1
|
Kushwaha AC, Mrunalini B, Malhotra P, Karmakar S, Roy Choudhury S. Bone-Marrow-Targeted Nanocomposite Abrogates C-Myb-Survivin Cross Talk in MLL-AF9-Rearranged Acute Myeloid Leukemia in In Vitro and In Vivo Patient-Derived Xenograft Models. ACS APPLIED MATERIALS & INTERFACES 2025; 17:711-724. [PMID: 39711012 DOI: 10.1021/acsami.4c18737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
The heterogeneous form of malignancy in the myeloid lineage of normal hematopoietic stem cells (HSCs) is characterized as acute myeloid leukemia (AML). The t(9;11) reciprocal translocation (p22;q23) generates MLL-AF9 oncogene, which results in myeloid-based monoblastic AML with frequent relapse and poor survival. MLL-AF9 binds with the C-Myb promoter and regulates AML onset, maintenance, and survival. The bone marrow microenvironment (BMM) protects leukemia stem cells (LSCs) from therapeutic agents, which can lead to relapsed condition. Targeting leukemia BMM can be a viable therapeutics approach for AML treatment, wherein bone homing bisphosphonate, ibandronic acid (IBD), can localize to the BMM. In order to target the BMM of AML, C-Myb siRNA was entrapped in Vitamin D nanoemulsion-functionalized with BMM-targeted IBD, which exhibited binding with ex vivo bone slices and localization into mice bone marrow. IBD functionalization and C-Myb siRNA nanotherapy enhanced the suppression of LSCs (c-Kit+) and the upregulation of myeloid differentiation markers CD11b and Gr-1 in peripheral blood and bone marrow of athymic nude mice and patient-derived xenograft models. IBD functionalization enhanced the downregulation of C-Myb and C-Myb-Survivin cross talk in bone marrow and spleen tissue responsible for AML onset, maintenance, and pathogenesis. Further C-Myb binding to Survivin promoter was abrogated by the present bone-marrow-targeted nanotherapy, signifying its translational potential for AML therapeutics.
Collapse
MESH Headings
- Humans
- Animals
- Proto-Oncogene Proteins c-myb/metabolism
- Proto-Oncogene Proteins c-myb/genetics
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/genetics
- Mice
- Myeloid-Lymphoid Leukemia Protein/genetics
- Myeloid-Lymphoid Leukemia Protein/metabolism
- Survivin/metabolism
- Survivin/genetics
- Bone Marrow/pathology
- Bone Marrow/metabolism
- Bone Marrow/drug effects
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Nanocomposites/chemistry
- Xenograft Model Antitumor Assays
- Cell Line, Tumor
- Mice, Nude
- RNA, Small Interfering/metabolism
- RNA, Small Interfering/chemistry
Collapse
Affiliation(s)
- Avinash Chandra Kushwaha
- Epigenetics Research Laboratory, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali, Punjab 140306, India
| | - Boddu Mrunalini
- Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali, Punjab 140306, India
| | - Pankaj Malhotra
- Department of Clinical Hematology and Medical Oncology, Nehru Hospital, Post-Graduate Institute of Medical Education and Research, Room No. 18, Fourth Level, F Block, Chandigarh 160020, India
| | - Surajit Karmakar
- Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali, Punjab 140306, India
| | - Subhasree Roy Choudhury
- Epigenetics Research Laboratory, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali, Punjab 140306, India
| |
Collapse
|
2
|
Ceolin V, Spadea M, Apolito V, Saglio F, Fagioli F. Emerging CART Therapies for Pediatric Acute Myeloid Leukemia. J Pediatr Hematol Oncol 2024; 46:393-403. [PMID: 39469946 DOI: 10.1097/mph.0000000000002956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 09/16/2024] [Indexed: 10/30/2024]
Abstract
The prognosis of children with acute myeloid leukemia (AML) has improved incrementally over the last decades. However, at relapse, overall survival (OS) ∼40% to 50% and is even lower for patients with chemorefractory disease. Effective and less-toxic therapies are urgently needed for these children. In the last years, immune-directed therapies such as chimeric antigen receptor (CAR)-T cells were introduced, which showed outstanding clinical activity against B-cell malignancies. CART therapies are being developed for AML on the basis of the results obtained for other hematologic malignancies. The biggest challenge of CART therapy for AML is to identify a specific target antigen, since antigens expressed in AML cells are usually shared with healthy hematopoietic stem cells. An overview of prospects of CART in pediatric AML, focused on the common antigens targeted by CART in AML that have been tested or are currently under investigation, is provided in this manuscript.
