1
|
Zhe N, Li Q, Huang N, Li H, Chen H, Zhu P. Hotspots evolution and frontiers of immunotherapy for the treatment of acute myeloid leukemia: A bibliometric analysis. Hum Vaccin Immunother 2025; 21:2448888. [PMID: 39819314 DOI: 10.1080/21645515.2024.2448888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 12/10/2024] [Accepted: 12/29/2024] [Indexed: 01/19/2025] Open
Abstract
Given the growing significance of immunotherapy in addressing the limitations of conventional acute myeloid leukemia (AML) treatments, this study aimed to elucidate the hotspot evolution and frontiers of immunotherapy in AML using bibliometric analysis. With a strict retrieval strategy applied in the Web of Science Core Collection, 2411 publications were obtained and exported. The temporal and geographical distributions of these publications and the countries, institutions, journals, and authors who contributed to the field were investigated. An in-depth content analysis was performed. The United States had various research institutions dedicated to AML immunotherapy. Frontiers in Immunology had the highest number of publications, but Blood had the highest H-index. Marion Subklewe was the most productive author. The current research hotspots of AML immunotherapy included chimeric antigen receptor-T-cell therapy, antibody-based immunotherapies, immune checkpoint blockade, and combination therapy, highlighting the key aspects of immunotherapy for AML treatment and providing comprehensive insights into the research status and advances in this field. Novel immunotherapies combined with chemotherapy may become the primary focus of AML treatment.
Collapse
Affiliation(s)
- Nana Zhe
- Department of Hematology, The First Peoples' Hospital of Zunyi(The Third Affiliated Hospital of Zunyi Medical University), Zunyi, Guizhou, China
| | - Qiang Li
- Department of Hematology, The First Peoples' Hospital of Zunyi(The Third Affiliated Hospital of Zunyi Medical University), Zunyi, Guizhou, China
| | - Nanqu Huang
- Department of Pharmacy, The First Peoples' Hospital of Zunyi(The Third Affiliated Hospital of Zunyi Medical University), Zunyi, Guizhou, China
| | - Hang Li
- Department of Hematology, The First Peoples' Hospital of Zunyi(The Third Affiliated Hospital of Zunyi Medical University), Zunyi, Guizhou, China
| | - Hongyun Chen
- Department of Dermatology, The First Peoples' Hospital of Zunyi(The Third Affiliated Hospital of Zunyi Medical University), Zunyi, Guizhou, China
| | - Pinwei Zhu
- Department of Hematology, The First Peoples' Hospital of Zunyi(The Third Affiliated Hospital of Zunyi Medical University), Zunyi, Guizhou, China
| |
Collapse
|
2
|
Al Malki MM, Minden MD, Rich ES, Hill JE, Gill SC, Fan A, Fredericks CE, Fathi AT, Abdul-Hay M. Safety, tolerability, and pharmacokinetics of ASP1235 in relapsed or refractory acute myeloid leukemia: A phase 1 study. Leuk Res 2025; 152:107690. [PMID: 40209615 DOI: 10.1016/j.leukres.2025.107690] [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: 11/21/2024] [Revised: 03/21/2025] [Accepted: 04/01/2025] [Indexed: 04/12/2025]
Abstract
Acute myeloid leukemia (AML) is an aggressive hematologic malignancy. Although new agents including targeted therapies for relapsed or refractory (R/R) AML have been introduced, poor outcomes remain, requiring the need for novel approaches. One novel approach is the use of antibody-drug conjugates (ADCs). We conducted an early phase clinical trial with ASP1235, an ADC targeting FMS-like tyrosine kinase 3. In total, 43 patients with R/R AML were treated with ASP1235. The most common adverse events (AEs) included elevated liver transaminase levels, ocular toxicity, and muscular weakness. Ocular treatment-emergent AEs (TEAEs) were observed in 53 % of patients; most were mild or moderate in severity. The most common ocular TEAEs were blurred vision, dry eye, keratitis, photophobia, and reduced visual acuity. Serious (grade ≥3) ocular TEAEs occurred in 16.3 % of patients, with only 1 patient experiencing grade 4 keratitis. Six patients achieved composite complete remission (complete remission [CR] + CR with incomplete hematologic recovery + CR with incomplete platelet recovery), 2 of whom proceeded to hematopoietic cell transplantation with long-term leukemia-free survival. This trial was registered at www.clinicaltrials.gov as #NCT02864290.
Collapse
Affiliation(s)
- Monzr M Al Malki
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA.
| | - Mark D Minden
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | | | - Jason E Hill
- Astellas Pharma Global Development, Inc., Northbrook, IL, USA
| | - Stanley C Gill
- Astellas Pharma Global Development, Inc., Northbrook, IL, USA
| | - Alan Fan
- Astellas Pharma Global Development, Inc., Northbrook, IL, USA
| | | | | | - Maher Abdul-Hay
- Laura and Isaac Perlmutter Cancer Center at NYU Langone, New York, NY, USA
| |
Collapse
|
3
|
Oh Y, An DE, Park J, Koh B, Cho KJ, Jeon H. Synthesis and evaluation of KX-01 analogs with an exploration of linker attachment points for antibody-drug conjugates. Bioorg Med Chem Lett 2025; 120:130114. [PMID: 39880175 DOI: 10.1016/j.bmcl.2025.130114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 12/26/2024] [Accepted: 01/26/2025] [Indexed: 01/31/2025]
Abstract
KX-01 (tirbanibulin, Klisyri®) is a recently FDA-approved drug for treating actinic keratosis, with a distinct dual mechanism of action combining microtubule disruption and non-ATP-competitive Src inhibition. This unique mechanism and novel chemotype highlight KX-01's potential as a payload for antibody-drug conjugates. In this study, we synthesized and evaluated KX-01 derivatives to enhance anticancer potency and explore functional groups suitable for antibody conjugation. Notably, replacing the morpholine group with an N-benzoylpiperazine scaffold resulted in an analog with significantly improved in vitro antiproliferative activity, attributed to enhanced microtubule disruption and Src inhibition. Furthermore, introducing a phenol or aniline functionality as a common linker attachment point preserved substantial cytotoxicity. These results suggest the potential of KX-01 derivatives for future use as ADC payloads.
Collapse
Affiliation(s)
- Yeju Oh
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114 Republic of Korea; Medicinal Chemistry & Pharmacology, University of Science & Technology, Daejeon 34113 Republic of Korea
| | - Da Eun An
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114 Republic of Korea
| | - Jaebeom Park
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114 Republic of Korea
| | - Byumseok Koh
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114 Republic of Korea; Medicinal Chemistry & Pharmacology, University of Science & Technology, Daejeon 34113 Republic of Korea
| | - Kyung-Jin Cho
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114 Republic of Korea.
| | - Hongjun Jeon
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114 Republic of Korea; Medicinal Chemistry & Pharmacology, University of Science & Technology, Daejeon 34113 Republic of Korea.
| |
Collapse
|
4
|
Cole FM, Laszlo GS, Lunn-Halbert MC, Kehret AR, Zweidler-McKay PA, Rodríguez-Arbolí E, Wu D, Nyberg K, Li J, Lim SYT, Pettenger-Willey CM, Lakshmikanthan S, Walter RB. Preclinical characterization of the anti-leukemia activity of the CD123 antibody-drug conjugate, pivekimab sunirine (IMGN632). Leukemia 2025:10.1038/s41375-025-02571-0. [PMID: 40119158 DOI: 10.1038/s41375-025-02571-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/21/2025] [Accepted: 03/13/2025] [Indexed: 03/24/2025]
Affiliation(s)
- Frances M Cole
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - George S Laszlo
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Margaret C Lunn-Halbert
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Allie R Kehret
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Eduardo Rodríguez-Arbolí
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Hematology, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS/CSIC), University of Seville, Seville, Spain
| | - David Wu
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Kyle Nyberg
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Junyang Li
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Sheryl Y T Lim
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | | | - Roland B Walter
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
5
|
Galati D, Zanotta S, Florio F, Mele S, De Filippi R, Pinto A. Immunotherapies Targeting CD123 and CD303: A New Frontier in Treating Blastic Plasmacytoid Dendritic Cell Neoplasm. Int J Mol Sci 2025; 26:2732. [PMID: 40141368 PMCID: PMC11942551 DOI: 10.3390/ijms26062732] [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: 02/04/2025] [Revised: 03/13/2025] [Accepted: 03/16/2025] [Indexed: 03/28/2025] Open
Abstract
Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a rare and aggressive hematologic malignancy characterized by the overexpression of CD123 and CD303 surface antigens. These molecular markers play a crucial role in diagnosing diseases and developing targeted therapies. Traditional treatment options for BPDCN have demonstrated limited effectiveness, highlighting the need for new and innovative therapeutic strategies. Recent advances in immunotherapy, particularly therapeutic monoclonal antibodies, bispecific T-cell engagers, and CAR T-cell therapy, have provided promising alternatives. Tagraxofusp, the first FDA-approved CD123-targeted therapy, has significantly improved patient outcomes. Additionally, emerging CD303-targeting strategies offer the potential for further advancements. Despite these breakthroughs, challenges such as treatment resistance and toxicity remain. This review explores the latest developments in BPDCN treatment, emphasizing the potential of CD123 and CD303 as targets for precision medicine interventions. The ongoing evolution of targeted immunotherapies holds promise for improving patient survival and redefining treatment paradigms in hematologic malignancies.
Collapse
Affiliation(s)
- Domenico Galati
- Hematology-Oncology and Stem-Cell Transplantation Unit, Department of Onco-Hematology and Innovative Diagnostics, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, 80131 Napoli, Italy; (S.Z.); (F.F.); (S.M.); (A.P.)
| | - Serena Zanotta
- Hematology-Oncology and Stem-Cell Transplantation Unit, Department of Onco-Hematology and Innovative Diagnostics, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, 80131 Napoli, Italy; (S.Z.); (F.F.); (S.M.); (A.P.)
| | - Fabrizia Florio
- Hematology-Oncology and Stem-Cell Transplantation Unit, Department of Onco-Hematology and Innovative Diagnostics, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, 80131 Napoli, Italy; (S.Z.); (F.F.); (S.M.); (A.P.)
| | - Sara Mele
- Hematology-Oncology and Stem-Cell Transplantation Unit, Department of Onco-Hematology and Innovative Diagnostics, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, 80131 Napoli, Italy; (S.Z.); (F.F.); (S.M.); (A.P.)
| | - Rosaria De Filippi
- Department of Clinical Medicine and Surgery, Università degli Studi di Napoli Federico II, 80131 Napoli, Italy;
| | - Antonio Pinto
- Hematology-Oncology and Stem-Cell Transplantation Unit, Department of Onco-Hematology and Innovative Diagnostics, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, 80131 Napoli, Italy; (S.Z.); (F.F.); (S.M.); (A.P.)
| |
Collapse
|
6
|
Wang Y, Yang X, Liu Y, Li Y. A review of common immunotherapy and nano immunotherapy for acute myeloid leukemia. Front Immunol 2025; 16:1505247. [PMID: 40129984 PMCID: PMC11931025 DOI: 10.3389/fimmu.2025.1505247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 02/24/2025] [Indexed: 03/26/2025] Open
Abstract
Acute myeloid leukemia (AML) is a highly aggressive hematological malignancy. Traditional chemotherapy methods not only bring serious side effects, but also lead to high recurrence rate and drug resistance in some patients. However, as an emerging therapeutic strategy, immunotherapy has shown great potential in the field of AML treatment in recent years. At present, common immunotherapy methods for AML include monoclonal antibodies, CAR-T cell therapy, and immune checkpoint inhibitors. With the deepening of research and technological progress, especially the application of nanotechnology in medicine, new immunotherapy is expected to become one of the important means for the treatment of acute myeloid leukemia in the future.
Collapse
Affiliation(s)
- Yaoyao Wang
- Department of Pediatrics of Yantai Affiliated Hospital, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong, China
| | - Xiancong Yang
- Laboratory Department, Qilu Hospital of ShanDong University Dezhou Hospital, Dezhou, Shandong, China
| | - Yalin Liu
- Department of Pediatrics of Yantai Affiliated Hospital, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Youjie Li
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong, China
| |
Collapse
|
7
|
Able M, Kasper MA, Vick B, Schwach J, Gao X, Schmitt S, Tizazu B, Fischer A, Künzl S, Leilich M, Mai I, Ochtrop P, Stengl A, de Geus MAR, von Bergwelt-Baildon M, Schumacher D, Helma J, Hackenberger CPR, Götze KS, Jeremias I, Leonhardt H, Feuring M, Spiekermann K. Effective eradication of acute myeloid leukemia stem cells with FLT3-directed antibody-drug conjugates. Leukemia 2025; 39:632-642. [PMID: 39870768 PMCID: PMC11879846 DOI: 10.1038/s41375-024-02510-5] [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: 05/14/2024] [Revised: 12/09/2024] [Accepted: 12/23/2024] [Indexed: 01/29/2025]
Abstract
Refractory disease and relapse are major challenges in acute myeloid leukemia (AML) therapy attributed to survival of leukemic stem cells (LSC). To target LSCs, antibody-drug conjugates (ADCs) provide an elegant solution, combining the specificity of antibodies with highly potent payloads. We aimed to investigate if FLT3-20D9h3-ADCs delivering either the DNA-alkylator duocarmycin (DUBA) or the microtubule-toxin monomethyl auristatin F (MMAF) can eradicate quiescent LSCs. We show here that DUBA more potently kills cell-cycle arrested AML cells compared to microtubule-targeting auristatins. Due to limited stability of 20D9h3-DUBA ADC in vivo, we analyzed both ADCs in advanced in vitro stem cell assays. 20D9h3-DUBA successfully eliminated leukemic progenitors in vitro in colony-forming unit and long-term culture initiating cell assays, both in patient cells and in patient-derived xenograft (PDX) cells. Further, it completely prevented engraftment of AML PDX leukemia-initiating cells in NSG mice. 20D9h3-MMAF had a similar effect in engraftment assays, but a less prominent effect in colony assays. Both ADCs did not affect healthy stem and progenitor cells at comparable doses providing the rationale for FLT3 as therapeutic LSC target. Collectively, we show that FLT3-directed ADCs with DUBA or MMAF have potent activity against AML LSCs and represent promising candidates for further clinical development.
