1
|
Li L, Xu D, Huang X. SERCA-mediated endoplasmic reticulum stress facilitates hematopoietic stem cell mobilization. Stem Cell Res Ther 2025; 16:208. [PMID: 40275396 PMCID: PMC12023629 DOI: 10.1186/s13287-025-04345-y] [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: 10/22/2024] [Accepted: 04/15/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Hematopoietic stem cell (HSC) transplantation is widely recognized as an effective treatment for various malignant diseases. Enhancing HSC mobilization can improve transplantation outcomes and ultimately increase patient survival rates. Recent studies suggest that mild endoplasmic reticulum (ER) stress promotes HSC self-renewal, anti-apoptotic, and anti-aging capabilities. This led us to investigate whether inducing mild ER stress could facilitate HSC mobilization. METHODS The phenotype changes in cells treated with ER stress inducers and Sarco/endoplasmic reticulum Ca²⁺-ATPase (SERCA) inhibitors were assessed using flow cytometry. The efficacy of these agents on HSC mobilization was evaluated in C57Bl/6 mice, with colony forming unit (CFU) assays used for quantification. Knockdown Jurkat cell lines were constructed to validate the role of SERCA in the mobilization mechanism. Molecular and protein expression levels associated with the pathway were analyzed through quantitative reverse-transcription PCR and western blotting. RESULTS Our findings revealed that BHQ, a SERCA inhibitor, efficiently enhanced HSC mobilization in vivo. Mechanistically, BHQ regulated the CaMKII-STAT3-CXCR4 pathway by suppressing SERCA activity. This inhibition led to a reduction in CXCR4 expression on the surface of HSCs, facilitating their migration from the bone marrow into peripheral circulation. CONCLUSIONS Our study provides novel insights into the role of the SERCA-ER stress pathway in HSC mobilization. By targeting SERCA activity with BHQ, we observed a significant enhancement in the mobilization of HSCs, facilitated by the modulation of the CaMKII-STAT3-CXCR4 signaling pathway. This research highlights the potential of utilizing mild ER stress as a strategy to promote HSC mobilization, with significant implications for improving stem cell-based therapies, including those used in HSC transplantation.
Collapse
Affiliation(s)
- Lijun Li
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, and The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Danhua Xu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, and The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Xinxin Huang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, and The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
2
|
Jantunen E, Turunen A, Partanen A. Autograft composition and outcome-towards an optimal graft? Cytotherapy 2025; 27:493-499. [PMID: 39755979 DOI: 10.1016/j.jcyt.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/12/2024] [Accepted: 12/20/2024] [Indexed: 01/07/2025]
Abstract
The amount of CD34+ cells has been for decades the most important marker of autologous graft quality, but other graft cells, including various lymphocyte subsets, have gained some interest. This review attempts to summarize what is known about autograft cellular composition regarding post-transplant outcomes. The amount of CD34+ cells in the graft is associated with tempo of platelet recovery. It also has been associated with improved progression-free (PFS) and overall survival (OS) in many studies in patients with non-Hodgkin lymphoma and in some studies in patients with multiple myeloma. A greater number of lymphocytes in the graft has been linked with earlier lymphocyte recovery, which on the other hand has been associated with better post-transplant outcomes. In prospective studies, a greater number of T lymphocytes has been found to correlate with better PFS and OS in patients with non-Hodgkin lymphoma and multiple myeloma. Some studies also indicate that the number of natural killer cells in grafts is prognostically important. At present, it is challenging to define a so-called optimal graft in the autologous setting. In addition to adequate CD34+ cell counts, more lymphocytes also should be collected to achieve immune autografts, which may translate to improved patient outcomes. More data are needed regarding the functional status of various lymphocyte subset for post-transplant outcomes.
Collapse
Affiliation(s)
- Esa Jantunen
- Clinical Medicine/University of Eastern Finland, Kuopio, Finland; Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Antti Turunen
- Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Anu Partanen
- Department of Medicine, Kuopio University Hospital, Kuopio, Finland.
| |
Collapse
|
3
|
Wu M, Koester DC, Walkinshaw G, Ng D, Zhou X, Ho A, Tsao J, Barnes M, Brenner MC, Spong S, Nelson G, Gervasi DC, Vaisberg E, Sternlicht M, Sidhu P, Lin J, Ibrahim M, Thompson MD, Chou J, Pangilinan G, Makwana O, Wei Z, Signore PE, Del Balzo U, Hoch U, Ramurthy S. Discovery of Novel, Potent, Orally Bioavailable and Efficacious, Hypoxia-Inducible Factor Prolyl Hydroxylase Inhibitors for Hematopoietic Stem Cell Mobilization. J Med Chem 2025; 68:6386-6406. [PMID: 40047531 DOI: 10.1021/acs.jmedchem.4c02889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Hematopoietic stem cell (HSC) mobilization is often difficult to achieve in patients suffering from multiple myeloma and non-Hodgkin's lymphoma. Granulocyte-colony stimulating factor (G-CSF) therapy alone has often not led to the desired outcomes. Herein, we describe the discovery of 7-cyclohexyl-4-hydroxy-8-oxo-N-(pyridazin-4-ylmethyl)-7,8-dihydro-2,7-naphthyridine-3-carboxamide 13, a hypoxia-inducible factor prolyl hydroxylase (HIF-PH) inhibitor, which was discovered by focusing on drug-like properties. Building on a previous discovery that HIF-PH inhibitors can enhance HSC mobilization in combination with G-CSF, we optimized 13 to exhibit high PHD2 potency, improved solubility, and an optimized PK profile. 13 was effective at enhancing G-CSF-induced HSC mobilization in mice at a dose of 2 mg/kg.
Collapse
Affiliation(s)
- Min Wu
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Dennis C Koester
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Gail Walkinshaw
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Danny Ng
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Xiaoti Zhou
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Angel Ho
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Jenny Tsao
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Michael Barnes
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Mitchell C Brenner
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Suzanne Spong
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Grace Nelson
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - David C Gervasi
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Elena Vaisberg
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Mark Sternlicht
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Parmjeet Sidhu
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Jack Lin
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Mohamed Ibrahim
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Michael D Thompson
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - James Chou
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Gerardo Pangilinan
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Om Makwana
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Zhihua Wei
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Pierre E Signore
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Ughetta Del Balzo
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Ute Hoch
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Savithri Ramurthy
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| |
Collapse
|
4
|
Xiao W, Jiang W, Chen Z, Huang Y, Mao J, Zheng W, Hu Y, Shi J. Advance in peptide-based drug development: delivery platforms, therapeutics and vaccines. Signal Transduct Target Ther 2025; 10:74. [PMID: 40038239 DOI: 10.1038/s41392-024-02107-5] [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: 03/07/2024] [Revised: 11/01/2024] [Accepted: 12/13/2024] [Indexed: 03/06/2025] Open
Abstract
The successful approval of peptide-based drugs can be attributed to a collaborative effort across multiple disciplines. The integration of novel drug design and synthesis techniques, display library technology, delivery systems, bioengineering advancements, and artificial intelligence have significantly expedited the development of groundbreaking peptide-based drugs, effectively addressing the obstacles associated with their character, such as the rapid clearance and degradation, necessitating subcutaneous injection leading to increasing patient discomfort, and ultimately advancing translational research efforts. Peptides are presently employed in the management and diagnosis of a diverse array of medical conditions, such as diabetes mellitus, weight loss, oncology, and rare diseases, and are additionally garnering interest in facilitating targeted drug delivery platforms and the advancement of peptide-based vaccines. This paper provides an overview of the present market and clinical trial progress of peptide-based therapeutics, delivery platforms, and vaccines. It examines the key areas of research in peptide-based drug development through a literature analysis and emphasizes the structural modification principles of peptide-based drugs, as well as the recent advancements in screening, design, and delivery technologies. The accelerated advancement in the development of novel peptide-based therapeutics, including peptide-drug complexes, new peptide-based vaccines, and innovative peptide-based diagnostic reagents, has the potential to promote the era of precise customization of disease therapeutic schedule.