Collapse
Affiliation(s)
- Valeria Ceolin
- Department of Pediatric Oncology/Hematology, Regina Margherita Children's Hospital
| | - Manuela Spadea
- Department of Pediatric Oncology/Hematology, Regina Margherita Children's Hospital
- Department of Pediatric Oncology/Hematology, University of Turin, Turin, Italy
| | - Vincenzo Apolito
- Department of Pediatric Oncology/Hematology, Regina Margherita Children's Hospital
| | - Francesco Saglio
- Department of Pediatric Oncology/Hematology, Regina Margherita Children's Hospital
| | - Franca Fagioli
- Department of Pediatric Oncology/Hematology, Regina Margherita Children's Hospital
- Department of Pediatric Oncology/Hematology, University of Turin, Turin, Italy
| |
Collapse
|
3
|
Hill TF, Narvekar P, Asher GD, Edelstein JN, Camp ND, Grimm A, Thomas KR, Leiken MD, Molloy KM, Cook PJ, Arlauckas SP, Morgan RA, Tasian SK, Rawlings DJ, James RG. Human plasma cells engineered to secrete bispecifics drive effective in vivo leukemia killing. Mol Ther 2024; 32:2676-2691. [PMID: 38959896 PMCID: PMC11405176 DOI: 10.1016/j.ymthe.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/09/2024] [Accepted: 06/04/2024] [Indexed: 07/05/2024] Open
Abstract
Bispecific antibodies are an important tool for the management and treatment of acute leukemias. As a next step toward clinical translation of engineered plasma cells, we describe approaches for secretion of bispecific antibodies by human plasma cells. We show that human plasma cells expressing either fragment crystallizable domain-deficient anti-CD19 × anti-CD3 (blinatumomab) or anti-CD33 × anti-CD3 bispecific antibodies mediate T cell activation and direct T cell killing of B acute lymphoblastic leukemia or acute myeloid leukemia cell lines in vitro. We demonstrate that knockout of the self-expressed antigen, CD19, boosts anti-CD19-bispecific secretion by plasma cells and prevents self-targeting. Plasma cells secreting anti-CD19-bispecific antibodies elicited in vivo control of acute lymphoblastic leukemia patient-derived xenografts in immunodeficient mice co-engrafted with autologous T cells. In these studies, we found that leukemic control elicited by engineered plasma cells was similar to CD19-targeted chimeric antigen receptor-expressing T cells. Finally, the steady-state concentration of anti-CD19 bispecifics in serum 1 month after cell delivery and tumor eradication was comparable with that observed in patients treated with a steady-state infusion of blinatumomab. These findings support further development of ePCs for use as a durable delivery system for the treatment of acute leukemias, and potentially other cancers.
Collapse
Affiliation(s)
- Tyler F Hill
- University of Washington, Medical Scientist Training Program, Seattle, WA, USA; Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA
| | - Parnal Narvekar
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA
| | - Gregory D Asher
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA
| | | | - Nathan D Camp
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA
| | - Annaiz Grimm
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA
| | - Kerri R Thomas
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA
| | | | | | - Peter J Cook
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA
| | | | | | - Sarah K Tasian
- Children's Hospital of Philadelphia, Division of Oncology and Center for Childhood Cancer Research, Philadelphia, PA, USA; Department of Pediatrics and Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - David J Rawlings
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA; University of Washington, Departments of Pediatrics and Immunology, Seattle, WA, USA
| | - Richard G James
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA; University of Washington, Departments of Pediatrics and Pharmacology, Seattle, WA, USA.
| |
Collapse
|
4
|
Hill TF, Narvekar P, Asher G, Camp N, Thomas KR, Tasian SK, Rawlings DJ, James RG. Human plasma cells engineered to secrete bispecifics drive effective in vivo leukemia killing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.24.554523. [PMID: 37662410 PMCID: PMC10473709 DOI: 10.1101/2023.08.24.554523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Bispecific antibodies are an important tool for the management and treatment of acute leukemias. Advances in genome-engineering have enabled the generation of human plasma cells that secrete therapeutic proteins and are capable of long-term in vivo engraftment in humanized mouse models. As a next step towards clinical translation of engineered plasma cells (ePCs) towards cancer therapy, here we describe approaches for the expression and secretion of bispecific antibodies by human plasma cells. We show that human ePCs expressing either fragment crystallizable domain deficient anti-CD19 × anti-CD3 (blinatumomab) or anti-CD33 × anti-CD3 bispecific antibodies mediate T cell activation and direct T cell killing of specific primary human cell subsets and B-acute lymphoblastic leukemia or acute myeloid leukemia cell lines in vitro. We demonstrate that knockout of the self-expressed antigen, CD19, boosts anti-CD19 bispecific secretion by ePCs and prevents self-targeting. Further, anti-CD19 bispecific-ePCs elicited tumor eradication in vivo following local delivery in flank-implanted Raji lymphoma cells. Finally, immunodeficient mice engrafted with anti-CD19 bispecific-ePCs and autologous T cells potently prevented in vivo growth of CD19+ acute lymphoblastic leukemia in patient-derived xenografts. Collectively, these findings support further development of ePCs for use as a durable, local delivery system for the treatment of acute leukemias, and potentially other cancers.