Collapse
Affiliation(s)
- Marina Able
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
| | - Marc-André Kasper
- Chemical Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Campus Berlin, Berlin, Germany
- Department of Chemistry, Humboldt Universität zu Berlin, Berlin, Germany
- Tubulis GmbH, Munich, Germany
| | - Binje Vick
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and LMU University Hospital, Munich, Germany
- Research Unit Apoptosis in Hematopoietic Stem Cells (AHS), Helmholtz Munich, German Research Center for Environmental Health (HMGU), Munich, Germany
| | - Jonathan Schwach
- Faculty of Biology, Human Biology and BioImaging, LMU Munich, Planegg-Martinsried, Germany
| | - Xiang Gao
- Department of Internal Medicine III, University Hospital Ulm, Ulm, Germany
| | | | - Belay Tizazu
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
| | - Amrei Fischer
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and LMU University Hospital, Munich, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sarah Künzl
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
| | - Marit Leilich
- Technical University of Munich School of Medicine and Health, Department of Medicine III, Technical University of Munich (TUM), Munich, Germany
| | | | - Philipp Ochtrop
- Chemical Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Campus Berlin, Berlin, Germany
- Tubulis GmbH, Munich, Germany
| | - Andreas Stengl
- Faculty of Biology, Human Biology and BioImaging, LMU Munich, Planegg-Martinsried, Germany
| | - Mark A R de Geus
- Chemical Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Campus Berlin, Berlin, Germany
| | - Michael von Bergwelt-Baildon
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and LMU University Hospital, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Dominik Schumacher
- Chemical Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Campus Berlin, Berlin, Germany
- Department of Chemistry, Humboldt Universität zu Berlin, Berlin, Germany
- Tubulis GmbH, Munich, Germany
| | - Jonas Helma
- Tubulis GmbH, Munich, Germany
- Faculty of Biology, Human Biology and BioImaging, LMU Munich, Planegg-Martinsried, Germany
| | - Christian P R Hackenberger
- Chemical Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Campus Berlin, Berlin, Germany
- Department of Chemistry, Humboldt Universität zu Berlin, Berlin, Germany
| | - Katharina S Götze
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and LMU University Hospital, Munich, Germany
- Technical University of Munich School of Medicine and Health, Department of Medicine III, Technical University of Munich (TUM), Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Irmela Jeremias
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and LMU University Hospital, Munich, Germany
- Research Unit Apoptosis in Hematopoietic Stem Cells (AHS), Helmholtz Munich, German Research Center for Environmental Health (HMGU), Munich, Germany
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU, Munich, Germany
| | - Heinrich Leonhardt
- Faculty of Biology, Human Biology and BioImaging, LMU Munich, Planegg-Martinsried, Germany
| | - Michaela Feuring
- Department of Internal Medicine III, University Hospital Ulm, Ulm, Germany
| | - Karsten Spiekermann
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany.
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and LMU University Hospital, Munich, Germany.
- Bavarian Cancer Research Center (BZKF), Munich, Germany.
| |
Collapse
|
8
|
Hochman MJ, Muniz JP, Papadantonakis N. Precision Medicine in Myeloid Neoplasia: Challenges and Opportunities. J Pers Med 2025; 15:49. [PMID: 39997326 PMCID: PMC11856194 DOI: 10.3390/jpm15020049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/12/2025] [Accepted: 01/21/2025] [Indexed: 02/26/2025] Open
Abstract
High-risk myeloid neoplasms encompass a group of hematologic malignancies known to cause significant cytopenias, which are accompanied by the risk of end-organ damage. They tend to have an aggressive clinical course and limit life expectancy in the absence of effective treatments. The adoption of precision medicine approaches has been limited by substantive diversity in somatic mutations, limited fraction of patients with targetable genetic lesions, and the prolonged turnaround times of pertinent genetic tests. Efforts to incorporate targeted agents into first-line treatment, rapidly determine pre-treatment molecular or cytogenetic aberrations, and evaluate functional vulnerabilities ex vivo hold promise for advancing the use of precision medicine in these malignancies. Given the relative accessibility of malignant cells from blood and bone marrow, precision medicine strategies hold great potential to shape future standard-of-care approaches to patients with high-risk myeloid malignancies. This review aims to summarize the development of the targeted therapies currently available to treat these blood cancers, most notably acute myeloid leukemia, and also evaluate future opportunities and challenges related to the integration of personalized approaches.
Collapse
Affiliation(s)
- Michael J. Hochman
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| | - Joshua P. Muniz
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
- Aflac Cancer & Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, GA 30329, USA
| | - Nikolaos Papadantonakis
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
9
|
Watts B, Smith CM, Evans K, Gifford AJ, Mohamed SMA, Erickson SW, Earley EJ, Neuhauser S, Stearns TM, Philip VM, Chuang JH, Zweidler-McKay PA, Lakshmikanthan S, Jocoy EL, Bult CJ, Teicher BA, Smith MA, Lock RB. The CD123 antibody-drug conjugate pivekimab sunirine exerts profound activity in preclinical models of pediatric acute lymphoblastic leukemia. Hemasphere 2025; 9:e70063. [PMID: 39830370 PMCID: PMC11739898 DOI: 10.1002/hem3.70063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/11/2024] [Accepted: 11/17/2024] [Indexed: 01/22/2025] Open
Abstract
Antibody-drug conjugates (ADCs) combining monoclonal antibodies with cytotoxic payloads are a rapidly emerging class of immune-based therapeutics with the potential to improve the treatment of cancer, including children with relapse/refractory acute lymphoblastic leukemia (ALL). CD123, the α subunit of the interleukin-3 receptor, is overexpressed in ALL and is a potential therapeutic target. Here, we show that pivekimab sunirine (PVEK), a recently developed ADC comprising the CD123-targeting antibody, G4723A, and the cytotoxic payload, DGN549, was highly effective in vivo against a large panel of pediatric ALL patient-derived xenograft (PDX) models (n = 39). PVEK administered once weekly for 3 weeks resulted in a median event-free survival (EFS) of 57.2 days across all PDXs. CD123 mRNA and protein expression was significantly higher in B-lineage (n = 65) compared with T-lineage (n = 25) ALL PDXs (p < 0.0001), and mice engrafted with B-lineage PDXs achieved significantly longer EFS than those engrafted with T-lineage PDXs (p < 0.0001). PVEK treatment also resulted in significant clearance of human leukemia cells in hematolymphoid organs in mice engrafted with B-ALL PDXs. Notably, our results showed no direct correlation between CD123 expression and mouse EFS, indicating that CD123 is necessary but not sufficient for in vivo PVEK activity. Importantly, a PDX with very high CD123 cell surface expression but resistant to in vivo PVEK treatment, failed to internalize the G4723A antibody while remaining sensitive to the PVEK payload, DGN549, suggesting a novel mechanism of resistance. In conclusion, PVEK was highly effective against a large panel of B-ALL PDXs supporting its clinical translation for B-lineage pediatric ALL.
Collapse
Affiliation(s)
- Ben Watts
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Centre for Childhood Cancer Research UNSW Sydney Sydney New South Wales Australia
| | - Christopher M Smith
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Centre for Childhood Cancer Research UNSW Sydney Sydney New South Wales Australia
| | - Kathryn Evans
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Centre for Childhood Cancer Research UNSW Sydney Sydney New South Wales Australia
| | - Andrew J Gifford
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Centre for Childhood Cancer Research UNSW Sydney Sydney New South Wales Australia
- Anatomical Pathology, NSW Health Pathology Prince of Wales Hospital Randwick New South Wales Australia
- School of Clinical Medicine, UNSW Medicine & Health UNSW Sydney Sydney New South Wales Australia
| | - Sara M A Mohamed
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Centre for Childhood Cancer Research UNSW Sydney Sydney New South Wales Australia
- Department of Pharmaceutics and Industrial Pharmacy Faculty of Pharmacy, Ain Shams University Cairo Egypt
| | | | - Eric J Earley
- RTI International Research Triangle Park North Carolina USA
| | - Steven Neuhauser
- The Jackson Laboratory for Mammalian Genetics Bar Harbor Maine USA
| | | | - Vivek M Philip
- The Jackson Laboratory for Mammalian Genetics Bar Harbor Maine USA
| | - Jeffrey H Chuang
- The Jackson Laboratory for Genomic Medicine Farmington Connecticut USA
| | | | | | | | - Carol J Bult
- The Jackson Laboratory for Mammalian Genetics Bar Harbor Maine USA
| | | | | | - Richard B Lock
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Centre for Childhood Cancer Research UNSW Sydney Sydney New South Wales Australia
| |
Collapse
|
10
|
Pemmaraju N. BPDCN: state of the art. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2024; 2024:279-286. [PMID: 39644068 DOI: 10.1182/hematology.2024000553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2024]
Abstract
The emergence of blastic plasmacytoid dendritic cell neoplasm (BPDCN) as its own distinct entity within the pantheon of hematologic malignancies is due to the growing understanding of its unique multiorgan clinical presentation and characteristic skin lesions. The occurrence of BPDCN is generally heralded by a multicompartmental presentation of violaceous cutaneous lesions, involvement by bone marrow and/or blood, lymph node invasion, and an inclination toward extramedullary organ involvement, including, most remarkably, central nervous system (CNS)/cerebrospinal fluid positivity. With a median age historically of ≥ 70 years and up to 5:1 male predominance in most of the field's earlier studies, the most notable development in the modern era is the recognition of emerging important groups with BPDCN, such as female, pediatric, and adolescent/young adult patients; CNS + BPDCN patients; and an increasing number of cases being diagnosed worldwide. These trends are in line with the increased educational and research efforts, greater international collaboration, and markedly improved diagnostic tools and clinical approaches among hematology/oncology, hematopathology, dermatology, and dermatopathology teams around the world. Now, with over 5 years since the first commercially approved targeted agent specifically dedicated for BPDCN, the CD123-targeted agent tagraxofusp, improvements have been demonstrated particularly in the frontline setting for patients with BPDCN. The field is abundant with hope, as it has experienced advancements including greater molecular characterization, expanded identification of potential targets for therapy beyond CD123, advent of combination therapies, improving parameters for stem cell transplantation, and novel clinical trials specifically available for patients with BPDCN.
Collapse
Affiliation(s)
- Naveen Pemmaraju
- Department of Leukemia, University of Texas, MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
11
|
Bawek S, Gurusinghe S, Burwinkel M, Przespolewski A. Updates in novel immunotherapeutic strategies for relapsed/refractory AML. Front Oncol 2024; 14:1374963. [PMID: 39697225 PMCID: PMC11652486 DOI: 10.3389/fonc.2024.1374963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 11/11/2024] [Indexed: 12/20/2024] Open
Abstract
Acute myeloid leukemia (AML) is a severe hematological malignancy with poor outcomes, particularly in older adults. Traditional treatment options like high-dose chemotherapy often lead to refractory or relapsed AML, with even worse outcomes. New therapies for relapsed and refractory AML are needed, and this review explores the most recent advancements in immunotherapy in AML. Checkpoint Inhibitors utilizing innate or adaptive immune targeting have shown potential to improve AML outcomes when combined with hypomethylating agents and chemotherapy. The use of adoptive cell therapy in AML demonstrates promising early data, however, there is a need for better target selection. Although early in development, both vaccine therapy as well as stimulator of interferon genes (STING) agonists have potential to enhance the innate immune response to overcome AML's immune evasion. Immunotherapy has become a promising approach for AML treatment, especially in refractory and relapsed AML, especially in patients who are not eligible for allogeneic stem cell transplants. Future research should focus on a deeper understanding of the immune microenvironment to identify the most critical targets for optimization, as well as personalized therapeutic combination strategies. Here we present a comprehensive overview of the recent developments in immunotherapy for relapsed and refractory AML.
