markedly superior, those for adults remain grim. Nevertheless, younger adults with ALL have experienced a steady improvement in long-term survival in the last few decades. This is significantly different for older ALL patients, for whom long-term outcomes remain poor. Conventional chemotherapies are associated with sub-optimal outcomes and increased toxicity in this population. However, several emerging therapies, including antibody-drug conjugates, bi-specific engagers, and chimeric antigen receptor (CAR) T cells, have demonstrated much promise and are either incorporated into the existing therapeutic paradigms or being actively investigated to improve outcomes.
Collapse
Affiliation(s)
- Victor D Fedorov
- Stanbury Service PSP Group, Department of Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Vivek A Upadhyay
- Department of Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Amir T Fathi
- Harvard Medical School, Massachusetts General Hospital, Division of Hematology and Medical Oncology, Zero Emerson Place, Suite 118, Boston, MA, 02114, USA.
| |
Collapse
|
36
|
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: 134] [Impact Index Per Article: 19.1] [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
|
37
|
Lopez-Albaitero A, Xu H, Guo H, Wang L, Wu Z, Tran H, Chandarlapaty S, Scaltriti M, Janjigian Y, de Stanchina E, Cheung NKV. Overcoming resistance to HER2-targeted therapy with a novel HER2/CD3 bispecific antibody. Oncoimmunology 2017; 6:e1267891. [PMID: 28405494 PMCID: PMC5384386 DOI: 10.1080/2162402x.2016.1267891] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/21/2016] [Accepted: 11/24/2016] [Indexed: 01/21/2023] Open
Abstract
T-cell-based therapies have emerged as one of the most clinically effective ways to target solid and non-solid tumors. HER2 is responsible for the oncogenesis and treatment resistance of several human solid tumors. As a member of the HER family of tyrosine kinase receptors, its over-activity confers unfavorable clinical outcome. Targeted therapies directed at this receptor have achieved responses, although development of resistance is common. We explored a novel HER2/CD3 bispecific antibody (HER2-BsAb) platform that while preserving the anti-proliferative effects of trastuzumab, it recruits and activates non-specific circulating T-cells, promoting T cell tumor infiltration and ablating HER2(+) tumors, even when these are resistant to standard HER2-targeted therapies. Its in vitro tumor cytotoxicity, when expressed as EC50, correlated with the surface HER2 expression in a large panel of human tumor cell lines, irrespective of lineage or tumor type. HER2-BsAb-mediated cytotoxicity was relatively insensitive to PD-1/PD-L1 immune checkpoint inhibition. In four separate humanized mouse models of human breast cancer and ovarian cancer cell line xenografts, as well as human breast cancer and gastric cancer patient-derived xenografts (PDXs), HER2-BsAb was highly effective in promoting T cell infiltration and suppressing tumor growth when used in the presence of human peripheral blood mononuclear cells (PBMC) or activated T cells (ATC). The in vivo and in vitro antitumor properties of this BsAb support its further clinical development as a cancer immunotherapeutic.
Collapse
Affiliation(s)
| | - Hong Xu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hongfen Guo
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Linlin Wang
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zhihao Wu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hoa Tran
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sarat Chandarlapaty
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maurizio Scaltriti
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yelena Janjigian
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elisa de Stanchina
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nai-Kong V. Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
38
|
Godar M, Morello V, Sadi A, Hultberg A, De Jonge N, Basilico C, Hanssens V, Saunders M, Lambrecht BN, El Khattabi M, de Haard H, Michieli P, Blanchetot C. Dual anti-idiotypic purification of a novel, native-format biparatopic anti-MET antibody with improved in vitro and in vivo efficacy. Sci Rep 2016; 6:31621. [PMID: 27546726 PMCID: PMC4992859 DOI: 10.1038/srep31621] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 07/22/2016] [Indexed: 12/15/2022] Open
Abstract
Bispecific antibodies are of great interest due to their ability to simultaneously bind and engage different antigens or epitopes. Nevertheless, it remains a challenge to assemble, produce and/or purify them. Here we present an innovative dual anti-idiotypic purification process, which provides pure bispecific antibodies with native immunoglobulin format. Using this approach, a biparatopic IgG1 antibody targeting two distinct, HGF-competing, non-overlapping epitopes on the extracellular region of the MET receptor, was purified with camelid single-domain antibody fragments that bind specifically to the correct heavy chain/light chain pairings of each arm. The purity and functionality of the anti-MET biparatopic antibody was then confirmed by mass spectrometry and binding experiments, demonstrating its ability to simultaneously target the two epitopes recognized by the parental monoclonal antibodies. The improved MET-inhibitory activity of the biparatopic antibody compared to the parental monoclonal antibodies, was finally corroborated in cell-based assays and more importantly in a tumor xenograft mouse model. In conclusion, this approach is fast and specific, broadly applicable and results in the isolation of a pure, novel and native-format anti-MET biparatopic antibody that shows superior biological activity over the parental monospecific antibodies both in vitro and in vivo.