Collapse
Affiliation(s)
- Tyler F. Hill
- University of Washington, Medical Scientist Training Program, Seattle WA
- Seattle Children’s Research Institute, Center for Immunity and Immunotherapy, Seattle WA
| | - Parnal Narvekar
- Seattle Children’s Research Institute, Center for Immunity and Immunotherapy, Seattle WA
| | - Gregory Asher
- Seattle Children’s Research Institute, Center for Immunity and Immunotherapy, Seattle WA
| | - Nathan Camp
- Seattle Children’s Research Institute, Center for Immunity and Immunotherapy, Seattle WA
| | - Kerri R. Thomas
- Seattle Children’s Research Institute, Center for Immunity and Immunotherapy, Seattle WA
| | - Sarah K. Tasian
- Children’s Hospital of Philadelphia, Division of Oncology and Center for Childhood Cancer Research, Philadelphia PA
- Department of Pediatrics and Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia PA
| | - David J. Rawlings
- Seattle Children’s Research Institute, Center for Immunity and Immunotherapy, Seattle WA
- University of Washington, Departments of Pediatrics and Immunology, Seattle WA
| | - Richard G. James
- Seattle Children’s Research Institute, Center for Immunity and Immunotherapy, Seattle WA
- University of Washington, Departments of Pediatrics and Pharmacology, Seattle WA
| |
Collapse
|
5
|
Li Y, Xue M, Deng X, Dong L, Nguyen LXT, Ren L, Han L, Li C, Xue J, Zhao Z, Li W, Qing Y, Shen C, Tan B, Chen Z, Leung K, Wang K, Swaminathan S, Li L, Wunderlich M, Mulloy JC, Li X, Chen H, Zhang B, Horne D, Rosen ST, Marcucci G, Xu M, Li Z, Wei M, Tian J, Shen B, Su R, Chen J. TET2-mediated mRNA demethylation regulates leukemia stem cell homing and self-renewal. Cell Stem Cell 2023; 30:1072-1090.e10. [PMID: 37541212 PMCID: PMC11166201 DOI: 10.1016/j.stem.2023.07.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 05/10/2023] [Accepted: 07/03/2023] [Indexed: 08/06/2023]
Abstract
TET2 is recurrently mutated in acute myeloid leukemia (AML) and its deficiency promotes leukemogenesis (driven by aggressive oncogenic mutations) and enhances leukemia stem cell (LSC) self-renewal. However, the underlying cellular/molecular mechanisms have yet to be fully understood. Here, we show that Tet2 deficiency significantly facilitates leukemogenesis in various AML models (mediated by aggressive or less aggressive mutations) through promoting homing of LSCs into bone marrow (BM) niche to increase their self-renewal/proliferation. TET2 deficiency in AML blast cells increases expression of Tetraspanin 13 (TSPAN13) and thereby activates the CXCR4/CXCL12 signaling, leading to increased homing/migration of LSCs into BM niche. Mechanistically, TET2 deficiency results in the accumulation of methyl-5-cytosine (m5C) modification in TSPAN13 mRNA; YBX1 specifically recognizes the m5C modification and increases the stability and expression of TSPAN13 transcripts. Collectively, our studies reveal the functional importance of TET2 in leukemogenesis, leukemic blast cell migration/homing, and LSC self-renewal as an mRNA m5C demethylase.
Collapse
Affiliation(s)
- Yangchan Li
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA; Department of Radiation Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Meilin Xue
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA; Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaolan Deng
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
| | - Lei Dong
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
| | - Le Xuan Truong Nguyen
- Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA 91010, USA; Department of Hematological Malignancies Translational Science, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
| | - Lili Ren
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA; Department of Pathology, Harbin Medical University, Harbin 150081, China
| | - Li Han
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA; Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110001, Liaoning, China
| | - Chenying Li
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA; Department of Hematology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 31003, Zhejiang, China
| | - Jianhuang Xue
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
| | - Zhicong Zhao
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
| | - Wei Li
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
| | - Ying Qing
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
| | - Chao Shen
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
| | - Brandon Tan
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
| | - Zhenhua Chen
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
| | - Keith Leung
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
| | - Kitty Wang
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
| | - Srividya Swaminathan
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA; Department of Pediatrics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Ling Li
- Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA 91010, USA; Department of Hematological Malignancies Translational Science, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA; City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Mark Wunderlich
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - James C Mulloy
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Xiaobo Li
- Department of Pathology, Harbin Medical University, Harbin 150081, China
| | - Hao Chen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Bin Zhang
- Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA 91010, USA; Department of Hematological Malignancies Translational Science, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA; City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - David Horne
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Steven T Rosen
- Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA 91010, USA; City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; Department of Hematology/Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA
| | - Guido Marcucci
- Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA 91010, USA; Department of Hematological Malignancies Translational Science, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA; City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Mingjiang Xu
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Zejuan Li
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110001, Liaoning, China
| | - Jingyan Tian
- State Key Laboratory of Medical Genomics, Clinical Trial Center, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Baiyong Shen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Rui Su
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA; City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA.
| | - Jianjun Chen
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA; Gehr Family Center for Leukemia Research, City of Hope, Duarte, CA 91010, USA; City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA.
