1
|
Sun L, Jiang G, Ng YY, Xiao L, Du Z, Wang S, Zhu J. T cells with split CARs specific for NKG2D ligands and PD-L1 exhibit improved selectivity towards monocyte-derived cells while effective in eliminating acute myeloid leukaemia in vivo. J Cancer Res Clin Oncol 2023; 149:10189-10201. [PMID: 37270461 DOI: 10.1007/s00432-023-04865-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 05/16/2023] [Indexed: 06/05/2023]
Abstract
PURPOSE The expression of NKG2D ligands and PD-L1 has been detected on acute myeloid leukaemia (AML) cells, as well as normal cells of the myeloid lineage. To target leukemic cells while minimizing collateral damage to normal cells, we constructed a split dual CAR system based on the AND-gate logic. METHODS The NKG2D extracellular domain linked with DAP12 without a co-stimulatory signal was used for the basal activation of T cells, and used together with the PD-L1-specific chimeric costimulatory receptor containing the 4-1BB activating domain for co-stimulatory signal 2 input. This dual CAR displayed cell-type specificity and activity similar as a 2nd generation NKG2D ligand-specific CAR. RESULTS When compared to CD64 and PD-L1-specific 2nd generation CARs, we observed that the split dual CAR offered an improved myeloid cell type selectivity. For example, PD-L1-specific CAR-T cells lysed all tested myeloid cell types that expressed PD-L1, including M0 macrophages (Mø0), LPS-polarized Mø1, IFN-γ polarized Mø1, IL-4 polarized Mø2, monocytes, immature dendritic cells (imDCs), mature DCs, as well as KG-1 AML cells, while the dual CAR-T cells displaying killing activity only towards LPS polarized Mø1, mature DCs and KG-1 cells that expressed both NKG2D ligands and PD-L1. In a mouse liquid tumor model, the dual CAR-T cells were effective in eradicating established KG-1 AML xenografts. CONCLUSION The improved cell type specificity offered by our split dual CAR-T cell system targeting paired antigens would favour the reduction of the on-target off-tumor toxicity towards normal myeloid cells during the treatment of myeloid leukaemia.
Collapse
Affiliation(s)
- Lu Sun
- Department of Gynaecologic Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, People's Republic of China
| | - Guangyi Jiang
- Department of Gynaecologic Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, People's Republic of China
| | - Yu Yang Ng
- Department of Biological Sciences, National University of Singapore, Singapore, 117543, Singapore
| | - Lin Xiao
- CNK Cell Therapeutics, #501, No 2 Avenue, Hangzhou, 310018, Zhejiang, China
| | - Zhicheng Du
- CNK Cell Therapeutics, #501, No 2 Avenue, Hangzhou, 310018, Zhejiang, China
| | - Shu Wang
- Department of Gynaecologic Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, People's Republic of China.
- Department of Biological Sciences, National University of Singapore, Singapore, 117543, Singapore.
- CNK Cell Therapeutics, #501, No 2 Avenue, Hangzhou, 310018, Zhejiang, China.
| | - Jianqing Zhu
- Department of Gynaecologic Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, People's Republic of China.
