1
|
Carvalho AM, Greene MK, Smyth P, Mutch A, McLaughlin KM, Cairns LV, Mills KI, McCloskey KD, Scott CJ. Development of CD33-Targeted Dual Drug-Loaded Nanoparticles for the Treatment of Pediatric Acute Myeloid Leukemia. Biomacromolecules 2024; 25:6503-6514. [PMID: 39235263 PMCID: PMC11480974 DOI: 10.1021/acs.biomac.4c00672] [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: 05/20/2024] [Revised: 08/19/2024] [Accepted: 08/19/2024] [Indexed: 09/06/2024]
Abstract
Paediatric acute myeloid leukemia (AML) is a heterogeneous hematological malignancy still heavily reliant on traditional chemotherapeutic approaches. Combination treatments have shown to be a superior approach, but their success is often hindered by side effects and different drugs' pharmacokinetics. Here, we investigated ABT-737 and Purvalanol A as a potential drug pairing for pediatric AML and described the development of CD33-targeted polymeric nanoparticles (NPs) to enable their simultaneous targeted codelivery. Separate drug encapsulation within poly(lactic-co-glycolic acid) (PLGA) NPs was optimized prior to coencapsulation of both drugs at a synergistic ratio in PEGylated PLGA NPs. The therapeutic effects of formulations were evaluated in a panel of pediatric AML cells, and dual drug-loaded NPs (dual NPs) demonstrated significantly enhanced apoptotic cell death. Moreover, conjugation to gemtuzumab resulted in improved NP binding and internalization in CD33-positive cells. Finally, CD33-targeted dual-loaded NPs showed enhanced cytotoxicity to CD33-positive AML cells via CD33-mediated targeted drug delivery.
Collapse
Affiliation(s)
- Ana M. Carvalho
- The Patrick G Johnston Centre
for Cancer Research, School of Medicine, Dentistry and Biomedical
Sciences, Queen’s University Belfast, Belfast BT9 7AE, U.K.
| | - Michelle K. Greene
- The Patrick G Johnston Centre
for Cancer Research, School of Medicine, Dentistry and Biomedical
Sciences, Queen’s University Belfast, Belfast BT9 7AE, U.K.
| | - Peter Smyth
- The Patrick G Johnston Centre
for Cancer Research, School of Medicine, Dentistry and Biomedical
Sciences, Queen’s University Belfast, Belfast BT9 7AE, U.K.
| | - Alexander Mutch
- The Patrick G Johnston Centre
for Cancer Research, School of Medicine, Dentistry and Biomedical
Sciences, Queen’s University Belfast, Belfast BT9 7AE, U.K.
| | - Kirsty M. McLaughlin
- The Patrick G Johnston Centre
for Cancer Research, School of Medicine, Dentistry and Biomedical
Sciences, Queen’s University Belfast, Belfast BT9 7AE, U.K.
| | - Lauren V. Cairns
- The Patrick G Johnston Centre
for Cancer Research, School of Medicine, Dentistry and Biomedical
Sciences, Queen’s University Belfast, Belfast BT9 7AE, U.K.
| | - Ken I. Mills
- The Patrick G Johnston Centre
for Cancer Research, School of Medicine, Dentistry and Biomedical
Sciences, Queen’s University Belfast, Belfast BT9 7AE, U.K.
| | - Karen D. McCloskey
- The Patrick G Johnston Centre
for Cancer Research, School of Medicine, Dentistry and Biomedical
Sciences, Queen’s University Belfast, Belfast BT9 7AE, U.K.
| | - Christopher J. Scott
- The Patrick G Johnston Centre
for Cancer Research, School of Medicine, Dentistry and Biomedical
Sciences, Queen’s University Belfast, Belfast BT9 7AE, U.K.
| |
Collapse
|
2
|
Omer MH, Shafqat A, Ahmad O, Alkattan K, Yaqinuddin A, Damlaj M. Bispecific Antibodies in Hematological Malignancies: A Scoping Review. Cancers (Basel) 2023; 15:4550. [PMID: 37760519 PMCID: PMC10526328 DOI: 10.3390/cancers15184550] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
Bispecific T-cell engagers (BiTEs) and bispecific antibodies (BiAbs) have revolutionized the treatment landscape of hematological malignancies. By directing T cells towards specific tumor antigens, BiTEs and BiAbs facilitate the T-cell-mediated lysis of neoplastic cells. The success of blinatumomab, a CD19xCD3 BiTE, in acute lymphoblastic leukemia spearheaded the expansive development of BiTEs/BiAbs in the context of hematological neoplasms. Nearly a decade later, numerous BiTEs/BiAbs targeting a range of tumor-associated antigens have transpired in the treatment of multiple myeloma, non-Hodgkin's lymphoma, acute myelogenous leukemia, and acute lymphoblastic leukemia. However, despite their generally favorable safety profiles, particular toxicities such as infections, cytokine release syndrome, myelosuppression, and neurotoxicity after BiAb/BiTE therapy raise valid concerns. Moreover, target antigen loss and the immunosuppressive microenvironment of hematological neoplasms facilitate resistance towards BiTEs/BiAbs. This review aims to highlight the most recent evidence from clinical trials evaluating the safety and efficacy of BiAbs/BiTEs. Additionally, the review will provide mechanistic insights into the limitations of BiAbs whilst outlining practical applications and strategies to overcome these limitations.