| |
Collapse
|
6
|
Leukemic Stem Cells as a Target for Eliminating Acute Myeloid Leukemia: Gaps in Translational Research. Crit Rev Oncol Hematol 2022; 175:103710. [DOI: 10.1016/j.critrevonc.2022.103710] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 04/29/2022] [Accepted: 05/11/2022] [Indexed: 12/26/2022] Open
|
7
|
Pearson AD, Rossig C, Mackall C, Shah NN, Baruchel A, Reaman G, Ricafort R, Heenen D, Bassan A, Berntgen M, Bird N, Bleickardt E, Bouchkouj N, Bross P, Brownstein C, Cohen SB, de Rojas T, Ehrlich L, Fox E, Gottschalk S, Hanssens L, Hawkins DS, Horak ID, Taylor DH, Johnson C, Karres D, Ligas F, Ludwinski D, Mamonkin M, Marshall L, Masouleh BK, Matloub Y, Maude S, McDonough J, Minard-Colin V, Norga K, Nysom K, Pappo A, Pearce L, Pieters R, Pule M, Quintás-Cardama A, Richardson N, Schüßler-Lenz M, Scobie N, Sersch MA, Smith MA, Sterba J, Tasian SK, Weigel B, Weiner SL, Zwaan CM, Lesa G, Vassal G. Paediatric Strategy Forum for medicinal product development of chimeric antigen receptor T-cells in children and adolescents with cancer: ACCELERATE in collaboration with the European Medicines Agency with participation of the Food and Drug Administration. Eur J Cancer 2021; 160:112-133. [PMID: 34840026 DOI: 10.1016/j.ejca.2021.10.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 10/13/2021] [Indexed: 12/30/2022]
Abstract
The seventh multi-stakeholder Paediatric Strategy Forum focused on chimeric antigen receptor (CAR) T-cells for children and adolescents with cancer. The development of CAR T-cells for patients with haematological malignancies, especially B-cell precursor acute lymphoblastic leukaemia (BCP-ALL), has been spectacular. However, currently, there are scientific, clinical and logistical challenges for use of CAR T-cells in BCP-ALL and other paediatric malignancies, particularly in acute myeloid leukaemia (AML), lymphomas and solid tumours. The aims of the Forum were to summarise the current landscape of CAR T-cell therapy development in paediatrics, too identify current challenges and future directions, with consideration of other immune effector modalities and ascertain the best strategies to accelerate their development and availability to children. Although the effect is of limited duration in about half of the patients, anti-CD19 CAR T-cells produce high response rates in relapsed/refractory BCP-ALL and this has highlighted previously unknown mechanisms of relapse. CAR T-cell treatment as first- or second-line therapy could also potentially benefit patients whose disease has high-risk features associated with relapse and failure of conventional therapies. Identifying patients with very early and early relapse in whom CAR T-cell therapy may replace haematopoietic stem cell transplantation and be definitive therapy versus those in whom it provides a more effective bridge to haematopoietic stem cell transplantation is a very high priority. Development of approaches to improve persistence, either by improving T cell fitness or using more humanised/fully humanised products and co-targeting of multiple antigens to prevent antigen escape, could potentially further optimise therapy. Many differences exist between paediatric B-cell non-Hodgkin lymphomas (B-NHL) and BCP-ALL. In view of the very small patient numbers with relapsed lymphoma, careful prioritisation is needed to evaluate CAR T-cells in children with Burkitt lymphoma, primary mediastinal B cell lymphoma and other NHL subtypes. Combination trials of alternative targets to CD19 (CD20 or CD22) should also be explored as a priority to improve efficacy in this population. Development of CD30 CAR T-cell immunotherapy strategies in patients with relapsed/refractory Hodgkin lymphoma will likely be most efficiently accomplished by joint paediatric and adult trials. CAR T-cell approaches are early in development for AML and T-ALL, given the unique challenges of successful immunotherapy actualisation in these diseases. At this time, CD33 and CD123 appear to be the most universal targets in AML and CD7 in T-ALL. The results of ongoing or planned first-in-human studies are required to facilitate further understanding. There are promising early results in solid tumours, particularly with GD2 targeting cell therapies in neuroblastoma and central nervous system gliomas that represent significant unmet clinical needs. Further understanding of biology is critical to success. The comparative benefits of autologous versus allogeneic CAR T-cells, T-cells engineered with T cell receptors T-cells engineered with T cell receptor fusion constructs, CAR Natural Killer (NK)-cell products, bispecific T-cell engager antibodies and antibody-drug conjugates require evaluation in paediatric malignancies. Early and proactive academia and multi-company engagement are mandatory to advance cellular immunotherapies in paediatric oncology. Regulatory advice should be sought very early in the design and preparation of clinical trials of innovative medicines, for which regulatory approval may ultimately be sought. Aligning strategic, scientific, regulatory, health technology and funding requirements from the inception of a clinical trial is especially important as these are very expensive therapies. The model for drug development for cell therapy in paediatric oncology could also involve a 'later stage handoff' to industry after early development in academic hands. Finally, and very importantly, strategies must evolve to ensure appropriate ease of access for children who need and could potentially benefit from these therapies.
