1
|
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
|
2
|
Bianchi M, Reichen C, Croset A, Fischer S, Eggenschwiler A, Grübler Y, Marpakwar R, Looser T, Spitzli P, Herzog C, Villemagne D, Schiegg D, Abduli L, Iss C, Neculcea A, Franchini M, Lekishvili T, Ragusa S, Zitt C, Kaufmann Y, Auge A, Hänggi M, Ali W, Frasconi TM, Wullschleger S, Schlegel I, Matzner M, Lüthi U, Schlereth B, Dawson KM, Kirkin V, Ochsenbein AF, Grimm S, Reschke N, Riether C, Steiner D, Leupin N, Goubier A. The CD33xCD123xCD70 Multispecific CD3-Engaging DARPin MP0533 Induces Selective T Cell-Mediated Killing of AML Leukemic Stem Cells. Cancer Immunol Res 2024; 12:921-943. [PMID: 38683145 PMCID: PMC11217734 DOI: 10.1158/2326-6066.cir-23-0692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 01/04/2024] [Accepted: 04/19/2024] [Indexed: 05/01/2024]
Abstract
The prognosis of patients with acute myeloid leukemia (AML) is limited, especially for elderly or unfit patients not eligible for hematopoietic stem cell (HSC) transplantation. The disease is driven by leukemic stem cells (LSCs), which are characterized by clonal heterogeneity and resistance to conventional therapy. These cells are therefore believed to be a major cause of progression and relapse. We designed MP0533, a multispecific CD3-engaging designed ankyrin repeat protein (DARPin) that can simultaneously bind to three antigens on AML cells (CD33, CD123, and CD70), aiming to enable avidity-driven T cell-mediated killing of AML cells coexpressing at least two of the antigens. In vitro, MP0533 induced selective T cell-mediated killing of AML cell lines, as well as patient-derived AML blasts and LSCs, expressing two or more target antigens, while sparing healthy HSCs, blood, and endothelial cells. The higher selectivity also resulted in markedly lower levels of cytokine release in normal human blood compared to single antigen-targeting T-cell engagers. In xenograft AML mice models, MP0533 induced tumor-localized T-cell activation and cytokine release, leading to complete eradication of the tumors while having no systemic adverse effects. These studies show that the multispecific-targeting strategy used with MP0533 holds promise for improved selectivity toward LSCs and efficacy against clonal heterogeneity, potentially bringing a new therapeutic option to this group of patients with a high unmet need. MP0533 is currently being evaluated in a dose-escalation phase 1 study in patients with relapsed or refractory AML (NCT05673057).
Collapse
Affiliation(s)
| | | | - Amelie Croset
- Molecular Partners AG, Zurich-Schlieren, Switzerland.
| | | | | | | | | | - Thamar Looser
- Molecular Partners AG, Zurich-Schlieren, Switzerland.
| | | | | | | | | | | | - Chloé Iss
- Molecular Partners AG, Zurich-Schlieren, Switzerland.
| | | | | | | | - Simone Ragusa
- Molecular Partners AG, Zurich-Schlieren, Switzerland.
| | - Christof Zitt
- Molecular Partners AG, Zurich-Schlieren, Switzerland.
| | | | - Alienor Auge
- Molecular Partners AG, Zurich-Schlieren, Switzerland.
| | - Martin Hänggi
- Molecular Partners AG, Zurich-Schlieren, Switzerland.
| | - Waleed Ali
- Molecular Partners AG, Zurich-Schlieren, Switzerland.
| | | | | | - Iris Schlegel
- Molecular Partners AG, Zurich-Schlieren, Switzerland.
| | | | - Ursina Lüthi
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| | | | | | | | - Adrian F. Ochsenbein
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| | | | - Nina Reschke
- Molecular Partners AG, Zurich-Schlieren, Switzerland.
| | - Carsten Riether
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| | | | | | - Anne Goubier
- Molecular Partners AG, Zurich-Schlieren, Switzerland.
| |
Collapse
|
3
|
Jen WY, Konopleva M, Pemmaraju N. Tagraxofusp, a first-in-class CD123-targeted agent: Five-year postapproval comprehensive review of the literature. Cancer 2024; 130:2260-2271. [PMID: 38620053 DOI: 10.1002/cncr.35315] [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: 02/22/2024] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 04/17/2024]
Abstract
Tagraxofusp is a first-in-class CD123-directed conjugate of an amended diphtheria toxin platform and recombinant interleukin 3. Binding and subsequent internalization of the drug result in cell death via disruption of intracellular protein synthesis. CD123 is a surface marker that is expressed in several hematological malignancies, especially blastic plasmacytoid dendritic cell neoplasm (BPDCN), where its expression is ubiquitous. A pivotal study of tagraxofusp in BPDCN resulted in its approval for the treatment of BPDCN, the first treatment approved for this indication. Since the introduction of tagraxofusp, research has focused on the management of adverse effects, combination therapy to improve outcomes in fit patients, and dosing and combination strategies to mitigate toxicities while preserving efficacy, especially among older patients. The successful targeting of CD123 in BPDCN has also encouraged research into a variety of other CD123-positive hematological neoplasms, including acute myeloid leukemia (AML), and informed the development of other novel agents targeting CD123. This review examines the clinical data leading to the development and approval of tagraxofusp in BPDCN, how it is being used in combination to improve outcomes in BPDCN and AML, and its developing role in other hematological malignancies.
Collapse
Affiliation(s)
- Wei-Ying Jen
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Marina Konopleva
- Department of Oncology, Montefiore Einstein Comprehensive Cancer Center, Bronx, New York, USA
| | - Naveen Pemmaraju
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
4
|
Thompson EJ, Escarbe S, Tvorogov D, Farshid G, Gregory PA, Khew-Goodall Y, Madden S, Ingman WV, Lindeman GJ, Lim E, Lopez AF, Bonder CS. Interleukin-3 production by basal-like breast cancer cells is associated with poor prognosis. Growth Factors 2024; 42:49-61. [PMID: 38299881 DOI: 10.1080/08977194.2023.2297693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 12/07/2023] [Indexed: 02/02/2024]
Abstract
Breast cancer represents a collection of pathologies with different molecular subtypes, histopathology, risk factors, clinical behavior, and responses to treatment. "Basal-like" breast cancers predominantly lack the receptors for estrogen and progesterone (ER/PR), lack amplification of human epidermal growth factor receptor 2 (HER2) but account for 10-15% of all breast cancers, are largely insensitive to targeted treatment and represent a disproportionate number of metastatic cases and deaths. Analysis of interleukin (IL)-3 and the IL-3 receptor subunits (IL-3RA + CSF2RB) reveals elevated expression in predominantly the basal-like group. Further analysis suggests that IL-3 itself, but not the IL-3 receptor subunits, associates with poor patient outcome. Histology on patient-derived xenografts supports the notion that breast cancer cells are a significant source of IL-3 that may promote disease progression. Taken together, these observations suggest that IL-3 may be a useful marker in solid tumors, particularly triple negative breast cancer, and warrants further investigation into its contribution to disease pathogenesis.
Collapse
Affiliation(s)
- Emma J Thompson
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
| | - Samantha Escarbe
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
| | - Denis Tvorogov
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
| | - Gelareh Farshid
- BreastScreen SA and SA Pathology, Adelaide, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Philip A Gregory
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Yeesim Khew-Goodall
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, Australia
- School of Biological Sciences, University of Adelaide, Australia
| | | | - Wendy V Ingman
- Adelaide Medical School, University of Adelaide, Adelaide, Australia
- The Robinson Research Institute, University of Adelaide, Adelaide, Australia
| | - Geoffrey J Lindeman
- Cancer Biology and Stem Cells Division, Walter, Eliza Hall Institute of Medical Research, Melbourne, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, Australia
| | - Elgene Lim
- Garvan Institute of Medical Research and St. Vincent"s Clinical School, University of New South Wales, Darlinghurst,Australia
| | - Angel F Lopez
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Claudine S Bonder
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, Australia
| |
Collapse
|
5
|
Hachem CE, Marschall P, Hener P, Karnam A, Bonam SR, Meyer P, Flatter E, Birling MC, Bayry J, Li M. IL-3 produced by T cells is crucial for basophil extravasation in hapten-induced allergic contact dermatitis. Front Immunol 2023; 14:1151468. [PMID: 37180157 PMCID: PMC10169741 DOI: 10.3389/fimmu.2023.1151468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/06/2023] [Indexed: 05/15/2023] Open
Abstract
Basophils have been recognized as a characterized cellular player for Th2 immune responses implicated in allergic diseases, but the mechanisms responsible for basophil recruitment to allergic skin remain not well understood. Using a hapten fluorescein isothiocyanate (FITC)-induced allergic contact dermatitis (ACD) mouse model, we show that basophils in FITC-treated IL-3-knockout mice are defective in crossing the vascular endothelium to enter the inflamed skin. By generating mice in which IL-3 is selectively ablated in T cells, we further demonstrate that IL-3 produced by T cells mediates basophil extravasation. Moreover, basophils sorted from FITC-treated IL-3-knockout mice exhibit a decreased expression of integrins Itgam, Itgb2, Itga2b and Itgb7, which are potentially implicated in extravasation process. Interestingly, we observed that these basophils had a reduced expression of retinaldehyde dehydrogenase 1 family member A2 (Aldh1a2), an enzyme responsible for the production of retinoic acid (RA), and administration of all-trans RA restored partially the extravasation of basophils in IL-3-knockout mice. Finally, we validate that IL-3 induces the expression of ALDH1A2 in primary human basophils, and provide further evidence that IL-3 stimulation induces the expression of integrins particularly ITGB7 in an RA-dependent manner. Together, our data propose a model that IL-3 produced by T cells activates ALDH1A2 expression by basophils, leading to the production of RA, which subsequently induces the expression of integrins crucially implicated in basophil extravasation to inflamed ACD skin.
Collapse
Affiliation(s)
- Carole El Hachem
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique UMR7104, Institut National de la Santé et de la Recherche Médicale U1258, Université de Strasbourg, Illkirch, France
| | - Pierre Marschall
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique UMR7104, Institut National de la Santé et de la Recherche Médicale U1258, Université de Strasbourg, Illkirch, France
| | - Pierre Hener
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique UMR7104, Institut National de la Santé et de la Recherche Médicale U1258, Université de Strasbourg, Illkirch, France
| | - Anupama Karnam
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
| | - Srinivasa Reddy Bonam
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
| | - Pierre Meyer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique UMR7104, Institut National de la Santé et de la Recherche Médicale U1258, Université de Strasbourg, Illkirch, France
| | - Eric Flatter
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique UMR7104, Institut National de la Santé et de la Recherche Médicale U1258, Université de Strasbourg, Illkirch, France
| | | | - Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
- Department of Biological Sciences & Engineering, Indian Institute of Technology Palakkad, Palakkad, India
| | - Mei Li
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique UMR7104, Institut National de la Santé et de la Recherche Médicale U1258, Université de Strasbourg, Illkirch, France
| |
Collapse
|
6
|
Woll PS, Yoshizato T, Hellström‐Lindberg E, Fioretos T, Ebert BL, Jacobsen SEW. Targeting stem cells in myelodysplastic syndromes and acute myeloid leukemia. J Intern Med 2022; 292:262-277. [PMID: 35822488 PMCID: PMC9544124 DOI: 10.1111/joim.13535] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The genetic architecture of cancer has been delineated through advances in high-throughput next-generation sequencing, where the sequential acquisition of recurrent driver mutations initially targeted towards normal cells ultimately leads to malignant transformation. Myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) are hematologic malignancies frequently initiated by mutations in the normal hematopoietic stem cell compartment leading to the establishment of leukemic stem cells. Although the genetic characterization of MDS and AML has led to identification of new therapeutic targets and development of new promising therapeutic strategies, disease progression, relapse, and treatment-related mortality remain a major challenge in MDS and AML. The selective persistence of rare leukemic stem cells following therapy-induced remission implies unique resistance mechanisms of leukemic stem cells towards conventional therapeutic strategies and that leukemic stem cells represent the cellular origin of relapse. Therefore, targeted surveillance of leukemic stem cells following therapy should, in the future, allow better prediction of relapse and disease progression, but is currently challenged by our restricted ability to distinguish leukemic stem cells from other leukemic cells and residual normal cells. To advance current and new clinical strategies for the treatment of MDS and AML, there is a need to improve our understanding and characterization of MDS and AML stem cells at the cellular, molecular, and genetic levels. Such work has already led to the identification of promising new candidate leukemic stem cell molecular targets that can now be exploited in preclinical and clinical therapeutic strategies, towards more efficient and specific elimination of leukemic stem cells.