Collapse
Affiliation(s)
- Marie Godar
- argenx BVBA, Industriepark Zwijnaarde 7, Building C, 9052 Zwijnaarde, Belgium.,VIB Inflammation Research Center 9052 Zwijnaarde, Belgium.,Department of Internal Medicine, Ghent University, 9000 Ghent, Belgium
| | - Virginia Morello
- Department of Oncology, University of Torino Medical School, 10060 Candiolo, Turin, Italy.,Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Turin, Italy
| | - Ava Sadi
- QVQ BV, Yalelaan 1 Androclus building, 3584 CL Utrecht, The Netherlands
| | - Anna Hultberg
- argenx BVBA, Industriepark Zwijnaarde 7, Building C, 9052 Zwijnaarde, Belgium
| | - Natalie De Jonge
- argenx BVBA, Industriepark Zwijnaarde 7, Building C, 9052 Zwijnaarde, Belgium
| | | | - Valérie Hanssens
- argenx BVBA, Industriepark Zwijnaarde 7, Building C, 9052 Zwijnaarde, Belgium
| | - Michael Saunders
- argenx BVBA, Industriepark Zwijnaarde 7, Building C, 9052 Zwijnaarde, Belgium
| | - Bart N Lambrecht
- VIB Inflammation Research Center 9052 Zwijnaarde, Belgium.,Department of Internal Medicine, Ghent University, 9000 Ghent, Belgium.,Department of Pulmonary Medicine, ErasmusMC, 3015 GE Rotterdam, The Netherlands
| | | | - Hans de Haard
- argenx BVBA, Industriepark Zwijnaarde 7, Building C, 9052 Zwijnaarde, Belgium
| | - Paolo Michieli
- Department of Oncology, University of Torino Medical School, 10060 Candiolo, Turin, Italy.,Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Turin, Italy
| | | |
Collapse
|
39
|
Reusch U, Duell J, Ellwanger K, Herbrecht C, Knackmuss SH, Fucek I, Eser M, McAleese F, Molkenthin V, Gall FL, Topp M, Little M, Zhukovsky EA. A tetravalent bispecific TandAb (CD19/CD3), AFM11, efficiently recruits T cells for the potent lysis of CD19(+) tumor cells. MAbs 2016; 7:584-604. [PMID: 25875246 DOI: 10.1080/19420862.2015.1029216] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
To harness the potent tumor-killing capacity of T cells for the treatment of CD19(+) malignancies, we constructed AFM11, a humanized tetravalent bispecific CD19/CD3 tandem diabody (TandAb) consisting solely of Fv domains. The molecule exhibits good manufacturability and stability properties. AFM11 has 2 binding sites for CD3 and 2 for CD19, an antigen that is expressed from early B cell development through differentiation into plasma cells, and is an attractive alternative to CD20 as a target for the development of therapeutic antibodies to treat B cell malignancies. Comparison of the binding and cytotoxicity of AFM11 with those of a tandem scFv bispecific T cell engager (BiTE) molecule targeting the same antigens revealed that AFM11 elicited more potent in vitro B cell lysis. Though possessing high affinity to CD3, the TandAb mediates serial-killing of CD19(+) cells with little dependence of potency or efficacy upon effector:target ratio, unlike the BiTE. The advantage of the TandAb over the BiTE was most pronounced at lower effector:target ratios. AFM11 mediated strictly target-dependent T cell activation evidenced by CD25 and CD69 induction, proliferation, and cytokine release, notwithstanding bivalent CD3 engagement. In a NOD/scid xenograft model, AFM11 induced dose-dependent growth inhibition of Raji tumors in vivo, and radiolabeled TandAb exhibited excellent localization to tumor but not to normal tissue. After intravenous administration in mice, half-life ranged from 18.4 to 22.9 h. In a human ex vivo B-cell chronic lymphocytic leukemia study, AFM11 exhibited substantial cytotoxic activity in an autologous setting. Thus, AFM11 may represent a promising therapeutic for treatment of CD19(+) malignancies with an advantageous safety risk profile and anticipated dosing regimen.