Collapse
Affiliation(s)
- Sawyer Bawek
- Department of Internal Medicine, University at Buffalo, Buffalo, NY, United States
- Leukemia Service, Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Sayuri Gurusinghe
- Department of Internal Medicine, University at Buffalo, Buffalo, NY, United States
| | - Matthew Burwinkel
- Department of Internal Medicine, University at Buffalo, Buffalo, NY, United States
| | - Amanda Przespolewski
- Leukemia Service, Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
- Cell Therapy, Bristol Myers Squibb, Princeton, NJ, United States
| |
Collapse
|
12
|
Stajer M, Horacek JM, Kupsa T, Zak P. The role of chemokines and interleukins in acute lymphoblastic leukemia: a systematic review. J Appl Biomed 2024; 22:165-184. [PMID: 40033805 DOI: 10.32725/jab.2024.024] [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: 08/10/2024] [Accepted: 11/15/2024] [Indexed: 03/05/2025] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common childhood hematological malignancy, but it also affects adult patients with worse prognosis and outcomes. Leukemic cells benefit from protective mechanisms, which are mediated by intercellular signaling molecules - cytokines. Through these signals, cytokines modulate the biology of leukemic cells and their surroundings, enhancing the proliferation, survival, and chemoresistance of the disease. This ultimately leads to disease progression, refractoriness, and relapse, decreasing the chances of curability and overall survival of the patients. Targeting and modulating these pathological processes without affecting the healthy physiology is desirable, offering more possibilities for the treatment of ALL patients, which still remains unsatisfactory in certain cases. In this review, we comprehensively analyze the existing literature and ongoing trials regarding the role of chemokines and interleukins in the biology of ALL. Focusing on the functional pathways, genetic background, and critical checkpoints, we constructed a summary of molecules that are promising for prognostic stratification and mainly therapeutic use. Targeted therapy, including chemokine and interleukin pathways, is a new and promising approach to the treatment of cancer. With the expansion of our knowledge, we are able to uncover a spectrum of new potential checkpoints in order to modulate the disease biology. Several cytokine-related targets are advancing toward clinical application, offering the hope of higher disease response rates to treatment.
Collapse
Affiliation(s)
- Martin Stajer
- University of Defence, Military Faculty of Medicine, Department of Military Internal Medicine and Military Hygiene, Hradec Kralove, Czech Republic
- University Hospital Hradec Kralove and Charles University, Faculty of Medicine in Hradec Kralove, Department of Internal Medicine IV - Hematology, Hradec Kralove, Czech Republic
| | - Jan M Horacek
- University of Defence, Military Faculty of Medicine, Department of Military Internal Medicine and Military Hygiene, Hradec Kralove, Czech Republic
- University Hospital Hradec Kralove and Charles University, Faculty of Medicine in Hradec Kralove, Department of Internal Medicine IV - Hematology, Hradec Kralove, Czech Republic
| | - Tomas Kupsa
- University of Defence, Military Faculty of Medicine, Department of Military Internal Medicine and Military Hygiene, Hradec Kralove, Czech Republic
- University Hospital Hradec Kralove and Charles University, Faculty of Medicine in Hradec Kralove, Department of Internal Medicine IV - Hematology, Hradec Kralove, Czech Republic
| | - Pavel Zak
- University Hospital Hradec Kralove and Charles University, Faculty of Medicine in Hradec Kralove, Department of Internal Medicine IV - Hematology, Hradec Kralove, Czech Republic
| |
Collapse
|
13
|
Lee H, Ko N, Namgoong S, Ham S, Koo J. Recent advances in and applications of ex vivo drug sensitivity analysis for blood cancers. Blood Res 2024; 59:37. [PMID: 39503808 PMCID: PMC11541977 DOI: 10.1007/s44313-024-00032-8] [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: 07/31/2024] [Accepted: 09/06/2024] [Indexed: 11/09/2024] Open
Abstract
Blood cancers, including leukemia, multiple myeloma, and lymphoma, pose significant challenges owing to their heterogeneous nature and the limitations of traditional treatments. Precision medicine has emerged as a transformative approach that offers tailored therapeutic strategies based on individual patient profiles. Ex vivo drug sensitivity analysis is central to this advancement, which enables testing of patient-derived cancer cells against a panel of therapeutic agents to predict clinical responses. This review provides a comprehensive overview of the latest advancements in ex vivo drug sensitivity analyses and their application in blood cancers. We discuss the development of more comprehensive drug response metrics and the evaluation of drug combinations to identify synergistic interactions. Additionally, we present evaluation of the advanced therapeutics such as antibody-drug conjugates using ex vivo assays. This review describes the critical role of ex vivo drug sensitivity analyses in advancing precision medicine by examining technological innovations and clinical applications. Ultimately, these innovations are paving the way for more effective and individualized treatments, improving patient outcomes, and establishing new standards for the management of blood cancers.
Collapse
Affiliation(s)
- Haeryung Lee
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea
| | - Nahee Ko
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea
| | - Sujin Namgoong
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea
| | - Seunghyok Ham
- ImpriMedKorea, Inc., Seoul, 03920, Republic of Korea
| | - Jamin Koo
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea.
- ImpriMedKorea, Inc., Seoul, 03920, Republic of Korea.
| |
Collapse
|
14
|
Valsasina B, Orsini P, Terenghi C, Ocana A. Present Scenario and Future Landscape of Payloads for ADCs: Focus on DNA-Interacting Agents. Pharmaceuticals (Basel) 2024; 17:1338. [PMID: 39458979 PMCID: PMC11510327 DOI: 10.3390/ph17101338] [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: 08/08/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/28/2024] Open
Abstract
ADCs have emerged as a promising class of therapeutics, combining the targeting specificity of monoclonal antibodies with the cytotoxic potency of small-molecule drugs. Although the majority of approved ADCs are still based on microtubule binder payloads, the recent success of topoisomerase I inhibitors has revitalized interest in the identification of novel agents overcoming present limitations in the field including narrow therapeutic window and chemoresistance. The success of DNA binders as payload for ADCs has been very limited, up to now, due, among other factors, to high hydrophobicity and planar chemical structures resulting in most cases in ADCs with a strong tendency to aggregate, poor plasma stability, and limited therapeutic index. Some of these molecules, however, continue to be of interest due to their favorable properties in terms of cytotoxic potency even in chemoresistant settings, bystander and immunogenic cell death effects, and known combinability with approved drugs. We critically evaluated several clinically tested ADCs containing DNA binders, focusing on payload physicochemical properties, cytotoxic potency, and obtained clinical results. Our analysis suggests that further exploration of certain chemical classes, specifically anthracyclines and duocarmycins, based on the optimization of physicochemical parameters, reduction of cytotoxic potency, and careful design of targeting molecules is warranted. This approach will possibly result in a novel generation of payloads overcoming the limitations of clinically validated ADCs.
Collapse
Affiliation(s)
| | - Paolo Orsini
- Nerviano Medical Sciences, Viale Pasteur 10, 20014 Nerviano, Italy
| | - Chiara Terenghi
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Alberto Ocana
- Experimental Therapeutics Unit, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria San Carlos (IdISSC), 2546 Madrid, Spain
- START Madrid-FJD, Hospital Fundación Jiménez Díaz, 2546 Madrid, Spain
- Breast Cancer, Centro de Investigación Biomédica en Red en Oncología (CIBERONC), 2546 Madrid, Spain
| |
Collapse
|
15
|
Wu X, Wang F, Yang X, Gong Y, Niu T, Chu B, Qu Y, Qian Z. Advances in Drug Delivery Systems for the Treatment of Acute Myeloid Leukemia. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2403409. [PMID: 38934349 DOI: 10.1002/smll.202403409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 06/06/2024] [Indexed: 06/28/2024]
Abstract
Acute myeloid leukemia (AML) is a common and catastrophic hematological neoplasm with high mortality rates. Conventional therapies, including chemotherapy, hematopoietic stem cell transplantation (HSCT), immune therapy, and targeted agents, have unsatisfactory outcomes for AML patients due to drug toxicity, off-target effects, drug resistance, drug side effects, and AML relapse and refractoriness. These intrinsic limitations of current treatments have promoted the development and application of nanomedicine for more effective and safer leukemia therapy. In this review, the classification of nanoparticles applied in AML therapy, including liposomes, polymersomes, micelles, dendrimers, and inorganic nanoparticles, is reviewed. In addition, various strategies for enhancing therapeutic targetability in nanomedicine, including the use of conjugating ligands, biomimetic-nanotechnology, and bone marrow targeting, which indicates the potential to reverse drug resistance, are discussed. The application of nanomedicine for assisting immunotherapy is also involved. Finally, the advantages and possible challenges of nanomedicine for the transition from the preclinical phase to the clinical phase are discussed.
Collapse
Affiliation(s)
- Xia Wu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Fangfang Wang
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Xijing Yang
- The Experimental Animal Center of West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Yuping Gong
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Ting Niu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Bingyang Chu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Ying Qu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Zhiyong Qian
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| |
Collapse
|
16
|
Gill SK, Sugiman-Marangos SN, Beilhartz GL, Mei E, Taipale M, Melnyk RA. An enhanced intracellular delivery platform based on a distant diphtheria toxin homolog that evades pre-existing antitoxin antibodies. EMBO Mol Med 2024; 16:2638-2651. [PMID: 39160301 PMCID: PMC11473700 DOI: 10.1038/s44321-024-00116-z] [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: 05/02/2024] [Revised: 07/15/2024] [Accepted: 07/18/2024] [Indexed: 08/21/2024] Open
Abstract
Targeted intracellular delivery of therapeutic proteins remains a significant unmet challenge in biotechnology. A promising approach is to leverage the intrinsic capabilities of bacterial toxins like diphtheria toxin (DT) to deliver a potent cytotoxic enzyme into cells with an associated membrane translocation moiety. Despite showing promising clinical efficacy, widespread deployment of DT-based therapeutics is complicated by the prevalence of pre-existing antibodies in the general population arising from childhood DT toxoid vaccinations, which impact the exposure, efficacy, and safety of these potent molecules. Here, we describe the discovery and characterization of a distant DT homolog from the ancient reptile pathogen Austwickia chelonae that we have dubbed chelona toxin (ACT). We show that ACT is comparable to DT structure and function in all respects except that it is not recognized by pre-existing anti-DT antibodies circulating in human sera. Furthermore, we demonstrate that ACT delivers heterologous therapeutic cargos into target cells more efficiently than DT. Our findings highlight ACT as a promising new chassis for building next-generation immunotoxins and targeted delivery platforms with improved pharmacokinetic and pharmacodynamic properties.
Collapse
Affiliation(s)
- Shivneet K Gill
- Department of Biochemistry, University of Toronto, Toronto, ON, M5S1A8, Canada
- Molecular Medicine Program, The Hospital for Sick Children Research Institute, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
| | - Seiji N Sugiman-Marangos
- Molecular Medicine Program, The Hospital for Sick Children Research Institute, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
| | - Greg L Beilhartz
- Molecular Medicine Program, The Hospital for Sick Children Research Institute, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
| | - Elizabeth Mei
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Mikko Taipale
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Roman A Melnyk
- Department of Biochemistry, University of Toronto, Toronto, ON, M5S1A8, Canada.
- Molecular Medicine Program, The Hospital for Sick Children Research Institute, 686 Bay Street, Toronto, ON, M5G 0A4, Canada.
| |
Collapse
|
17
|
Mosna F. The Immunotherapy of Acute Myeloid Leukemia: A Clinical Point of View. Cancers (Basel) 2024; 16:2359. [PMID: 39001421 PMCID: PMC11240611 DOI: 10.3390/cancers16132359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 06/16/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
The potential of the immune system to eradicate leukemic cells has been consistently demonstrated by the Graft vs. Leukemia effect occurring after allo-HSCT and in the context of donor leukocyte infusions. Various immunotherapeutic approaches, ranging from the use of antibodies, antibody-drug conjugates, bispecific T-cell engagers, chimeric antigen receptor (CAR) T-cells, and therapeutic infusions of NK cells, are thus currently being tested with promising, yet conflicting, results. This review will concentrate on various types of immunotherapies in preclinical and clinical development, from the point of view of a clinical hematologist. The most promising therapies for clinical translation are the use of bispecific T-cell engagers and CAR-T cells aimed at lineage-restricted antigens, where overall responses (ORR) ranging from 20 to 40% can be achieved in a small series of heavily pretreated patients affected by refractory or relapsing leukemia. Toxicity consists mainly in the occurrence of cytokine-release syndrome, which is mostly manageable with step-up dosing, the early use of cytokine-blocking agents and corticosteroids, and myelosuppression. Various cytokine-enhanced natural killer products are also being tested, mainly as allogeneic off-the-shelf therapies, with a good tolerability profile and promising results (ORR: 20-37.5% in small trials). The in vivo activation of T lymphocytes and NK cells via the inhibition of their immune checkpoints also yielded interesting, yet limited, results (ORR: 33-59%) but with an increased risk of severe Graft vs. Host disease in transplanted patients. Therefore, there are still several hurdles to overcome before the widespread clinical use of these novel compounds.
Collapse
Affiliation(s)
- Federico Mosna
- Hematology and Bone Marrow Transplantation Unit (BMTU), Hospital of Bolzano (SABES-ASDAA), Teaching Hospital of Paracelsus Medical University (PMU), 39100 Bolzano, Italy
| |
Collapse
|
18
|
Brivio E, Bautista F, Zwaan CM. Naked antibodies and antibody-drug conjugates: targeted therapy for childhood acute lymphoblastic leukemia. Haematologica 2024; 109:1700-1712. [PMID: 38832425 PMCID: PMC11141655 DOI: 10.3324/haematol.2023.283815] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 03/15/2024] [Indexed: 06/05/2024] Open
Abstract
The treatment of childhood acute lymphoblastic leukemia (ALL) has reached overall survival rates exceeding 90%. The present and future challenges are to cure the remainder of patients still dying from disease, and to reduce morbidity and mortality in those who can be cured with standard-of-care chemotherapy by replacing toxic chemotherapy elements while retaining cure rates. With the novel therapeutic options introduced in the last years, including immunotherapies and targeted antibodies, the treatment of ALL is undergoing major changes. For B-cell precursor ALL, blinatumomab, an anti-CD19 bispecific antibody, has established its role in the consolidation treatment for both high- and standard-risk first relapse of ALL, in the presence of bone marrow involvement, and may also have an impact on the outcome of high-risk subsets such as infant ALL and Philadelphia chromosome-positive ALL. Inotuzumab ozogamicin, an anti-CD22 drug conjugated antibody, has demonstrated high efficacy in inducing complete remission in relapsed ALL, even in the presence of high tumor burden, but randomized phase III trials are still ongoing. For T-ALL the role of CD38-directed treatment, such as daratumumab, is gaining interest, but randomized data are needed to assess its specific benefit. These antibodies are currently being tested in patients with newly diagnosed ALL and may lead to major changes in the present paradigm of treatment of pediatric ALL. Unlike the past, lessons may be learned from innovations in adult ALL, in which more drastic changes are piloted that may need to be translated to pediatrics.