Collapse
Affiliation(s)
| | - Claudia Rossig
- University Children´s Hospital Muenster, Pediatric Hematology and Oncology, Germany
| | - Crystal Mackall
- Department of Pediatrics and Medicine, Stanford University, Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford, CA, USA
| | - Nirali N Shah
- Pediatric Oncology Branch, National Cancer Institute, USA
| | - Andre Baruchel
- Hôpital Universitaire Robert Debré (APHP) and Université de Paris, France
| | | | | | | | | | - Michael Berntgen
- Scientific Evidence Generation Department, Human Medicines Division, European Medicines Agency (EMA), Amsterdam, Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Dominik Karres
- Paediatric Medicines Office, Scientific Evidence Generation Department, Human Medicines Division, European Medicines Agency (EMA), Amsterdam, Netherlands
| | - Franca Ligas
- Paediatric Medicines Office, Scientific Evidence Generation Department, Human Medicines Division, European Medicines Agency (EMA), Amsterdam, Netherlands
| | | | | | - Lynley Marshall
- The Royal Marsden Hospital and the Institute of Cancer Research, London, UK
| | | | | | - Shannon Maude
- Children's Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, USA
| | | | - Veronique Minard-Colin
- Department of Pediatric and Adolescent Oncology, INSERM U1015, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Koen Norga
- Antwerp University Hospital, Paediatric Committee of the European Medicines Agency, Federal Agency for Medicines and Health Products, Belgium
| | | | | | | | - Rob Pieters
- Princess Maxima Center for Pediatric Oncology, Netherlands
| | | | | | | | - Martina Schüßler-Lenz
- Chair of CAT (Committee for Advanced Therapies), European Medicines Agency (EMA), Amsterdam, Netherlands; Paul-Ehrlich-Institut, Germany
| | | | | | | | - Jaroslav Sterba
- University Hospital Brno, Masaryk University, Brno, Czech Republic
| | - Sarah K Tasian
- Children's Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, USA
| | | | | | - Christian Michel Zwaan
- Princess Maxima Center for Pediatric Oncology, Netherlands; Haematological Malignancies Co-Chair Innovative Therapies for Children with Cancer Consortium (ITCC), Europe; Erasmus University Medical Center Rotterdam, Netherlands
| | - Giovanni Lesa
- Paediatric Medicines Office, Scientific Evidence Generation Department, Human Medicines Division, European Medicines Agency (EMA), Amsterdam, Netherlands
| | - Gilles Vassal
- ACCELERATE, Europe; Department of Pediatric and Adolescent Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
8
|
Sharkeev YP, Komarova EG, Chebodaeva VV, Sedelnikova MB, Zakharenko AM, Golokhvast KS, Litvinova LS, Khaziakhmatova OG, Malashchenko VV, Yurova KA, Gazatova ND, Kozlov IG, Khlusova MY, Zaitsev KV, Khlusov IA. Amorphous-Crystalline Calcium Phosphate Coating Promotes In Vitro Growth of Tumor-Derived Jurkat T Cells Activated by Anti-CD2/CD3/CD28 Antibodies. MATERIALS (BASEL, SWITZERLAND) 2021; 14:3693. [PMID: 34279263 PMCID: PMC8269898 DOI: 10.3390/ma14133693] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 12/29/2022]
Abstract
A modern trend in traumatology, orthopedics, and implantology is the development of materials and coatings with an amorphous-crystalline structure that exhibits excellent biocopatibility. The structure and physico-chemical and biological properties of calcium phosphate (CaP) coatings deposited on Ti plates using the micro-arc oxidation (MAO) method under different voltages (200, 250, and 300 V) were studied. Amorphous, nanocrystalline, and microcrystalline statesof CaHPO4 and β-Ca2P2O7 were observed in the coatings using TEM and XRD. The increase in MAO voltage resulted in augmentation of the surface roughness Ra from 2.5 to 6.5 µm, mass from 10 to 25 mg, thickness from 50 to 105 µm, and Ca/P ratio from 0.3 to 0.6. The electrical potential (EP) of the CaP coatings changed from -456 to -535 mV, while the zeta potential (ZP) decreased from -53 to -40 mV following an increase in the values of the MAO voltage. Numerous correlations of physical and chemical indices of CaP coatings were estimated. A decrease in the ZP magnitudes of CaP coatings deposited at 200-250 V was strongly associated with elevated hTERT expression in tumor-derived Jurkat T cells preliminarily activated with anti-CD2/CD3/CD28 antibodies and then contacted in vitro with CaP-coated samples for 14 days. In turn, in vitro survival of CD4+ subsets was enhanced, with proinflammatory cytokine secretion of activated Jurkat T cells. Thus, the applied MAO voltage allowed the regulation of the physicochemical properties of amorphous-crystalline CaP-coatings on Ti substrates to a certain extent. This method may be used as a technological mechanism to trigger the behavior of cells through contact with micro-arc CaP coatings. The possible role of negative ZP and Ca2+ as effectors of the biological effects of amorphous-crystalline CaP coatings is discussed. Micro-arc CaP coatings should be carefully tested to determine their suitability for use in patients with chronic lymphoid malignancies.