Collapse
Affiliation(s)
- Wenjing Xiao
- Department of Pharmacy, The General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Wenjie Jiang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Zheng Chen
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yu Huang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Junyi Mao
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Wei Zheng
- Department of Integrative Medicine, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Yonghe Hu
- School of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
5
|
Khan S, Bergstrom DJ, Côté J, Kotb R, LeBlanc R, Louzada ML, Mian HS, Othman I, Colasurdo G, Visram A. First Line Treatment of Newly Diagnosed Transplant Eligible Multiple Myeloma Recommendations From a Canadian Consensus Guideline Consortium. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2025; 25:e151-e172. [PMID: 39567294 DOI: 10.1016/j.clml.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/02/2024] [Accepted: 10/17/2024] [Indexed: 11/22/2024]
Abstract
The availability of effective therapies for multiple myeloma (MM) has sparked debate on the role of first line autologous stem cell transplantation (ASCT), particularly in standard-risk patients. However, treatment for individuals with high-risk disease continues to display suboptimal outcomes. With novel therapies used earlier, practice is changing rapidly in the field of MM. Presently, quadruplet induction therapy incorporating an anti-CD38 monoclonal antibody to a proteasome inhibitor and an immunomodulatory drug prior to ASCT followed by maintenance therapy stands as the foremost strategy for attaining deep and sustained responses in transplant eligible MM (TEMM). This Canadian Consensus Guideline Consortium (CGC) proposes consensus recommendations for the first line treatment of TEMM. To address the needs of physicians and people diagnosed with MM, this document focuses on ASCT eligibility, induction therapy, mobilization and collection, conditioning, consolidation, and maintenance therapy, as well as, high-risk populations, management of adverse events, assessment of treatment response, and monitoring for disease relapse. The CGC will periodically review the recommendations herein and update as necessary.
Collapse
Affiliation(s)
- Sahar Khan
- Windsor Regional Hospital, University of Western Ontario, Windsor, Ontario, Canada.
| | - Debra J Bergstrom
- Division of Hematology, Memorial University of Newfoundland, Newfoundland and Labrador, Canada
| | - Julie Côté
- Centre Hospitalier Universitaire de Québec, Quebec, Quebec, Canada
| | - Rami Kotb
- Department of Medical Oncology and Hematology, CancerCare Manitoba, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Richard LeBlanc
- Hôpital Maisonneuve-Rosemont, University of Montreal, Montreal, Quebec, Canada
| | - Martha L Louzada
- London Health Sciences Centre, Western University, London, Ontario, Canada
| | - Hira S Mian
- Juravinski Cancer Centre, McMaster University, Hamilton, Ontario, Canada
| | - Ibraheem Othman
- Allan Blair Cancer Centre, University of Saskatchewan, Regina, Saskatchewan, Canada
| | | | - Alissa Visram
- The Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
6
|
Sharma A. How I treat sickle cell disease with gene therapy. Blood 2024; 144:2693-2705. [PMID: 39356871 PMCID: PMC11830977 DOI: 10.1182/blood.2024024519] [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: 06/20/2024] [Revised: 09/03/2024] [Accepted: 09/08/2024] [Indexed: 10/04/2024] Open
Abstract
ABSTRACT In 2023, 2 different gene therapies were approved for individuals with severe sickle cell disease (SCD). The small number of patients treated on the pivotal clinical trials that led to these approvals have experienced dramatic short-term reductions in the occurrence of painful vaso-occlusive crises, but the long-term safety and efficacy of these genetic therapies are yet to be ascertained. Several challenges and treatment-related concerns have emerged in regard to administering these therapies in clinical practice. This article discusses the selection and preparation of individuals with SCD who wish to receive autologous gene therapy, as well as the salient features of the care needed to support them through a long and arduous treatment process. I specifically focus on postinfusion care, as it relates to immune monitoring and infection prevention. Compared with allogeneic hematopoietic cell transplantation, delivering autologous gene therapy to an individual with SCD has distinct nuances that require awareness and special interventions. Using clinical vignettes derived from real-life patients, I provide perspectives on the complex decision-making process for gene therapy for SCD based on currently available data and make recommendations for evaluating and supporting these patients.
Collapse
Affiliation(s)
- Akshay Sharma
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN
| |
Collapse
|
7
|
Liu Y, Geng N, Huang X. Molecular regulators of chemotaxis in human hematopoietic stem cells. Biochem Soc Trans 2024; 52:2427-2437. [PMID: 39584478 DOI: 10.1042/bst20240288] [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: 07/26/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 11/26/2024]
Abstract
Hematopoietic stem cells (HSCs), essential for lifelong blood cell regeneration, are clinically utilized to treat various hematological disorders. These cells originate in the aorta-gonad-mesonephros region, expand in the fetal liver, and mature in the bone marrow. Chemotaxis, involving gradient sensing, polarization, and migration, directs HSCs and is crucial for their homing and mobilization. The molecular regulation of HSC chemotaxis involves chemokines, chemokine receptors, signaling pathways, and cytoskeletal proteins. Recent advances in understanding these regulatory mechanisms have deepened insights into HSC development and hematopoiesis, offering new avenues for therapeutic innovations. Strategies including glucocorticoid receptor activation, modulation of histone acetylation, stimulation of nitric oxide signaling, and interference with m6A RNA modification have shown potential in enhancing CXCR4 expression, thereby improving the chemotactic response and homing capabilities of human HSCs. This review synthesizes current knowledge on the molecular regulation of human HSC chemotaxis and its implications for health and disease.
Collapse
Affiliation(s)
- Yining Liu
- Zhongshan-Xuhui Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Nanxi Geng
- Zhongshan-Xuhui Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Xinxin Huang
- Zhongshan-Xuhui Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
8
|
Sidana S, Bankova AK, Hosoya H, Kumar SK, Holmes TH, Tamaresis J, Le A, Muffly LS, Maysel-Auslender S, Johnston L, Arai S, Lowsky R, Meyer E, Rezvani A, Weng WK, Frank MJ, Shiraz P, Maecker HT, Lu Y, Miklos DB, Shizuru JA. Phase II study of novel CXCR2 agonist and Plerixafor for rapid stem cell mobilization in patients with multiple myeloma. Blood Cancer J 2024; 14:173. [PMID: 39384609 PMCID: PMC11464886 DOI: 10.1038/s41408-024-01152-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 09/13/2024] [Accepted: 09/23/2024] [Indexed: 10/11/2024] Open
Abstract
MGTA-145 or GROβT, a CXCR2 agonist, has shown promising activity for hematopoietic stem cell (HSC) mobilization with plerixafor in pre-clinical studies and healthy volunteers. Twenty-five patients with multiple myeloma enrolled in a phase 2 trial evaluating MGTA-145 and plerixafor for HSC mobilization (NCT04552743). Plerixafor was given subcutaneously followed 2 h later by MGTA-145 (0.03 mg/kg) intravenously with same day apheresis. Mobilization/apheresis could be repeated for a second day in patients who collected <6 ×106 CD34+ cells/kg. Lenalidomide and anti-CD38 antibody were part of induction therapy in 92% (n = 23) and 24% (n = 6) of patients, respectively. Median total HSC cell yield (CD34+ cells/kg × 106) was 5.0 (range: 1.1-16.2) and day 1 yield was 3.4 (range: 0.3-16.2). 88% (n = 22) of patients met the primary endpoint of collecting 2 ×106 CD34+ cells/kg in ≤ two days, 68% (n = 17) in one day. Secondary endpoints of collecting 4 and 6 × 106 CD34+ cells/kg in ≤ two days were met in 68% (n = 17) and 40% (n = 10) patients. Grade 1 or 2 adverse events (AE) were seen in 60% of patients, the most common AE being grade 1 pain, usually self-limited. All 19 patients who underwent transplant with MGTA-145 and plerixafor mobilized HSCs engrafted successfully, with durable engraftment at day 100. 74% (17 of 23) of grafts with this regimen were minimal residual disease negative by next generation flow cytometry. Graft composition for HSCs and immune cells were similar to a contemporaneous cohort mobilized with G-CSF and plerixafor.