Collapse
Key Words
- ALL
- AUCtot, total area under the curve
- B-ALL, B-precursor acute lymphoblastic leukemia
- BBB, blood-brain barrier
- BiTE, bispecific T cell engager
- CAR, chimeric antigen receptor
- CCS, cell culture supernatant
- CD, cluster of differentiation
- CD3
- CDR, complementarity determining region
- CHO, Chinese hamster ovary
- CL, clearance
- CLL, chronic lymphocytic leukemia
- CNS, central nervous system
- Cmax, maximal concentration
- DMSO, dimethyl sulfoxide
- E:T, effector:target
- EC50, half maximal effective concentration
- ECL, electrochemiluminescence
- F, fluorescence
- FACS, fluorescence-activated cell sorting
- FCS, fetal calf serum
- FR, framework region
- Fab, fragment antigen-binding
- Fc, fragment crystallizable
- FcRn, neonatal Fc receptor
- FcgR, Fc gamma receptor
- Fv, variable fragment
- HMF, high molecular weight forms
- HSA, human serum albumin
- His, histidine
- IFN, interferon
- IL, interleukin
- IgG, immunoglobulin G
- KD, dissociation constant
- LMF, low molecular weight forms
- MSD, MesoScale Discovery
- MWCO, molecular weight cut-off
- NHL, non-Hodgkin lymphoma
- NK, natural killer
- NOD/scid, nonobese diabetic/severe combined immunodeficiency
- Non-Hodgkin lymphoma
- ORR, overall response rate
- PBMC, peripheral blood mononuclear cell
- PBS, phosphate buffered saline
- PES, polyethersulfone
- PHA, phytohemagglutinin
- PI, propidium iodide
- SABC, standardized antibody binding capacity
- SD, standard deviation
- SDS-PAGE, sodium dodecyl sulfate polyacrylamide gel electrophoresis
- SE-HPLC, size exclusion high-pressure liquid chromatography
- SEC, size exclusion chromatography
- SPR, surface plasmon resonance
- T cells
- TNF, tumor necrosis factor
- TandAb, tandem diabody
- VH, variable heavy
- VL, variable light
- Vss, volume of distribution at steady state
- WBA, whole body autoradiography
- bispecific antibodies
- ctrl., control
- i.v., intravenous
- ka, association rate constant
- kd, dissociation rate constant
- s.c., subcutaneous
- scFv, single-chain variable fragment
- t1/2, terminal elimination half-life
- w/o, without
Collapse
Affiliation(s)
- Uwe Reusch
- a Affimed Therapeutics AG ; Heidelberg , Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Blinatumomab (BLINCYTO™) is a novel, bispecific T-cell engaging antibody that binds cluster of differentiation (CD) 19 antigens on blast cells while also binding and activating the CD3/T cell receptor complex, causing cell lysis. The antibody is being developed by Amgen as a treatment for haematological cancers that originate from B cell lines. Blinatumomab was approved by the US FDA in December 2014 for the treatment of adults with Philadelphia chromosome (Ph)-negative relapsed/refractory B-cell precursor acute lymphoblastic leukaemia (BCP-ALL). It is awaiting approval for this indication in the EU and is in phase III development in various countries. This article summarizes the milestones in the development of blinatumomab leading to its first approval for the treatment of Ph-negative BCP-ALL.