Collapse
Affiliation(s)
- Mohamed H. Omer
- School of Medicine, Cardiff University, Cardiff CF14 4YS, UK
| | - Areez Shafqat
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.S.); (O.A.); (K.A.); (A.Y.)
| | - Omar Ahmad
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.S.); (O.A.); (K.A.); (A.Y.)
| | - Khaled Alkattan
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.S.); (O.A.); (K.A.); (A.Y.)
| | - Ahmed Yaqinuddin
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.S.); (O.A.); (K.A.); (A.Y.)
| | - Moussab Damlaj
- Department of Hematology & Oncology, Sheikh Shakhbout Medical City, Abu Dhabi P.O. Box 11001, United Arab Emirates;
- College of Medicine, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| |
Collapse
|
3
|
Espinosa-Cotton M, Guo HF, Cheung NKV. Tracking Bispecific Antibody-Induced T Cell Trafficking Using Luciferase-Transduced Human T Cells. J Vis Exp 2023:10.3791/64390. [PMID: 37246883 PMCID: PMC10999115 DOI: 10.3791/64390] [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] [Indexed: 05/30/2023] Open
Abstract
T cell-engaging bispecific antibodies (T-BsAbs) are in various stages of preclinical development and clinical testing for solid tumors. Factors such as valency, spatial arrangement, interdomain distance, and Fc mutations affect the anti-tumor efficacy of these therapies, commonly by influencing the homing of T cells to tumors, which remains a major challenge. Here, we describe a method to transduce activated human T cells with luciferase, allowing in vivo tracking of T cells during T-BsAb therapy studies. The ability of T-BsAbs to redirect T cells to tumors can be quantitatively evaluated at multiple time points during treatment, allowing researchers to correlate the anti-tumor efficacy of T-BsAbs and other interventions with the persistence of T cells in tumors. This method alleviates the need to sacrifice animals during treatment to histologically assess T cell infiltration and can be repeated at multiple time points to determine the kinetics of T cell trafficking during and after treatment.
Collapse
Affiliation(s)
| | - Hong-Fen Guo
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center
| | | |
Collapse
|
4
|
Espinosa-Cotton M, Guo HF, Tickoo SK, Cheung NKV. Identification of immunotherapy and radioimmunotherapy targets on desmoplastic small round cell tumors. Front Oncol 2023; 13:1104693. [PMID: 37091153 PMCID: PMC10119788 DOI: 10.3389/fonc.2023.1104693] [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: 11/21/2022] [Accepted: 03/23/2023] [Indexed: 04/25/2023] Open
Abstract
Background Development of successful antibody-based immunotherapeutic and radioimmunotherapeutic strategies rely on the identification of cell surface tumor-associated antigens (TAA) with restricted expression on normal tissues. Desmoplastic small round cell tumor (DSRCT) is a rare and generally neglected malignancy that primarily affects adolescent and young adult males. New therapies capable of treating disseminated disease are needed for DSRCT, which is often widespread at diagnosis. Methods We used immunohistochemistry (IHC) on fresh frozen surgical specimens and patient-derived xenograft (PDX) tumors and flow cytometry on DSRCT cell lines to evaluate expression of TAAs in these tumors. In vitro cytotoxicity assays were used to evaluate the efficacy of T cell-engaging bispecific antibodies (T-BsAbs) directed at these targets. In vivo, we used an intraperitoneal xenograft mouse model of DSRCT to test T-BsAbs against several TAAs. Results In DSRCT specimens we found widespread expression of B7-H3, EGFR, GD2, HER2, mesothelin, and polysialic acid, clinical targets for which specific antibody therapeutics are available. The expression of B7-H3, EGFR, HER2, and mesothelin was confirmed on the cell surface of DSRCT cell lines. In vitro cytotoxicity assays confirmed the efficacy of T cell-engaging bispecific antibodies (T-BsAbs) directed at these targets against DSRCT cells. Remarkably, a HER2xCD3 T-BsAb was capable of completely shrinking established tumors in an intraperitoneal mouse model of DSRCT. Conclusions We propose that these TAAs should be further investigated in preclinical models as targets for immunotherapy and radioimmunotherapy with the hope of providing a rationale to extend these therapies to patients with advanced DSRCT.