Collapse
Affiliation(s)
- Surbhi Sidana
- Division of BMT and Cell Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, USA.
| | - Andriyana K Bankova
- Division of BMT and Cell Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, USA
- National Specialized Hospital for Hematological Diseases, Sofia, Bulgaria
| | - Hitomi Hosoya
- Division of BMT and Cell Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, USA
| | - Shaji K Kumar
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Tyson H Holmes
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, USA
| | - John Tamaresis
- Department of Biostatistics, Stanford University School of Medicine, Stanford, USA
| | - Anne Le
- Division of BMT and Cell Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, USA
- Jasper Therapeutics, Stanford, USA
| | - Lori S Muffly
- Division of BMT and Cell Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, USA
| | - Sofia Maysel-Auslender
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, USA
| | - Laura Johnston
- Division of BMT and Cell Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, USA
| | - Sally Arai
- Division of BMT and Cell Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, USA
| | - Robert Lowsky
- Division of BMT and Cell Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, USA
| | - Everett Meyer
- Division of BMT and Cell Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, USA
| | - Andrew Rezvani
- Division of BMT and Cell Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, USA
| | - Wen-Kai Weng
- Division of BMT and Cell Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, USA
| | - Matthew J Frank
- Division of BMT and Cell Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, USA
| | - Parveen Shiraz
- Division of BMT and Cell Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, USA
| | - Holden T Maecker
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, USA
| | - Ying Lu
- Department of Biostatistics, Stanford University School of Medicine, Stanford, USA
| | - David B Miklos
- Division of BMT and Cell Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, USA
| | - Judith A Shizuru
- Division of BMT and Cell Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, USA
| |
Collapse
|
9
|
Oshi M, Wu R, Khoury T, Gandhi S, Yan L, Yamada A, Ishikawa T, Endo I, Takabe K. Infiltration of Common Myeloid Progenitor (CMP) Cells is Associated With Less Aggressive Tumor Biology, Lower Risk of Brain Metastasis, Better Response to Immunotherapy, and Higher Patient Survival in Breast Cancer. Ann Surg 2024; 280:557-569. [PMID: 38946549 PMCID: PMC11797412 DOI: 10.1097/sla.0000000000006428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
OBJECTIVE To investigate the clinical relevance of common myeloid progenitor (CMP) cells in breast tumor microenvironment (TME). BACKGROUND The role of rare cells in TME is less studied. In Silico transcriptomic analyses of real-world data enable us to detect and quantify rare cells, including CMP cells. METHODS A total of 5176 breast cancer (BC) patients from SCAN-B, METABRIC, and 5 single-cell sequence cohorts were analyzed using the xCell algorithm. The high group was defined as more than two-thirds of the CMP scores in each cohort. RESULTS CMP cells consist of 0.07% to 0.25% of bulk breast tumor cells, more in estrogen receptor-positive (ER+) compared with triple-negative (TN) subtype (0.1% to 0.75%, 0.18% to 0.33% of immune cells, respectively). CMP cells did not correlate with any of the myeloid lineages or stem cells in TME. CMP infiltration was higher in smaller tumors, with lower Nottingham grade, and in ER+/HER2- than in TNBC consistently in both SCAN-B and METABRIC cohorts. High CMP was significantly associated with a lower risk of brain metastasis and with better survival, particularly in ER+/HER2-. High CMP enriched epithelial-to-mesenchymal transition and angiogenesis pathways, and less cell proliferation and DNA repair gene sets. High CMP ER+/HER2- was associated with less immune cell infiltration and cytolytic activity ( P <0.001). CMP infiltration correlated with neoadjuvant chemoimmunotherapy response for both ER+/HER2- and TNBC in the ISPY-2 cohort (AUC=0.69 and 0.74, respectively). CONCLUSIONS CMP in BC is inversely associated with cell proliferation and brain metastasis, better response to immunotherapy, and survival. This is the first to report the clinical relevance of CMP infiltration in BC.
Collapse
Affiliation(s)
- Masanori Oshi
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Rongrong Wu
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo 160-8402, Japan
| | - Thaer Khoury
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Shipra Gandhi
- Department of Medical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Li Yan
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Akimitsu Yamada
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Takashi Ishikawa
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo 160-8402, Japan
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Kazuaki Takabe
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa 236-0004, Japan
- Department of Surgery, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY 14263, USA
- Department of Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- Department of Breast Surgery, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
10
|
Yang J, Tian E, Chen L, Liu Z, Ren Y, Mao W, Zhang Y, Zhang J. Development and therapeutic perspectives of CXCR4 antagonists for disease therapy. Eur J Med Chem 2024; 275:116594. [PMID: 38879970 DOI: 10.1016/j.ejmech.2024.116594] [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: 04/07/2024] [Revised: 06/10/2024] [Accepted: 06/10/2024] [Indexed: 06/18/2024]
Abstract
Chemokine receptor 4 (CXCR4) is a subtype receptor protein of the GPCR family with a seven-transmembrane structure widely distributed in human tissues. CXCR4 is involved in diseases (e.g., HIV-1 infection), cancer proliferation and metastasis, inflammation signaling pathways, and leukemia, making it a promising drug target. Clinical trials on CXCR4 antagonists mainly focused on peptides and antibodies, with a few small molecule compounds, such as AMD11070 (2) and MSX-122 (3), showing promise in cancer treatment. This perspective discusses the structure-activity relationship (SAR) of CXCR4 and its role in diseases, mainly focusing on the SAR of CXCR4 antagonists. It also explores the standard structural features and target interactions of CXCR4 binding in different disease categories. Furthermore, it investigates various modification strategies to propose potential improvements in the effectiveness of CXCR4 drugs.
Collapse
Affiliation(s)
- Jun Yang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center and Institute of Respiratory Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Erkang Tian
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Li Chen
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center and Institute of Respiratory Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zihang Liu
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center and Institute of Respiratory Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yijiu Ren
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Wuyu Mao
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center and Institute of Respiratory Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Yiwen Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center and Institute of Respiratory Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Jifa Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center and Institute of Respiratory Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
11
|
Butt H, Tisdale JF. Gene therapies on the horizon for sickle cell disease: a clinician's perspective. Expert Rev Hematol 2024; 17:555-566. [PMID: 39076056 DOI: 10.1080/17474086.2024.2386366] [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: 04/12/2024] [Revised: 06/20/2024] [Accepted: 07/26/2024] [Indexed: 07/31/2024]
Abstract
INTRODUCTION Sickle cell disease (SCD) is a monogenic disorder that exerts several detrimental health effects on those affected, ultimately resulting in significant morbidity and early mortality. There are millions of individuals globally impacted by this disease. Research in gene therapy has been growing significantly over the past decade, now with two FDA approved products, aiming to find another cure for this complex disease. AREAS COVERED This perspective article aims to provide a clinician's insight into the current landscape of gene therapies, exploring the novel approaches, clinical advances, and potential impact on the management and prognosis of SCD. A comprehensive literature search encompassing databases such as PubMed, Web of Science and Google Scholar was employed. The search covered literature published from 1980 to 2024, focusing on SCD and curative therapy. EXPERT OPINION After careful evaluation of the risks and benefits associated with the use of gene therapy for affected patients, the need for a cure outweighs the risks associated with treatment in most cases of SCD. With advances in current technologies, gene therapies can increase access to cures for patients with SCD.