Collapse
|
41
|
Increase of PD-L1 expressing B-precursor ALL cells in a patient resistant to the CD19/CD3-bispecific T cell engager antibody blinatumomab. J Hematol Oncol 2015; 8:111. [PMID: 26449653 PMCID: PMC4599591 DOI: 10.1186/s13045-015-0213-6] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 09/28/2015] [Indexed: 01/19/2023] Open
Abstract
The bispecific T cell engager blinatumomab has shown encouraging clinical activity in B-precursor acute lymphoblastic leukemia (ALL). However, about half of relapsed/refractory patients do not respond to therapy. Here, we present the case of a 32-year-old male patient with refractory B-precursor ALL who was resistant to treatment with blinatumomab. Bone marrow immunohistochemistry revealed T cell infiltrates and an increase in programmed death-ligand 1 (PD-L1)-positive ALL cells as a potential immune escape mechanism. We were able to recapitulate the clinical observation in vitro by showing that blinatumomab was not able to mediate cytotoxicity of CD19-positive ALL cells using autologous T cells. In contrast, the addition of healthy donor T cells led to lysis of ALL cells. These results strongly encourage further systematic evaluation of checkpoint molecules in cases of blinatumomab treatment failure and might highlight a possible mechanism to overcome resistance to this otherwise highly effective treatment.
Collapse
|
42
|
Suzuki M, Curran KJ, Cheung NKV. Chimeric antigen receptors and bispecific antibodies to retarget T cells in pediatric oncology. Pediatr Blood Cancer 2015; 62:1326-36. [PMID: 25832831 PMCID: PMC4976492 DOI: 10.1002/pbc.25513] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 02/20/2015] [Indexed: 12/22/2022]
Abstract
Cancer immunotherapy using antigen-specific T cells has broad therapeutic potential. Chimeric antigen receptors and bispecific antibodies can redirect T cells to kill tumors without human leukocyte antigens (HLA) restriction. Key determinants of clinical potential include the choice of target antigen, antibody specificity, antibody affinity, tumor accessibility, T cell persistence, and tumor immune evasion. For pediatric cancers, additional constraints include their propensity for bulky metastatic disease and the concern for late toxicities from treatment. Nonetheless, the recent preclinical and clinical developments of these T cell based therapies are highly encouraging.
Collapse
Affiliation(s)
- Maya Suzuki
- Department of Pediatrics; Memorial Sloan Kettering Cancer Center; 1275 York Avenue, New York 10065 NY
| | - Kevin J. Curran
- Department of Pediatrics; Memorial Sloan Kettering Cancer Center; 1275 York Avenue, New York 10065 NY
| | - Nai-Kong V. Cheung
- Department of Pediatrics; Memorial Sloan Kettering Cancer Center; 1275 York Avenue, New York 10065 NY
| |
Collapse
|
43
|
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: 4.0] [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
|
44
|
Exploiting light chains for the scalable generation and platform purification of native human bispecific IgG. Nat Commun 2015; 6:6113. [PMID: 25672245 PMCID: PMC4339886 DOI: 10.1038/ncomms7113] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 12/15/2014] [Indexed: 12/23/2022] Open
Abstract
Bispecific antibodies enable unique therapeutic approaches but it remains a challenge to produce them at the industrial scale, and the modifications introduced to achieve bispecificity often have an impact on stability and risk of immunogenicity. Here we describe a fully human bispecific IgG devoid of any modification, which can be produced at the industrial scale, using a platform process. This format, referred to as a κλ-body, is assembled by co-expressing one heavy chain and two different light chains, one κ and one λ. Using ten different targets, we demonstrate that light chains can play a dominant role in mediating specificity and high affinity. The κλ-bodies support multiple modes of action, and their stability and pharmacokinetic properties are indistinguishable from therapeutic antibodies. Thus, the κλ-body represents a unique, fully human format that exploits light-chain variable domains for antigen binding and light-chain constant domains for robust downstream processing, to realize the potential of bispecific antibodies. Bispecific antibodies allow for novel therapeutic approaches but industrial-scale production and immunogenicity represent significant challenges. Here Fischer et al. describe a unique human bispecific antibody format that exploits differing light chains to overcome these obstacles.