Collapse
Affiliation(s)
- Petter S. Woll
- Department of Medicine HuddingeCenter for Hematology and Regenerative MedicineKarolinska InstitutetStockholmSweden
| | - Tetsuichi Yoshizato
- Department of Medicine HuddingeCenter for Hematology and Regenerative MedicineKarolinska InstitutetStockholmSweden
| | - Eva Hellström‐Lindberg
- Department of Medicine HuddingeCenter for Hematology and Regenerative MedicineKarolinska InstitutetStockholmSweden
- Department of HematologyKarolinska University HospitalStockholmSweden
| | - Thoas Fioretos
- Division of Clinical GeneticsDepartment of Laboratory MedicineLund UniversityLundSweden
- Division of Laboratory MedicineDepartment of Clinical Genetics and PathologyLundSweden
| | - Benjamin L. Ebert
- Department of Medical OncologyDana–Farber Cancer InstituteBostonMassachusettsUSA
- Broad Institute of Harvard and MITCambridgeMassachusettsUSA
- Howard Hughes Medical InstituteBostonMassachusettsUSA
| | - Sten Eirik W. Jacobsen
- Department of Medicine HuddingeCenter for Hematology and Regenerative MedicineKarolinska InstitutetStockholmSweden
- Department of HematologyKarolinska University HospitalStockholmSweden
- Department of Cell and Molecular BiologyKarolinska InstitutetStockholmSweden
- MRC Molecular Haematology UnitMRC Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordUK
| |
Collapse
|
7
|
Meyer JE, Loff S, Dietrich J, Spehr J, Jurado Jiménez G, von Bonin M, Ehninger G, Cartellieri M, Ehninger A. Evaluation of switch-mediated costimulation in trans on universal CAR-T cells (UniCAR) targeting CD123-positive AML. Oncoimmunology 2021; 10:1945804. [PMID: 34290907 PMCID: PMC8274446 DOI: 10.1080/2162402x.2021.1945804] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Chimeric antigen receptor T cells (CAR-T) targeting CD19 have achieved significant success in patients with B cell malignancies. To date, implementation of CAR-T in other indications remains challenging due to the lack of truly tumor-specific antigens as well as control of CAR-T activity in patients. CD123 is highly expressed in acute myeloid leukemia (AML) blasts including leukemia-initiating cells making it an attractive immunotherapeutic target. However, CD123 expression in normal hematopoietic progenitor cells and endothelia bears the risk of severe toxicities and may limit CAR-T applications lacking fine-tuned control mechanisms. Therefore, we recently developed a rapidly switchable universal CAR-T platform (UniCAR), in which CAR-T activity depends on the presence of a soluble adapter called targeting module (TM), and confirmed clinical proof-of-concept for targeting CD123 in AML with improved safety. As costimulation via 4–1BB ligand (4–1BBL) can enhance CAR-T expansion, persistence, and effector functions, a novel CD123-specific TM variant (TM123-4-1BBL) comprising trimeric single-chain 4–1BBL was developed for transient costimulation of UniCAR-T cells (UniCAR-T) at the leukemic site in trans. TM123-4-1BBL-directed UniCAR-T efficiently eradicated CD123-positive AML cells in vitro and in a CDX in vivo model. Moreover, additional costimulation via TM123-4-1BBL enabled enhanced expansion and persistence with a modulated UniCAR-T phenotype. In addition, the increased hydrodynamic volume of TM123-4-1BBL prolonged terminal plasma half-life and ensured a high total drug exposure in vivo. In conclusion, expanding the soluble adapter optionality for CD123-directed UniCAR-T maintains the platforms high anti-leukemic efficacy and immediate control mechanism for a flexible, safe, and individualized CAR-T therapy of AML patients.
Collapse
Affiliation(s)
| | | | | | | | | | - Malte von Bonin
- Division of Hematology, Oncology and Stem Cell Transplantation, Medical Clinic I, Department of Medicine I, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | | | | | | |
Collapse
|
8
|
Yoshikawa S, Miyake K, Kamiya A, Karasuyama H. The role of basophils in acquired protective immunity to tick infestation. Parasite Immunol 2021; 43:e12804. [PMID: 33124059 PMCID: PMC8244031 DOI: 10.1111/pim.12804] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 10/26/2020] [Indexed: 01/22/2023]
Abstract
Ticks are blood-feeding ectoparasites that transmit a variety of pathogens to host animals and humans, causing severe infectious diseases such as Lyme disease. In a certain combination of animal and tick species, tick infestation elicits acquired immunity against ticks in the host, which can reduce the ability of ticks to feed on blood and to transmit pathogens in the following tick infestations. Therefore, our understanding of the cellular and molecular mechanisms of acquired tick resistance (ATR) can advance the development of anti-tick vaccines to prevent tick infestation and tick-borne diseases. Basophils are a minor population of white blood cells circulating in the bloodstream and are rarely observed in peripheral tissues under steady-state conditions. Basophils have been reported to accumulate at tick-feeding sites during re-infestation in cattle, rabbits, guinea pigs and mice. Selective ablation of basophils resulted in a loss of ATR in guinea pigs and mice, illuminating the essential role of basophils in the manifestation of ATR. In this review, we discuss the recent advance in the elucidation of the cellular and molecular mechanisms underlying basophil recruitment to the tick-feeding site and basophil-mediated ATR.
Collapse
Affiliation(s)
- Soichiro Yoshikawa
- Department of Cellular PhysiologyOkayama University Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayamaJapan
| | - Kensuke Miyake
- Inflammation, Infection and Immunity LaboratoryTMDU Advanced Research InstituteTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Atsunori Kamiya
- Department of Cellular PhysiologyOkayama University Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayamaJapan
| | - Hajime Karasuyama
- Inflammation, Infection and Immunity LaboratoryTMDU Advanced Research InstituteTokyo Medical and Dental University (TMDU)TokyoJapan
| |
Collapse
|
9
|
Fraser DD, Patterson EK, Daley M, Cepinskas G. Case Report: Inflammation and Endothelial Injury Profiling of COVID-19 Pediatric Multisystem Inflammatory Syndrome (MIS-C). Front Pediatr 2021; 9:597926. [PMID: 33898353 PMCID: PMC8060468 DOI: 10.3389/fped.2021.597926] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 03/09/2021] [Indexed: 12/26/2022] Open
Abstract
Introduction: COVID-19 is associated with a novel multi-system inflammatory syndrome that shares some characteristics with Kawasaki's Disease. The syndrome manifestation is delayed relative to COVID-19 onset, with a spectrum of clinical severity. Clinical signs may include persistent fever, gastrointestinal symptoms, cardiac inflammation and/or shock. Case Presentation: We measured 59 inflammatory and endothelial injury plasma analytes in an adolescent girl that presented with malaise, fever, cough, strawberry tongue and jaundice. Her COVID-19 status was positive with detection of 2 SARS-CoV-2 viral genes using polymerase chain reaction. She was treated with intravenous immunoglobulin prior to blood draw, but our plasma measurements suggested a unique analyte expression pattern associated with inflammation, endothelial injury and microvascular glycocalyx degradation. Conclusions: COVID-19 is associated with a multi-system inflammatory syndrome and a unique inflammatory and endothelial injury signature. Summary: Analyte markers of inflammation and endothelial cell injury might serve as putative biomarkers and/or be investigated further as potential therapeutic targets.
Collapse
Affiliation(s)
- Douglas D. Fraser
- Lawson Health Research Institute, London, ON, Canada
- Pediatrics, Western University, London, ON, Canada
| | | | - Mark Daley
- Lawson Health Research Institute, London, ON, Canada
- Computer Science, Western University, London, ON, Canada
| | - Gediminas Cepinskas
- Lawson Health Research Institute, London, ON, Canada
- Medical Biophysics, Western University, London, ON, Canada
| |
Collapse
|
10
|
Perriello VM, Gionfriddo I, Rossi R, Milano F, Mezzasoma F, Marra A, Spinelli O, Rambaldi A, Annibali O, Avvisati G, Di Raimondo F, Ascani S, Falini B, Martelli MP, Brunetti L. CD123 Is Consistently Expressed on NPM1-Mutated AML Cells. Cancers (Basel) 2021; 13:cancers13030496. [PMID: 33525388 PMCID: PMC7865228 DOI: 10.3390/cancers13030496] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary One-third of adult acute myeloid leukemia (AML) harbors NPM1 mutations. A deep knowledge of the distribution of selected antigens on the surface of NPM1-mutated AML cells may help optimizing new therapies for this frequent AML subtype. CD123 is known to be expressed on leukemic cells but also on healthy hematopoietic and endothelial cells, although at lower levels. Differences in antigen densities between AML and healthy cells may enlighten therapeutic windows, where targeting CD123 could be effective without triggering “on-target off-tumor” toxicities. Here, we perform a thorough analysis of CD123 expression demonstrating high expression of this antigen on both NPM1-mutated bulk leukemic cells and CD34+CD38− cells. Abstract NPM1-mutated (NPM1mut) acute myeloid leukemia (AML) comprises about 30% of newly diagnosed AML in adults. Despite notable advances in the treatment of this frequent AML subtype, about 50% of NPM1mut AML patients treated with conventional treatment die due to disease progression. CD123 has been identified as potential target for immunotherapy in AML, and several anti-CD123 therapeutic approaches have been developed for AML resistant to conventional therapies. As this antigen has been previously reported to be expressed by NPM1mut cells, we performed a deep flow cytometry analysis of CD123 expression in a large cohort of NPM1mut and wild-type samples, examining the whole blastic population, as well as CD34+CD38− leukemic cells. We demonstrate that CD123 is highly expressed on NPM1mut cells, with particularly high expression levels showed by CD34+CD38− leukemic cells. Additionally, CD123 expression was further enhanced by FLT3 mutations, which frequently co-occur with NPM1 mutations. Our results identify NPM1-mutated and particularly NPM1/FLT3 double-mutated AML as disease subsets that may benefit from anti-CD123 targeted therapies.
Collapse
Affiliation(s)
- Vincenzo Maria Perriello
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
| | - Ilaria Gionfriddo
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
| | - Roberta Rossi
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
| | - Francesca Milano
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
| | - Federica Mezzasoma
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
| | - Andrea Marra
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
| | - Orietta Spinelli
- Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, 24127 Bergamo, Italy; (O.S.); (A.R.)
| | - Alessandro Rambaldi
- Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, 24127 Bergamo, Italy; (O.S.); (A.R.)