Collapse
Affiliation(s)
- Madelyn Espinosa-Cotton
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Hong-Fen Guo
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Satish K. Tickoo
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Nai-Kong V. Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
5
|
Lanier OL, Pérez-Herrero E, Andrea APD, Bahrami K, Lee E, Ward DM, Ayala-Suárez N, Rodríguez-Méndez SM, Peppas NA. Immunotherapy approaches for hematological cancers. iScience 2022; 25:105326. [PMID: 36325064 PMCID: PMC9619355 DOI: 10.1016/j.isci.2022.105326] [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] [Indexed: 12/24/2022] Open
Abstract
Hematological cancers such as leukemia, lymphoma, and multiple myeloma have traditionally been treated with chemo and radiotherapy approaches. Introduction of immunotherapies for treatment of these diseases has led to patient remissions that would not have been possible with traditional approaches. In this critical review we identify main disease characteristics, symptoms, and current treatment options. Five common immunotherapies, namely checkpoint inhibitors, vaccines, cell-based therapies, antibodies, and oncolytic viruses, are described, and their applications in hematological cancers are critically discussed.
Collapse
Affiliation(s)
- Olivia L. Lanier
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
| | - Edgar Pérez-Herrero
- Departamento de Ingeniería Química y Tecnología Farmacéutica, Universidad de La Laguna, La Laguna, 38206 Tenerife, Spain
- Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, La Laguna, 38206 Tenerife, Spain
- Instituto Universitario de Tecnologías Biomédicas, Universidad de La Laguna, La Laguna, 38200 Tenerife, Spain
| | - Abielle P. D.’ Andrea
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
| | - Kiana Bahrami
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
| | - Elaine Lee
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
| | - Deidra M. Ward
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
| | - Nilaya Ayala-Suárez
- Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, La Laguna, 38206 Tenerife, Spain
| | - Sheyla M. Rodríguez-Méndez
- Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, La Laguna, 38206 Tenerife, Spain
| | - Nicholas A. Peppas
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
- Department of Surgery and Perioperative Care, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
6
|
Pediatric Mixed-Phenotype Acute Leukemia: What's New? Cancers (Basel) 2021; 13:cancers13184658. [PMID: 34572885 PMCID: PMC8469808 DOI: 10.3390/cancers13184658] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/09/2021] [Accepted: 09/12/2021] [Indexed: 11/23/2022] Open
Abstract
Simple Summary Pediatric mixed-phenotype leukemia is a rare form of blood cancer in children. In this review, we cover both the evolution of treatment over the past several years and outline new emerging concepts in this disease. Abstract Mixed-phenotype acute leukemias (MPAL) are rare in children and often lack consensus on optimal management. This review examines the current controversies and emerging paradigms in the management of pediatric MPAL. We examine risk stratification, outcomes of recent retrospective and prospective collaborative trials, and the role of transplantation and precision genomics, and outline emerging targets and concepts in this rare entity.
Collapse
|
7
|
Lin TY, Park JA, Long A, Guo HF, Cheung NKV. Novel potent anti-STEAP1 bispecific antibody to redirect T cells for cancer immunotherapy. J Immunother Cancer 2021; 9:jitc-2021-003114. [PMID: 34497115 PMCID: PMC8438958 DOI: 10.1136/jitc-2021-003114] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/17/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The prognosis for metastatic Ewing sarcoma family of tumors (EFT) is still poor despite high-dose chemotherapy and radiation treatment. Immunotherapies hold promise, but cancer antigen-targeting immunotherapies have largely failed to induce effective T cell receptor-mediated antitumor response. However, T cell-engaging bispecific antibodies (T-BsAbs) have yet to be adequately explored. METHODS Rehumanized STEAP1-IgG was used to build T-BsAb (named BC261) using the 2+2 IgG-[L]-scFv platform carrying the anti-CD3 huOKT3 scFv as the second specificity. Its binding epitope mapping, species cross-reactivity, tumor cell line staining, and in vitro cytotoxicity were investigated thoroughly. Its potency in driving tumor-infiltrating lymphocytes (TILs) was quantified using bioluminescence, correlated with in vivo antitumor response against cell line-derived or patient-derived xenografts (CDXs or PDXs) and compared with anti-STEAP1 T-BsAbs built on representative antibody platforms. RESULTS BC261 binding epitope was mapped to its second extracellular domain of STEAP1 shared among canine and primate orthologs. BC261 induced potent cytotoxicity against panels of EFT, prostate cancer, and canine osteosarcoma cell lines despite their low antigen density. BC261 drove significantly more TILs into tumors (30-fold) and exerted superior antitumor effects compared with the other standard BsAb platforms. The antitumor efficacy of BC261 was consistent against EFT and prostate cancer CDXs and PDXs. CONCLUSIONS BC261 was highly efficient in driving T cell infiltration and tumor ablation. Either as stand-alone therapeutics or for ex vivo armed T cells, this novel anti-STEAP1 T-BsAb BC261 has therapeutic potential.
Collapse
Affiliation(s)
- Tsung-Yi Lin
- Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jeong A Park
- Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Alan Long
- Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Hong-Fen Guo
- Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Nai-Kong V Cheung
- Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|