Collapse
Affiliation(s)
- Yurii P Sharkeev
- Laboratory of Physics of Nanostructured Biocomposites, Institute of Strength Physics and Materials Science, Siberian Branch of Russian Academy of Sciences, 634055 Tomsk, Russia
- Research School of High-Energy Physics, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Ekaterina G Komarova
- Laboratory of Physics of Nanostructured Biocomposites, Institute of Strength Physics and Materials Science, Siberian Branch of Russian Academy of Sciences, 634055 Tomsk, Russia
| | - Valentina V Chebodaeva
- Laboratory of Physics of Nanostructured Biocomposites, Institute of Strength Physics and Materials Science, Siberian Branch of Russian Academy of Sciences, 634055 Tomsk, Russia
| | - Mariya B Sedelnikova
- Laboratory of Physics of Nanostructured Biocomposites, Institute of Strength Physics and Materials Science, Siberian Branch of Russian Academy of Sciences, 634055 Tomsk, Russia
| | | | - Kirill S Golokhvast
- School of Engineering, Far Eastern Federal University, 690090 Vladivostok, Russia
| | - Larisa S Litvinova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236029 Kaliningrad, Russia
| | - Olga G Khaziakhmatova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236029 Kaliningrad, Russia
| | - Vladimir V Malashchenko
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236029 Kaliningrad, Russia
| | - Kristina A Yurova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236029 Kaliningrad, Russia
| | - Natalia D Gazatova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236029 Kaliningrad, Russia
| | - Ivan G Kozlov
- Department of Organization and Management in the Sphere of Circulation of Medicines, Institute of Postgraduate Education, I.M. Sechenov Federal State Autonomous Educational University of Higher Education-First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119991 Moscow, Russia
| | - Marina Y Khlusova
- Department of Pathophysiology, Siberian State Medical University, 634050 Tomsk, Russia
| | - Konstantin V Zaitsev
- Siberian Federal Scientific and Clinical Center of the Federal Medical-Biological Agency, 636070 Seversk, Russia
| | - Igor A Khlusov
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236029 Kaliningrad, Russia
- Research School of Chemistry and Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia
- Department of Morphology and General Pathology, Siberian State Medical University, 634050 Tomsk, Russia
| |
Collapse
|
9
|
Eldesouki RE, Wu C, Saleh FM, Mohammed EAM, Younes S, Hassan NE, Brown TC, Alt EU, Robinson JE, Badr FM, Braun SE. Identification and Targeting of Thomsen-Friedenreich and IL1RAP Antigens on Chronic Myeloid Leukemia Stem Cells Using Bi-Specific Antibodies. Onco Targets Ther 2021; 14:609-621. [PMID: 33519209 PMCID: PMC7837560 DOI: 10.2147/ott.s255299] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
Introduction Quiescent leukemia stem cells (LSCs) play a major role in therapeutic resistance and disease progression of chronic myeloid leukemia (CML). LSCs belong to the primitive population; CD34+CD38-Lin-, which does not distinguish normal hematopoietic stem cells (HSC) from CML LSCs. Because Thomsen–Friedenreich/CD176 antigen is expressed on CD34+ HSC and IL1RAP is tightly correlated to BCR-ABL expression, we sought to increase the specificity towards LSC by using additional biomarkers. Methods We evaluated the co-expression of both antigens on CD34+ peripheral blood mononuclear cells (PBMCs) from both healthy volunteers and CML patients, using flow cytometry. Then, we used site-directed mutagenesis to induce knob-in-hole mutations in the human IgG heavy chain and the human lambda light chain to generate the bi-specific antibody (Bis-Ab) TF/RAP that binds both antigens simultaneously. We measured complement-directed cytotoxicity (CDC) in CML samples with the Bis-Ab by flow cytometry. Results In contrast to healthy volunteers, CML samples displayed a highly significant co-expression of CD176 and IL1RAP. When either a double-positive cell line or CML samples were treated with increasing doses of Bis-Ab, increased binding and CDC was observed indicating co-operative binding of the Bis-Ab as compared to monoclonal antibodies. Discussion These results show that the bi-specific antibody is capable of targeting IL1RAP+ and CD176+ cell population among CML PBMCs, but not corresponding normal cells in CDC assay. We hereby offer a novel strategy for the depletion of CML stem cells from the bulk population in clinical hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Raghda E Eldesouki
- Genetics Unit, Department of Histology and Cell Biology, School of Medicine, Suez Canal University, Ismailia, Egypt.,Division of Immunology, Tulane National Primate Research Center, Covington, LA, USA
| | - Chengxiang Wu
- Division of Immunology, Tulane National Primate Research Center, Covington, LA, USA
| | - Fayez M Saleh
- Division of Immunology, Tulane National Primate Research Center, Covington, LA, USA.,Department of Medical Microbiology, Faculty of Medicine, University of Tabuk, Tabuk, Kingdom of Saudi Arabia
| | - Eman Abdel-Moemen Mohammed
- Genetics Unit, Department of Histology and Cell Biology, School of Medicine, Suez Canal University, Ismailia, Egypt
| | - Soha Younes
- Department of Clinical pathology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | | | - Theresa C Brown
- Hayward Genetics Center, Tulane University School of Medicine, New Orleans, LA, USA
| | - Eckhard U Alt
- Applied Stem Cell Laboratory, Departments of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - James E Robinson
- Sections of Infectious Disease, Departments of Pediatrics and Internal Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Fouad Mohamed Badr
- Genetics Unit, Department of Histology and Cell Biology, School of Medicine, Suez Canal University, Ismailia, Egypt
| | - Stephen E Braun
- Division of Immunology, Tulane National Primate Research Center, Covington, LA, USA.,Departments of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
10
|
Li J, Zhang Y, Cui Y, Jin H, Lin Z, Piao Y, Jin J. CD44 enhances adriamycin resistance in chronic myelogenous leukaemia cells K562. Int J Lab Hematol 2021; 43:983-989. [PMID: 33411349 DOI: 10.1111/ijlh.13455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 11/23/2020] [Accepted: 12/16/2020] [Indexed: 11/27/2022]
Abstract
INTRODUCTION To investigate CD44 effects on the adriamycin-resistant in chronic myelogenous leukaemia cells K562, we explored the role of CD44 in the K562 cells migration and apoptosis. METHODS GeneChip® screening is used for elucidating various chemoresistance-related gene expression in the adriamycin-resistant leukaemia cells K562/ADR. We constructed K562/CD44 cells by transfection of an EGFP-SV40-CD44 plasmid, and adriamycin-resistant ability was confirmed by detecting migration and apoptosis-related proteins and mRNA expression using Western blotting and Real-time PCR respectively. RESULTS K562/CD44 cells were generated by the transfection of an EGFP-SV40-CD44 plasmid with high CD44 expression. mRNA expression levels of CD44 and P-glycoprotein (P-gp), along with the proliferation rate, were increased, while the apoptosis rate of K562/CD44 cells was decreased. Migration-associated proteins such as MMP-2 and MMP-9 were upregulated, whereas apoptosis-related protein Bax was downregulated and Bcl-2 protein was not significantly altered in the K562/CD44 cells. CONCLUSIONS CD44 might be involved in adriamycin resistance via regulation of P-gp, MMP-2, MMP-9, and Bcl-2/Bax.