Collapse
Affiliation(s)
- Henna Butt
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Center for Cancer and Blood Disorders, Children's National Hospital, Washington, DC, USA
| | - John F Tisdale
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
12
|
Dietz L, Marini J, Hashmi H, McGann M. A comparative review of Aphexda and Mozobil for mobilization prior to stem cell collection. J Oncol Pharm Pract 2024; 30:1073-1077. [PMID: 38629183 DOI: 10.1177/10781552241247472] [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] [Indexed: 10/12/2024]
Abstract
High-dose chemotherapy followed by autologous hematopoietic stem cell transplantation is the standard of care for eligible patients with newly diagnosed multiple myeloma leading to prolonged progression free and overall survival. Successful engraftment following hematopoietic stem cell infusion requires adequate stem cell doses. Current mobilization regimens include granulocyte colony-stimulating factor (G-CSF) with or without plerixafor. Motixafortide is a recently approved agent that can be used in combination with G-CSF for mobilization. In the absence of any head-to-head trials comparing the two products, this article aims to outline the similarities and differences of these two agents. Though moxitafortide has a more favorable pharmacokinetic profile in comparison to plerixafor, in clinical trials, the agents demonstrated similar efficacy. In addition, the use of motixafortide in clinical practice may be limited by product cost as well as administration and monitoring requirements.
Collapse
Affiliation(s)
- Lauren Dietz
- Department of Pharmacy, Medical University of South Carolina Health, Charleston, SC, US
| | - Jessica Marini
- Department of Pharmacy, Medical University of South Carolina Health, Charleston, SC, US
| | - Hamza Hashmi
- Division of Hematology Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY, US
| | - Mary McGann
- Department of Pharmacy, Medical University of South Carolina Health, Charleston, SC, US
| |
Collapse
|
13
|
Ruminski PG, Rettig MP, DiPersio JF. Development of VLA4 and CXCR4 Antagonists for the Mobilization of Hematopoietic Stem and Progenitor Cells. Biomolecules 2024; 14:1003. [PMID: 39199390 PMCID: PMC11353233 DOI: 10.3390/biom14081003] [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: 06/28/2024] [Revised: 07/29/2024] [Accepted: 08/02/2024] [Indexed: 09/01/2024] Open
Abstract
The treatment of patients diagnosed with hematologic malignancies typically includes hematopoietic stem cell transplantation (HSCT) as part of a therapeutic standard of care. The primary graft source of hematopoietic stem and progenitor cells (HSPCs) for HSCT is mobilized from the bone marrow into the peripheral blood of allogeneic donors or patients. More recently, these mobilized HSPCs have also been the source for gene editing strategies to treat diseases such as sickle-cell anemia. For a HSCT to be successful, it requires the infusion of a sufficient number of HSPCs that are capable of adequate homing to the bone marrow niche and the subsequent regeneration of stable trilineage hematopoiesis in a timely manner. Granulocyte-colony-stimulating factor (G-CSF) is currently the most frequently used agent for HSPC mobilization. However, it requires five or more daily infusions to produce an adequate number of HSPCs and the use of G-CSF alone often results in suboptimal stem cell yields in a significant number of patients. Furthermore, there are several undesirable side effects associated with G-CSF, and it is contraindicated for use in sickle-cell anemia patients, where it has been linked to serious vaso-occlusive and thrombotic events. The chemokine receptor CXCR4 and the cell surface integrin α4β1 (very late antigen 4 (VLA4)) are both involved in the homing and retention of HSPCs within the bone marrow microenvironment. Preclinical and/or clinical studies have shown that targeted disruption of the interaction of the CXCR4 or VLA4 receptors with their endogenous ligands within the bone marrow niche results in the rapid and reversible mobilization of HSPCs into the peripheral circulation and is synergistic when combined with G-CSF. In this review, we discuss the roles CXCR4 and VLA4 play in bone marrow homing and retention and will summarize more recent development of small-molecule CXCR4 and VLA4 inhibitors that, when combined, can synergistically improve the magnitude, quality and convenience of HSPC mobilization for stem cell transplantation and ex vivo gene therapy after the administration of just a single dose. This optimized regimen has the potential to afford a superior alternative to G-CSF for HSPC mobilization.
Collapse
Affiliation(s)
| | | | - John F. DiPersio
- Division of Oncology, Department of Medicine, Washington University School of Medicine, 660 S. Euclid Ave., St Louis, MO 63105, USA
| |
Collapse
|
14
|
Zhu Z, Li X, Yuan X, Chen X, Lin T, Guo X, Li N. Efficacy and safety of stem cell mobilization with etoposide +cytarabine plus G-CSF in poor mobilizers with relapsed or refractory lymphoma. Front Immunol 2024; 15:1439253. [PMID: 39091501 PMCID: PMC11291192 DOI: 10.3389/fimmu.2024.1439253] [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: 05/27/2024] [Accepted: 07/08/2024] [Indexed: 08/04/2024] Open
Abstract
Background Autologous stem cell transplantation (ASCT) is a potentially curative strategy for relapse or refractory(r/r) aggressive lymphoma. However, a proportion of lymphoma patients who are at high risk of mobilization failure fail to mobilize stem cells and cannot proceed to ASCT. The aim of this study is to explore the efficacy and safety of Etoposide combined with Cytarabine (EA) plus G-CSF mobilization in poor mobilizers (PMs) with r/r aggressive lymphoma. Methods This retrospective study analyzed the outcomes of chemo-mobilization based on EA (Etoposide 0.1 g/m2, qd d1~3; AraC 0.5 g/m2, q12h d1~3) in 98 patients with r/r aggressive lymphoma. Of these, 39 patients met the criteria for predicted PMs as proposed by the Gruppo Italiano Trapianto di Midollo Osseo working group. Results Of the 39 PMs, 38(97.4%) patents harvested adequate mobilization (≥2×106 CD34+ cells/kg), while 31(79.5%) patients achieved optimal mobilization (≥5×106 CD34+ cells/kg). Overall, the mean number of CD34+ cells/kg collected was 17.99(range: 1.08~83.07) ×106 with an average of 1.4 apheresis sessions, and the number was 15.86(range: 0.37~83.07) ×106 for the first apheresis, respectively. A single apheresis procedure was sufficient to reach the target yield of adequate mobilization in 35(89.7%) PMs, while 76.9% of PMs achieved optimal collection within two apheresis sessions. We observed acceptable hematological toxicity and antibiotic usage exposure in 26 patients with a mean duration of 3.6 days. No grade 4 infection or mobilization-related mortality was recorded. Most patients underwent ASCT and achieved successful hematopoietic recovery with prompt engraftment duration, except for one NK/T-cell lymphoma patient who succumbed to severe septicemia after receiving conditioning chemotherapy. Conclusion Our findings indicate that EA plus G-CSF is an effective and tolerable CD34+ stem cell mobilization strategy for patients with r/r lymphoma, including those predicted to be PMs. This regimen could be an option for patients with r/r lymphoma, particularly those undergoing mobilization for salvage ASCT therapy.