- Department of Oncology and Hematology, University of Milan, 20122 Milan, Italy
| | - Ombretta Annibali
- Hematology and Stem Cell Transplant Unit, Campus Biomedico University Hospital, 00128 Rome, Italy; (O.A.); (G.A.)
| | - Giuseppe Avvisati
- Hematology and Stem Cell Transplant Unit, Campus Biomedico University Hospital, 00128 Rome, Italy; (O.A.); (G.A.)
| | - Francesco Di Raimondo
- Hematology and Bone Marrow Transplant Unit, Catania University Hospital, 95125 Catania, Italy;
| | - Stefano Ascani
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
- Hematology and Bone Marrow Transplant Unit, Santa Maria della Misericordia Hospital, 06131 Perugia, Italy
- Pathology, Santa Maria Hospital, 05100 Terni, Italy
| | - Brunangelo Falini
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
- Hematology and Bone Marrow Transplant Unit, Santa Maria della Misericordia Hospital, 06131 Perugia, Italy
| | - Maria Paola Martelli
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
- Hematology and Bone Marrow Transplant Unit, Santa Maria della Misericordia Hospital, 06131 Perugia, Italy
- Correspondence: (M.P.M.); (L.B.)
| | - Lorenzo Brunetti
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
- Hematology and Bone Marrow Transplant Unit, Santa Maria della Misericordia Hospital, 06131 Perugia, Italy
- Correspondence: (M.P.M.); (L.B.)
| |
Collapse
|
11
|
Karasuyama H, Miyake K, Yoshikawa S. Immunobiology of Acquired Resistance to Ticks. Front Immunol 2020; 11:601504. [PMID: 33154758 PMCID: PMC7591762 DOI: 10.3389/fimmu.2020.601504] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 09/30/2020] [Indexed: 11/13/2022] Open
Abstract
Ticks are blood-sucking arthropods of great importance in the medical and veterinary fields worldwide. They are considered second only to mosquitos as vectors of pathogenic microorganisms that can cause serious infectious disorders, such as Lyme borreliosis and tick-borne encephalitis. Hard (Ixodid) ticks feed on host animals for several days and inject saliva together with pathogens to hosts during blood feeding. Some animal species can acquire resistance to blood-feeding by ticks after a single or repeated tick infestation, resulting in decreased weights and numbers of engorged ticks or the death of ticks in subsequent infestations. Importantly, this acquired tick resistance (ATR) can reduce the risk of pathogen transmission from pathogen-infected ticks to hosts. This is the basis for the development of tick antigen-targeted vaccines to forestall tick infestation and tick-borne diseases. Accumulation of basophils is detected in the tick re-infested skin lesion of animals showing ATR, and the ablation of basophils abolishes ATR in mice and guinea pigs, illustrating the critical role for basophils in the expression of ATR. In this review article, we provide a comprehensive overview of recent advances in our understanding of the cellular and molecular mechanisms responsible for the development and manifestation of ATR, with a particular focus on the role of basophils.
Collapse
Affiliation(s)
- Hajime Karasuyama
- Inflammation, Infection and Immunity Laboratory, TMDU Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kensuke Miyake
- Inflammation, Infection and Immunity Laboratory, TMDU Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Soichiro Yoshikawa
- Department of Cellular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
12
|
Sugita M, Guzman ML. CD123 as a Therapeutic Target Against Malignant Stem Cells. Hematol Oncol Clin North Am 2020; 34:553-564. [DOI: 10.1016/j.hoc.2020.01.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
13
|
Loff S, Dietrich J, Meyer JE, Riewaldt J, Spehr J, von Bonin M, Gründer C, Swayampakula M, Franke K, Feldmann A, Bachmann M, Ehninger G, Ehninger A, Cartellieri M. Rapidly Switchable Universal CAR-T Cells for Treatment of CD123-Positive Leukemia. MOLECULAR THERAPY-ONCOLYTICS 2020; 17:408-420. [PMID: 32462078 PMCID: PMC7240059 DOI: 10.1016/j.omto.2020.04.009] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 04/23/2020] [Indexed: 12/11/2022]
Abstract
Chimeric antigen receptor T cells (CAR-T) targeting CD19 or B cell maturation antigen (BCMA) are highly effective against B cell malignancies. However, application of CAR-T to less differentially expressed targets remains a challenge due to lack of tumor-specific antigens and CAR-T controllability. CD123, a highly promising leukemia target, is expressed not only by leukemic and leukemia-initiating cells, but also by myeloid, hematopoietic progenitor, and certain endothelial cells. Thus, CAR-T lacking fine-tuned control mechanisms pose a high toxicity risk. To extend the CAR-T target landscape and widen the therapeutic window, we adapted our rapidly switchable universal CAR-T platform (UniCAR) to target CD123. UniCAR-T efficiently eradicated CD123+ leukemia in vitro and in vivo. Activation, cytolytic response, and cytokine release were strictly dependent on the presence of the CD123-specific targeting module (TM123) with comparable efficacy to CD123-specific CAR-T in vitro. We further demonstrated a pre-clinical proof of concept for the safety-switch mechanism using a hematotoxicity mouse model wherein TM123-redirected UniCAR-T showed reversible toxicity toward hematopoietic cells compared to CD123 CAR-T. In conclusion, UniCAR-T maintain full anti-leukemic efficacy, while ensuring rapid controllability to improve safety and versatility of CD123-directed immunotherapy. The safety and efficacy of UniCAR-T in combination with TM123 will now be assessed in a phase I clinical trial (ClinicalTrials.gov: NCT04230265).
Collapse
Affiliation(s)
- Simon Loff
- GEMoaB Monoclonals GmbH, 01307 Dresden, Germany
| | | | | | | | | | - Malte von Bonin
- Medical Clinic and Policlinic I, University Hospital "Carl Gustav Carus," TU Dresden, 01307 Dresden, Germany.,German Cancer Consortium "Carl Gustav Carus," TU Dresden, 01307 Dresden, Germany
| | | | | | | | - Anja Feldmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany
| | - Michael Bachmann
- University Cancer Center "Carl Gustav Carus," TU Dresden, Tumor Immunology, 01307 Dresden, Germany.,Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany.,German Cancer Consortium "Carl Gustav Carus," TU Dresden, 01307 Dresden, Germany.,National Center for Tumor Diseases, "Carl Gustav Carus," TU Dresden, 01307 Dresden, Germany
| | - Gerhard Ehninger
- GEMoaB Monoclonals GmbH, 01307 Dresden, Germany.,Cellex Patient Treatment GmbH, 01307 Dresden, Germany
| | - Armin Ehninger
- GEMoaB Monoclonals GmbH, 01307 Dresden, Germany.,Cellex Patient Treatment GmbH, 01307 Dresden, Germany
| | - Marc Cartellieri
- GEMoaB Monoclonals GmbH, 01307 Dresden, Germany.,Cellex Patient Treatment GmbH, 01307 Dresden, Germany
| |
Collapse
|
14
|
Kumar A, Rani L, Mhaske ST, Pote ST, Behera S, Mishra GC, Wani MR. IL-3 Receptor Expression on Activated Human Th Cells Is Regulated by IL-4, and IL-3 Synergizes with IL-4 to Enhance Th2 Cell Differentiation. THE JOURNAL OF IMMUNOLOGY 2020; 204:819-831. [DOI: 10.4049/jimmunol.1801629] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 11/21/2019] [Indexed: 12/15/2022]
|
15
|
Yao S, Jianlin C, Yarong L, Botao L, Qinghan W, Hongliang F, Lu Z, Hongmei N, Pin W, Hu C, Liangding H, Bin Z. Donor-Derived CD123-Targeted CAR T Cell Serves as a RIC Regimen for Haploidentical Transplantation in a Patient With FUS-ERG+ AML. Front Oncol 2019; 9:1358. [PMID: 31850234 PMCID: PMC6901822 DOI: 10.3389/fonc.2019.01358] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 11/18/2019] [Indexed: 01/16/2023] Open
Abstract
Background: Allogeneic hematopoietic stem cell transplantation (allo-HSCT) following chemotherapy is part of standard treatment protocol for patients with acute myeloid leukemia (AML). FUS-ERG+ AML is rare but has an extremely poor prognosis even with allo-HSCT in remission, possibly due to its a leukemia stem cell (LSC)-driven disease resulting in chemotherapy resistance and a novel therapy is urgently required. It has been reported that FUS-ERG-positive AML expresses CD123, a marker of LSC, in some cases. CD123-targeted CAR T cell (CART123) is promising immunotherapy, but how to improve the complete remission (CR) rate and rescue potential hematopoietic toxicity still need to explore. Case Presentation: We used donor-derived CART123 as part of conditioning regimen for haploidentical HSCT (haplo-HSCT) in a patient with FUS-ERG+ AML who relapsed after allogeneic transplantation within 3 months, resists to multi-agent chemotherapy and donor lymphocyte infusion (DLI) and remained non-remission, aiming to reduce these chemotherapy-resistant blasts and rescue potential hematopoietic toxicity. The blasts in BM were reduced within 2 weeks and coincided with CAR copies expansion after CART123 infusion. The patient achieved full donor chimerism, CR with incomplete blood count recovery, and myeloid implantation. Conclusion: Our results hints that CART123 reduces the chemotherapy-resistant AML blasts for FUS-ERG+ AML without affecting the full donor chimerism and myeloid implantation.
Collapse
Affiliation(s)
- Sun Yao
- Department of Hematopoietic Stem Cell Transplantation, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of PLA), The Research Institute of Hematopoietic Stem Cell of the People's Liberation Army, Beijing, China
| | - Chen Jianlin
- Department of Hematopoietic Stem Cell Transplantation, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of PLA), The Research Institute of Hematopoietic Stem Cell of the People's Liberation Army, Beijing, China
| | - Liu Yarong
- R&D Department, HRAIN Biotechnology Co., Ltd., Shanghai, China
| | - Li Botao
- Department of Hematopoietic Stem Cell Transplantation, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of PLA), The Research Institute of Hematopoietic Stem Cell of the People's Liberation Army, Beijing, China
| | - Wang Qinghan
- Department of Hematopoietic Stem Cell Transplantation, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of PLA), The Research Institute of Hematopoietic Stem Cell of the People's Liberation Army, Beijing, China
| | - Fang Hongliang
- R&D Department, HRAIN Biotechnology Co., Ltd., Shanghai, China
| | - Zhang Lu
- R&D Department, HRAIN Biotechnology Co., Ltd., Shanghai, China
| | - Ning Hongmei
- Department of Hematopoietic Stem Cell Transplantation, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of PLA), The Research Institute of Hematopoietic Stem Cell of the People's Liberation Army, Beijing, China
| | - Wang Pin
- R&D Department, HRAIN Biotechnology Co., Ltd., Shanghai, China.,Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, United States.,Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, United States.,Department of Pharmaceutical Sciences and Pharmacology, University of Southern California, Los Angeles, CA, United States
| | - Chen Hu
- Department of Hematopoietic Stem Cell Transplantation, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of PLA), The Research Institute of Hematopoietic Stem Cell of the People's Liberation Army, Beijing, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Therapy and Transformation Research, Department of Hematopoietic Stem Cell Transplantation, The Cell and Gene Therapy Center, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of PLA), The Research Institute of Hematopoietic Stem Cell of the People's Liberation Army, Beijing, China
| | - Hu Liangding
- Department of Hematopoietic Stem Cell Transplantation, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of PLA), The Research Institute of Hematopoietic Stem Cell of the People's Liberation Army, Beijing, China
| | - Zhang Bin
- Beijing Key Laboratory of Hematopoietic Stem Cell Therapy and Transformation Research, Department of Hematopoietic Stem Cell Transplantation, The Cell and Gene Therapy Center, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of PLA), The Research Institute of Hematopoietic Stem Cell of the People's Liberation Army, Beijing, China
| |
Collapse
|
16
|
Sun Y, Wang S, Zhao L, Zhang B, Chen H. IFN-γ and TNF-α aggravate endothelial damage caused by CD123-targeted CAR T cell. Onco Targets Ther 2019; 12:4907-4925. [PMID: 31417286 PMCID: PMC6600319 DOI: 10.2147/ott.s205678] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 05/13/2019] [Indexed: 01/08/2023] Open
Abstract
Background: CD123-targeted chimeric antigen receptor (CAR) T cell (CART123) for the treatment of acute myeloid leukemia (AML) and blastic plasmacytoid dendritic cell neoplasm has exhibited potential in clinical trials. However, capillary leakage syndrome, which is associated with endothelial cells damage, is under intensive focus in CART123 therapy. Purpose: The present study aimed to explore the change in CD123 in endothelial cells and the injury to endothelial cells caused by CART123. Methods: The expression of CD123 and cytotoxicity were assessed by flow cytometry. Cytokine release was assessed by ELISA. An in vitro co-culture model was designed to mimic the status, wherein CART123 was stimulated and cytokines were released. Results: In the current study, CART123 exhibited cytotoxicity and the effects of cytokine production on endothelium, and the upregulation of CD123 enhanced the cytotoxicity. The addition of interferon (IFN)-γ and tumor necrosis factor (TNF)-α neutralizing antibodies can effectively reverse the upregulation of CD123 on the endothelial cells caused by CART123, while the cytotoxicity of CART123 in AML cell lines was not affected in vitro. Second, we proved that CD123 expresses in CART123 and would be upregulated after activation, putatively causing an overactivated and fratricide effect. Conclusion: In summary, this study identified that the expression of CD123 on endothelial cells could be upregulated when co-cultured with CART123. Furthermore, IFN-γ and TNF-α could aggravate endothelial damage caused by CART123 in vitro.