| |
Collapse
|
2
|
Gómez-Llobell M, Peleteiro Raíndo A, Climent Medina J, Gómez Centurión I, Mosquera Orgueira A. Immune Checkpoint Inhibitors in Acute Myeloid Leukemia: A Meta-Analysis. Front Oncol 2022; 12:882531. [PMID: 35530329 PMCID: PMC9069679 DOI: 10.3389/fonc.2022.882531] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 03/21/2022] [Indexed: 12/03/2022] Open
Abstract
Background Experience with immune checkpoint inhibitors (ICIs) in the treatment of acute myeloid leukemia (AML) is still limited and based on early clinical trials, with no reported randomized clinical data. In this study, we reviewed the available evidence on the use of ICIs, either in monotherapy or in combination with other treatments, in different AML settings, including newly diagnosed AML, relapsed or refractory (R/R) AML and maintenance treatment after allogeneic-HSCT (allo-HSCT). Materials and Methods A systematic literature review was conducted using PubMed electronic database as primary source to identify the studies involving immune checkpoint inhibitors in first-line and R/R AML. We recorded Overall Response (ORR), Complete Response (CR) and Complete Response with incomplete count recovery (CRi) rates, overall survival (OS) and immune-related adverse events ≥ grade 3 (irAEs). Hereafter, we analyzed the overall profile of these ICIs by performing a meta-analysis of the reported outcomes. Results A total of 13 studies were identified where ICI was used in patients with AML. ORR across these studies was 42% (IC95%, 31% - 54%) and CR/CRi was 33% (IC95%, 22%-45%). Efficacy was also assessed considering the AML setting (first-line vs. relapsed/refractory) and results pointed to higher response rates in first-line, compared to R/R. Mean overall survival was 8.9 months [median 8 months, (IC95%, 3.9 - 15.5)]. Differences between first line and R/R settings were observed, since average overall survival in first line was 12.0 months, duplicating the OS in R/R which was 7.3 months. Additionally, the most specific adverse events (AEs) of these therapies are immune-related adverse events (irAEs), derived from their inflammatory effects. Grade ≥3 irAEs rate was low and similar among studies [12% (95%CI 8% - 16%)]. Conclusion ICIs in combination with intensive chemotherapy, hypomethylating agents or other targeted therapies are gaining interest in the management of hematological malignancies such as AML. However, results obtained from clinical trials are modest and limited by both, the type of design and the clinical trial phase. Hopefully, the prospective study of these therapies in late-stage development could help to identify patients who may benefit from ICI therapy.
Collapse
Affiliation(s)
- Marina Gómez-Llobell
- Hematology Department, Medical University General Hospital Gregorio Marañon, Madrid, Spain
| | - Andrés Peleteiro Raíndo
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Division of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS) University Hospital of Santiago de Compostela (SERGAS), Department of Hematology, Santiago de Compostela, Spain
| | | | | | - Adrián Mosquera Orgueira
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Division of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS) University Hospital of Santiago de Compostela (SERGAS), Department of Hematology, Santiago de Compostela, Spain
| |
Collapse
|
3
|
Gopalakrishnapillai A, Correnti CE, Pilat K, Lin I, Chan MK, Bandaranayake AD, Mehlin C, Kisielewski A, Hamill D, Kaeding AJ, Meshinchi S, Olson JM, Kolb EA, Barwe SP. Immunotherapeutic Targeting of Mesothelin Positive Pediatric AML Using Bispecific T Cell Engaging Antibodies. Cancers (Basel) 2021; 13:5964. [PMID: 34885074 PMCID: PMC8657033 DOI: 10.3390/cancers13235964] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 12/20/2022] Open
Abstract
Advances in the treatment of pediatric AML have been modest over the past four decades. Despite maximally intensive therapy, approximately 40% of patients will relapse. Novel targeted therapies are needed to improve outcomes. We identified mesothelin (MSLN), a well-validated target overexpressed in some adult malignancies, to be highly expressed on the leukemic cell surface in a subset of pediatric AML patients. The lack of expression on normal bone marrow cells makes MSLN a viable target for immunotherapies such as T-cell engaging bispecific antibodies (BsAbs) that combine two distinct antibody-variable regions into a single molecule targeting a cancer-specific antigen and the T-cell co-receptor CD3. Using antibody single-chain variable region (scFv) sequences derived from amatuximab-recognizing MSLN, and from either blinatumomab or AMG330 targeting CD3, we engineered and expressed two MSLN/CD3-targeting BsAbs: MSLNAMA-CD3L2K and MSLNAMA-CD3AMG, respectively. Both BsAbs promoted T-cell activation and reduced leukemic burden in MV4;11:MSLN xenografted mice, but not in those transplanted with MSLN-negative parental MV4;11 cells. MSLNAMA-CD3AMG induced complete remission in NTPL-146 and DF-5 patient-derived xenograft models. These data validate the in vivo efficacy and specificity of MSLN-targeting BsAbs. Because prior MSLN-directed therapies appeared safe in humans, MSLN-targeting BsAbs could be ideal immunotherapies for MSLN-positive pediatric AML patients.