Collapse
Affiliation(s)
- Erica Brivio
- Princess Máxima Center for Pediatric Oncology, Utrecht
| | | | - C. Michel Zwaan
- Princess Máxima Center for Pediatric Oncology, Utrecht
- Pediatric Oncology, Erasmus MC-Sophia Children’s Hospital, Rotterdam, the Netherlands
| |
Collapse
|
19
|
Journeaux T, Bernardes GJL. Homogeneous multi-payload antibody-drug conjugates. Nat Chem 2024; 16:854-870. [PMID: 38760431 DOI: 10.1038/s41557-024-01507-y] [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: 09/20/2023] [Accepted: 03/14/2024] [Indexed: 05/19/2024]
Abstract
Many systemic cancer chemotherapies comprise a combination of drugs, yet all clinically used antibody-drug conjugates (ADCs) contain a single-drug payload. These combination regimens improve treatment outcomes by producing synergistic anticancer effects and slowing the development of drug-resistant cell populations. In an attempt to replicate these regimens and improve the efficacy of targeted therapy, the field of ADCs has moved towards developing techniques that allow for multiple unique payloads to be attached to a single antibody molecule with high homogeneity. However, the methods for generating such constructs-homogeneous multi-payload ADCs-are both numerous and complex owing to the plethora of reactive functional groups that make up the surface of an antibody. Here, by summarizing and comparing the methods of both single- and multi-payload ADC generation and their key preclinical and clinical results, we provide a timely overview of this relatively new area of research. The methods discussed range from branched linker installation to the incorporation of unnatural amino acids, with a generalized comparison tool of the most promising modification strategies also provided. Finally, the successes and challenges of this rapidly growing field are critically evaluated, and from this, future areas of research and development are proposed.
Collapse
Affiliation(s)
- Toby Journeaux
- Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Gonçalo J L Bernardes
- Department of Chemistry, University of Cambridge, Cambridge, UK.
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
20
|
Massone C, Rivoli G, Sola S, Angelucci E. Blastic plasmacytoid dendritic cell neoplasm: a short review and update. Dermatol Reports 2024; 16:9781. [PMID: 39295884 PMCID: PMC11406213 DOI: 10.4081/dr.2023.9781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 07/01/2023] [Indexed: 09/21/2024] Open
Abstract
Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a rare hematologic neoplasm (less than 1% of primary cutaneous lymphomas and acute leukemia) with a highly aggressive clinical course and frequent skin, bone marrow and central nervous system involvement. Even though there is often an early response to chemotherapy, leukemic dissemination relapses are very common and result in poor outcomes, with a median overall survival of 8 to 14 months in the first-line setting using standard combination chemotherapy regimens. Almost 90% of patients experience skin involvement as their initial site of infection, where BPDCN may stay restricted for weeks or even months until a swift secondary phase involving multiple organs takes place. Consequently, it is crucial to suspect and identify early skin lesions, as well as to conduct and report a skin biopsy as soon as possible. In order to diagnose and treat BPDCN, a multidisciplinary strategy involving collaboration between pathologists, hematologists, and dermatologists is unquestionably essential.
Collapse
Affiliation(s)
- Cesare Massone
- Dermatology Unit and Scientific Directorate, Galliera Hospital, Genoa
| | - Giulia Rivoli
- Hematology and Cellular Therapies Operational Unit, IRCCS San Martino Polyclinic Hospital, Genoa
| | - Simona Sola
- Department of Surgical Pathology, Galliera Hospital, Genoa, Italy
| | - Emanuele Angelucci
- Hematology and Cellular Therapies Operational Unit, IRCCS San Martino Polyclinic Hospital, Genoa
| |
Collapse
|
21
|
Pemmaraju N, Deconinck E, Mehta P, Walker I, Herling M, Garnache-Ottou F, Gabarin N, Campbell CJV, Duell J, Moshe Y, Mughal T, Mohty M, Angelucci E. Recent Advances in the Biology and CD123-Directed Treatment of Blastic Plasmacytoid Dendritic Cell Neoplasm. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:e130-e137. [PMID: 38267355 DOI: 10.1016/j.clml.2023.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/05/2023] [Accepted: 12/14/2023] [Indexed: 01/26/2024]
Abstract
Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is an aggressive myeloid malignancy of the dendritic cell lineage that affects patients of all ages, though the incidence appears to be highest in patients over the age of 60 years. Diagnosis is based on the presence of plasmacytoid dendritic cell precursors expressing CD123, the interleukin-3 (IL-3) receptor alpha, and a distinct histologic appearance. Timely diagnosis remains a challenge, due to lack of disease awareness and overlapping biologic and clinical features with other hematologic malignancies. Prognosis is poor with a median overall survival of 8 to 14 months, irrespective of disease presentation pattern. Historically, the principal treatment was remission induction therapy followed by a stem cell transplant (SCT) in eligible patients. However, bridging to SCT is often not achieved with induction chemotherapy regimens. The discovery that CD123 is universally expressed in BPDCN and is considered to have a pathogenetic role in its development paved the way for the successful introduction of tagraxofusp, a recombinant human IL-3 fused to a truncated diphtheria toxin payload, as an initial treatment for BPDCN. Tagraxofusp was approved in 2018 by the United States Food and Drug Administration for the treatment of patients aged 2 years and older with newly diagnosed and relapsed/refractory BPDCN, and by the European Medicines Agency in 2021 for first-line treatment of adults. The advent of tagraxofusp has opened a new era of precision oncology in the treatment of BPDCN. Herein, we present an overview of BPDCN biology, its diagnosis, and treatment options, illustrated by clinical cases.
Collapse
Affiliation(s)
- Naveen Pemmaraju
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX.
| | - Eric Deconinck
- Department of Hematology, CHU Besançon, Besançon Cedex, France; INSERM, UMR1098 RIGHT, Franche-Comté University, Établissement Français du Sang, Besançon, France
| | - Priyanka Mehta
- Department of Haematology, University Hospitals of Bristol and Weston, NHS Foundation Trust, Bristol, United Kingdom
| | - Irwin Walker
- Department of Medicine, McMaster University, Hamilton, ON, Canada; Juravinski Hospital and Cancer Centre, Hamilton, ON, Canada
| | - Marco Herling
- Department of Hematology, Cellular Therapy, and Hemostaseology, University of Leipzig, Leipzig, Germany
| | - Francine Garnache-Ottou
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France; Etablissement Français du Sang Bourgogne Franche-Comté, Laboratoire d'Hématologie et d'Immunologie Régional, Besançon, France
| | - Nadia Gabarin
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Clinton J V Campbell
- Juravinski Hospital and Cancer Centre, Hamilton, ON, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Johannes Duell
- Medizinische Klinik und Poliklinik II des Universitätsklinikums, Zentrum Innere Medizin (ZIM), Würzburg, Germany
| | - Yakir Moshe
- Department of Hematology and Bone Marrow Transplantation, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Tariq Mughal
- Division of Hematology-Oncology, Tufts University School of Medicine, Boston, MA; Consultant to Stemline Therapeutics Inc, New York, NY
| | - Mohamad Mohty
- Department of Hematology and Cellular Therapy, Saint-Antoine Hospital, Sorbonne University, Paris, France
| | - Emanuele Angelucci
- Hematology and Cellular Therapy, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
22
|
Daver NG, Montesinos P, DeAngelo DJ, Wang ES, Papadantonakis N, Todisco E, Sweet KL, Pemmaraju N, Lane AA, Torres-Miñana L, Thompson JE, Konopleva MY, Sloss CM, Watkins K, Bedse G, Du Y, Malcolm KE, Zweidler-McKay PA, Kantarjian HM. Pivekimab sunirine (IMGN632), a novel CD123-targeting antibody-drug conjugate, in relapsed or refractory acute myeloid leukaemia: a phase 1/2 study. Lancet Oncol 2024; 25:388-399. [PMID: 38423051 PMCID: PMC11103591 DOI: 10.1016/s1470-2045(23)00674-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/14/2023] [Accepted: 12/19/2023] [Indexed: 03/02/2024]
Abstract
BACKGROUND Pivekimab sunirine (IMGN632) is a first-in-class antibody-drug conjugate comprising a high-affinity CD123 antibody, cleavable linker, and novel indolinobenzodiazepine pseudodimer payload. CD123 is overexpressed in several haematological malignancies, including acute myeloid leukaemia. We present clinical data on pivekimab sunirine in relapsed or refractory acute myeloid leukaemia. METHODS This first-in-human, phase 1/2 dose-escalation and dose-expansion study enrolled participants aged 18 years or older at nine hospitals in France, Italy, Spain, and the USA with CD123+ haematological malignancies (Eastern Cooperative Oncology Group performance status of 0-1); participants reported here were in a cohort of participants with acute myeloid leukaemia who were refractory to or had relapsed on one or more previous treatments for acute myeloid leukaemia. The 3 + 3 dose-escalation phase evaluated two dosing schedules: schedule A (once every 3 weeks, on day 1 of a 3-week cycle) and fractionated schedule B (days 1, 4, and 8 of a 3-week cycle). The dose-expansion phase evaluated two cohorts: one cohort given 0·045 mg/kg of bodyweight (schedule A) and one cohort given 0·090 mg/kg of bodyweight (schedule A). The primary endpoints were the maximum tolerated dose and the recommended phase 2 dose. Antileukaemia activity (overall response and a composite complete remission assessment) was a secondary endpoint. The study is ongoing and registered with ClinicalTrials.gov, NCT03386513. FINDINGS Between Dec 29, 2017, and May 27, 2020, 91 participants were enrolled (schedule A, n=68; schedule B, n=23). 30 (44%) of schedule A participants were female and 38 (56%) were male; 60 (88%) were White, six (9%) were Black or African American, and two (3%) were other races. Pivekimab sunirine at doses of 0·015 mg/kg to 0·450 mg/kg in schedule A was administered in six escalating doses with no maximum tolerated dose defined; three dose-limiting toxicities were observed (reversible veno-occlusive disease; 0·180 mg/kg, n=1 and 0·450 mg/kg, n=1; and neutropenia; 0·300 mg/kg, n=1). Schedule B was not pursued further on the basis of comparative safety and antileukaemia findings with schedule A. The recommended phase 2 dose was selected as 0·045 mg/kg once every 3 weeks. At the recommended phase 2 dose (n=29), the most common grade 3 or worse treatment-related adverse events were febrile neutropenia (three [10%]), infusion-related reactions (two [7%]), and anaemia (two [7%]). Treatment-related serious adverse events occurring in 5% or more of participants treated at the recommended phase 2 dose were febrile neutropenia (two [7%]) and infusion-related reactions (two [7%]). Among 68 participants who received schedule A, one death (1%) was considered to be treatment-related (cause unknown; 0·300 mg/kg cohort). At the recommended phase 2 dose, the overall response rate was 21% (95% CI 8-40; six of 29) and the composite complete remission rate was 17% (95% CI 6-36; five of 29). INTERPRETATION Pivekimab sunirine showed single-agent activity across multiple doses, with a recommended phase 2 dose of 0·045 mg/kg once every 3 weeks. These findings led to a phase 1b/2 study of pivekimab sunirine plus azacitidine and venetoclax in patients with CD123-positive acute myeloid leukaemia. FUNDING ImmunoGen.
Collapse
Affiliation(s)
| | - Pau Montesinos
- Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | | | - Eunice S Wang
- Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Kayser S. Antibody-drug conjugates in acute myeloid leukaemia: more research needed. Lancet Oncol 2024; 25:272-273. [PMID: 38423042 DOI: 10.1016/s1470-2045(24)00014-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/03/2024] [Accepted: 01/09/2024] [Indexed: 03/02/2024]
Affiliation(s)
- Sabine Kayser
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany; NCT Trial Centre, National Centre of Tumor Diseases, German Cancer Research Center, Heidelberg, Germany.
| |
Collapse
|
24
|
Pemmaraju N, Madanat YF, Rizzieri D, Fazal S, Rampal R, Mannis G, Wang ES, Foran J, Lane AA. Treatment of patients with blastic plasmacytoid dendritic cell neoplasm (BPDCN): focus on the use of tagraxofusp and clinical considerations. Leuk Lymphoma 2024:1-12. [PMID: 38391126 DOI: 10.1080/10428194.2024.2305288] [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: 06/09/2023] [Accepted: 01/09/2024] [Indexed: 02/24/2024]
Abstract
BPDCN is an aggressive myeloid malignancy with a poor prognosis. It derives from the precursors of plasmacytoid dendritic cells and is characterized by CD123 overexpression, which is seen in all patients with BPDCN. The CD123-directed therapy tagraxofusp is the only approved treatment for BPDCN; it was approved in the US as monotherapy for the treatment of patients aged ≥2 years with treatment-naive or relapsed/refractory BPDCN. Herein, we review the available data supporting the utility of tagraxofusp in treating patients with BPDCN. In addition, we present best practices and real-world insights from clinicians in academic and community settings in the US on how they use tagraxofusp to treat BPDCN. Several case studies illustrate the efficacy of tagraxofusp and discuss its safety profile, as well as the prevention, mitigation, and management of anticipated adverse events.