Collapse
Affiliation(s)
- Zhijuan Zhu
- Hematopoietic Stem Cell Transplantation Center, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Department of Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaofan Li
- Hematopoietic Stem Cell Transplantation Center, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Department of Hematology, Fujian Medical University Union Hospital, Fuzhou, China
- Translational Medicine Center on Hematology, Fujian Medical University, Fuzhou, China
| | - Xiaohong Yuan
- Hematopoietic Stem Cell Transplantation Center, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Department of Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xianling Chen
- Hematopoietic Stem Cell Transplantation Center, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Department of Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ting Lin
- Hematopoietic Stem Cell Transplantation Center, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Department of Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiangli Guo
- Hematopoietic Stem Cell Transplantation Center, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Department of Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Nainong Li
- Hematopoietic Stem Cell Transplantation Center, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Department of Hematology, Fujian Medical University Union Hospital, Fuzhou, China
- Translational Medicine Center on Hematology, Fujian Medical University, Fuzhou, China
| |
Collapse
|
15
|
Worel N. How to manage poor mobilisers. Transfus Apher Sci 2024; 63:103934. [PMID: 38678982 DOI: 10.1016/j.transci.2024.103934] [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] [Indexed: 05/01/2024]
Abstract
Autologous hematopoietic progenitor cell transplantation (ASCT) has been used for more than five decades to treat malignant and non-malignant diseases. Successful engraftment after high-dose chemotherapy relies on the ability to collect sufficient CD34 + hematopoietic progenitor cells (HPCs), typically from peripheral blood after mobilization. Commonly, either granulocyte colony-stimulating factor (G-CSF) alone as a single agent (i.e. steady-state mobilization) or G-CSF after chemotherapy is administered to collect adequate numbers of HPCs (minimum ≥2 × 106 CD34 + cells/kg for one ASCT; optimal up to 5 × 106 CD34 + cells/kg). However, a significant proportion of patients fail successful HPC mobilization, which is commonly defined as a CD34+ cell count below 10-15/µL after at least 4 days of 10 µg/kg b.w. G-CSF alone, or after chemo-mobilization in combination with 5-10 µg/kg b.w. G-CSF. In these situations plerixafor, a chemokine receptor inhibitor (CXCR4) can be used to enhance HPC collection in patients with multiple myeloma and malignant lymphoma whose cells mobilize poorly. Risk factors for poor mobilization have been evaluated and several strategies (e.g. plerixafor to rescue the mobilization approach or pre-emptive use) have been suggested to optimize mobilization, especially in patients at risk. This manuscript discusses the risk factors of poor CD34+ mobilization and summarizes the current strategies to optimize mobilization and HPC collection.
Collapse
Affiliation(s)
- Nina Worel
- Department of Transfusion Medicine and Cell Therapy, Medical University Vienna, Waehringer Guertel 18-29, A-1090 Vienna, Austria.
| |
Collapse
|
16
|
Javanbakht A, Stringer S, Anderson H, Hamilton E, Philip A, Waller EK, Langston AA, Joseph N, Roback JD, Schneider T, Sullivan HC, Hendrickson JE. Optimizing autologous stem cell collections for patients with multiple myeloma receiving G-CSF and Plerixafor: A single center project. J Clin Apher 2024; 39:e22127. [PMID: 38803152 DOI: 10.1002/jca.22127] [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: 12/18/2023] [Revised: 03/05/2024] [Accepted: 05/01/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Increasing indications for cellular therapy collections have stressed our healthcare system, with autologous collections having a longer than desired wait time until apheresis collection. This quality improvement initiative was undertaken to accommodate more patients within existing resources. STUDY DESIGN AND METHODS Patients with multiple myeloma who underwent autologous peripheral blood stem cell collection from October 2022 to April 2023 were included. Demographic, mobilization, laboratory, and apheresis data were retrospectively collected from the medical record. RESULTS This cohort included 120 patients (49.2% male), with a median age of 60 years. All received G-CSF and 95% received pre-emptive Plerixafor approximately 18 hours pre-collection. Most (79%) had collection goals of at least 8 × 106/kg CD34 cells, with 63% over 70 years old having this high collection goal (despite 20 years of institutional data showing <1% over 70 years old have a second transplant). With collection efficiencies of 55.9%, 44% of patients achieved their collection goal in a single day apheresis collection. A platelet count <150 × 103/μL on the day of collection was a predictor for poor mobilization; among 27 patients with a low baseline platelet count, 17 did not achieve the collection goal and 2 failed to collect a transplantable dose. CONCLUSIONS With minor collection goal adjustments, 15% of all collection appointments could have been avoided over this 6-month period. Other strategies to accommodate more patients include mobilization modifications (Plerixafor timing or substituting a longer acting drug), utilizing platelet counts to predict mobilization, and modifying apheresis collection volumes or schedule templates.
Collapse
Affiliation(s)
- Ayda Javanbakht
- Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Stephanie Stringer
- Center for Transfusion and Cellular Therapies, Emory Healthcare, Atlanta, Georgia, USA
| | - Hollie Anderson
- Center for Transfusion and Cellular Therapies, Emory Healthcare, Atlanta, Georgia, USA
| | - Ellie Hamilton
- Center for Transfusion and Cellular Therapies, Emory Healthcare, Atlanta, Georgia, USA
| | - Anisha Philip
- Center for Transfusion and Cellular Therapies, Emory Healthcare, Atlanta, Georgia, USA
| | - Edmund K Waller
- Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Amelia A Langston
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Nisha Joseph
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - John D Roback
- Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Thomas Schneider
- Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - H Cliff Sullivan
- Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jeanne E Hendrickson
- Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
17
|
Chabannon C, Visentin S, Granata A, Thuret I. Mobilization and collection of CD34 + cells in patients with globin disorders: Providing the starting material to manufacturers of autologous gene therapies. Transfus Apher Sci 2024; 63:103926. [PMID: 38670856 DOI: 10.1016/j.transci.2024.103926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Affiliation(s)
- Christian Chabannon
- Centre de Thérapie Cellulaire, Département de Biologie du Cancer, Institut Paoli-Calmettes Comprehensive Cancer, Marseille, France; Module Biothérapies du Centre d'Investigations Cliniques de Marseille, CBT-1409 Inserm - Aix-Marseille Université - Assistance Publique des Hôpitaux de Marseille - Institut Paoli-Calmettes, Marseille, France.
| | - Sandrine Visentin
- Département d'Hématologie et d'Oncologie Pédiatriques, Hôpital de la Timone-Enfants, Assistance Publique des Hôpitaux de Marseille / Hôpitaux Universitaires de Marseille, Marseille, France
| | - Angéla Granata
- Centre de Thérapie Cellulaire, Département de Biologie du Cancer, Institut Paoli-Calmettes Comprehensive Cancer, Marseille, France
| | - Isabelle Thuret
- Département d'Hématologie et d'Oncologie Pédiatriques, Hôpital de la Timone-Enfants, Assistance Publique des Hôpitaux de Marseille / Hôpitaux Universitaires de Marseille, Marseille, France
| |
Collapse
|
18
|
Sanchis-Pascual D, Del Olmo-García MI, Prado-Wohlwend S, Zac-Romero C, Segura Huerta Á, Hernández-Gil J, Martí-Bonmatí L, Merino-Torres JF. CXCR4: From Signaling to Clinical Applications in Neuroendocrine Neoplasms. Cancers (Basel) 2024; 16:1799. [PMID: 38791878 PMCID: PMC11120359 DOI: 10.3390/cancers16101799] [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: 04/12/2024] [Revised: 04/30/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
There are several well-described molecular mechanisms that influence cell growth and are related to the development of cancer. Chemokines constitute a fundamental element that is not only involved in local growth but also affects angiogenesis, tumor spread, and metastatic disease. Among them, the C-X-C motif chemokine ligand 12 (CXCL12) and its specific receptor the chemokine C-X-C motif receptor 4 (CXCR4) have been widely studied. The overexpression in cell membranes of CXCR4 has been shown to be associated with the development of different kinds of histological malignancies, such as adenocarcinomas, epidermoid carcinomas, mesenchymal tumors, or neuroendocrine neoplasms (NENs). The molecular synapsis between CXCL12 and CXCR4 leads to the interaction of G proteins and the activation of different intracellular signaling pathways in both gastroenteropancreatic (GEP) and bronchopulmonary (BP) NENs, conferring greater capacity for locoregional aggressiveness, the epithelial-mesenchymal transition (EMT), and the appearance of metastases. Therefore, it has been hypothesized as to how to design tools that target this receptor. The aim of this review is to focus on current knowledge of the relationship between CXCR4 and NENs, with a special emphasis on diagnostic and therapeutic molecular targets.