Collapse
Affiliation(s)
- Anilkumar Gopalakrishnapillai
- Nemours Centers for Childhood Cancer Research & Cancer and Blood Disorders, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA; (A.G.); (A.K.); (D.H.); (E.A.K.)
| | - Colin E. Correnti
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (C.E.C.); (K.P.); (I.L.); (M.K.C.); (A.D.B.); (C.M.); (A.J.K.); (S.M.); (J.M.O.)
| | - Kristina Pilat
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (C.E.C.); (K.P.); (I.L.); (M.K.C.); (A.D.B.); (C.M.); (A.J.K.); (S.M.); (J.M.O.)
| | - Ida Lin
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (C.E.C.); (K.P.); (I.L.); (M.K.C.); (A.D.B.); (C.M.); (A.J.K.); (S.M.); (J.M.O.)
| | - Man Kid Chan
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (C.E.C.); (K.P.); (I.L.); (M.K.C.); (A.D.B.); (C.M.); (A.J.K.); (S.M.); (J.M.O.)
| | - Ashok D. Bandaranayake
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (C.E.C.); (K.P.); (I.L.); (M.K.C.); (A.D.B.); (C.M.); (A.J.K.); (S.M.); (J.M.O.)
| | - Christopher Mehlin
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (C.E.C.); (K.P.); (I.L.); (M.K.C.); (A.D.B.); (C.M.); (A.J.K.); (S.M.); (J.M.O.)
| | - Anne Kisielewski
- Nemours Centers for Childhood Cancer Research & Cancer and Blood Disorders, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA; (A.G.); (A.K.); (D.H.); (E.A.K.)
| | - Darcy Hamill
- Nemours Centers for Childhood Cancer Research & Cancer and Blood Disorders, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA; (A.G.); (A.K.); (D.H.); (E.A.K.)
| | - Allison J. Kaeding
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (C.E.C.); (K.P.); (I.L.); (M.K.C.); (A.D.B.); (C.M.); (A.J.K.); (S.M.); (J.M.O.)
| | - Soheil Meshinchi
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (C.E.C.); (K.P.); (I.L.); (M.K.C.); (A.D.B.); (C.M.); (A.J.K.); (S.M.); (J.M.O.)
| | - James M. Olson
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (C.E.C.); (K.P.); (I.L.); (M.K.C.); (A.D.B.); (C.M.); (A.J.K.); (S.M.); (J.M.O.)
| | - Edward Anders Kolb
- Nemours Centers for Childhood Cancer Research & Cancer and Blood Disorders, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA; (A.G.); (A.K.); (D.H.); (E.A.K.)
| | - Sonali P. Barwe
- Nemours Centers for Childhood Cancer Research & Cancer and Blood Disorders, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA; (A.G.); (A.K.); (D.H.); (E.A.K.)
| |
Collapse
|
4
|
Westermann J, Bullinger L. Precision medicine in myeloid malignancies. Semin Cancer Biol 2021; 84:153-169. [PMID: 33895273 DOI: 10.1016/j.semcancer.2021.03.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 03/28/2021] [Accepted: 03/29/2021] [Indexed: 12/13/2022]
Abstract
Myeloid malignancies have always been at the forefront of an improved understanding of the molecular pathogenesis of cancer. In accordance, over the last years, basic research focusing on the aberrations underlying malignant transformation of myeloid cells has provided the basis for precision medicine approaches and subsequently has led to the development of powerful therapeutic strategies. In this review article, we will recapitulate what has happened since in the 1980s the use of all-trans retinoic acid (ATRA), as a first targeted cancer therapy, has changed one of the deadliest leukemia subtypes, acute promyelocytic leukemia (APL), into one that can be cured without classical chemotherapy today. Similarly, imatinib, the first molecularly designed cancer therapy, has revolutionized the management of chronic myeloid leukemia (CML). Thus, targeted treatment approaches have become the paradigm for myeloid malignancy, but many questions still remain unanswered, especially how identical mutations can be associated with different phenotypes. This might be linked to the impact of the cell of origin, gene-gene interactions, or the tumor microenvironment including the immune system. Continuous research in the field of myeloid neoplasia has started to unravel the molecular pathways that are not only crucial for initial treatment response, but also resistance of leukemia cells under therapy. Ongoing studies focusing on leukemia cell vulnerabilities do already point to novel (targetable) "Achilles heels" that can further improve myeloid cancer therapy.