Collapse
Affiliation(s)
- Yao Sun
- Academy of Military Medical Sciences , Beijing 100071, People's Republic of China.,Department of Hematopoietic Stem Cell Transplantation, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of PLA), the Research Institute of Hematopoietic Stem Cell of the People's Liberation Army, Beijing 100071, People's Republic of China.,Beijing Key Laboratory of Hematopoietic Stem Cell Therapy and Transformation Research , Beijing 100071, People's Republic of China
| | - Shenyu Wang
- Academy of Military Medical Sciences , Beijing 100071, People's Republic of China.,Department of Hematopoietic Stem Cell Transplantation, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of PLA), the Research Institute of Hematopoietic Stem Cell of the People's Liberation Army, Beijing 100071, People's Republic of China.,Beijing Key Laboratory of Hematopoietic Stem Cell Therapy and Transformation Research , Beijing 100071, People's Republic of China
| | - Long Zhao
- Department of Hematopoietic Stem Cell Transplantation, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of PLA), the Research Institute of Hematopoietic Stem Cell of the People's Liberation Army, Beijing 100071, People's Republic of China.,Beijing Key Laboratory of Hematopoietic Stem Cell Therapy and Transformation Research , Beijing 100071, People's Republic of China
| | - Bin Zhang
- Department of Hematopoietic Stem Cell Transplantation, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of PLA), the Research Institute of Hematopoietic Stem Cell of the People's Liberation Army, Beijing 100071, People's Republic of China.,Beijing Key Laboratory of Hematopoietic Stem Cell Therapy and Transformation Research , Beijing 100071, People's Republic of China
| | - Hu Chen
- Department of Hematopoietic Stem Cell Transplantation, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of PLA), the Research Institute of Hematopoietic Stem Cell of the People's Liberation Army, Beijing 100071, People's Republic of China.,Beijing Key Laboratory of Hematopoietic Stem Cell Therapy and Transformation Research , Beijing 100071, People's Republic of China
| |
Collapse
|
17
|
Zaidi N, Quezada SA, Kuroiwa JM, Zhang L, Jaffee EM, Steinman RM, Wang B. Anti-CTLA-4 synergizes with dendritic cell-targeted vaccine to promote IL-3-dependent CD4 + effector T cell infiltration into murine pancreatic tumors. Ann N Y Acad Sci 2019; 1445:62-73. [PMID: 30945313 PMCID: PMC6557673 DOI: 10.1111/nyas.14049] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 02/09/2019] [Accepted: 02/15/2019] [Indexed: 12/22/2022]
Abstract
One successful class of cancer immunotherapies, immune checkpoint inhibitory antibodies, disrupts key pathways that regulate immune checkpoints, such as cytotoxic T lymphocyte-associated antigen-4 (CTLA-4). These agents unleash the potency of antigen-experienced T cells that have already been induced as a consequence of the existing tumor. But only 20% of cancers naturally induce T cells. For most cancers, vaccines are require to induce and mobilize T effector cells (Teffs ) to traffick into tumors. We evaluated the effects of anti-CTLA-4 given in combination with an antigen-specific dendritic cell vaccine on intratumoral Teffs in a murine pancreatic cancer model. The dendritic cell-targeted tumor antigen plus anti-CTLA-4 significantly increased the number of vaccine-induced CD4+ Teffs within the tumor. This increase was accompanied by a reduction in the size of the peripheral CD4+ Teff pool. We also found that IL-3 production by activated CD4+ T cells was significantly increased with this combination. Importantly, the CD4+ Teff response was attenuated in Il3-/- mice, suggesting mediation of the effect by IL-3. Finally, the induced T cell infiltration was associated with activation of the tumor endothelium by T cell-derived IL-3. Our findings collectively provide a new insight into the mechanism driving Teff infiltration and vascular activation in a murine pancreatic cancer model, specifically identifying a new role for IL-3 in the anticancer immune response.
Collapse
Affiliation(s)
- Neeha Zaidi
- Laboratory of Cellular Physiology and Immunology and Chris Browne Center of Immunology and Immune Disease, The Rockefeller University, New York
- The Sidney Kimmel Comprehensive Cancer Center, The Skip Viragh Center for Pancreatic Cancer, The Bloomberg–Kimmel Institute for Cancer Immunotherapy, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sergio A. Quezada
- University College London Cancer Institute, Research Department of Haematology, London, United Kingdom
| | - Janelle M.Y. Kuroiwa
- Laboratory of Cellular Physiology and Immunology and Chris Browne Center of Immunology and Immune Disease, The Rockefeller University, New York
| | - Li Zhang
- Laboratory of Cellular Physiology and Immunology and Chris Browne Center of Immunology and Immune Disease, The Rockefeller University, New York
| | - Elizabeth M. Jaffee
- The Sidney Kimmel Comprehensive Cancer Center, The Skip Viragh Center for Pancreatic Cancer, The Bloomberg–Kimmel Institute for Cancer Immunotherapy, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ralph M. Steinman
- Laboratory of Cellular Physiology and Immunology and Chris Browne Center of Immunology and Immune Disease, The Rockefeller University, New York
| | - Bei Wang
- Laboratory of Cellular Physiology and Immunology and Chris Browne Center of Immunology and Immune Disease, The Rockefeller University, New York
| |
Collapse
|
18
|
Krawczyk E, Zolov SN, Huang K, Bonifant CL. T-cell Activity against AML Improved by Dual-Targeted T Cells Stimulated through T-cell and IL7 Receptors. Cancer Immunol Res 2019; 7:683-692. [PMID: 30782669 PMCID: PMC8186236 DOI: 10.1158/2326-6066.cir-18-0748] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/22/2018] [Accepted: 02/14/2019] [Indexed: 12/13/2022]
Abstract
The development of engineered T cells to treat acute myeloid leukemia (AML) is challenging due to difficulty in target selection and the need for robust T-cell expansion and persistence. We designed a T cell stimulated to kill AML cells based on recognition of the AML-associated surface marker CLEC12A, via secretion of a CLEC12AxCD3 bispecific "engager" molecule (CLEC12A-ENG). CLEC12A-ENG T cells are specifically activated by CLEC12A, are not toxic to hematopoietic progenitor cells, and exhibit antigen-dependent AML killing. Next, we coupled stimulation of T-cell survival to triggering of a chimeric IL7 receptor with an ectodomain that binds a second AML-associated surface antigen, CD123. The resulting T cells, identified as CLEC12A-ENG.CD123IL7Rα T cells, demonstrate improved activation upon dual target recognition, kill AML, and exhibit antitumor activity in xenograft models. Enhanced T-cell activation conferred by CD123.IL7Rα was dependent both on recognition of the CD123 target and on IL7Rα-mediated downstream signaling. Expression of a chimeric IL7R targeted to a second tumor-associated antigen (TAA) should improve T-cell activity not only against hematologic malignancies, but perhaps against all cancers.
Collapse
Affiliation(s)
- Eric Krawczyk
- Department of Pediatrics and Communicable Diseases, Division of Pediatric Hematology and Oncology, University of Michigan, Ann Arbor, Michigan
| | - Sergey N Zolov
- Department of Pediatrics and Communicable Diseases, Division of Pediatric Hematology and Oncology, University of Michigan, Ann Arbor, Michigan
| | - Kevin Huang
- Department of Pediatrics and Communicable Diseases, Division of Pediatric Hematology and Oncology, University of Michigan, Ann Arbor, Michigan
| | - Challice L Bonifant
- Department of Pediatrics and Communicable Diseases, Division of Pediatric Hematology and Oncology, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
19
|
Anzai A, Mindur JE, Halle L, Sano S, Choi JL, He S, McAlpine CS, Chan CT, Kahles F, Valet C, Fenn AM, Nairz M, Rattik S, Iwamoto Y, Fairweather D, Walsh K, Libby P, Nahrendorf M, Swirski FK. Self-reactive CD4 + IL-3 + T cells amplify autoimmune inflammation in myocarditis by inciting monocyte chemotaxis. J Exp Med 2019; 216:369-383. [PMID: 30670465 PMCID: PMC6363430 DOI: 10.1084/jem.20180722] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 12/07/2018] [Accepted: 01/07/2019] [Indexed: 12/29/2022] Open
Abstract
Acquisition of self-reactive effector CD4+ T cells is a major component of the autoimmune response that can occur during myocarditis, an inflammatory form of cardiomyopathy. Although the processes by which self-reactive T cells gain effector function have received considerable attention, how these T cells contribute to effector organ inflammation and damage is less clear. Here, we identified an IL-3-dependent amplification loop that exacerbates autoimmune inflammation. In experimental myocarditis, we show that effector organ-accumulating autoreactive IL-3+ CD4+ T cells stimulate IL-3R+ tissue macrophages to produce monocyte-attracting chemokines. The newly recruited monocytes differentiate into antigen-presenting cells that stimulate local IL-3+ CD4+ T cell proliferation, thereby amplifying organ inflammation. Consequently, Il3 -/- mice resist developing robust autoimmune inflammation and myocardial dysfunction, whereas therapeutic IL-3 targeting ameliorates disease. This study defines a mechanism that orchestrates inflammation in myocarditis, describes a previously unknown function for IL-3, and identifies IL-3 as a potential therapeutic target in patients with myocarditis.
Collapse
Affiliation(s)
- Atsushi Anzai
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA.,Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - John E Mindur
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Lennard Halle
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Soichi Sano
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA
| | - Jennifer L Choi
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Shun He
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Cameron S McAlpine
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Christopher T Chan
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Florian Kahles
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Colin Valet
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Ashley M Fenn
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Manfred Nairz
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Sara Rattik
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Yoshiko Iwamoto
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | | | - Kenneth Walsh
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA
| | - Peter Libby
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA.,Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Filip K Swirski
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA .,Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
20
|
Karasuyama H, Tabakawa Y, Ohta T, Wada T, Yoshikawa S. Crucial Role for Basophils in Acquired Protective Immunity to Tick Infestation. Front Physiol 2018; 9:1769. [PMID: 30581391 PMCID: PMC6293010 DOI: 10.3389/fphys.2018.01769] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 11/23/2018] [Indexed: 11/21/2022] Open
Abstract
Ticks are blood-sucking arthropods that can transmit various pathogenic organisms to host animals and humans, causing serious infectious diseases including Lyme disease. Tick feeding induces innate and acquired immune responses in host animals, depending on the combination of different species of animals and ticks. Acquired tick resistance (ATR) can diminish the chance of pathogen transmission from infected ticks to the host. Hence, the elucidation of cellular and molecular mechanism underlying ATR is important for the development of efficient anti-tick vaccines. In this review article, we briefly overview the history of studies on ATR and summarize recent findings, particularly focusing on the role for basophils in the manifestation of ATR. In several animal species, including cattle, guinea pigs, rabbits and mice, basophil accumulation is observed at the tick re-infestation site, even though the frequency of basophils among cellular infiltrates varies in different animal species, ranging from approximately 3% in mice to 70% in guinea pigs. Skin-resident, memory CD4+ T cells contribute to the recruitment of basophils to the tick re-infestation site through production of IL-3 in mice. Depletion of basophils before the tick re-infestation abolishes ATR in guinea pigs infested with Amblyomma americanum and mice infested with Haemaphysalis longicornis, demonstrating the crucial role of basophils in the manifestation of ATR. The activation of basophils via IgE and its receptor FcεRI is essential for ATR in mice. Histamine released from activated basophils functions as an important effector molecule in murine ATR, probably through promotion of epidermal hyperplasia which interferes with tick attachment or blood feeding in the skin. Accumulating evidence suggests the following scenario. The 1st tick infestation triggers the production of IgE against tick saliva antigens in the host, and blood-circulating basophils bind such IgE on the cell surface via FcεRI. In the 2nd infestation, IgE-armed basophils are recruited to tick-feeding sites and stimulated by tick saliva antigens to release histamine that promotes epidermal hyperplasia, contributing to ATR. Further studies are needed to clarify whether this scenario in mice can be applied to ATR in other animal species and humans.