Collapse
|
45
|
Naddafi F, Davami F. Anti-CD19 Monoclonal Antibodies: a New Approach to Lymphoma Therapy. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2015; 4:143-51. [PMID: 26629482 PMCID: PMC4644525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
CD19 is expressed on B- lineage cells and follicular dendritic cells and plays a key role in B cell malignancies and autoimmune diseases. Thus, it has been considered as potential target for several monoclonal antibodies (mAbs). For decades, chemotherapy has been known as one of the major antitumor therapies eradicating high proliferative tumor cells. But, anti- CD19 mAbs developed for treating CD19- positive lymphomas and autoimmune diseases would rank among the most novel area of research and development in the pharmaceutical industry. Moreover, several anti- CD19 mAbs are currently being tested in various clinical trials and this review provides an overview of the research accomplished so far.
Collapse
Affiliation(s)
- Fatemeh Naddafi
- Pharmaceutical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Fatemeh Davami
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran. ,Corresponding author: Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran. E-mail:
| |
Collapse
|
46
|
The role of Pax5 in leukemia: diagnosis and prognosis significance. Med Oncol 2014; 32:360. [PMID: 25428382 DOI: 10.1007/s12032-014-0360-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 11/12/2014] [Indexed: 12/12/2022]
Abstract
Pax5 transcription factor, also known as B-cell specific activator protein (BSAP), plays a dual role in the hematopoietic system. Pax5 expression is essential in B-cell precursors for normal differentiation and maturation of B-cells. On the other hand, it inhibits the differentiation and progress toward other lineages. The expression of this factor is involved in several aspects of B-cell differentiation, including commitment, immunoglobulin gene rearrangement, BCR signal transduction and B-cell survival, so that the deletion or inactivating mutations of Pax5 cause cell arrest in Pro-B-cell stage. In recent years, point mutations, deletions and various rearrangements in Pax5 gene have been reported in several types of human cancers. However, no clear relationship has been found between these aberrations and disease prognosis. Specific expression of Pax5 in B-cells can raise it as a marker for the diagnosis and differentiation of B-cell leukemias and lymphomas as well as account for remission or relapse. Extensive studies on Pax5 along with other genes and immunomarkers are necessary for decisive results in this regard.
Collapse
|
47
|
Magge RS, DeAngelis LM. The double-edged sword: Neurotoxicity of chemotherapy. Blood Rev 2014; 29:93-100. [PMID: 25445718 DOI: 10.1016/j.blre.2014.09.012] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 09/17/2014] [Accepted: 09/19/2014] [Indexed: 02/06/2023]
Abstract
The number of available therapies for hematologic malignancies continues to grow at a rapid pace. Unfortunately, many of these treatments carry both central and peripheral nervous system toxicities, potentially limiting a patient's ability to tolerate a full course of treatment. Neurotoxicity with chemotherapy is common and second only to myelosuppression as a reason to limit dosing. This review addresses the neurotoxicity of newly available therapeutic agents including brentuximab vedotin and blinatumomab as well as classic ones such as methotrexate, vinca alkaloids and platinums. Although peripheral neuropathy is common with many drugs, other complications such as seizures and encephalopathy may require more immediate attention. Rapid recognition of adverse neurologic effects may lead to earlier treatment and appropriate adjustment of dosing regimens. In addition, knowledge of common toxicities may help differentiate chemotherapy-related symptoms from actual progression of cancer into the CNS.