Collapse
Affiliation(s)
- David Sanchis-Pascual
- Endocrinology and Nutrition Department, University and Politecnic Hospital La Fe (Valencia), 46026 Valencia, Spain; (M.I.D.O.-G.); (J.F.M.-T.)
| | - María Isabel Del Olmo-García
- Endocrinology and Nutrition Department, University and Politecnic Hospital La Fe (Valencia), 46026 Valencia, Spain; (M.I.D.O.-G.); (J.F.M.-T.)
- Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, Health Research Institute La Fe, 46026 Valencia, Spain
| | - Stefan Prado-Wohlwend
- Nuclear Medicine Department, University and Politecnic Hospital La Fe (Valencia), 46026 Valencia, Spain;
| | - Carlos Zac-Romero
- Patholoy Department, University and Politecnic Hospital La Fe (Valencia), 46026 Valencia, Spain;
| | - Ángel Segura Huerta
- Medical Oncology Department, University and Politecnic Hospital La Fe (Valencia), 46026 Valencia, Spain;
| | - Javier Hernández-Gil
- Instituto de Tecnología Química, Universitat Politècnica de València, Consejo Superior de Investigaciones Científicas, 46022 Valencia, Spain;
| | - Luis Martí-Bonmatí
- Medical Imaging Department, Biomedical Imaging Research Group, Health Research Institute, University and Politecnic Hospital La Fe, 46026 Valencia, Spain;
| | - Juan Francisco Merino-Torres
- Endocrinology and Nutrition Department, University and Politecnic Hospital La Fe (Valencia), 46026 Valencia, Spain; (M.I.D.O.-G.); (J.F.M.-T.)
- Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, Health Research Institute La Fe, 46026 Valencia, Spain
- Department of Medicine, University of Valencia, 46010 Valencia, Spain
| |
Collapse
|
19
|
Cohen I, Vaxman I, Gertz MA. Historical Perspective of High-Dose Therapy Followed by Autologous Stem Cell Transplantation in Multiple Myeloma. Acta Haematol 2024:1-10. [PMID: 38710160 DOI: 10.1159/000539225] [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: 02/27/2024] [Accepted: 05/03/2024] [Indexed: 05/08/2024]
Abstract
BACKGROUND High-dose therapy (HDT) followed by autologous stem cell transplantation (ASCT) has become part of standard of care (SOC) in newly diagnosed multiple myeloma. In this review, we provide a historical perspective on ASCT since its introduction in the 1990s. SUMMARY Overall survival (OS) benefit for HDT followed by ASCT was demonstrated in studies comparing HDT with ASCT to standard-dose therapy (SDT) before the era of novel agents. Conditioning is done with melphalan 200 mg/m2. Lower doses (MEL140, MEL150) for older patients with comorbidities are safe and have comparable results. The addition of busulfan to melphalan improves progression-free survival (PFS) but not OS. HDT with ASCT after induction with novel agents prolongs PFS but not OS compared to SDT alone. The benefit is more evident in patients with high-risk cytogenetics. Mobilization can be achieved with granulocyte colony-stimulating factor alone, but is improved with the addition of chemotherapy. Plerixafor reduces mobilization failure and enables sufficient stem cell collection after induction with novel agents. ASCT is safe with a low rate of mortality (1%), and selected patients can be managed as outpatients. KEY MESSAGES HDT followed by ASCT remains part of SOC due to its PFS benefit and relatively low toxicity.
Collapse
Affiliation(s)
- Inbar Cohen
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel,
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel,
| | - Iuliana Vaxman
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| | - Morie A Gertz
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
20
|
Sharma A, Singh LR. An insight into the pharmacology of cysteine/methionine containing peptide drugs. Eur J Med Chem 2024; 271:116456. [PMID: 38691890 DOI: 10.1016/j.ejmech.2024.116456] [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: 01/16/2024] [Revised: 04/13/2024] [Accepted: 04/16/2024] [Indexed: 05/03/2024]
Abstract
Since last century, peptides have emerged as potential drugs with >90 FDA approvals for various targets with several in the pipeline. Sulphur, in peptides is present either as thiol (-SH) from Cys or thioether from Met. In this review, all the peptides approved by FDA since 2000 containing sulphur have been included. Among them ∼50 % contains disulphide bridges. This clearly demonstrates the significance of disulphide bonds in peptide drugs. This can be achieved synthetically by using orthogonal protecting groups (PGs) for -SH. These PGs are compatible with Solid Phase Peptide Synthesis (SPPS), which is still the method of choice for peptide synthesis. The orthogonal PGs used for Cys thiol side chain protecting for disulphide bond formation have been included which are currently in use both by academia and industry from small scale to large scale synthesis. In addition, the details of the FDA approved drugs containing Cys and Met (or both) have also been discussed.
Collapse
Affiliation(s)
- Anamika Sharma
- Department of Natural Products and Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Chemical Science Division, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| | - L Ravithej Singh
- Chemical Science Division, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Fluoro-Agrochemicals Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India.
| |
Collapse
|
21
|
Kayki-Mutlu G, Aksoyalp ZS, Wojnowski L, Michel MC. A year in pharmacology: new drugs approved by the US Food and Drug Administration in 2023. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:2949-2970. [PMID: 38530400 PMCID: PMC11074039 DOI: 10.1007/s00210-024-03063-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 03/19/2024] [Indexed: 03/28/2024]
Abstract
With 54 new drugs and seven cellular and gene therapy products, the approvals by the US Food and Drug Administration (FDA) recovered 2023 from the 2022 dent back to the levels of 2020-2021. As in previous years of this annual review, we assign these new drugs to one of three levels of innovation: first drug against a condition ("first-in-indication"), first drug using a novel molecular mechanism ("first-in-class"), and "next-in-class," i.e., a drug using an already exploited molecular mechanism. We identify four (7%) "first-in-indication," 22 (36%) "first-in-class," and 35 (57%) "next-in-class" drugs. By treatment area, rare diseases (54%) and cancer drugs (23%) were once again the most prevalent (and partly overlapping) therapeutic areas. Other continuing trends were the use of accelerated regulatory approval pathways and the reliance on biopharmaceuticals (biologics). 2023 marks the approval of a first therapy based on CRISPR/Cas9 gene editing.
Collapse
Affiliation(s)
- Gizem Kayki-Mutlu
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, Ankara, Türkiye
| | - Zinnet Sevval Aksoyalp
- Department of Pharmacology, Faculty of Pharmacy, Izmir Katip Celebi University, Izmir, Türkiye
| | - Leszek Wojnowski
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University, Langenbeckstr. 1, 55118, Mainz, Germany
| | - Martin C Michel
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University, Langenbeckstr. 1, 55118, Mainz, Germany.
| |
Collapse
|
22
|
Leonard A, Weiss MJ. Hematopoietic stem cell collection for sickle cell disease gene therapy. Curr Opin Hematol 2024; 31:104-114. [PMID: 38359264 PMCID: PMC11414477 DOI: 10.1097/moh.0000000000000807] [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] [Indexed: 02/17/2024]
Abstract
PURPOSE OF REVIEW Gene therapy for sickle cell disease (SCD) is advancing rapidly, with two transformative products recently approved by the US Food and Drug Administration and numerous others under study. All current gene therapy protocols require ex vivo modification of autologous hematopoietic stem cells (HSCs). However, several SCD-related problems impair HSC collection, including a stressed and damaged bone marrow, potential cytotoxicity by the major therapeutic drug hydroxyurea, and inability to use granulocyte colony stimulating factor, which can precipitate severe vaso-occlusive events. RECENT FINDINGS Peripheral blood mobilization of HSCs using the CXCR4 antagonist plerixafor followed by apheresis collection was recently shown to be safe and effective for most SCD patients and is the current strategy for mobilizing HSCs. However, exceptionally large numbers of HSCs are required to manufacture an adequate cellular product, responses to plerixafor are variable, and most patients require multiple mobilization cycles, increasing the risk for adverse events. For some, gene therapy is prohibited by the failure to obtain adequate numbers of HSCs. SUMMARY Here we review the current knowledge on HSC collection from individuals with SCD and potential improvements that may enhance the safety, efficacy, and availability of gene therapy for this disorder.