Collapse
Affiliation(s)
- Jörg Westermann
- Department of Hematology, Oncology and Tumor Immunology, Charité University Medicine Berlin, Campus Virchow Clinic, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Lars Bullinger
- Department of Hematology, Oncology and Tumor Immunology, Charité University Medicine Berlin, Campus Virchow Clinic, Augustenburger Platz 1, 13353 Berlin, Germany.
| |
Collapse
|
5
|
Bialek-Waldmann JK, Domning S, Esser R, Glienke W, Mertens M, Aleksandrova K, Arseniev L, Kumar S, Schneider A, Koenig J, Theobald SJ, Tsay HC, Cornelius ADA, Bonifacius A, Eiz-Vesper B, Figueiredo C, Schaudien D, Talbot SR, Bleich A, Spineli LM, von Kaisenberg C, Clark C, Blasczyk R, Heuser M, Ganser A, Köhl U, Farzaneh F, Stripecke R. Induced dendritic cells co-expressing GM-CSF/IFN-α/tWT1 priming T and B cells and automated manufacturing to boost GvL. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 21:621-641. [PMID: 34095345 PMCID: PMC8142053 DOI: 10.1016/j.omtm.2021.04.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 04/03/2021] [Indexed: 12/13/2022]
Abstract
Acute myeloid leukemia (AML) patients with minimal residual disease and receiving allogeneic hematopoietic stem cell transplantation (HCT) have poor survival. Adoptive administration of dendritic cells (DCs) presenting the Wilms tumor protein 1 (WT1) leukemia-associated antigen can potentially stimulate de novo T and B cell development to harness the graft-versus-leukemia (GvL) effect after HCT. We established a simple and fast genetic modification of monocytes for simultaneous lentiviral expression of a truncated WT1 antigen (tWT1), granulocyte macrophage-colony-stimulating factor (GM-CSF), and interferon (IFN)-α, promoting their self-differentiation into potent “induced DCs” (iDCtWT1). A tricistronic integrase-defective lentiviral vector produced under good manufacturing practice (GMP)-like conditions was validated. Transduction of CD14+ monocytes isolated from peripheral blood, cord blood, and leukapheresis material effectively induced their self-differentiation. CD34+ cell-transplanted Nod.Rag.Gamma (NRG)- and Nod.Scid.Gamma (NSG) mice expressing human leukocyte antigen (HLA)-A∗0201 (NSG-A2)-immunodeficient mice were immunized with autologous iDCtWT1. Both humanized mouse models showed improved development and maturation of human T and B cells in the absence of adverse effects. Toward clinical use, manufacturing of iDCtWT1 was up scaled and streamlined using the automated CliniMACS Prodigy system. Proof-of-concept clinical-scale runs were feasible, and the 38-h process enabled standardized production and high recovery of a cryopreserved cell product with the expected identity characteristics. These results advocate for clinical trials testing iDCtWT1 to boost GvL and eradicate leukemia.