Collapse
Affiliation(s)
- Naveen Pemmaraju
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - David Rizzieri
- Novant Health Cancer Institute, Winston Salem, North Carolina, USA
| | - Salman Fazal
- Allegheny Health Network, Pittsburgh, Pennsylvania, USA
| | - Raajit Rampal
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Eunice S Wang
- Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - James Foran
- Mayo Clinic Cancer Center, Jacksonville, Florida, USA
| | - Andrew A Lane
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| |
Collapse
|
25
|
Song J, Li W, Bai Y, Zhou P, Niu J, Niu X, Liu Y, Liu X, Drokow EK, Sun K, Zhou H. A blastic plasmacytoid dendritic cell neoplasm-like immunophenotype is negatively associated with CEBPA bZIP mutation and predicts unfavorable prognosis in acute myeloid leukemia. Ann Hematol 2024; 103:463-473. [PMID: 38183444 DOI: 10.1007/s00277-023-05594-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 12/17/2023] [Indexed: 01/08/2024]
Abstract
Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a rare and aggressive myeloid malignancy which characteristically expresses an atypical phenotype including CD123+, CD56+, and CD4+. We are aimed to investigate the clinical and prognostic characteristics of AML patients exhibiting BPDCN-like immunophenotype and provide additional insights for risk stratification of AML. A total of 241 newly diagnosed AML patients were enrolled in this retrospective study and categorized into BPDCN-like positive (n = 125)/negative (n = 116) groups, determined by the present with CD123+ along with either CD56+ or CD4+, or both. Subsequently, an analysis was conducted to examine the general clinical characteristics, genetic profiles, and prognosis of the two respective groups. Patients with BPDCN-like immunophenotype manifested higher frequencies of acute myelomonocytic leukemia and acute monoblastic leukemia. Surprisingly, the presence of the BPDCN-like immunophenotype exhibited an inverse relationship with CEBPA bZIP mutation. Notably, patients with BPDCN-like phenotype had both worse OS and EFS compared to those without BPDCN-like phenotype. In the CN-AML subgroups, the BPDCN-like phenotype was associated with worse EFS. Similarly, a statistically significant disparity was observed in both OS and EFS within the favorable-risk subgroup, while only OS was significant within the adverse-risk subgrouMoreover, patients possessing favorable-risk genetics without BPDCN-like phenotype had the longest survival, whereas those who had both adverse-risk genetics and BPDCN-like phenotype exhibited the worst survival. Our study indicated that BPDCN-like phenotype negatively associated with CEBPA bZIP mutation and revealed a significantly poor prognosis in AML. Moreover, the 2022 ELN classification, in combination with the BPDCN-like phenotype, may better distinguish between different risk groups.
Collapse
Affiliation(s)
- Juanjuan Song
- Department of Hematology, Affiliated Tumor Hospital of Zhengzhou University, Tumor Hospital of Henan Province, Institute of Hematology of Henan Province, No.127 Dongming Road, Jinshui District, Zhengzhou, 450000, Henan, People's Republic of China
| | - Weiya Li
- Department of Hematology, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan, 450003, People's Republic of China
| | - Yanliang Bai
- Department of Hematology, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan, 450003, People's Republic of China
| | - Pan Zhou
- Department of Hematology, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan, 450003, People's Republic of China
| | - Junwei Niu
- Department of Hematology, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan, 450003, People's Republic of China
| | - Xiaona Niu
- Department of Hematology, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan, 450003, People's Republic of China
| | - Ying Liu
- Department of Hematology, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan, 450003, People's Republic of China
| | - Xiaobo Liu
- Department of Hematology, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan, 450003, People's Republic of China
| | - Emmanuel Kwateng Drokow
- Department of Hematology, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan, 450003, People's Republic of China
| | - Kai Sun
- Department of Hematology, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan, 450003, People's Republic of China.
| | - Hu Zhou
- Department of Hematology, Affiliated Tumor Hospital of Zhengzhou University, Tumor Hospital of Henan Province, Institute of Hematology of Henan Province, No.127 Dongming Road, Jinshui District, Zhengzhou, 450000, Henan, People's Republic of China.
| |
Collapse
|
26
|
Wu Y, Li Y, Gao Y, Zhang P, Jing Q, Zhang Y, Jin W, Wang Y, Du J, Wu G. Immunotherapies of acute myeloid leukemia: Rationale, clinical evidence and perspective. Biomed Pharmacother 2024; 171:116132. [PMID: 38198961 DOI: 10.1016/j.biopha.2024.116132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 12/28/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024] Open
Abstract
Acute myeloid leukemia (AML) is a prevalent hematological malignancy that exhibits a wide array of molecular abnormalities. Although traditional treatment modalities such as chemotherapy and allogeneic stem cell transplantation (HSCT) have become standard therapeutic approaches, a considerable number of patients continue to face relapse and encounter a bleak prognosis. The emergence of immune escape, immunosuppression, minimal residual disease (MRD), and other contributing factors collectively contribute to this challenge. Recent research has increasingly highlighted the notable distinctions between AML tumor microenvironments and those of healthy individuals. In order to investigate the potential therapeutic mechanisms, this study examines the intricate transformations occurring between leukemic cells and their surrounding cells within the tumor microenvironment (TME) of AML. This review classifies immunotherapies into four distinct categories: cancer vaccines, immune checkpoint inhibitors (ICIs), antibody-based immunotherapies, and adoptive T-cell therapies. The results of numerous clinical trials strongly indicate that the identification of optimal combinations of novel agents, either in conjunction with each other or with chemotherapy, represents a crucial advancement in this field. In this review, we aim to explore the current and emerging immunotherapeutic methodologies applicable to AML patients, identify promising targets, and emphasize the crucial requirement to augment patient outcomes. The application of these strategies presents substantial therapeutic prospects within the realm of precision medicine for AML, encompassing the potential to ameliorate patient outcomes.
Collapse
Affiliation(s)
- Yunyi Wu
- Department of Central Laboratory, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China; Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yanchun Li
- Department of Central Laboratory, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Yan Gao
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ping Zhang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qiangan Jing
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yinhao Zhang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Weidong Jin
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ying Wang
- Department of Central Laboratory, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China.
| | - Jing Du
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Gongqiang Wu
- Department of Hematology, Dongyang Hospitai Affiliated to Wenzhou Medical University, Dongyang People's Hospital, Dongyang, Zhejiang, China.
| |
Collapse
|
27
|
Zanotta S, Galati D, De Filippi R, Pinto A. Breakthrough in Blastic Plasmacytoid Dendritic Cell Neoplasm Cancer Therapy Owing to Precision Targeting of CD123. Int J Mol Sci 2024; 25:1454. [PMID: 38338733 PMCID: PMC10855071 DOI: 10.3390/ijms25031454] [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: 01/02/2024] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a rare and aggressive hematologic cancer originating from the malignant transformation of plasmacytoid dendritic cell precursors. This malignancy progresses rapidly, with frequent relapses and a poor overall survival rate, underscoring the urgent need for effective treatments. However, diagnosing and treating BPDCN have historically been challenging due to its rarity and the lack of standardized approaches. The recognition of BPDCN as a distinct disease entity is recent, and standardized treatment protocols are yet to be established. Traditionally, conventional chemotherapy and stem cell transplantation have been the primary methods for treating BPDCN patients. Advances in immunophenotyping and molecular profiling have identified potential therapeutic targets, leading to a shift toward CD123-targeted immunotherapies in both clinical and research settings. Ongoing developments with SL-401, IMGN632, CD123 chimeric antigen receptor (CAR) T-cells, and bispecific antibodies (BsAb) show promising advancements. However, the therapeutic effectiveness of CD123-targeting treatments needs improvement through innovative approaches and combinations of treatments with other anti-leukemic drugs. The exploration of combinations such as CD123-targeted immunotherapies with azacitidine and venetoclax is suggested to enhance antineoplastic responses and improve survival rates in BPDCN patients. In conclusion, this multifaceted approach offers hope for more effective and tailored therapeutic interventions against this challenging hematologic malignancy.
Collapse
Affiliation(s)
- Serena Zanotta
- Hematology-Oncology and Stem-Cell Transplantation Unit, Department of Onco-Hematology and Innovative Diagnostics, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (S.Z.); (A.P.)
| | - Domenico Galati
- Hematology-Oncology and Stem-Cell Transplantation Unit, Department of Onco-Hematology and Innovative Diagnostics, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (S.Z.); (A.P.)
| | - Rosaria De Filippi
- Department of Clinical Medicine and Surgery, Università degli Studi di Napoli Federico II, 80131 Napoli, Italy;
| | - Antonio Pinto
- Hematology-Oncology and Stem-Cell Transplantation Unit, Department of Onco-Hematology and Innovative Diagnostics, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (S.Z.); (A.P.)
| |
Collapse
|
28
|
Chan O, Walker AR. Novel therapies upon failure of HMA plus venetoclax. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2023; 2023:702-708. [PMID: 38066883 PMCID: PMC10727075 DOI: 10.1182/hematology.2023000456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
The efficacy and tolerability of the combination of hypomethylating agents with venetoclax (HMA-VEN) in patients with newly diagnosed acute myeloid leukemia has been a practice-changing milestone in the field. However, treatment failure and relapse remain major barriers to prolonged survival. TP53 mutation is a predictor of primary induction failure and portends especially poor outcomes. Prelinical data suggest that VEN resistance stems from these genetic changes, which lead to increases in antiapoptotic proteins such as MCL-1 and BCLXL. For patients who discontinue HMA-VEN for reasons other than disease progression, such as post allotransplantation, infection, and personal preference, rechallenge with HMA-VEN at the time of relapse may be considered. For those who progress on HMA-VEN, clinical trials with novel agents or rational drug combinations are preferred if available. If no trial option is available, fit patients may benefit from intensive chemotherapy. Emerging therapies aim to overcome venetoclax resistance, target interactions that promote leukemogenesis, and harness the immune system to irradicate leukemic blasts and stem cells.
Collapse
Affiliation(s)
- Onyee Chan
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL
| | - Alison R Walker
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL
| |
Collapse
|
29
|
Murphy LA, Winters AC. Emerging and Future Targeted Therapies for Pediatric Acute Myeloid Leukemia: Targeting the Leukemia Stem Cells. Biomedicines 2023; 11:3248. [PMID: 38137469 PMCID: PMC10741170 DOI: 10.3390/biomedicines11123248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/01/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
Acute myeloid leukemia (AML) is a rare subtype of acute leukemia in the pediatric and adolescent population but causes disproportionate morbidity and mortality in this age group. Standard chemotherapeutic regimens for AML have changed very little in the past 3-4 decades, but the addition of targeted agents in recent years has led to improved survival in select subsets of patients as well as a better biological understanding of the disease. Currently, one key paradigm of bench-to-bedside practice in the context of adult AML is the focus on leukemia stem cell (LSC)-targeted therapies. Here, we review current and emerging immunotherapies and other targeted agents that are in clinical use for pediatric AML through the lens of what is known (and not known) about their LSC-targeting capability. Based on a growing understanding of pediatric LSC biology, we also briefly discuss potential future agents on the horizon.
Collapse
Affiliation(s)
- Lindsey A. Murphy
- Department of Pediatrics, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA;
| | - Amanda C. Winters
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| |
Collapse
|
30
|
Marone R, Landmann E, Devaux A, Lepore R, Seyres D, Zuin J, Burgold T, Engdahl C, Capoferri G, Dell’Aglio A, Larrue C, Simonetta F, Rositzka J, Rhiel M, Andrieux G, Gallagher DN, Schröder MS, Wiederkehr A, Sinopoli A, Do Sacramento V, Haydn A, Garcia-Prat L, Divsalar C, Camus A, Xu L, Bordoli L, Schwede T, Porteus M, Tamburini J, Corn JE, Cathomen T, Cornu TI, Urlinger S, Jeker LT. Epitope-engineered human hematopoietic stem cells are shielded from CD123-targeted immunotherapy. J Exp Med 2023; 220:e20231235. [PMID: 37773046 PMCID: PMC10541312 DOI: 10.1084/jem.20231235] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/01/2023] [Accepted: 09/08/2023] [Indexed: 09/30/2023] Open
Abstract
Targeted eradication of transformed or otherwise dysregulated cells using monoclonal antibodies (mAb), antibody-drug conjugates (ADC), T cell engagers (TCE), or chimeric antigen receptor (CAR) cells is very effective for hematologic diseases. Unlike the breakthrough progress achieved for B cell malignancies, there is a pressing need to find suitable antigens for myeloid malignancies. CD123, the interleukin-3 (IL-3) receptor alpha-chain, is highly expressed in various hematological malignancies, including acute myeloid leukemia (AML). However, shared CD123 expression on healthy hematopoietic stem and progenitor cells (HSPCs) bears the risk for myelotoxicity. We demonstrate that epitope-engineered HSPCs were shielded from CD123-targeted immunotherapy but remained functional, while CD123-deficient HSPCs displayed a competitive disadvantage. Transplantation of genome-edited HSPCs could enable tumor-selective targeted immunotherapy while rebuilding a fully functional hematopoietic system. We envision that this approach is broadly applicable to other targets and cells, could render hitherto undruggable targets accessible to immunotherapy, and will allow continued posttransplant therapy, for instance, to treat minimal residual disease (MRD).