Collapse
Affiliation(s)
- Alexis Leonard
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | |
Collapse
|
23
|
Giannakoulas A, Nikolaidis M, Amoutzias GD, Giannakoulas N. A comparative analysis of transcriptomics of newly diagnosed multiple myeloma: exploring drug repurposing. Front Oncol 2024; 14:1390105. [PMID: 38690165 PMCID: PMC11058662 DOI: 10.3389/fonc.2024.1390105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/01/2024] [Indexed: 05/02/2024] Open
Abstract
Multiple myeloma (MM) is an incurable malignant plasma cell disorder characterized by the infiltration of clonal plasma cells in the bone marrow compartment. Gene Expression Profiling (GEP) has emerged as a powerful investigation tool in modern myeloma research enabling the dissection of the molecular background of MM and allowing the identification of gene products that could potentially serve as targets for therapeutic intervention. In this study we investigated shared transcriptomic abnormalities across newly diagnosed multiple myeloma (NDMM) patient cohorts. In total, publicly available transcriptomic data of 7 studies from CD138+ cells from 281 NDMM patients and 44 healthy individuals were integrated and analyzed. Overall, we identified 28 genes that were consistently differentially expressed (DE) between NDMM patients and healthy donors (HD) across various studies. Of those, 9 genes were over/under-expressed in more than 75% of NDMM patients. In addition, we identified 4 genes (MT1F, PURPL, LINC01239 and LINC01480) that were not previously considered to participate in MM pathogenesis. Meanwhile, by mining three drug databases (ChEMBL, IUPHAR/BPS and DrugBank) we identified 31 FDA-approved and 144 experimental drugs that target 8 of these 28 over/under-expressed MM genes. Taken together, our study offers new insights in MM pathogenesis and importantly, it reveals potential new treatment options that need to be further investigated in future studies.
Collapse
Affiliation(s)
- Angelos Giannakoulas
- Department of Hematology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Marios Nikolaidis
- Bioinformatics Laboratory, Department of Biochemistry & Biotechnology, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Grigorios D. Amoutzias
- Bioinformatics Laboratory, Department of Biochemistry & Biotechnology, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Nikolaos Giannakoulas
- Department of Hematology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| |
Collapse
|
24
|
Kim H, Taslakjian B, Kim S, Tirrell MV, Guler MO. Therapeutic Peptides, Proteins and their Nanostructures for Drug Delivery and Precision Medicine. Chembiochem 2024; 25:e202300831. [PMID: 38408302 DOI: 10.1002/cbic.202300831] [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: 12/08/2023] [Revised: 02/05/2024] [Accepted: 02/22/2024] [Indexed: 02/28/2024]
Abstract
Peptide and protein nanostructures with tunable structural features, multifunctionality, biocompatibility and biomolecular recognition capacity enable development of efficient targeted drug delivery tools for precision medicine applications. In this review article, we present various techniques employed for the synthesis and self-assembly of peptides and proteins into nanostructures. We discuss design strategies utilized to enhance their stability, drug-loading capacity, and controlled release properties, in addition to the mechanisms by which peptide nanostructures interact with target cells, including receptor-mediated endocytosis and cell-penetrating capabilities. We also explore the potential of peptide and protein nanostructures for precision medicine, focusing on applications in personalized therapies and disease-specific targeting for diagnostics and therapeutics in diseases such as cancer.
Collapse
Affiliation(s)
- HaRam Kim
- The Pritzker School of Molecular Engineering, The University of Chicago, 5640 S. Ellis Ave., Chicago, 60637, IL, USA
| | - Boghos Taslakjian
- The Pritzker School of Molecular Engineering, The University of Chicago, 5640 S. Ellis Ave., Chicago, 60637, IL, USA
| | - Sarah Kim
- The Pritzker School of Molecular Engineering, The University of Chicago, 5640 S. Ellis Ave., Chicago, 60637, IL, USA
| | - Matthew V Tirrell
- The Pritzker School of Molecular Engineering, The University of Chicago, 5640 S. Ellis Ave., Chicago, 60637, IL, USA
| | - Mustafa O Guler
- The Pritzker School of Molecular Engineering, The University of Chicago, 5640 S. Ellis Ave., Chicago, 60637, IL, USA
| |
Collapse
|
25
|
MacLean MR, Walker OL, Arun RP, Fernando W, Marcato P. Informed by Cancer Stem Cells of Solid Tumors: Advances in Treatments Targeting Tumor-Promoting Factors and Pathways. Int J Mol Sci 2024; 25:4102. [PMID: 38612911 PMCID: PMC11012648 DOI: 10.3390/ijms25074102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/30/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Cancer stem cells (CSCs) represent a subpopulation within tumors that promote cancer progression, metastasis, and recurrence due to their self-renewal capacity and resistance to conventional therapies. CSC-specific markers and signaling pathways highly active in CSCs have emerged as a promising strategy for improving patient outcomes. This review provides a comprehensive overview of the therapeutic targets associated with CSCs of solid tumors across various cancer types, including key molecular markers aldehyde dehydrogenases, CD44, epithelial cellular adhesion molecule, and CD133 and signaling pathways such as Wnt/β-catenin, Notch, and Sonic Hedgehog. We discuss a wide array of therapeutic modalities ranging from targeted antibodies, small molecule inhibitors, and near-infrared photoimmunotherapy to advanced genetic approaches like RNA interference, CRISPR/Cas9 technology, aptamers, antisense oligonucleotides, chimeric antigen receptor (CAR) T cells, CAR natural killer cells, bispecific T cell engagers, immunotoxins, drug-antibody conjugates, therapeutic peptides, and dendritic cell vaccines. This review spans developments from preclinical investigations to ongoing clinical trials, highlighting the innovative targeting strategies that have been informed by CSC-associated pathways and molecules to overcome therapeutic resistance. We aim to provide insights into the potential of these therapies to revolutionize cancer treatment, underscoring the critical need for a multi-faceted approach in the battle against cancer. This comprehensive analysis demonstrates how advances made in the CSC field have informed significant developments in novel targeted therapeutic approaches, with the ultimate goal of achieving more effective and durable responses in cancer patients.
Collapse
Affiliation(s)
- Maya R. MacLean
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.M.); (O.L.W.); (R.P.A.); (W.F.)
| | - Olivia L. Walker
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.M.); (O.L.W.); (R.P.A.); (W.F.)
| | - Raj Pranap Arun
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.M.); (O.L.W.); (R.P.A.); (W.F.)
| | - Wasundara Fernando
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.M.); (O.L.W.); (R.P.A.); (W.F.)
- Department of Biology, Acadia University, Wolfville, NS B4P 2R6, Canada
| | - Paola Marcato
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.M.); (O.L.W.); (R.P.A.); (W.F.)
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Nova Scotia Health Authority, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
26
|
Abstract
No therapy in multiple myeloma has been as extensively investigated as stem cell transplantation following high-dose chemotherapy. A search of the national library of medicine in February 2023 revealed over 27,000 publications covering stem cell transplantation. No other treatment for multiple myeloma has been so vigorously investigated. However, given the rapid advances seen in the treatment of multiple myeloma, it is legitimate to ask whether the technique first introduced in 1983 by Thomas McIlwain still has relevance. In 1984,Barlogie introduced infusional vincristine, doxorubicin, and dexamethasone and in 1986 published a first series on high-dose therapy with autologous marrow-derived stem cells. At this point, the only available therapies were melphalan, prednisone, other intensive steroids such as methylprednisolone, and interferon. Cyclophosphamide was used both orally and parenterally. VBMCP was introduced as a combination therapy at Memorial Hospital subsequently shown not to be superior to melphalan and prednisone.