Collapse
Affiliation(s)
- Rajiv S Magge
- Department of Neurology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| | - Lisa M DeAngelis
- Department of Neurology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Department of Neurology, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
48
|
A novel strategy inducing autophagic cell death in Burkitt's lymphoma cells with anti-CD19-targeted liposomal rapamycin. Blood Cancer J 2014; 4:e180. [PMID: 24510029 PMCID: PMC3944660 DOI: 10.1038/bcj.2014.2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 11/20/2013] [Accepted: 12/31/2013] [Indexed: 11/18/2022] Open
Abstract
Relapsed or refractory Burkitt's lymphoma often has a poor prognosis in spite of intensive chemotherapy that induces apoptotic and/or necrotic death of lymphoma cells. Rapamycin (Rap) brings about autophagy, and could be another treatment. Further, anti-CD19-targeted liposomal delivery may enable Rap to kill lymphoma cells specifically. Rap was encapsulated by anionic liposome and conjugated with anti-CD19 antibody (CD19-GL-Rap) or anti-CD2 antibody (CD2-GL-Rap) as a control. A fluorescent probe Cy5.5 was also liposomized in the same way (CD19 or CD2-GL-Cy5.5) to examine the efficacy of anti-CD19-targeted liposomal delivery into CD19-positive Burkitt's lymphoma cell line, SKW6.4. CD19-GL-Cy5.5 was more effectively uptaken into SKW6.4 cells than CD2-GL-Cy5.5 in vitro. When the cells were inoculated subcutaneously into nonobese diabetic/severe combined immunodeficiency mice, intravenously administered CD19-GL-Cy5.5 made the subcutaneous tumor fluorescent, while CD2-GL-Cy5.5 did not. Further, CD19-GL-Rap had a greater cytocidal effect on not only SKW6.4 cells but also Burkitt's lymphoma cells derived from patients than CD2-GL-Rap in vitro. The specific toxicity of CD19-GL-Rap was cancelled by neutralizing anti-CD19 antibody. The survival period of mice treated with intravenous CD19-GL-Rap was significantly longer than that of mice treated with CD2-GL-Rap after intraperitoneal inoculation of SKW6.4 cells. Anti-CD19-targeted liposomal Rap could be a promising lymphoma cell-specific treatment inducing autophagic cell death.
Collapse
|
49
|
Kraft JC, Freeling JP, Wang Z, Ho RJY. Emerging research and clinical development trends of liposome and lipid nanoparticle drug delivery systems. J Pharm Sci 2014; 103:29-52. [PMID: 24338748 PMCID: PMC4074410 DOI: 10.1002/jps.23773] [Citation(s) in RCA: 360] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 10/11/2013] [Accepted: 10/14/2013] [Indexed: 12/13/2022]
Abstract
Liposomes are spherical-enclosed membrane vesicles mainly constructed with lipids. Lipid nanoparticles are loaded with therapeutics and may not contain an enclosed bilayer. The majority of those clinically approved have diameters of 50-300 nm. The growing interest in nanomedicine has fueled lipid-drug and lipid-protein studies, which provide a foundation for developing lipid particles that improve drug potency and reduce off-target effects. Integrating advances in lipid membrane research has enabled therapeutic development. At present, about 600 clinical trials involve lipid particle drug delivery systems. Greater understanding of pharmacokinetics, biodistribution, and disposition of lipid-drug particles facilitated particle surface hydration technology (with polyethylene glycol) to reduce rapid clearance and provide sufficient blood circulation time for drug to reach target tissues and cells. Surface hydration enabled the liposome-encapsulated cancer drug doxorubicin (Doxil) to gain clinical approval in 1995. Fifteen lipidic therapeutics are now clinically approved. Although much research involves attaching lipid particles to ligands selective for occult cells and tissues, preparation procedures are often complex and pose scale-up challenges. With emerging knowledge in drug target and lipid-drug distribution in the body, a systems approach that integrates knowledge to design and scale lipid-drug particles may further advance translation of these systems to improve therapeutic safety and efficacy.
Collapse
Affiliation(s)
- John C Kraft
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| | | | | | | |
Collapse
|
50
|
Frankel AE. Special issue of clinical pharmacology: advances and applications in new protein therapeutics modulating tumor immunity. Clin Pharmacol 2013; 5:1-3. [PMID: 24353448 PMCID: PMC3860356 DOI: 10.2147/cpaa.s55613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Affiliation(s)
- Arthur E Frankel
- Department of Internal Medicine, Simmons Comprehensive Cancer Center, the University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|