Collapse
Affiliation(s)
- Hajime Karasuyama
- Department of Immune Regulation, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yuya Tabakawa
- Department of Immune Regulation, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takuya Ohta
- Department of Immune Regulation, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takeshi Wada
- Department of Immune Regulation, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Division of Molecular Medicine, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| | - Soichiro Yoshikawa
- Department of Immune Regulation, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
21
|
Kooijman S, Brummelman J, van Els CACM, Marino F, Heck AJR, van Riet E, Metz B, Kersten GFA, Pennings JLA, Meiring HD. Vaccine antigens modulate the innate response of monocytes to Al(OH)3. PLoS One 2018; 13:e0197885. [PMID: 29813132 PMCID: PMC5973561 DOI: 10.1371/journal.pone.0197885] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 04/10/2018] [Indexed: 12/12/2022] Open
Abstract
Aluminum-based adjuvants have widely been used in human vaccines since 1926. In the absence of antigens, aluminum-based adjuvants can initiate the inflammatory preparedness of innate cells, yet the impact of antigens on this response has not been investigated so far. In this study, we address the modulating effect of vaccine antigens on the monocyte-derived innate response by comparing processes initiated by Al(OH)3 and by Infanrix, an Al(OH)3-adjuvanted trivalent combination vaccine (DTaP), containing diphtheria toxoid (D), tetanus toxoid (T) and acellular pertussis (aP) vaccine antigens. A systems-wide analysis of stimulated monocytes was performed in which full proteome analysis was combined with targeted transcriptome analysis and cytokine analysis. This comprehensive study revealed four major differences in the monocyte response, between plain Al(OH)3 and DTaP stimulation conditions: (I) DTaP increased the anti-inflammatory cytokine IL-10, whereas Al(OH)3 did not; (II) Al(OH)3 increased the gene expression of IFNγ, IL-2 and IL-17a in contrast to the limited induction or even downregulation by DTaP; (III) increased expression of type I interferons-induced proteins was not observed upon DTaP stimulation, but was observed upon Al(OH)3 stimulation; (IV) opposing regulation of protein localization pathways was observed for Al(OH)3 and DTaP stimulation, related to the induction of exocytosis by Al(OH)3 alone. This study highlights that vaccine antigens can antagonize Al(OH)3-induced programming of the innate immune responses at the monocyte level.
Collapse
Affiliation(s)
- Sietske Kooijman
- Intravacc, Bilthoven, The Netherlands
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Science Faculty, Utrecht University, Utrecht, The Netherlands
| | - Jolanda Brummelman
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Cécile A. C. M. van Els
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Fabio Marino
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Science Faculty, Utrecht University, Utrecht, The Netherlands
- Netherlands Proteomics Centre, Utrecht, The Netherlands
| | - Albert J. R. Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Science Faculty, Utrecht University, Utrecht, The Netherlands
- Netherlands Proteomics Centre, Utrecht, The Netherlands
| | | | | | - Gideon F. A. Kersten
- Intravacc, Bilthoven, The Netherlands
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Jeroen L. A. Pennings
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | | |
Collapse
|
22
|
Ohta T, Yoshikawa S, Tabakawa Y, Yamaji K, Ishiwata K, Shitara H, Taya C, Oh-Hora M, Kawano Y, Miyake K, Yamanishi Y, Yonekawa H, Watanabe N, Kanuka H, Karasuyama H. Skin CD4 + Memory T Cells Play an Essential Role in Acquired Anti-Tick Immunity through Interleukin-3-Mediated Basophil Recruitment to Tick-Feeding Sites. Front Immunol 2017; 8:1348. [PMID: 29085376 PMCID: PMC5650685 DOI: 10.3389/fimmu.2017.01348] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/03/2017] [Indexed: 12/14/2022] Open
Abstract
Ticks, blood-sucking arthropods, serve as vectors for transmission of infectious diseases including Lyme borreliosis. After tick infestation, several animal species can develop resistance to subsequent infestations, reducing the risk of transmission. In a mouse model, basophils reportedly infiltrate tick-feeding sites during the second but not first infestation and play a crucial role in the expression of acquired tick resistance. However, the mechanism underlying basophil recruitment to the second tick-feeding site remains ill-defined. Here, we investigated cells and their products responsible for the basophil recruitment. Little or no basophil infiltration was detected in T-cell-deficient mice, and adoptive transfer of CD4+ but not CD8+ T cells reconstituted it. Il3 gene expression was highly upregulated at the second tick-feeding site, and adoptive transfer of interleukin-3 (IL-3)-sufficient but not IL-3-deficient CD4+ T cells conferred the basophil infiltration on T-cell-deficient mice, indicating that the CD4+ T-cell-derived IL-3 is essential for the basophil recruitment. Notably, IL-3+ resident CD4+ memory T cells were detected even before the second infestation in previously uninfested skin distant from the first tick-feeding site. Taken together, IL-3 produced locally by skin CD4+ memory T cells appears to play a crucial role in basophil recruitment to the second tick-feeding site.
Collapse
Affiliation(s)
- Takuya Ohta
- Department of Immune Regulation, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Soichiro Yoshikawa
- Department of Immune Regulation, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yuya Tabakawa
- Department of Immune Regulation, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kayoko Yamaji
- Department of Tropical Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Kenji Ishiwata
- Department of Tropical Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Hiroshi Shitara
- Laboratory for Transgenic Technology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Choji Taya
- Laboratory for Transgenic Technology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Masatsugu Oh-Hora
- Division of Molecular Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Yohei Kawano
- Department of Immune Regulation, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kensuke Miyake
- Department of Immune Regulation, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshinori Yamanishi
- Department of Immune Regulation, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiromichi Yonekawa
- Laboratory for Transgenic Technology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Bio-Oriented Technology Research Advancement Institution (BRAIN), Saitama, Japan
| | - Naohiro Watanabe
- Department of Tropical Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Hirotaka Kanuka
- Department of Tropical Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Hajime Karasuyama
- Department of Immune Regulation, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
23
|
Han L, Jorgensen JL, Brooks C, Shi C, Zhang Q, Nogueras González GM, Cavazos A, Pan R, Mu H, Wang SA, Zhou J, Ai-Atrash G, Ciurea SO, Rettig M, DiPersio JF, Cortes J, Huang X, Kantarjian HM, Andreeff M, Ravandi F, Konopleva M. Antileukemia Efficacy and Mechanisms of Action of SL-101, a Novel Anti-CD123 Antibody Conjugate, in Acute Myeloid Leukemia. Clin Cancer Res 2017; 23:3385-3395. [PMID: 28096272 DOI: 10.1158/1078-0432.ccr-16-1904] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 12/13/2016] [Accepted: 12/29/2016] [Indexed: 12/11/2022]
Abstract
Purpose: The persistence of leukemia stem cells (LSC)-containing cells after induction therapy may contribute to minimal residual disease (MRD) and relapse in acute myeloid leukemia (AML). We investigated the clinical relevance of CD34+CD123+ LSC-containing cells and antileukemia potency of a novel antibody conjugate SL-101 in targeting CD123+ LSCs.Experimental Methods and Results: In a retrospective study on 86 newly diagnosed AML patients, we demonstrated that a higher proportion of CD34+CD123+ LSC-containing cells in remission was associated with persistent MRD and predicted shorter relapse-free survival in patients with poor-risk cytogenetics. Using flow cytometry, we explored the potential benefit of therapeutic targeting of CD34+CD38-CD123+ cells by SL-101, a novel antibody conjugate comprising an anti-CD123 single-chain Fv fused to Pseudomonas exotoxin A The antileukemia potency of SL-101 was determined by the expression levels of CD123 antigen in a panel of AML cell lines. Colony-forming assay established that SL-101 strongly and selectively suppressed the function of leukemic progenitors while sparing normal counterparts. The internalization, protein synthesis inhibition, and flow cytometry assays revealed the mechanisms underlying the cytotoxic activities of SL-101 involved rapid and efficient internalization of antibody, sustained inhibition of protein synthesis, induction of apoptosis, and blockade of IL3-induced p-STAT5 and p-AKT signaling pathways. In a patient-derived xenograft model using NSG mice, the repopulating capacity of LSCs pretreated with SL-101 in vitro was significantly impaired.Conclusions: Our data define the mechanisms by which SL-101 targets AML and warrant further investigation of the clinical application of SL-101 and other CD123-targeting strategies in AML. Clin Cancer Res; 23(13); 3385-95. ©2017 AACR.
Collapse
Affiliation(s)
- Lina Han
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jeffrey L Jorgensen
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Ce Shi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qi Zhang
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Antonio Cavazos
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rongqing Pan
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hong Mu
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sa A Wang
- Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jin Zhou
- Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Gheath Ai-Atrash
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stefan O Ciurea
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mike Rettig
- Bone Marrow Transplantation and Leukemia Program, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - John F DiPersio
- Bone Marrow Transplantation and Leukemia Program, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Jorge Cortes
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xuelin Huang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hagop M Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael Andreeff
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
24
|
Rotiroti MC, Arcangeli S, Casucci M, Perriello V, Bondanza A, Biondi A, Tettamanti S, Biagi E. Acute Myeloid Leukemia Targeting by Chimeric Antigen Receptor T Cells: Bridging the Gap from Preclinical Modeling to Human Studies. Hum Gene Ther 2016; 28:231-241. [PMID: 27967241 DOI: 10.1089/hum.2016.092] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Acute myeloid leukemia (AML) still represents an unmet clinical need for adult and pediatric high-risk patients, thus demanding advanced and personalized therapies. In this regard, different targeted immunotherapeutic approaches are available, ranging from naked monoclonal antibodies (mAb) to conjugated and multifunctional mAbs (i.e., BiTEs and DARTs). Recently, researchers have focused their attention on novel techniques of genetic manipulation specifically to redirect cytotoxic T cells endowed with chimeric antigen receptors (CARs) toward selected tumor associated antigens. So far, CAR T cells targeting the CD19 antigen expressed by B-cell origin hematological cancers have gained impressive clinical results, leading to the possibility of translating the CAR platform to treat other hematological malignancies such as AML. However, one of the main concerns in the field of AML CAR immunotherapy is the identification of an ideal target cell surface antigen, being highly expressed on tumor cells but minimally present on healthy tissues, together with the design of an anti-AML CAR appropriately balancing efficacy and safety profiles. The current review focuses mainly on AML target antigens and the related immunotherapeutic approaches developed so far, deeply dissecting methods of CAR T cell safety improvements, when designing novel CARs approaching human studies.