Collapse
Affiliation(s)
- Julia K Bialek-Waldmann
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, 30625 Hannover, Germany.,Laboratory of Regenerative Immune Therapies Applied, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Sabine Domning
- Molecular Medicine Group, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, Kings College London, London, UK
| | - Ruth Esser
- Institute of Cellular Therapeutics, Hannover Medical School, 30625 Hannover, Germany
| | - Wolfgang Glienke
- Institute of Cellular Therapeutics, Hannover Medical School, 30625 Hannover, Germany
| | - Mira Mertens
- Institute of Cellular Therapeutics, Hannover Medical School, 30625 Hannover, Germany
| | | | - Lubomir Arseniev
- Institute of Cellular Therapeutics, Hannover Medical School, 30625 Hannover, Germany
| | - Suresh Kumar
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, 30625 Hannover, Germany.,Laboratory of Regenerative Immune Therapies Applied, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Andreas Schneider
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, 30625 Hannover, Germany.,Laboratory of Regenerative Immune Therapies Applied, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Johannes Koenig
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, 30625 Hannover, Germany.,Laboratory of Regenerative Immune Therapies Applied, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany.,German Centre for Infection Research (DZIF), Partner site Hannover, 30625 Hannover, Germany
| | - Sebastian J Theobald
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, 30625 Hannover, Germany.,Laboratory of Regenerative Immune Therapies Applied, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany.,German Centre for Infection Research (DZIF), Partner site Hannover, 30625 Hannover, Germany
| | - Hsin-Chieh Tsay
- Laboratory of Regenerative Immune Therapies Applied, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Angela D A Cornelius
- Laboratory of Regenerative Immune Therapies Applied, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Agnes Bonifacius
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, Germany
| | - Britta Eiz-Vesper
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, Germany
| | - Constanca Figueiredo
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, Germany
| | - Dirk Schaudien
- Fraunhofer Institute for Toxicology and Experimental Medicine, 30625 Hannover, Germany
| | - Steven R Talbot
- Institute for Laboratory Animal Science, Hannover Medical School, 30625 Hannover, Germany
| | - Andre Bleich
- Institute for Laboratory Animal Science, Hannover Medical School, 30625 Hannover, Germany
| | - Loukia M Spineli
- Department of Obstetrics, Gynecology and Reproductive Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Constantin von Kaisenberg
- Department of Obstetrics, Gynecology and Reproductive Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Caren Clark
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany
| | - Rainer Blasczyk
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, Germany
| | - Michael Heuser
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, 30625 Hannover, Germany
| | - Arnold Ganser
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, 30625 Hannover, Germany
| | - Ulrike Köhl
- Institute of Cellular Therapeutics, Hannover Medical School, 30625 Hannover, Germany.,Fraunhofer Institute for Cell Therapy and Immunology IZI and University of Leipzig, 04103 Leipzig, Germany
| | - Farzin Farzaneh
- Molecular Medicine Group, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, Kings College London, London, UK
| | - Renata Stripecke
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, 30625 Hannover, Germany.,Laboratory of Regenerative Immune Therapies Applied, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany.,German Centre for Infection Research (DZIF), Partner site Hannover, 30625 Hannover, Germany
| |
Collapse
|
6
|
Roussel X, Daguindau E, Berceanu A, Desbrosses Y, Warda W, Neto da Rocha M, Trad R, Deconinck E, Deschamps M, Ferrand C. Acute Myeloid Leukemia: From Biology to Clinical Practices Through Development and Pre-Clinical Therapeutics. Front Oncol 2020; 10:599933. [PMID: 33363031 PMCID: PMC7757414 DOI: 10.3389/fonc.2020.599933] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 11/02/2020] [Indexed: 12/19/2022] Open
Abstract
Recent studies have provided several insights into acute myeloid leukemia. Studies based on molecular biology have identified eight functional mutations involved in leukemogenesis, including driver and passenger mutations. Insight into Leukemia stem cells (LSCs) and assessment of cell surface markers have enabled characterization of LSCs from hematopoietic stem and progenitor cells. Clonal evolution has been described as having an effect similar to that of microenvironment alterations. Such biological findings have enabled the development of new targeted drugs, including drug inhibitors and monoclonal antibodies with blockage functions. Some recently approved targeted drugs have resulted in new therapeutic strategies that enhance standard intensive chemotherapy regimens as well as supportive care regimens. Besides the progress made in adoptive immunotherapy, since allogenic hematopoietic stem cell transplantation enabled the development of new T-cell transfer therapies, such as chimeric antigen receptor T-cell and transgenic TCR T-cell engineering, new promising strategies that are investigated.