Collapse
Affiliation(s)
- Juan Li
- Department of Internal Medicine, The Affiliated Hospital of Yanbian University, Yanji, China.,Department of Rheumatology and Immunology, The First Affiliated Hospital of Luohe Medical College (Luohe Central Hospital), Luohe, China
| | - Yanfang Zhang
- Department of Internal Medicine, The Affiliated Hospital of Yanbian University, Yanji, China.,Department of Rheumatology and Immunology, Zhejiang Hospital, Hangzhou, China
| | - Yubo Cui
- Department of Internal Medicine, The Affiliated Hospital of Yanbian University, Yanji, China.,Department of Respiratory, Yanbian No. 2 People's Hospital, Yanji, China
| | - Honghua Jin
- Department of Internal Medicine, The Affiliated Hospital of Yanbian University, Yanji, China
| | - Zhenhua Lin
- Department of Internal Medicine, The Affiliated Hospital of Yanbian University, Yanji, China.,Key Laboratory of the Science and Technology Department (Jilin Province), Cancer Research Center, Yanbian University Medical College, Yanji, China
| | - Yingshi Piao
- Key Laboratory of the Science and Technology Department (Jilin Province), Cancer Research Center, Yanbian University Medical College, Yanji, China
| | - Jingchun Jin
- Department of Internal Medicine, The Affiliated Hospital of Yanbian University, Yanji, China.,Key Laboratory of the Science and Technology Department (Jilin Province), Cancer Research Center, Yanbian University Medical College, Yanji, China
| |
Collapse
|
11
|
Krüger T, Middeke JM, Stölzel F, Mütherig A, List C, Brandt K, Heidrich K, Teipel R, Ordemann R, Schuler U, Oelschlägel U, Wermke M, Kräter M, Herbig M, Wehner R, Schmitz M, Bornhäuser M, von Bonin M. Reliable isolation of human mesenchymal stromal cells from bone marrow biopsy specimens in patients after allogeneic hematopoietic cell transplantation. Cytotherapy 2019; 22:21-26. [PMID: 31883948 DOI: 10.1016/j.jcyt.2019.10.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/25/2019] [Accepted: 10/28/2019] [Indexed: 12/28/2022]
Abstract
Isolation of mesenchymal stromal cells (MSCs) from pretreated, hematologic patients is challenging. Especially after allogeneic hematopoietic cell transplantation (HCT), standard protocols using bone marrow aspirates fail to reliably recover sufficient cell numbers. Because MSCs are considered to contribute to processes that mainly affect the outcome after transplantation, such as an efficient lymphohematopoietic recovery, extent of graft-versus-host disease as well as the occurrence of leukemic relapse, it is of great clinical relevance to investigate MSC function in this context. Previous studies showed that MSCs can be isolated by collagenase digestion of large bone fragments of hematologically healthy patients undergoing hip replacement or knee surgeries. We have now further developed this procedure for the isolation of MSCs from hematologic patients after allogeneic HCT by using trephine biopsy specimens obtained during routine examinations. Comparison of aspirates and trephine biopsy specimens from patients after allogeneic HCT revealed a significantly higher frequency of clonogenic MSCs (colony-forming unit-fibroblast [CFU-F]) in trephine biopsy specimens (mean, 289.8 ± standard deviation 322.5 CFU-F colonies/1 × 106 total nucleated cells versus 4.2 ± 9.9; P < 0.0001). Subsequent expansion of functional MSCs isolated from trephine biopsy specimen was more robust and led to a significantly higher yield compared with control samples expanded from aspirates (median, 1.6 × 106; range, 0-2.3 × 107 P0 MSCs versus 5.4 × 104; range, 0-8.9 × 106; P < 0.0001). Using trephine biopsy specimens as MSC source facilitates the investigation of various clinical questions.