Collapse
Affiliation(s)
- Maria Caterina Rotiroti
- 1 Molecular Therapy Unit, Tettamanti Research Center, Pediatric Clinic, University of Milano Bicocca , San Gerardo Hospital/MBBM Foundation, Monza, Italy
| | - Silvia Arcangeli
- 1 Molecular Therapy Unit, Tettamanti Research Center, Pediatric Clinic, University of Milano Bicocca , San Gerardo Hospital/MBBM Foundation, Monza, Italy
| | - Monica Casucci
- 2 Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Hospital Scientific Institute, Vita-Salute San Raffaele University , Milan, Italy
| | - Vincenzo Perriello
- 1 Molecular Therapy Unit, Tettamanti Research Center, Pediatric Clinic, University of Milano Bicocca , San Gerardo Hospital/MBBM Foundation, Monza, Italy
| | - Attilio Bondanza
- 2 Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Hospital Scientific Institute, Vita-Salute San Raffaele University , Milan, Italy
| | - Andrea Biondi
- 1 Molecular Therapy Unit, Tettamanti Research Center, Pediatric Clinic, University of Milano Bicocca , San Gerardo Hospital/MBBM Foundation, Monza, Italy
| | - Sarah Tettamanti
- 1 Molecular Therapy Unit, Tettamanti Research Center, Pediatric Clinic, University of Milano Bicocca , San Gerardo Hospital/MBBM Foundation, Monza, Italy
| | - Ettore Biagi
- 1 Molecular Therapy Unit, Tettamanti Research Center, Pediatric Clinic, University of Milano Bicocca , San Gerardo Hospital/MBBM Foundation, Monza, Italy
| |
Collapse
|
25
|
Finotti G, Tamassia N, Cassatella MA. Synergistic production of TNFα and IFNα by human pDCs incubated with IFNλ3 and IL-3. Cytokine 2016; 86:124-131. [PMID: 27513213 DOI: 10.1016/j.cyto.2016.08.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 08/03/2016] [Accepted: 08/03/2016] [Indexed: 12/14/2022]
Abstract
In this study, we investigated whether IFNλ3 and IL-3 reciprocally influence their capacity to activate various functions of human plasmacytoid dendritic cells (pDCs). In fact, we preliminarily observed that IFNλ3 upregulates the expression of the IL-3Rα (CD123), while IL-3 augments the expression of IFNλR1 in pDCs. As a result, we found that combination of IFNλ3 and IL-3 induces a strong potentiation in the production of TNFα, IFNα, as well as in the expression of Interferon-Stimulated Gene (ISG) mRNAs by pDCs, as compared to either IFNλ3 or IL-3 alone. In such regard, we found that endogenous IFNα autocrinally promotes the expression of ISG mRNAs in IL-3-, but not in IFNλ3 plus IL-3-, treated pDCs. Moreover, we uncovered that the production of IFNα by IFNλ3 plus IL-3-treated pDCs is mostly dependent on endogenously produced TNFα. Altogether, our data demonstrate that IFNλ3 and IL-3 collaborate to promote, at maximal levels, discrete functional responses of human pDCs.
Collapse
Affiliation(s)
- Giulia Finotti
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Nicola Tamassia
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Marco A Cassatella
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy.
| |
Collapse
|
26
|
CD123-Engager T Cells as a Novel Immunotherapeutic for Acute Myeloid Leukemia. Mol Ther 2016; 24:1615-26. [PMID: 27401038 DOI: 10.1038/mt.2016.116] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 06/01/2016] [Indexed: 01/10/2023] Open
Abstract
Immunotherapy with CD123-specific T-cell engager proteins or with T cells expressing CD123-specific chimeric antigen receptors is actively being pursued for acute myeloid leukemia. T cells secreting bispecific engager molecules (ENG-T cells) may present a promising alternative to these approaches. To evaluate therapeutic potential, we generated T cells to secrete CD123/CD3-bispecific engager molecules. CD123-ENG T cells recognized primary acute myeloid leukemia (AML) cells and cell lines in an antigen-dependent manner as judged by cytokine production and/or tumor killing, and redirected bystander T cells to AML cells. Infusion of CD123-ENG T cells resulted in regression of AML in xenograft models conferring a significant survival advantage of treated mice in comparison to mice that received control T cells. At high effector to target ratios, CD123-ENG T cells recognized normal hematopoietic stem and progenitor cells (HSPCs) with preferential recognition of HSPCs from cord blood compared to bone marrow. We therefore introduced the CD20 suicide gene that can be targeted in vivo with rituximab into CD123-ENG T cells. The expression of CD20 did not diminish the anti-AML activity of CD123-ENG T cells, but allowed for rituximab-mediated ENG-T cell elimination. Thus, ENG-T cells coexpressing CD20 suicide and CD123 engager molecules may present a promising immunotherapeutic approach for AML.
Collapse
|
27
|
Activated Stat5 trafficking Via Endothelial Cell-derived Extracellular Vesicles Controls IL-3 Pro-angiogenic Paracrine Action. Sci Rep 2016; 6:25689. [PMID: 27157262 PMCID: PMC4860593 DOI: 10.1038/srep25689] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 04/21/2016] [Indexed: 12/17/2022] Open
Abstract
Soluble factors and cell-derived extracellular vesicles (EVs) control vascular cell fate during inflammation. The present study investigates the impact of Interleukin 3 (IL-3) on EV release by endothelial cells (ECs), the mechanisms involved in EV release and paracrine actions. We found that IL-3 increases EV release, which is prevented by IL-3Ralpha blockade. EVs released upon IL-3 stimulation were able to induce pro-angiogenic signals as shown by chromatin immunoprecipitation (ChIP) assay performed on the promoter region of cyclin D1 and tridimensional tube-like structure formation. We herein demonstrate that these effects rely on the transfer of miR-126-3p, pre-miR-126 and, more importantly, of activated signal transduction and activator of transcription 5 (pSTAT5) from IL-3-EV cargo into recipient ECs. We show, using the dominant negative form (ΔN)STAT5 and an activated STAT5 (1*6STAT5) constructs, that STAT5 drives IL-3-mediated EV release, miR-126-3p and pSTAT5 content. Finally, using EVs recovered from ΔNSTAT5 expressing ECs, we provide evidence that miR-126-3p and pSTAT5 trafficking is relevant for IL-3-mediated paracrine pro-angiogenic signals. These results indicate that IL-3 regulates EC-EV release, cargo and IL-3 angiogenic paracrine action via STAT5. Moreover, these results provide evidence that EC-derived IL-3-EVs can serve as pro-angiogenic clinical delivery wound healing devices.
Collapse
|
28
|
Chichili GR, Huang L, Li H, Burke S, He L, Tang Q, Jin L, Gorlatov S, Ciccarone V, Chen F, Koenig S, Shannon M, Alderson R, Moore PA, Johnson S, Bonvini E. A CD3xCD123 bispecific DART for redirecting host T cells to myelogenous leukemia: preclinical activity and safety in nonhuman primates. Sci Transl Med 2016; 7:289ra82. [PMID: 26019218 DOI: 10.1126/scitranslmed.aaa5693] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Current therapies for acute myeloid leukemia (AML) are largely ineffective, and AML patients may benefit from targeted immunotherapy approaches. MGD006 is a bispecific CD3xCD123 dual-affinity re-targeting (DART) molecule that binds T lymphocytes and cells expressing CD123, an antigen up-regulated in several hematological malignancies including AML. MGD006 mediates blast killing in AML samples, together with concomitant activation and expansion of residual T cells. MGD006 is designed to be rapidly cleared, and therefore requires continuous delivery. In a mouse model of continuous administration, MGD006 eliminated engrafted KG-1a cells (an AML-M0 line) in human PBMC (peripheral blood mononuclear cell)-reconstituted NSG/β2m(-/-) mice at doses as low as 0.5 μg/kg per day for ~7 days. MGD006 binds to human and cynomolgus monkey antigens with similar affinities and redirects T cells from either species to kill CD123-expressing target cells. MGD006 was well tolerated in monkeys continuously infused with 0.1 μg/kg per day escalated weekly to up to 1 μg/kg per day during a 4-week period. Depletion of circulating CD123-positive cells was observed as early as 72 hours after treatment initiation and persisted throughout the infusion period. Cytokine release, observed after the first infusion, was reduced after subsequent administrations, even when the dose was escalated. T cells from animals with prolonged in vivo exposure exhibited unperturbed target cell lysis ex vivo, indicating no exhaustion. A transient decrease in red cell mass was observed, with no neutropenia or thrombocytopenia. These studies support clinical testing of MGD006 in hematological malignancies, including AML.
Collapse
Affiliation(s)
| | - Ling Huang
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA
| | - Hua Li
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA
| | - Steve Burke
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA
| | - Leilei He
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA
| | - Qin Tang
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA
| | - Linda Jin
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA
| | - Sergey Gorlatov
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA
| | | | - Francine Chen
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA
| | - Scott Koenig
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA
| | - Michele Shannon
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA
| | - Ralph Alderson
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA
| | - Paul A Moore
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA
| | - Syd Johnson
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA
| | - Ezio Bonvini
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA.
| |
Collapse
|
29
|
Valeta-Magara A, Hatami R, Axelrod D, Roses DF, Guth A, Formenti SC, Schneider RJ. Pro-oncogenic cytokines and growth factors are differentially expressed in the post-surgical wound fluid from malignant compared to benign breast lesions. SPRINGERPLUS 2015; 4:483. [PMID: 26361584 PMCID: PMC4560730 DOI: 10.1186/s40064-015-1260-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Accepted: 08/18/2015] [Indexed: 12/16/2022]
Abstract
Purpose The accumulation of wound fluid known as seroma in the chest cavity following breast surgery is a common occurrence that can persist for many weeks. While the pro-inflammatory composition of seroma is well established, there has been remarkably little research to determine whether seroma contains pro-oncogenic factors, and whether this is influenced by previous malignant disease. Methods We developed a clinical trial in which we obtained post-surgical seroma fluids from women with benign or malignant disease 1 or 2 weeks following lumpectomy or mastectomy. We conducted an analysis of more than 80 different cytokines, chemokines and growth factors. Results We found that surgical cavity seroma from breast cancer patients has a higher expression of key tumor-promoting cytokines and lower expression of important tumor-inhibiting factors when compared to benign lesions from non-cancer patients. Patients with high body mass index also had higher levels of leptin regardless of malignancy. Conclusions We conclude that the breast post-surgical tumor cavity contains factors that are pro-inflammatory regardless of malignant or benign disease, but in malignant disease there is significant enrichment of additional pro-oncogenic chemokines, cytokines and growth factors, and reduction in tumor-inhibiting factors. These results are consistent with tumor conditioning of surrounding normal stromal tissue and creation of a pro-oncogenic environment that persists long after surgical removal of the tumor. Electronic supplementary material The online version of this article (doi:10.1186/s40064-015-1260-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Raheleh Hatami
- Department of Microbiology, NYU School of Medicine, New York, NY 10016 USA
| | - Deborah Axelrod
- Department of Surgery, NYU School of Medicine, New York, NY 10016 USA ; NYU Perlmutter Cancer Institute, NYU School of Medicine, New York, NY 10016 USA
| | - Daniel F Roses
- Department of Surgery, NYU School of Medicine, New York, NY 10016 USA ; NYU Perlmutter Cancer Institute, NYU School of Medicine, New York, NY 10016 USA
| | - Amber Guth
- Department of Surgery, NYU School of Medicine, New York, NY 10016 USA ; NYU Perlmutter Cancer Institute, NYU School of Medicine, New York, NY 10016 USA
| | - Silvia C Formenti
- Department of Radiation Oncology, NYU School of Medicine, New York, NY 10016 USA ; Weill Cornell Medical College, New York, NY USA
| | - Robert J Schneider
- Department of Microbiology, NYU School of Medicine, New York, NY 10016 USA ; Department of Surgery, NYU School of Medicine, New York, NY 10016 USA ; NYU Perlmutter Cancer Institute, NYU School of Medicine, New York, NY 10016 USA ; Department of Radiation Oncology, NYU School of Medicine, New York, NY 10016 USA
| |
Collapse
|
30
|
Broughton SE, Nero TL, Dhagat U, Kan WL, Hercus TR, Tvorogov D, Lopez AF, Parker MW. The βc receptor family – Structural insights and their functional implications. Cytokine 2015; 74:247-58. [DOI: 10.1016/j.cyto.2015.02.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 02/06/2015] [Indexed: 11/25/2022]
|
31
|
Rollins-Raval MA, Marafioti T, Swerdlow SH, Roth CG. The number and growth pattern of plasmacytoid dendritic cells vary in different types of reactive lymph nodes: an immunohistochemical study. Hum Pathol 2013; 44:1003-10. [DOI: 10.1016/j.humpath.2012.08.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 08/24/2012] [Accepted: 08/29/2012] [Indexed: 10/27/2022]
|
32
|
Tettamanti S, Marin V, Pizzitola I, Magnani CF, Giordano Attianese GMP, Cribioli E, Maltese F, Galimberti S, Lopez AF, Biondi A, Bonnet D, Biagi E. Targeting of acute myeloid leukaemia by cytokine-induced killer cells redirected with a novel CD123-specific chimeric antigen receptor. Br J Haematol 2013; 161:389-401. [PMID: 23432359 DOI: 10.1111/bjh.12282] [Citation(s) in RCA: 175] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 01/11/2013] [Indexed: 12/13/2022]
Abstract
Current therapeutic regimens for acute myeloid leukaemia (AML) are still associated with high rates of relapse. Immunotherapy with T-cells genetically modified to express chimeric antigen receptors (CARs) represents an innovative approach. Here we investigated the targeting of the interleukin three receptor alpha (IL3RA; CD123) molecule, which is overexpressed on AML bulk population, CD34(+) leukaemia progenitors, and leukaemia stem cells (LSC) compared to normal haematopoietic stem/progenitor cells (HSPCs), and whose overexpression is associated with poor prognosis. Cytokine-induced killer (CIK) cells were transduced with SFG-retroviral-vector encoding an anti-CD123 CAR. Transduced cells were able to strongly kill CD123(+) cell lines, as well as primary AML blasts. Interestingly, secondary colony experiments demonstrated that anti-CD123.CAR preserved in vitro HSPCs, in contrast to a previously generated anti-CD33.CAR, while keeping an identical cytotoxicity profile towards AML. Furthermore, limited killing of normal monocytes and CD123-low-expressing endothelial cells was noted, thus indicating a low toxicity profile of the anti-CD123.CAR. Taken together, our results indicate that CD123-specific CARs strongly enhance anti-AML CIK functions, while sparing HSPCs and normal low-expressing antigen cells, paving the way to develop novel immunotherapy approaches for AML treatment.