Collapse
Affiliation(s)
- Xavier Roussel
- Inserm EFS BFC, UMR1098 RIGHT, University Bourgogne Franche-Comté, Besançon, France
- Department of Hematology, University Hospital of Besançon, Besançon, France
| | - Etienne Daguindau
- Inserm EFS BFC, UMR1098 RIGHT, University Bourgogne Franche-Comté, Besançon, France
- Department of Hematology, University Hospital of Besançon, Besançon, France
| | - Ana Berceanu
- Department of Hematology, University Hospital of Besançon, Besançon, France
| | - Yohan Desbrosses
- Department of Hematology, University Hospital of Besançon, Besançon, France
| | - Walid Warda
- Inserm EFS BFC, UMR1098 RIGHT, University Bourgogne Franche-Comté, Besançon, France
| | | | - Rim Trad
- Inserm EFS BFC, UMR1098 RIGHT, University Bourgogne Franche-Comté, Besançon, France
| | - Eric Deconinck
- Inserm EFS BFC, UMR1098 RIGHT, University Bourgogne Franche-Comté, Besançon, France
- Department of Hematology, University Hospital of Besançon, Besançon, France
| | - Marina Deschamps
- Inserm EFS BFC, UMR1098 RIGHT, University Bourgogne Franche-Comté, Besançon, France
| | - Christophe Ferrand
- Inserm EFS BFC, UMR1098 RIGHT, University Bourgogne Franche-Comté, Besançon, France
| |
Collapse
|
7
|
Stanchina M, Soong D, Zheng-Lin B, Watts JM, Taylor J. Advances in Acute Myeloid Leukemia: Recently Approved Therapies and Drugs in Development. Cancers (Basel) 2020; 12:E3225. [PMID: 33139625 PMCID: PMC7692236 DOI: 10.3390/cancers12113225] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 10/26/2020] [Accepted: 10/28/2020] [Indexed: 12/26/2022] Open
Abstract
Acute myeloid leukemia (AML) is a genetically heterogeneous malignancy comprised of various cytogenetic and molecular abnormalities that has notoriously been difficult to treat with an overall poor prognosis. For decades, treatment options were limited to either intensive chemotherapy with anthracycline and cytarabine-based regimens (7 + 3) or lower intensity regimens including hypomethylating agents or low dose cytarabine, followed by either allogeneic stem cell transplant or consolidation chemotherapy. Fortunately, with the influx of rapidly evolving molecular technologies and new genetic understanding, the treatment landscape for AML has dramatically changed. Advances in the formulation and delivery of 7 + 3 with liposomal cytarabine and daunorubicin (Vyxeos) have improved overall survival in secondary AML. Increased understanding of the genetic underpinnings of AML has led to targeting actionable mutations such as FLT3, IDH1/2 and TP53, and BCL2 or hedgehog pathways in more frail populations. Antibody drug conjugates have resurfaced in the AML landscape and there have been numerous advances utilizing immunotherapies including immune checkpoint inhibitors, antibody-drug conjugates, bispecific T cell engager antibodies, chimeric antigen receptor (CAR)-T therapy and the development of AML vaccines. While there are dozens of ongoing studies and new drugs in the pipeline, this paper serves as a review of the advances achieved in the treatment of AML in the last several years and the most promising future avenues of advancement.
Collapse
Affiliation(s)
- Michele Stanchina
- Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (M.S.); (D.S.)
| | - Deborah Soong
- Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (M.S.); (D.S.)
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Binbin Zheng-Lin
- Department of Medicine, Icahn School of Medicine Mount Sinai West-Morningside, New York, NY 10025, USA;
| | - Justin M. Watts
- Division of Hematology, Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Justin Taylor
- Division of Hematology, Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| |
Collapse
|
8
|
Reikvam H, Hemsing AL, Smith C. Therapy for acute myelogenous leukemia revisited: moving away from a one-size-fits-all approach. Expert Rev Anticancer Ther 2020; 21:5-8. [PMID: 33059507 DOI: 10.1080/14737140.2021.1838901] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Håkon Reikvam
- Department of Clinical Science, University of Bergen , Bergen, Norway.,Department of Medicine, Haukeland University Hospital , Bergen, Norway
| | - Anette Lodvir Hemsing
- Department of Clinical Science, University of Bergen , Bergen, Norway.,Department of Medicine, Haukeland University Hospital , Bergen, Norway
| | - Catherine Smith
- Department of Medicine, Division of Hematology/Oncology, UCSF , San Francisco, CA, USA
| |
Collapse
|