Collapse
Affiliation(s)
- Thomas Krüger
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Jan Moritz Middeke
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Friedrich Stölzel
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Anke Mütherig
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Catrin List
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Kalina Brandt
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Katharina Heidrich
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Raphael Teipel
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Rainer Ordemann
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Ulrich Schuler
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Uta Oelschlägel
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Martin Wermke
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany; University Cancer Centrum (UCC), Early Clinical Trial Unit (ECTU), University Hospital Carl Gustav Carus, Dresden, Germany
| | - Martin Kräter
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Maik Herbig
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany; Biotechnology Center, Center for Molecular and Cellular Bioengineering TU Dresden Tatzberg 47-49, Dresden, Germany
| | - Rebekka Wehner
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
| | - Marc Schmitz
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany; Center for Regenerative Therapies (CRTD), Dresden, Germany
| | - Martin Bornhäuser
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany; Center for Regenerative Therapies (CRTD), Dresden, Germany
| | - Malte von Bonin
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
12
|
Brennan L, Narendran A. Cancer Stem Cells in the Development of Novel Therapeutics for Refractory Pediatric Leukemia. Stem Cells Dev 2019; 28:1277-1287. [PMID: 31364487 DOI: 10.1089/scd.2019.0035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Although treatment strategies for pediatric leukemia have improved overall survival rates in the recent past, relapse rates in certain subgroups such as infant leukemia remain unacceptably high. Despite undergoing extensive chemotherapy designed to target the rapidly proliferating leukemia cells, many of these children experience relapse. In refractory leukemia, the existence of cell populations with stemness characteristics, termed leukemia stem cells (LSCs), which remain quiescent and subsequently replenish the blast population, has been described. A significant body of evidence exists, derived largely from xenograft models of adult acute myeloid leukemia, to support the idea that LSCs may play a fundamental role in refractory disease. In addition, clinical studies have also linked LSCs with increased minimal residual disease, higher relapse rate, and decreased survival rates in these patients. Recently, a number of reports have addressed effective ways to utilize new-generation genomic sequencing and transcriptomic analyses to identify targeted therapeutic agents aimed at LSCs, while sparing normal hematopoietic stem cells. These data underscore the value of timely translation of knowledge from adult studies to the unique molecular and physiological characteristics seen in pediatric leukemia. We aim to summarize this article in the rapidly expanding field of stem cell biology in hematopoietic malignancies, focusing particularly on relevant preclinical models and novel targeted therapeutics, and their applicability to childhood leukemia.
Collapse
Affiliation(s)
| | - Aru Narendran
- Division of Pediatric Hematology, Oncology and Transplant, POETIC Laboratory for Novel Therapeutics Discovery in Pediatric Oncology, Alberta Children's Hospital, Calgary, Canada
| |
Collapse
|
13
|
Yurova KA, Khaziakhmatova OG, Melashchenko ES, Malashchenko VV, Shunkin EO, Shupletsova VV, Ivanov PA, Khlusov IA, Litvinova LS. Cellular and Molecular Basis of Osteoblastic and Vascular Niches in the Processes of Hematopoiesis and Bone Remodeling (A Short Review of Modern Views). Curr Pharm Des 2019; 25:663-669. [PMID: 30931856 DOI: 10.2174/1381612825666190329153626] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 03/25/2019] [Indexed: 01/01/2023]
Abstract
In evolutionary processes, human bone marrow has formed as an organ depot of various types of cells that arise from hematopoietic stem cells (HSCs) and mesenchymal stem cells (MSCs). Vital HSC activity is controlled through molecular interactions with the niche microenvironment. The review describes current views on the formation of key molecular and cellular components of the HSC niche, which ensure maintenance of home ostasis in stem cell niches, obtained from studies of their role in regulating the proliferation and differentiation of HSCs, including the physiological, reparative and pathological remodeling of bone tissue. Due to rapid developments in biotechnology, tissue bioengineering, and regenerative medicine, information can be useful for developing biomimetic and bioinspired materials and implants that provide an effective bone/bone marrow recovery process after injuries and, to a greater extent, diseases of various etiologies.
Collapse
Affiliation(s)
- Kristina A Yurova
- Basic Laboratory of Immunology and Cell Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
| | - Olga G Khaziakhmatova
- Basic Laboratory of Immunology and Cell Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
| | - Elena S Melashchenko
- Center for Medical Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
| | - Vladimir V Malashchenko
- Center for Medical Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
| | - Egor O Shunkin
- Center for Medical Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
| | - Valeria V Shupletsova
- Basic Laboratory of Immunology and Cell Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation.,Center for Medical Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
| | - Pavel A Ivanov
- Basic Laboratory of Immunology and Cell Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
| | - Igor A Khlusov
- Morphology and General Pathology Department, Siberian State Medical University, Tomsk, Russian Federation.,Research School of Chemistry & Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk, Tomskaya oblast, Russian Federation
| | - Larisa S Litvinova
- Basic Laboratory of Immunology and Cell Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russian Federation
| |
Collapse
|
14
|
Leukemia Stem Cells in the Pathogenesis, Progression, and Treatment of Acute Myeloid Leukemia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1143:95-128. [DOI: 10.1007/978-981-13-7342-8_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|