Collapse
Affiliation(s)
- Sarah Tettamanti
- Centro di Ricerca Matilde Tettamanti, Department of Paediatrics, University of Milano-Bicocca, San Gerardo Hospital, Monza, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
|
34
|
Kim S, Prout M, Ramshaw H, Lopez AF, LeGros G, Min B. Cutting edge: basophils are transiently recruited into the draining lymph nodes during helminth infection via IL-3, but infection-induced Th2 immunity can develop without basophil lymph node recruitment or IL-3. THE JOURNAL OF IMMUNOLOGY 2009; 184:1143-7. [PMID: 20038645 DOI: 10.4049/jimmunol.0902447] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Basophils are recognized as immune modulators through their ability to produce IL-4, a key cytokine required for Th2 immunity. It has also recently been reported that basophils are transiently recruited into the draining lymph node (LN) after allergen immunization and that the recruited basophils promote the differentiation of naive CD4 T cells into Th2 effector cells. Using IL-3(-/-) and IL-3Rbeta(-/-) mice, we report in this study that the IL-3/IL-3R system is absolutely required to recruit circulating basophils into the draining LN following helminth infection. Unexpectedly, the absence of IL-3 or of basophil LN recruitment played little role in helminth-induced Th2 immune responses. Moreover, basophil depletion in infected mice did not diminish the development of IL-4-producing CD4 T cells. Our results reveal a previously unknown role of IL-3 in recruiting basophils to the LN and demonstrate that basophils are not necessarily associated with the development of Th2 immunity during parasite infection.
Collapse
Affiliation(s)
- Sohee Kim
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | | | | | | | | | | |
Collapse
|
35
|
Programmed assembly of 3-dimensional microtissues with defined cellular connectivity. Proc Natl Acad Sci U S A 2009; 106:4606-10. [PMID: 19273855 DOI: 10.1073/pnas.0900717106] [Citation(s) in RCA: 244] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Multicellular organs comprise differentiated cell types with discrete yet interdependent functions. The cells' spatial arrangements and interconnectivities, both critical elements of higher-order function, derive from complex developmental programs in vivo and are often difficult or impossible to emulate in vitro. Here, we report the bottom-up synthesis of microtissues composed of multiple cell types with programmed connectivity. We functionalized cells with short oligonucleotides to impart specific adhesive properties. Hybridization of complementary DNA sequences enabled the assembly of multicellular structures with defined cell-cell contacts. We demonstrated that the kinetic parameters of the assembly process depend on DNA sequence complexity, density, and total cell concentration. Thus, cell assembly can be highly controlled, enabling the design of microtissues with defined cell composition and stoichiometry. We used this strategy to construct a paracrine signaling network in isolated 3-dimensional microtissues.
Collapse
|
36
|
Shen T, Kim S, Do JS, Wang L, Lantz C, Urban JF, Le Gros G, Min B. T cell-derived IL-3 plays key role in parasite infection-induced basophil production but is dispensable for in vivo basophil survival. Int Immunol 2008; 20:1201-9. [DOI: 10.1093/intimm/dxn077] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
37
|
Du X, Ho M, Pastan I. New immunotoxins targeting CD123, a stem cell antigen on acute myeloid leukemia cells. J Immunother 2007; 30:607-13. [PMID: 17667524 DOI: 10.1097/cji.0b013e318053ed8e] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The specific alpha subunit of the interleukin-3 receptor (IL-3Ralpha, CD123) is strongly expressed in various leukemic blasts and leukemic stem cells and seems to be an excellent target for the therapy of leukemias. In this study, immunotoxins were developed to target CD123 only, which bypasses the dependence on other subunits to form intact IL-3R. Three anti-CD123 hybridomas (26292, 32701, and 32716) were selected on the basis of their affinity for CD123. Total RNAs were extracted from the 3 anti-CD123 hybridomas and used to clone the fragment of variable region (Fvs). The Fvs were assembled into single chain Fvs and fused to a 38-kd fragment of Pseudomonas exotoxin A to make recombinant immunotoxins. 26292(Fv)-PE38 was found to have the highest cytotoxic activity on the CD123 expressing leukemia cell line TF-1. It bound the cells with a kd of 3.5 nM. Another immunotoxin, 32716(Fv)-PE38, belonging to a different epitope group, had a similar binding ability but was less active, demonstrating the role of epitope selection in immunotoxin action. The cytotoxic activity of 26292(Fv)-PE38 was increased from 200 to about 40 ng/mL by mutating the REDLK sequence at the C terminus to KDEL. 26292(Fv)-PE38-KDEL was specifically cytotoxic to several CD123 expressing cell lines (TF-1, Molm-13, and Molm-14) with good CD123 expression but not to ML-1 or U937 with low or absent expression. In conclusion, 26292(Fv)-PE38-KDEL shows good cytotoxic activity against CD123 expressing cell lines, and merits further development for the possible treatment of acute myeloid leukemia and other CD123 expressing malignancies.
Collapse
Affiliation(s)
- Xing Du
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4264, USA
| | | | | |
Collapse
|
38
|
Jongbloed SL, Lebre MC, Fraser AR, Gracie JA, Sturrock RD, Tak PP, McInnes IB. Enumeration and phenotypical analysis of distinct dendritic cell subsets in psoriatic arthritis and rheumatoid arthritis. Arthritis Res Ther 2007; 8:R15. [PMID: 16507115 PMCID: PMC1526567 DOI: 10.1186/ar1864] [Citation(s) in RCA: 149] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2005] [Revised: 10/20/2005] [Accepted: 11/09/2005] [Indexed: 01/29/2023] Open
Abstract
Dendritic cells (DCs) comprise heterogeneous subsets of professional antigen-presenting cells, linking innate and adaptive immunity. Analysis of DC subsets has been hampered by a lack of specific DC markers and reliable quantitation assays. We characterised the immunophenotype and functional characteristics of psoriatic arthritis (PsA)-derived and rheumatoid arthritis (RA)-derived myeloid DCs (mDCs) and plasmacytoid DCs (pDCs) to evaluate their potential role in arthritis. Circulating peripheral blood (PB) pDC numbers were significantly reduced in PsA patients (P = 0.0098) and RA patients (P = 0.0194), and mDCs were significantly reduced in RA patients (P = 0.0086) compared with healthy controls. The number of circulating mDCs in RA PB was significantly inversely correlated to C-reactive protein (P = 0.021). The phenotype of both DC subsets in PsA PB and RA PB was immature as compared with healthy controls. Moreover, CD62L expression was significantly decreased on both mDCs (PsA, P = 0.0122; RA, P = 0.0371) and pDCs (PsA, P = 0.0373; RA, P = 0.0367) in PB. Both mDCs and pDCs were present in PsA synovial fluid (SF) and RA SF, with the mDC:pDC ratio significantly exceeding that in matched PB (PsA SF, P = 0.0453; RA SF, P = 0.0082). pDCs isolated from RA SF and PsA SF displayed an immature phenotype comparable with PB pDCs. RA and PsA SF mDCs, however, displayed a more mature phenotype (increased expression of CD80, CD83 and CD86) compared with PB mDCs. Functional analysis revealed that both SF DC subsets matured following toll-like receptor stimulation. pDCs from PB and SF produced interferon alpha and tumour necrosis factor alpha on TLR9 stimulation, but only SF pDCs produced IL-10. Similarly, mDCs from PB and SF produced similar tumour necrosis factor alpha levels to TLR2 agonism, whereas SF mDCs produced more IL-10 than PB controls. Circulating DC subset numbers are reduced in RA PB and PsA PB with reduced CD62L expression. Maturation is incomplete in the inflamed synovial compartment. Immature DCs in SF may contribute to the perpetuation of inflammation via sampling of the inflamed synovial environment, and in situ presentation of arthritogenic antigen.
Collapse
Affiliation(s)
- Sarah L Jongbloed
- Division of Immunology, Infection and Inflammation, 10 Alexandra Parade, Glasgow, G31 2ER, UK
| | - M Cristina Lebre
- Director, Academic Medical Center, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, F4-218 P.O. Box 22700, 1100 DE Amsterdam, The Netherlands
| | - Alasdair R Fraser
- Division of Immunology, Infection and Inflammation, 10 Alexandra Parade, Glasgow, G31 2ER, UK
| | - J Alastair Gracie
- Division of Immunology, Infection and Inflammation, 10 Alexandra Parade, Glasgow, G31 2ER, UK
| | - Roger D Sturrock
- Division of Immunology, Infection and Inflammation, 10 Alexandra Parade, Glasgow, G31 2ER, UK
| | - Paul P Tak
- Director, Academic Medical Center, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, F4-218 P.O. Box 22700, 1100 DE Amsterdam, The Netherlands
| | - Iain B McInnes
- Division of Immunology, Infection and Inflammation, 10 Alexandra Parade, Glasgow, G31 2ER, UK
| |
Collapse
|
39
|
Defilippi P, Rosso A, Dentelli P, Calvi C, Garbarino G, Tarone G, Pegoraro L, Brizzi MF. {beta}1 Integrin and IL-3R coordinately regulate STAT5 activation and anchorage-dependent proliferation. ACTA ACUST UNITED AC 2005; 168:1099-108. [PMID: 15795318 PMCID: PMC2171831 DOI: 10.1083/jcb.200405116] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We previously demonstrated that integrin-dependent adhesion activates STAT5A, a well known target of IL-3–mediated signaling. Here, we show that in endothelial cells the active β1 integrin constitutively associates with the unphosphorylated IL-3 receptor (IL-3R) β common subunit. This association is not sufficient for activating downstream signals. Indeed, only upon fibronectin adhesion is Janus Kinase 2 (JAK2) recruited to the β1 integrin–IL-3R complex and triggers IL-3R β common phosphorylation, leading to the formation of docking sites for activated STAT5A. These events are IL-3 independent but require the integrity of the IL-3R β common. IL-3 treatment increases JAK2 activation and STAT5A and STAT5B tyrosine and serine phosphorylation and leads to cell cycle progression in adherent cells. Expression of an inactive STAT5A inhibits cell cycle progression upon IL-3 treatment, identifying integrin-dependent STAT5A activation as a priming event for IL-3–mediated S phase entry. Consistently, overexpression of a constitutive active STAT5A leads to anchorage-independent cell cycle progression. Therefore, these data provide strong evidence that integrin-dependent STAT5A activation controls IL-3–mediated proliferation.