Collapse
Affiliation(s)
- Romina Marone
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
- Transplantation Immunology and Nephrology, Basel University Hospital, Basel, Switzerland
| | - Emmanuelle Landmann
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
- Transplantation Immunology and Nephrology, Basel University Hospital, Basel, Switzerland
| | - Anna Devaux
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
- Transplantation Immunology and Nephrology, Basel University Hospital, Basel, Switzerland
| | - Rosalba Lepore
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
- Transplantation Immunology and Nephrology, Basel University Hospital, Basel, Switzerland
- Cimeio Therapeutics AG, Basel, Switzerland
- Ridgeline Discovery GmbH, Basel, Switzerland
| | - Denis Seyres
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
- Transplantation Immunology and Nephrology, Basel University Hospital, Basel, Switzerland
| | - Jessica Zuin
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
- Transplantation Immunology and Nephrology, Basel University Hospital, Basel, Switzerland
| | - Thomas Burgold
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
- Transplantation Immunology and Nephrology, Basel University Hospital, Basel, Switzerland
| | - Corinne Engdahl
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
- Transplantation Immunology and Nephrology, Basel University Hospital, Basel, Switzerland
| | - Giuseppina Capoferri
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
- Transplantation Immunology and Nephrology, Basel University Hospital, Basel, Switzerland
| | - Alessandro Dell’Aglio
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
- Transplantation Immunology and Nephrology, Basel University Hospital, Basel, Switzerland
| | - Clément Larrue
- Translational Research Centre in Onco-Hematology, Faculty of Medicine, University of Geneva, and Swiss Cancer Center Leman, Geneva, Switzerland
| | - Federico Simonetta
- Division of Hematology, Department of Oncology, Geneva University Hospitals, Geneva, Switzerland
- Department of Medicine, Translational Research Center for Onco-Hematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Julia Rositzka
- Institute for Transfusion Medicine and Gene Therapy, Medical Center - University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Manuel Rhiel
- Institute for Transfusion Medicine and Gene Therapy, Medical Center - University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Geoffroy Andrieux
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Freiburg, Germany
| | - Danielle N. Gallagher
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland
| | - Markus S. Schröder
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland
| | | | | | | | - Anna Haydn
- Ridgeline Discovery GmbH, Basel, Switzerland
| | | | | | - Anna Camus
- Cimeio Therapeutics AG, Basel, Switzerland
| | - Liwen Xu
- Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA, USA
| | - Lorenza Bordoli
- Biozentrum, University of Basel, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Torsten Schwede
- Biozentrum, University of Basel, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Matthew Porteus
- Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA, USA
| | - Jérôme Tamburini
- Department of Medicine, Translational Research Center for Onco-Hematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Jacob E. Corn
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland
| | - Toni Cathomen
- Institute for Transfusion Medicine and Gene Therapy, Medical Center - University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tatjana I. Cornu
- Institute for Transfusion Medicine and Gene Therapy, Medical Center - University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stefanie Urlinger
- Cimeio Therapeutics AG, Basel, Switzerland
- Ridgeline Discovery GmbH, Basel, Switzerland
| | - Lukas T. Jeker
- Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
- Transplantation Immunology and Nephrology, Basel University Hospital, Basel, Switzerland
| |
Collapse
|
31
|
Peng J, He S, Yang X, Huang L, Wei J. Plasmacytoid dendritic cell expansion in myeloid neoplasms: A novel distinct subset of myeloid neoplasm? Crit Rev Oncol Hematol 2023; 192:104186. [PMID: 37863402 DOI: 10.1016/j.critrevonc.2023.104186] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 10/06/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023] Open
Abstract
Plasmacytoid dendritic cells (pDCs) are a specific dendritic cell type stemming from the myeloid lineage. Clinically and pathologically, neoplasms associated with pDCs are classified as blastic plasmacytoid dendritic cell neoplasm (BPDCN), mature plasmacytoid dendritic myeloid neoplasm (MPDMN) and pDC expansion in myeloid neoplasms (MNs). BPDCN was considered a rare and aggressive neoplasm in the 2016 World Health Organization (WHO) classification. MPDMN, known as mature pDC-derived neoplasm, is closely related to MNs and was first recognized in the latest 2022 WHO classification, proposing a new concept that acute myeloid leukemia cases could show clonally expanded pDCs (pDC-AML). With the advances in detection techniques, an increasing number of pDC expansion in MNs have been reported, but whether the pathogenesis is similar to that of MPDMN remains unclear. This review focuses on patient characteristics, diagnosis and treatment of pDC expansion in MNs to gain further insight into this novel and unique provisional subtype.
Collapse
Affiliation(s)
- Juan Peng
- Department of Hematology, Tongji Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan 430000, Hubei, China; Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei 430030, China
| | - Shaolong He
- Department of Hematology, Tongji Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan 430000, Hubei, China; Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China
| | - Xingcheng Yang
- Department of Hematology, Tongji Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan 430000, Hubei, China; Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei 430030, China
| | - Liang Huang
- Department of Hematology, Tongji Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan 430000, Hubei, China; Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei 430030, China.
| | - Jia Wei
- Department of Hematology, Tongji Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan 430000, Hubei, China; Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei 430030, China; Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China; Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, 030032 Taiyuan, Shanxi, China.
| |
Collapse
|
32
|
Cuglievan B, Connors J, He J, Khazal S, Yedururi S, Dai J, Garces S, Quesada AE, Roth M, Garcia M, McCall D, Gibson A, Ragoonanan D, Petropoulos D, Tewari P, Nunez C, Mahadeo KM, Tasian SK, Lamble AJ, Pawlowska A, Hammond D, Maiti A, Haddad FG, Senapati J, Daver N, Gangat N, Konopleva M, Meshinchi S, Pemmaraju N. Blastic plasmacytoid dendritic cell neoplasm: a comprehensive review in pediatrics, adolescents, and young adults (AYA) and an update of novel therapies. Leukemia 2023; 37:1767-1778. [PMID: 37452102 PMCID: PMC10457206 DOI: 10.1038/s41375-023-01968-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 06/23/2023] [Accepted: 06/30/2023] [Indexed: 07/18/2023]
Abstract
Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a rare hematologic malignancy that can involve the bone marrow, peripheral blood, skin, lymph nodes, and the central nervous system. Though more common in older adults, BPDCN has been reported across all age groups, including infants and children. The incidence of pediatric BPDCN is extremely low and little is known about the disease. Pediatric BPDCN is believed to be clinically less aggressive but often with more dissemination at presentation than adult cases. Unlike adults who almost always proceed to a hematopoietic stem cell transplantation in first complete remission if transplant-eligible, the majority of children can be cured with a high-risk acute lymphoblastic leukemia-like regimen. Hematopoietic stem cell transplantation is recommended for children with high-risk disease, the definition of which continues to evolve, or those in relapse and refractory settings where outcomes continue to be dismal. Novel agents used in other hematologic malignancies and CD123 targeted agents, including chimeric antigen receptor T-cells and monoclonal/bispecific antibodies, are being brought into research and practice. Our goal is to provide a comprehensive review of presentation, diagnosis, and treatment by review of pediatric cases reported for the last 20 years, and a review of novel targeted therapies and therapies under investigation for adult and pediatric patients.
Collapse
Affiliation(s)
- Branko Cuglievan
- Division of Pediatrics, Department of Pediatric Patient Care, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Jeremy Connors
- Division of Pediatrics, Department of Pediatric Patient Care, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jiasen He
- Division of Pediatrics, Department of Pediatric Patient Care, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sajad Khazal
- Division of Pediatrics, Department of Pediatric Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sireesha Yedururi
- Division of Radiology, Department of Abdominal Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Julia Dai
- Division of Internal Medicine, Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sofia Garces
- Division of Pathology, Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andres E Quesada
- Division of Pathology, Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael Roth
- Division of Pediatrics, Department of Pediatric Patient Care, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Miriam Garcia
- Division of Pediatrics, Department of Pediatric Patient Care, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David McCall
- Division of Pediatrics, Department of Pediatric Patient Care, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amber Gibson
- Division of Pediatrics, Department of Pediatric Patient Care, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dristhi Ragoonanan
- Division of Pediatrics, Department of Pediatric Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Demetrios Petropoulos
- Division of Pediatrics, Department of Pediatric Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Priti Tewari
- Division of Pediatrics, Department of Pediatric Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cesar Nunez
- Division of Pediatrics, Department of Pediatric Patient Care, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kris M Mahadeo
- Division of Pediatric Transplantation and Cellular Therapy, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Sarah K Tasian
- Division of Oncology and Center for Childhood Cancer Research, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Adam J Lamble
- Division of Hematology/Oncology, Seattle Children's Hospital, University of Washington, Seattle, WA, USA
| | - Anna Pawlowska
- Division of Pediatric Hematology/Oncology, and Hematopoietic Stem Cell Transplantation, City of Hope, Duarte, CA, USA
| | - Danielle Hammond
- Division of Cancer Medicine, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Abhishek Maiti
- Division of Cancer Medicine, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fadi G Haddad
- Division of Cancer Medicine, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jayatsu Senapati
- Division of Cancer Medicine, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naval Daver
- Division of Cancer Medicine, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naseema Gangat
- Department of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Marina Konopleva
- Department of Oncology, Montefiore Einstein Cancer Center, Bronx, NY, USA
| | | | - Naveen Pemmaraju
- Division of Cancer Medicine, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
33
|
Wu RH, Zhu CY, Yu PH, Ma Y, Hussain L, Naranmandura H, Wang QQ. The landscape of novel strategies for acute myeloid leukemia treatment: Therapeutic trends, challenges, and future directions. Toxicol Appl Pharmacol 2023; 473:116585. [PMID: 37302559 DOI: 10.1016/j.taap.2023.116585] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/13/2023]
Abstract
Acute myeloid leukemia (AML) is a highly heterogeneous subtype of hematological malignancies with a wide spectrum of cytogenetic and molecular abnormalities, which makes it difficult to manage and cure. Along with the deeper understanding of the molecular mechanisms underlying AML pathogenesis, a large cohort of novel targeted therapeutic approaches has emerged, which considerably expands the medical options and changes the therapeutic landscape of AML. Despite that, resistant and refractory cases caused by genomic mutations or bypass signalling activation remain a great challenge. Therefore, discovery of novel treatment targets, optimization of combination strategies, and development of efficient therapeutics are urgently required. This review provides a detailed and comprehensive discussion on the advantages and limitations of targeted therapies as a single agent or in combination with others. Furthermore, the innovative therapeutic approaches including hyperthermia, monoclonal antibody-based therapy, and CAR-T cell therapy are also introduced, which may provide safe and viable options for the treatment of patients with AML.
Collapse
Affiliation(s)
- Ri Han Wu
- College of Life Sciences, Changchun Normal University, Changchun 130032, China
| | - Chen Ying Zhu
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Pei Han Yu
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yafang Ma
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Liaqat Hussain
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Hua Naranmandura
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Qian Qian Wang
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
34
|
Shao R, Li Z, Xin H, Jiang S, Zhu Y, Liu J, Huang R, Xu K, Shi X. Biomarkers as targets for CAR-T/NK cell therapy in AML. Biomark Res 2023; 11:65. [PMID: 37330575 PMCID: PMC10276424 DOI: 10.1186/s40364-023-00501-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 05/11/2023] [Indexed: 06/19/2023] Open
Abstract
The most common kind of acute leukemia in adults is acute myeloid leukemia (AML), which is often treated with induction chemotherapy regimens followed by consolidation or allogeneic hematopoietic stem cell transplantation (HSCT). However, some patients continue to develop relapsed or refractory AML (R/R-AML). Small molecular targeted drugs require long-time administration. Not all the patients hold molecular targets. Novel medicines are therefore needed to enhance treatment outcomes. T cells and natural killer (NK) cells engineered with chimeric antigen receptors (CARs) that target antigens associated with AML have recently been produced and are currently being tested in both pre-clinical and clinical settings. This review provides an overview of CAR-T/NK treatments for AML.
Collapse
Affiliation(s)
- Ruonan Shao
- Second Affiliated Hospital of Nanjing Medical University, No.262, North Zhongshan Road, Nanjing, 210003, Jiangsu, China
| | - Zijian Li
- Second Affiliated Hospital of Nanjing Medical University, No.262, North Zhongshan Road, Nanjing, 210003, Jiangsu, China
| | - Honglei Xin
- Second Affiliated Hospital of Nanjing Medical University, No.262, North Zhongshan Road, Nanjing, 210003, Jiangsu, China
| | - Suyu Jiang
- Second Affiliated Hospital of Nanjing Medical University, No.262, North Zhongshan Road, Nanjing, 210003, Jiangsu, China
| | - Yilin Zhu
- Second Affiliated Hospital of Nanjing Medical University, No.262, North Zhongshan Road, Nanjing, 210003, Jiangsu, China
| | - Jingan Liu
- Second Affiliated Hospital of Nanjing Medical University, No.262, North Zhongshan Road, Nanjing, 210003, Jiangsu, China
| | - Rong Huang
- Second Affiliated Hospital of Nanjing Medical University, No.262, North Zhongshan Road, Nanjing, 210003, Jiangsu, China
| | - Kailin Xu
- Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| | - Xiaofeng Shi
- Second Affiliated Hospital of Nanjing Medical University, No.262, North Zhongshan Road, Nanjing, 210003, Jiangsu, China.
| |
Collapse
|
35
|
Turkalj S, Radtke FA, Vyas P. An Overview of Targeted Therapies in Acute Myeloid Leukemia. Hemasphere 2023; 7:e914. [PMID: 37304938 PMCID: PMC10256410 DOI: 10.1097/hs9.0000000000000914] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 05/01/2023] [Indexed: 06/13/2023] Open
Abstract
Acute myeloid leukemia (AML) is the most aggressive adult leukemia, characterized by clonal differentiation arrest of progenitor or precursor hematopoietic cells. Intense preclinical and clinical research has led to regulatory approval of several targeted therapeutics, administered either as single agents or as combination therapies. However, the majority of patients still face a poor prognosis and disease relapse frequently occurs due to selection of therapy-resistant clones. Hence, more effective novel therapies, most likely as innovative, rational combination therapies, are urgently needed. Chromosomal aberrations, gene mutations, and epigenetic alterations drive AML pathogenesis but concurrently provide vulnerabilities to specifically target leukemic cells. Other molecules, either aberrantly active and/or overexpressed in leukemic stem cells, may also be leveraged for therapeutic benefit. This concise review of targeted therapies for AML treatment, which are either approved or are being actively investigated in clinical trials or recent preclinical studies, provides a flavor of the direction of travel, but also highlights the current challenges in AML treatment.
Collapse
Affiliation(s)
- Sven Turkalj
- MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, United Kingdom
- Oxford Centre for Hematology, NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Felix A. Radtke
- MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, United Kingdom
- Oxford Centre for Hematology, NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
- Department of Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Paresh Vyas
- MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, United Kingdom
- Oxford Centre for Hematology, NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
- Department of Hematology, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| |
Collapse
|
36
|
Köseer AS, Di Gaetano S, Arndt C, Bachmann M, Dubrovska A. Immunotargeting of Cancer Stem Cells. Cancers (Basel) 2023; 15:1608. [PMID: 36900399 PMCID: PMC10001158 DOI: 10.3390/cancers15051608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/24/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
The generally accepted view is that CSCs hijack the signaling pathways attributed to normal stem cells that regulate the self-renewal and differentiation processes. Therefore, the development of selective targeting strategies for CSC, although clinically meaningful, is associated with significant challenges because CSC and normal stem cells share many important signaling mechanisms for their maintenance and survival. Furthermore, the efficacy of this therapy is opposed by tumor heterogeneity and CSC plasticity. While there have been considerable efforts to target CSC populations by the chemical inhibition of the developmental pathways such as Notch, Hedgehog (Hh), and Wnt/β-catenin, noticeably fewer attempts were focused on the stimulation of the immune response by CSC-specific antigens, including cell-surface targets. Cancer immunotherapies are based on triggering the anti-tumor immune response by specific activation and targeted redirecting of immune cells toward tumor cells. This review is focused on CSC-directed immunotherapeutic approaches such as bispecific antibodies and antibody-drug candidates, CSC-targeted cellular immunotherapies, and immune-based vaccines. We discuss the strategies to improve the safety and efficacy of the different immunotherapeutic approaches and describe the current state of their clinical development.
Collapse
Affiliation(s)
- Ayse Sedef Köseer
- National Center for Tumor Diseases (NCT), Partner Site Dresden: German Cancer Research Center (DKFZ), Heidelberg, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01307 Dresden, Germany
- OncoRay–National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01309 Dresden, Germany
| | - Simona Di Gaetano
- OncoRay–National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01309 Dresden, Germany
| | - Claudia Arndt
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany
- Mildred Scheel Early Career Center, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Michael Bachmann
- National Center for Tumor Diseases (NCT), Partner Site Dresden: German Cancer Research Center (DKFZ), Heidelberg, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01307 Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Anna Dubrovska
- National Center for Tumor Diseases (NCT), Partner Site Dresden: German Cancer Research Center (DKFZ), Heidelberg, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01307 Dresden, Germany
- OncoRay–National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01309 Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, 01328 Dresden, Germany
| |
Collapse
|
37
|
Wang Y, Xiao L, Yin L, Zhou L, Deng Y, Deng H. Diagnosis, treatment, and genetic characteristics of blastic plasmacytoid dendritic cell neoplasm: A review. Medicine (Baltimore) 2023; 102:e32904. [PMID: 36800625 PMCID: PMC9936012 DOI: 10.1097/md.0000000000032904] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/19/2023] Open
Abstract
Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a highly aggressive and extremely rare hematologic disease with a poor prognosis, involving mainly the skin and bone marrow. The immunophenotype of these tumor cells is characterized by the expression of CD4, CD56, CD123, TCL-1, and CD303. To date, no consensus has been reached on the standard of care for BPDCN. Currently, clinical treatment is mainly based on high-dose chemotherapy combined with hematopoietic stem cell transplantation. However, this treatment method has limitations for elderly, frail, and relapsed/refractory patients. In recent years, breakthroughs in molecular biology and genetics have not only provided new ideas for the diagnosis of BPDCN but also helped develop targeted treatment strategies for this disease. The emergence of targeted drugs has filled the gap left by traditional therapies and shown great clinical promise. This article focuses on the latest advances in genetics and targeted therapies for BPDCN, especially the emerging therapies that may provide new ideas for the clinical treatment of BPDCN.
Collapse
Affiliation(s)
- Yemin Wang
- Department of Pathology, Fourth Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Li Xiao
- Department of Pathology, Fourth Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Lili Yin
- Department of Pathology, Fourth Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Lv Zhou
- Department of Pathology, Fourth Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yanjuan Deng
- Department of Pathology, Fourth Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Mol. Med. & Genet. Center, Fourth Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Huan Deng
- Department of Pathology, Fourth Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Mol. Med. & Genet. Center, Fourth Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- * Correspondence: Huan Deng, Department of Pathology, Fourth Affiliated Hospital of Nanchang University, 133 South Guangchang Road, Nanchang, Jiangxi 330003, China (e-mail: )
| |
Collapse
|
38
|
Ciotti G, Marconi G, Sperotto A, Giannini MB, Gottardi M, Martinelli G. Biological therapy in elderly patients with acute myeloid leukemia. Expert Opin Biol Ther 2023; 23:175-194. [PMID: 36715330 DOI: 10.1080/14712598.2023.2174015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
INTRODUCTION The introduction of target molecules and immunological therapies is changing the treatment landscape of acute myeloid leukemia (AML). AREAS COVERED We recapitulate the biological therapies that can be employed in the treatment of elderly patients with AML. Alongside small molecules inhibitors that target specific gene mutations, antibodies, tumor microenvironment modulators, and cellular therapies are being developed for the cure of the disease. Here, we report the biological activities, the efficacy and toxicities of humanized antibodies and antibody-drug conjugates that targets surface antigens as CD33 (gemtuzumab ozogamicine) or CD123 (pivekimab sunirine). We further explore mechanisms and effectiveness of medications that modify the microenvironment, such as glasdegib, or that harness the immune system against leukemia, such as CD47 antibody magrolimab, PD1/PDL1 inhibitors pembrolizumab and nivolumab, TIM3 inhibitor sabatolimab, T-cell and NK-cell engagers. Cellular therapies are considered, even if a large trial is still pending for the feasibility of the approach. In this scenario, a brief overview of the mechanism of action of target agents is provided, particularly with respect to their biological mechanisms. EXPERT OPINION Overall, this therapeutic armamentarium will constitute the basis for multimodal and personalized combinations that, in the idea of precision medicine, will enormously benefit elderly AML patients.
Collapse
Affiliation(s)
- Giulia Ciotti
- Onco Hematology, Department of Oncology, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Giovanni Marconi
- IRCCS Istituto Romagnolo Per Lo Studio Dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Alessandra Sperotto
- Onco Hematology, Department of Oncology, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Maria B Giannini
- IRCCS Istituto Romagnolo Per Lo Studio Dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Michele Gottardi
- Onco Hematology, Department of Oncology, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Giovanni Martinelli
- IRCCS Istituto Romagnolo Per Lo Studio Dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| |
Collapse
|
39
|
Pelosi E, Castelli G, Testa U. CD123 a Therapeutic Target for Acute Myeloid Leukemia and Blastic Plasmocytoid Dendritic Neoplasm. Int J Mol Sci 2023; 24:2718. [PMID: 36769040 PMCID: PMC9917129 DOI: 10.3390/ijms24032718] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 02/04/2023] Open
Abstract
In spite of consistent progress at the level of basic research and of clinical treatment, acute myeloid leukemia (AML) still represents an unmet clinical need for adult and pediatric patients. To improve the outcomes of these patients, it is necessary to identify new therapeutic targets. IL3RA (CD123, alpha subunit of the interleukin 3 receptor) is a cell membrane protein overexpressed in several hematologic malignancies, including AML blastic plasmocytoid dendritic cell neoplasms (BPDCN). Given the higher expression of CD123 on leukemic cells compared to normal hematopoietic cells and its low/absent expression on normal hematopoietic stem cells, it appears as a suitable and attractive target for therapy. Various drugs targeting CD123 have been developed and evaluated at clinical level: interleukin-3 conjugated with diphtheria toxin; naked neutralizing anti-CD123 antibodies; drug-antibody conjugates; bispecific antibodies targeting both CD123 and CD3; and chimeric antigen receptor (CAR) T cells engineered to target CD123. Some of these agents have shown promising results at the clinical level, including tagraxofusp (CD123 conjugated with diphtheria toxin) for the treatment of BPDCN and IMGN632 (anti-CD123 drug-conjugate), and flotetuzumab (bispecific anti-CD123 and anti-CD3 monoclonal antibody) for the treatment of AML. However, the therapeutic efficacy of CD123-targeting treatments is still unsatisfactory and must be improved through new therapeutic strategies and combined treatments with other antileukemic drugs.
Collapse
Affiliation(s)
| | | | - Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
40
|
Morse JW, Rios M, Ye J, Rios A, Zhang CC, Daver NG, DiNardo CD, Zhang N, An Z. Antibody therapies for the treatment of acute myeloid leukemia: exploring current and emerging therapeutic targets. Expert Opin Investig Drugs 2023; 32:107-125. [PMID: 36762937 PMCID: PMC10031751 DOI: 10.1080/13543784.2023.2179482] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 02/08/2023] [Indexed: 02/11/2023]
Abstract
INTRODUCTION Acute myeloid leukemia (AML) is the most common and deadly type of leukemia affecting adults. It is typically managed with rounds of non-targeted chemotherapy followed by hematopoietic stem cell transplants, but this is only possible in patients who can tolerate these harsh treatments and many are elderly and frail. With the identification of novel tumor-specific cell surface receptors, there is great conviction that targeted antibody therapies will soon become available for these patients. AREAS COVERED In this review, we describe the current landscape of known target receptors for monospecific and bispecific antibody-based therapeutics for AML. Here, we characterize each of the receptors and targeted antibody-based therapeutics in development, illustrating the rational design behind each therapeutic compound. We then discuss the bispecific antibodies in development and how they improve immune surveillance of AML. For each therapeutic, we also summarize the available pre-clinical and clinical data, including data from discontinued trials. EXPERT OPINION One antibody-based therapeutic has already been approved for AML treatment, the CD33-targeting antibody-drug conjugate, gemtuzumab ozogamicin. Many more are currently in pre-clinical and clinical studies. These antibody-based therapeutics can perform tumor-specific, elaborate cytotoxic functions and there is growing confidence they will soon lead to personalized, safe AML treatment options that induce durable remissions.
Collapse
Affiliation(s)
- Joshua W Morse
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Margarita Rios
- Gorgas Memorial Institute of Health Studies, Panama City, Panama
| | - John Ye
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Adan Rios
- Division of Oncology, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Cheng Cheng Zhang
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Naval G Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Courtney D DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
41
|
Aru B, Pehlivanoğlu C, Dal Z, Dereli-Çalışkan NN, Gürlü E, Yanıkkaya-Demirel G. A potential area of use for immune checkpoint inhibitors: Targeting bone marrow microenvironment in acute myeloid leukemia. Front Immunol 2023; 14:1108200. [PMID: 36742324 PMCID: PMC9895857 DOI: 10.3389/fimmu.2023.1108200] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 01/03/2023] [Indexed: 01/22/2023] Open
Abstract
Acute myeloid leukemia (AML) arises from the cells of myeloid lineage and is the most frequent leukemia type in adulthood accounting for about 80% of all cases. The most common treatment strategy for the treatment of AML includes chemotherapy, in rare cases radiotherapy and stem cell and bone marrow transplantation are considered. Immune checkpoint proteins involve in the negative regulation of immune cells, leading to an escape from immune surveillance, in turn, causing failure of tumor cell elimination. Immune checkpoint inhibitors (ICIs) target the negative regulation of the immune cells and support the immune system in terms of anti-tumor immunity. Bone marrow microenvironment (BMM) bears various blood cell lineages and the interactions between these lineages and the noncellular components of BMM are considered important for AML development and progression. Administration of ICIs for the AML treatment may be a promising option by regulating BMM. In this review, we summarize the current treatment options in AML treatment and discuss the possible application of ICIs in AML treatment from the perspective of the regulation of BMM.