Collapse
Affiliation(s)
- Paola Defilippi
- Department of Genetics, Biology and Biochemistry, University of Torino, 10126, Torino, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Chen L, Martinez O, Overbergh L, Mathieu C, Prabhakar BS, Chan LS. Early up-regulation of Th2 cytokines and late surge of Th1 cytokines in an atopic dermatitis model. Clin Exp Immunol 2005; 138:375-87. [PMID: 15544612 PMCID: PMC1809236 DOI: 10.1111/j.1365-2249.2004.02649.x] [Citation(s) in RCA: 114] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
We investigated cytokine profiles in interleukin (IL)-4 transgenic (Tg) mice with a skin inflammatory disease resembling human atopic dermatitis. cDNA microarray revealed that the mRNAs encoding IL-1beta, IL-2, IL-3, IL-4, IL-5, IL-6, IL-10, IL-12p40, IL-13, tumour necrosis factor (TNF)-alpha, TNF-beta and interferon (IFN)-gamma were up-regulated in the skin of late lesion Tg mice and to a lesser degree in non-lesion Tg mice when compared to those of non-Tg mice. Real time reverse transcription-polymerase chain reaction (RT-PCR) analyses indicated that the cDNA copy numbers of IL-1beta, IL-4, IL-6, IL-10, TNF-alpha and IFN-gamma from the skin of late, early and non-lesions increased significantly compared to non-Tg mice. IL-2 and IL-12p40 cDNA copy numbers were increased significantly in early, but not late, lesions. Interestingly, IL-1beta, IL-3, IL-4, IL-5, IL-6, IL-10, IL-13, TNF-alpha, and IFN-gamma cDNAs were increased significantly the skin of before-onset and/or non-lesion mice. Flow cytometry analyses demonstrated an increased percentage of keratinocytes producing IL-4 as the disease progressed. The percentage of IL-2, IL-4, IL-10 and IFN-gamma-producing T cells and IL-12-producing antigen-presenting cells in skin-draining lymph nodes and inflammatory skin also increased, particularly in mice with late lesion. These results suggest that disease induction is primarily triggered by Th2 cytokines and that Th1, Th2 and non-Th proinflammatory cytokines are all involved in the disease process.
Collapse
Affiliation(s)
- L Chen
- Department of Dermatology, University of Illinois, Chicago, IL 60612, USA
| | | | | | | | | | | |
Collapse
|
41
|
Gröger M, Loewe R, Holnthoner W, Embacher R, Pillinger M, Herron GS, Wolff K, Petzelbauer P. IL-3 induces expression of lymphatic markers Prox-1 and podoplanin in human endothelial cells. THE JOURNAL OF IMMUNOLOGY 2005; 173:7161-9. [PMID: 15585837 DOI: 10.4049/jimmunol.173.12.7161] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Factors determining lymphatic differentiation in the adult organism are not yet well characterized. We have made the observation that mixed primary cultures of dermal blood endothelial cells (BEC) and lymphatic endothelial cells (LEC) grown under standard conditions change expression of markers during subculture: After passage 6, they uniformly express LEC-specific markers Prox-1 and podoplanin. Using sorted cells, we show that LEC but not BEC constitutively express IL-3, which regulates Prox-1 and podoplanin expression in LEC. The addition of IL-3 to the medium of BEC cultures induces Prox-1 and podoplanin. Blocking IL-3 activity in LEC cultures results in a loss of Prox-1 and podoplanin expression. In conclusion, endogenous IL-3 is required to maintain the LEC phenotype in culture, and the addition of IL-3 to BEC appears to induce transdifferentiation of BEC into LEC.
Collapse
Affiliation(s)
- Marion Gröger
- Department of Dermatology, Division of General Dermatology, Medical University of Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Thomas D, Vadas M, Lopez A. Regulation of haematopoiesis by growth factors - emerging insights and therapies. Expert Opin Biol Ther 2004; 4:869-79. [PMID: 15174969 DOI: 10.1517/14712598.4.6.869] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Haematopoiesis is regulated by a wide variety of glycoprotein hormones, including stem cell factor, granulocyte-macrophage colony-stimulating factor, thrombopoietin and IL-3. These haematopoietic growth factors (HGFs) share a number of properties, including redundancy, pleiotropy, autocrine and paracrine effects, receptor subunit oligomerisation and similar signal transduction mechanisms, yet each one has a unique spectrum of haematopoietic activity. Ongoing studies with knockout mice have discovered previously unrecognised physiological roles for HGFs, linking haematopoiesis to innate immunity, pulmonary physiology and bone metabolism. The regulation of stem cells by HGFs within niches of the bone marrow microenvironment is now well recognised and similar mechanisms appear to exist in the regulation of other stem cell compartments. Alternative signalling strategies, other than tyrosine kinase activation and phosphotyrosine cascades, may account for some of the more subtle differences between HGFs. Accumulating evidence suggests that some, but not all, HGF receptors can transduce a genuine lineage-determining signal at certain points in haematopoiesis. Further studies, primarily at the receptor level, are needed to determine the mechanisms of instructive signalling, which may include phosphoserine cascades. Novel haematopoietic regulators, as well as the development of biological therapies, including growth factor antagonists and peptide mimetics, are also discussed.
Collapse
Affiliation(s)
- Daniel Thomas
- The Hanson Institute, Division of Human Immunology, The Institute of Medical and Veterinary Science, Adelaide, SA, Australia
| | | | | |
Collapse
|
43
|
de Wilt JH, Bout A, Eggermont AM, van Tiel ST, de Vries MW, ten Hagen TL, de Roos WK, Valerio D, van der Kaaden ME. Adenovirus-mediated interleukin 3 beta gene transfer by isolated limb perfusion inhibits growth of limb sarcoma in rats. Hum Gene Ther 2001; 12:489-502. [PMID: 11268282 DOI: 10.1089/104303401300042384] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Cytokine gene transfer using (multiple) intratumoral injections can induce tumor regression in several animal models, but this administration technique limits the use for human gene therapy. In the present studies we describe tumor growth inhibition of established limb sarcomas after a single isolated limb perfusion (ILP) with recombinant adenoviral vectors harboring the rat IL-3 beta gene (IG.Ad.CMV.rIL-3 beta). In contrast, a single intratumoral injection or intravenous administration did not affect tumor growth. Dose-finding studies demonstrated a dose-dependent response with a loss of antitumor effect below 1 x 10(9) IU of IG.Ad.CMV.rIL-3 beta. Perfusions with adenoviral vectors bearing a weaker promoter (MLP promoter) driving the rIL-3 beta gene did not result in antitumor responses, suggesting that the rIL-3 beta-mediated antitumor effect depends on the amount of rIL-3 beta protein expressed by the infected cells. Furthermore, it was shown by direct comparison that ILP with IG.Ad.CMV.rIL-3 beta in the ROS-1 osteosarcoma model is at least as efficient as the established therapy with the combination of TNF-alpha and melphalan. Treatment with IG.Ad.CMV.rIL-3 beta induced a transient dose-dependent leukocytosis accompanied by an increase in peripheral blood levels of histamine. Leukocyte infiltrations were also histopathologically demonstrated in tumors after perfusion. These results demonstrate that ILP with recombinant adenoviral vectors carrying the IL-3 beta transgene inhibits tumor growth in rats and suggest that cytokine gene therapy using this administration technique might be beneficial for clinical cancer treatment.
Collapse
Affiliation(s)
- J H de Wilt
- Department of Surgical Oncology, University Hospital Rotterdam Dijkzigt/Daniel de Hoed Cancer Center, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
The importance of cytokines as mediators in numerous physiologic and pathologic processes became apparent in the early seventies. In a relatively short period of time the number of characterized and functionally defined interleukins increased rapidly. Concomitantly, the understanding of their role in various important reactions in different systems of the organism in general, and in the interaction between the immune, central nervous and neuroendocrine systems in particular increased. IL-3 is one of the cytokines of great significance for hematopoiesis. In addition, it was shown that interleukin-3 (IL-3) participates in the response of the organism to various types of stress. Surprisingly enough, the reports on its production and activation during or following stress are rather scarce. Therefore, the purpose of this review is to try and compile the information on IL-3 and its role in different types of stress, and also to contribute our own modest experience on the subject.
Collapse
Affiliation(s)
- H Bessler
- Hematology and Immunology Research Laboratory, Rabin Medical Center, Petah-Tiqva, Israel
| | | | | |
Collapse
|
45
|
Frankel AE, McCubrey JA, Miller MS, Delatte S, Ramage J, Kiser M, Kucera GL, Alexander RL, Beran M, Tagge EP, Kreitman RJ, Hogge DE. Diphtheria toxin fused to human interleukin-3 is toxic to blasts from patients with myeloid leukemias. Leukemia 2000; 14:576-85. [PMID: 10764142 DOI: 10.1038/sj.leu.2401743] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Leukemic blasts from patients with acute phase chronic myeloid leukemic and refractory acute myeloid leukemia are highly resistant to a number of cytotoxic drugs. To overcome multi-drug resistance, we engineered a diphtheria fusion protein by fusing human interleukin-3 (IL3) to a truncated form of diphtheria toxin (DT) with a (G4S)2 linker (L), expressed and purified the recombinant protein, and tested the cytotoxicity of the DTLIL3 molecule on human leukemias and normal progenitors. The DTLIL3 construct was more cytotoxic to interleukin-3 receptor (IL3R) bearing human myeloid leukemia cell lines than receptor-negative cell lines based on assays of cytotoxicity using thymidine incorporation, growth in semi-solid medium and induction of apoptosis. Exposure of mononuclear cells to 680 pM DTLIL3 for 48 h in culture reduced the number of cells capable of forming colonies in semi-solid medium (colony-forming units leukemia) > or =10-fold in 4/11 (36%) patients with myeloid acute phase chronic myeloid leukemia (CML) and 3/9 (33%) patients with acute myeloid leukemia (AML). Normal myeloid progenitors (colony-forming unit granulocyte-macrophage) from five different donors treated and assayed under identical conditions showed intermediate sensitivity with three- to five-fold reductions in colonies. The sensitivity to DTLIL3 of leukemic progenitors from a number of acute phase CML patients suggests that this agent could have therapeutic potential for some patients with this disease.
Collapse
Affiliation(s)
- A E Frankel
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Vallera DA, Seo SY, Panoskaltsis-Mortari A, Griffin JD, Blazar BR. Targeting myeloid leukemia with a DT(390)-mIL-3 fusion immunotoxin: ex vivo and in vivo studies in mice. PROTEIN ENGINEERING 1999; 12:779-85. [PMID: 10506288 DOI: 10.1093/protein/12.9.779] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The IL-3 receptor was expressed on a high frequency of myeloid leukemia cells and also on hematopoietic and vascular cells. We previously showed that a recombinant IL-3 fusion immunotoxin (DT(390)IL-3) expressed by splicing the murine IL-3 gene to a truncated diphtheria toxin (DT(390)) gene selectively killed IL-3R(+) expressing cells and was not uniformly toxic to uncommitted BM progenitor cells (Chan,C.-H., Blazar,B.R., Greenfield,L., Kreitman,R.J. and Vallera,D.A., 1996, Blood, 88, 1445-1456). Thus, we explored the feasibility of using DT(390)IL-3 as an anti-leukemia agent. DT(390)IL-3 was toxic when administered to mice at doses as low as 0.1 microg/day. The dose limiting toxicity appeared to be related to platelet and bleeding effects of the fusion toxin. Because of these effects, DT(390)IL-3 was studied ex vivo as a means of purging contaminating leukemia cells from BM grafts in a murine autologous BM transplantation. In this setting, as few as 1000 IL-3R-expressing, bcr/abl transformed myeloid 32Dp210 leukemia cells were lethal. An optimal purging interval of 10 nM/l for 8 h eliminated leukemia cells from 32Dp210/BM mixtures given to lethally irradiated (8 Gy) C3H/HeJ syngeneic mice. Mice given treated grafts containing BM and a lethal dose of 32Dp210 cells survived over 100 days while mice given untreated grafts did not survive (P < 0.00001). DT(390)IL-3 may prove highly useful for ex vivo purging of lethal malignant leukemia cells from autologous BM grafts.
Collapse
Affiliation(s)
- D A Vallera
- University of Minnesota Cancer Center, Department of Therapeutic Radiology (Section on Experimental Cancer Immunology) Minneapolis, MN 55455, USA
| | | | | | | | | |
Collapse
|