Collapse
Affiliation(s)
- Başak Aru
- Immunology Department, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Cemil Pehlivanoğlu
- Immunology Department, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Zeynep Dal
- School of Medicine, Yeditepe University, Istanbul, Türkiye
| | | | - Ege Gürlü
- School of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Gülderen Yanıkkaya-Demirel
- Immunology Department, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye,*Correspondence: Gülderen Yanıkkaya-Demirel,
| |
Collapse
|
42
|
Gołos A, Góra-Tybor J, Robak T. Experimental drugs in clinical trials for acute myeloid leukemia: innovations, trends, and opportunities. Expert Opin Investig Drugs 2023; 32:53-67. [PMID: 36669827 DOI: 10.1080/13543784.2023.2171860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Acute myeloid leukemia (AML) is a heterogeneous disease characterized by many cytogenetic and molecular alterations. Due to better knowledge of the molecular basis of AML, many targeted therapies have been introduced and registered, e.g. FMS-like tyrosine kinase 3 inhibitors, isocitrate dehydrogenase 1/2 mutation inhibitors, and Bcl-2 inhibitor. Despite that, the cure for AML remains an unmet clinical need in most patients. AREAS COVERED The review aims to present new, not yet registered drugs for AML. We searched the English literature for articles concerning AML, targeted drugs, menin inhibitors, DOT1L, BET, IDH inhibitors, FLT3, hedgehog inhibitors, Polo-like kinase inhibitors, RNA splicing, and immune therapies via PubMed. Publications from January 2000 to August 2022 were scrutinized. Additional relevant publications were obtained by reviewing the references from the chosen articles and Google search. Conference proceedings from the previous 5 years of The American Society of Hematology, the European Hematology Association, and the American Society of Clinical Oncology were searched manually. Additional relevant publications were obtained by reviewing the references. EXPERT OPINION For several years, the therapeutic approach in AML has become more individualized. Novel groups of drugs give hope for greater curability. High response rates have agents that restore the activity of the p53 protein. In addition, agents that work independently of a particular mutation seem promising for AML without any known mutation.
Collapse
Affiliation(s)
- Aleksandra Gołos
- Department of Hematooncology, Copernicus Memorial Hospital, Lodz, Poland
| | - Joanna Góra-Tybor
- Department of Hematooncology, Copernicus Memorial Hospital, Lodz, Poland.,Department of Hematology, Medical University of Lodz, Lodz, Poland
| | - Tadeusz Robak
- Department of Hematology, Medical University of Lodz, Lodz, Poland.,Department of General Hematology, Copernicus Memorial Hospital, Lodz, Poland
| |
Collapse
|
43
|
Abstract
Although complete remission could be achieved in about 60%-70% of acute myeloid leukemia (AML) patients after conventional chemotherapy, relapse and the state of being refractory to treatment remain the main cause of death. In addition, there is a great need for less intensive regimens for all medically frail patients (both due to age/comorbidity and treatment-related). Immune therapy anticipates improved prognosis and reduced toxicities, which may offer novel therapeutic rationales. However, one of the major difficulties in developing immune therapies against AML is that the target antigens are also significantly expressed on healthy hematopoietic stem cells; B-cell malignancies are different because CD20/CD19/healthy B-cells are readily replaceable. Only the anti-CD33 antibody-drug conjugate gemtuzumab-ozogamicin is approved by the FDA for AML. Thus, drug development remains extremely active, although it is still in its infancy. This review summarizes the clinical results of immune therapeutic agents for AML, such as antibody-based drugs, chimeric antigen receptor therapy, checkpoint inhibitors, and vaccines.
Collapse
|
44
|
Antibody-Drug Conjugates in Myeloid Leukemias. Cancer J 2022; 28:454-461. [DOI: 10.1097/ppo.0000000000000635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
45
|
Xu S, Zhang M, Fang X, Hu X, Xing H, Yang Y, Meng J, Wen T, Liu J, Wang J, Wang C, Xu H. CD123 Antagonistic Peptides Assembled with Nanomicelles Act as Monotherapeutics to Combat Refractory Acute Myeloid Leukemia. ACS APPLIED MATERIALS & INTERFACES 2022; 14:38584-38593. [PMID: 35977045 DOI: 10.1021/acsami.2c11538] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Acute myeloid leukemia (AML) is the most common type of acute leukemia in adults. Due to the development of drug resistance to traditional chemotherapies and high relapse rate, AML still has a low survival rate and there is in an urgent need for better treatment strategies. CD123 is widely expressed by AML cells, also associated with the poor prognosis of AML. In this study, we fabricated nanomicelles loaded with a lab-designed CD123 antagonistic peptide, which were referred to as mPO-6. The antagonistic and therapeutic effects were investigated with CD123+ AML cell lines and a refractory AML mouse (AE and CKITD816V) model. Results show that mPO-6 can specifically bind to the CD123+ AML cells and inhibit the cell viability effectively. Intravenous administration of mPO-6 significantly reduces the percentage of AML cells' infiltration and prolongs the median survival of AML mice. Further, the efficiency of mPO-6 is demonstrated to interfere with the axis of CD123/IL-3 via regulating the activation of STAT5, PI3K/AKT, and NF-κB signaling pathways related to cell proliferation or apoptosis at the level of mRNA and protein in vivo and in vitro. In conclusion, the novel CD123 antagonistic peptide micelle formulation mPO-6 can significantly enhance apoptosis and prolong the survival of AML mice by effectively interfering with the axis of CD123/IL-3 and therefore is a promising therapeutic candidate for the treatment of refractory AML.
Collapse
Affiliation(s)
- Shilin Xu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Meichen Zhang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Xiaocui Fang
- CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center of Nanoscience and Technology, Beijing 100190, China
- University of the Chinese Academy of Sciences, Beijing 100190, China
| | - Xuechun Hu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Haiyan Xing
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Yanlian Yang
- CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center of Nanoscience and Technology, Beijing 100190, China
- University of the Chinese Academy of Sciences, Beijing 100190, China
| | - Jie Meng
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Tao Wen
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Jian Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Jianxiang Wang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Chen Wang
- CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center of Nanoscience and Technology, Beijing 100190, China
- University of the Chinese Academy of Sciences, Beijing 100190, China
| | - Haiyan Xu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| |
Collapse
|
46
|
Poussard M, Angelot-Delettre F, Deconinck E. Conventional Therapeutics in BPDCN Patients-Do They Still Have a Place in the Era of Targeted Therapies? Cancers (Basel) 2022; 14:3767. [PMID: 35954431 PMCID: PMC9367503 DOI: 10.3390/cancers14153767] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/27/2022] [Accepted: 08/01/2022] [Indexed: 12/10/2022] Open
Abstract
No benchmark treatment exists for blastic plasmacytoid dendritic cell neoplasm (BPDCN). Since the malignancy is chemo-sensitive, chemotherapy followed by hematopoietic stem cell transplantation remains an effective treatment. However, relapses frequently occur with the development of resistance. New options arising with the development of therapies targeting signaling pathways and epigenetic dysregulation have shown promising results. In this review, we focus on conventional therapies used to treat BPDCN and the novel therapeutic approaches that guide us toward the future management of BPDCN.
Collapse
Affiliation(s)
- Margaux Poussard
- RIGHT Interactions Greffon-Hôte Tumeur/Ingénierie Cellulaire et Génique, UMR1098, EFS BFC, INSERM, University Bourgogne Franche-Comté, F-25000 Besançon, France; (M.P.); (F.A.-D.)
| | - Fanny Angelot-Delettre
- RIGHT Interactions Greffon-Hôte Tumeur/Ingénierie Cellulaire et Génique, UMR1098, EFS BFC, INSERM, University Bourgogne Franche-Comté, F-25000 Besançon, France; (M.P.); (F.A.-D.)
- Etablissement Français du Sang Bourgogne Franche-Comté, Laboratoire d’Immuno-Hématologie, F-25000 Besançon, France
| | - Eric Deconinck
- RIGHT Interactions Greffon-Hôte Tumeur/Ingénierie Cellulaire et Génique, UMR1098, EFS BFC, INSERM, University Bourgogne Franche-Comté, F-25000 Besançon, France; (M.P.); (F.A.-D.)
- Service d’Hématologie, CHRU Besançon, F-25000 Besançon, France
| |
Collapse
|
47
|
Zhang Y, Sokol L. Clinical Insights into the Management of Blastic Plasmacytoid Dendritic Cell Neoplasm. Cancer Manag Res 2022; 14:2107-2117. [PMID: 35789956 PMCID: PMC9250318 DOI: 10.2147/cmar.s330398] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 06/01/2022] [Indexed: 12/02/2022] Open
Abstract
Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is aggressive hematologic malignancy derived from plasmacytoid dendritic cell precursors of myeloid cell lineage. Patients frequently present with bruise-like skin lesions, which typically are followed months later by progressive cytopenias. Historically, BPDCN prognosis has been dismal, with median overall survival ranging from 9 to 13 months. In the past 2 decades, our understanding of BPDCN pathogenesis has led to the successful development of novel therapeutics. In December 2018, the FDA approved tagraxofusp-erzs for adults and pediatric patients older than 2 years who have either treatment-naïve or relapsed/refractory BPDCN. Acute lymphoblastic leukemia (ALL)-based chemotherapy regimens also provide comparable outcomes to tagraxofusp. In our practice, for patients with good performance status, we use tagraxofusp, ALL-based chemotherapy regimens, or clinical trials as frontline induction therapy, followed by consolidation with allogeneic stem cell transplant once the first complete response has been achieved. Our induction regimen also includes intrathecal chemotherapy for central nervous system prophylaxis. Patients with poor performance status who are treatment-naïve or patients with relapsed/refractory disease have limited therapeutic options, and we strongly recommend enrollment in clinical trials; several novel agents and combinations are currently under clinical investigation for both treatment-naïve and relapsed/refractory BPDCN.
Collapse
Affiliation(s)
- Yumeng Zhang
- Department of Internal Medicine, University of South Florida, Tampa, FL, USA.,Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, USA
| | - Lubomir Sokol
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
48
|
Angenendt L, Mikesch JH, Schliemann C. Emerging antibody-based therapies for the treatment of acute myeloid leukemia. Cancer Treat Rev 2022; 108:102409. [DOI: 10.1016/j.ctrv.2022.102409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 11/24/2022]
|
49
|
Thomas X, Elhamri M, Deloire A, Heiblig M. Antibody-based therapy for acute myeloid leukemia: a review of phase 2 and 3 trials. Expert Opin Emerg Drugs 2022; 27:169-185. [PMID: 35749672 DOI: 10.1080/14728214.2022.2094365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Despite recent advances in the treatment of adult acute myeloid leukemia (AML), the clinical outcome of patients continues to be unsatisfactory especially among older patients, those with a high-risk profile, and in the relapsed/refractory setting. For this reason, recent clinical trials have explored novel therapeutic agents either used alone or in combination with intensive chemotherapy or low-intensity treatments. AREAS COVERED The current paper reviews the clinical development of monoclonal antibody-based therapies in AML, their current status and phases 2 and 3 prospective trials. EXPERT OPINION Monoclonal antibody-based therapies demonstrated efficacy and tolerability in several clinical trials, especially when used in combination either with '3+7' chemotherapy or with low-intensity treatments. Additional studies are needed to determine new antigens for antibody-based therapies that target leukemia stem cells and spare normal hematopoiesis. Phase 2 and 3 additional clinical trial data are needed to assess the promise of first trials, especially regarding chimeric antigen receptor T cells redirected against myeloid antigens and immune checkpoint inhibitor therapies.
Collapse
Affiliation(s)
- Xavier Thomas
- Department of Clinical Hematology, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Pierre Bénite, France
| | - Mohamed Elhamri
- Department of Clinical Hematology, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Pierre Bénite, France
| | - Alexandre Deloire
- Department of Clinical Hematology, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Pierre Bénite, France
| | - Maël Heiblig
- Department of Clinical Hematology, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Pierre Bénite, France
| |
Collapse
|
50
|
Adimora IJ, Wilson NR, Pemmaraju N. Blastic plasmacytoid dendritic cell neoplasm (BPDCN): A promising future in the era of targeted therapeutics. Cancer 2022; 128:3019-3026. [PMID: 35726525 DOI: 10.1002/cncr.34345] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/21/2022] [Accepted: 05/20/2022] [Indexed: 11/09/2022]
Abstract
Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a rare hematologic malignancy arising from precursor dendritic cells. BPDCN cells characteristically express several markers on their cell surfaces including CD123, CD4, and CD56. Because of its rarity and challenging clinical presentation, there was no standard of care in managing BPDCN for decades and its prognosis overall was poor. However, as understanding of this rare neoplasm has increased, so have treatment options. The conventional cytotoxic chemotherapy regimens once used in the treatment of BPDCN were modest in their impact on disease relapse until paired with hematopoietic stem cell transplant. Although recent data suggest that there still remains a role for chemotherapeutic agents, targeted modalities have expanded the overall BPDCN treatment landscape. The CD123-targeted agent, tagraxofusp, was the first Food and Drug Administration-approved monotherapy in the treatment of BPDCN. Since its inception, several CD123-targeted and other cell-surface agents have been investigated, with many agents still in the preclinical stages. Although relapsed/refractory disease and central nervous system disease both remain formidable areas of research, there are several promising therapeutic approaches that could have a significant impact on the trajectory of treatment. This review will provide detailed insight on the novel drugs currently in use and those being explored in the management of BPDCN.
Collapse
Affiliation(s)
- Ijele J Adimora
- Department of Internal Medicine, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Nathaniel R Wilson
- Department of Internal Medicine, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Naveen Pemmaraju
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|