Collapse
Affiliation(s)
- Morie A Gertz
- Mayo Clinic, 200 SW First street, Rochester, MN 55905, USA.
| |
Collapse
|
27
|
Prisciandaro M, Santinelli E, Tomarchio V, Tafuri MA, Bonchi C, Palazzo G, Nobile C, Marinucci A, Mele M, Annibali O, Rigacci L, Vacca M. Stem Cells Collection and Mobilization in Adult Autologous/Allogeneic Transplantation: Critical Points and Future Challenges. Cells 2024; 13:586. [PMID: 38607025 PMCID: PMC11011310 DOI: 10.3390/cells13070586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/23/2024] [Accepted: 03/26/2024] [Indexed: 04/13/2024] Open
Abstract
Achieving successful hematopoietic stem cell transplantation (HSCT) relies on two fundamental pillars: effective mobilization and efficient collection through apheresis to attain the optimal graft dose. These cornerstones pave the way for enhanced patient outcomes. The primary challenges encountered by the clinical unit and collection facility within a transplant program encompass augmenting mobilization efficiency to optimize the harvest of target cell populations, implementing robust monitoring and predictive strategies for mobilization, streamlining the apheresis procedure to minimize collection duration while ensuring adequate yield, prioritizing patient comfort by reducing the overall collection time, guaranteeing the quality and purity of stem cell products to optimize graft function and transplant success, and facilitating seamless coordination between diverse entities involved in the HSCT process. In this review, we aim to address key questions and provide insights into the critical aspects of mobilizing and collecting hematopoietic stem cells for transplantation purposes.
Collapse
Affiliation(s)
- Michele Prisciandaro
- Operative Research Unit of Transfusion Medicine and Cellular Therapy, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Roma, Italy; (C.B.); (G.P.); (C.N.); (A.M.)
| | - Enrico Santinelli
- Operative Research Unit of Hematology and Stem Cell Transplantation, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Roma, Italy; (E.S.); (V.T.); (M.A.T.); (M.M.); (O.A.); (L.R.)
- Program in Immunology, Molecular Medicine and Applied Biotechnologies, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Valeria Tomarchio
- Operative Research Unit of Hematology and Stem Cell Transplantation, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Roma, Italy; (E.S.); (V.T.); (M.A.T.); (M.M.); (O.A.); (L.R.)
| | - Maria Antonietta Tafuri
- Operative Research Unit of Hematology and Stem Cell Transplantation, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Roma, Italy; (E.S.); (V.T.); (M.A.T.); (M.M.); (O.A.); (L.R.)
| | - Cecilia Bonchi
- Operative Research Unit of Transfusion Medicine and Cellular Therapy, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Roma, Italy; (C.B.); (G.P.); (C.N.); (A.M.)
| | - Gloria Palazzo
- Operative Research Unit of Transfusion Medicine and Cellular Therapy, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Roma, Italy; (C.B.); (G.P.); (C.N.); (A.M.)
| | - Carolina Nobile
- Operative Research Unit of Transfusion Medicine and Cellular Therapy, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Roma, Italy; (C.B.); (G.P.); (C.N.); (A.M.)
| | - Alessandra Marinucci
- Operative Research Unit of Transfusion Medicine and Cellular Therapy, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Roma, Italy; (C.B.); (G.P.); (C.N.); (A.M.)
| | - Marcella Mele
- Operative Research Unit of Hematology and Stem Cell Transplantation, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Roma, Italy; (E.S.); (V.T.); (M.A.T.); (M.M.); (O.A.); (L.R.)
| | - Ombretta Annibali
- Operative Research Unit of Hematology and Stem Cell Transplantation, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Roma, Italy; (E.S.); (V.T.); (M.A.T.); (M.M.); (O.A.); (L.R.)
| | - Luigi Rigacci
- Operative Research Unit of Hematology and Stem Cell Transplantation, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Roma, Italy; (E.S.); (V.T.); (M.A.T.); (M.M.); (O.A.); (L.R.)
| | - Michele Vacca
- Operative Research Unit of Transfusion Medicine and Cellular Therapy, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Roma, Italy; (C.B.); (G.P.); (C.N.); (A.M.)
| |
Collapse
|
28
|
Du J, Lin Z, Fu XH, Gu XR, Lu G, Hou J. Research progress of the chemokine/chemokine receptor axes in the oncobiology of multiple myeloma (MM). Cell Commun Signal 2024; 22:177. [PMID: 38475811 PMCID: PMC10935833 DOI: 10.1186/s12964-024-01544-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 02/25/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND The incidence of multiple myeloma (MM), a type of blood cancer affecting monoclonal plasma cells, is rising. Although new drugs and therapies have improved patient outcomes, MM remains incurable. Recent studies have highlighted the crucial role of the chemokine network in MM's pathological mechanism. Gaining a better understanding of this network and creating an overview of chemokines in MM could aid in identifying potential biomarkers and developing new therapeutic strategies and targets. PURPOSE To summarize the complicated role of chemokines in MM, discuss their potential as biomarkers, and introduce several treatments based on chemokines. METHODS Pubmed, Web of Science, ICTRP, and Clinical Trials were searched for articles and research related to chemokines. Publications published within the last 5 years are selected. RESULTS Malignant cells can utilize chemokines, including CCL2, CCL3, CCL5, CXCL7, CXCL8, CXCL12, and CXCL13 to evade apoptosis triggered by immune cells or medication, escape from bone marrow and escalate bone lesions. Other chemokines, including CXCL4, CCL19, and CXCL10, may aid in recruiting immune cells, increasing their cytotoxicity against cancer cells, and inducing apoptosis of malignant cells. CONCLUSION Utilizing anti-tumor chemokines or blocking pro-tumor chemokines may provide new therapeutic strategies for managing MM. Inspired by developed CXCR4 antagonists, including plerixafor, ulocuplumab, and motixafortide, more small molecular antagonists or antibodies for pro-tumor chemokine ligands and their receptors can be developed and used in clinical practice. Along with inhibiting pro-tumor chemokines, studies suggest combining chemokines with chimeric antigen receptor (CAR)-T therapy is promising and efficient.
Collapse
Affiliation(s)
- Jun Du
- Department of Hematology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Zheng Lin
- Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xue-Hang Fu
- Department of Hematology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Xiao-Ran Gu
- Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Guang Lu
- Department of Hematology, Shengli Oilfield Central Hospital, Dongying, 257099, China.
| | - Jian Hou
- Department of Hematology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
29
|
Zhu P, Yang L, Wu Y, Shi J, Lai X, Liu L, Ye Y, Yu J, Zhao Y, Yuan X, Fu H, Cai Z, Huang H, Luo Y. Graft CD8 T-cell-based risk system predicts survival in antithymocyte globulin-based myeloablative haploidentical peripheral blood stem cell transplantation. Clin Transl Immunology 2024; 13:e1484. [PMID: 38223258 PMCID: PMC10786671 DOI: 10.1002/cti2.1484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 12/04/2023] [Accepted: 01/04/2024] [Indexed: 01/16/2024] Open
Abstract
Objective This study investigated the cellular composition of peripheral blood grafts for anti-thymocyte globulin (ATG)-based myeloablative haploidentical haematopoietic stem cell transplantation (haplo-HSCT). Methods Clinical characteristics were retrospectively evaluated in a training cohort with ATG-based myeloablative haplo-HSCT between January 2016 and February 2020 and confirmed in a validation cohort between March 2020 and June 2021. Results A higher dose of graft CD8+ T cells (≥ 0.85 × 108 kg-1) was significantly improved overall survival (OS; hazard ratio [HR], 1.750; P = 0.002) and disease-free survival (DFS; HR, 1.751; P < 0.001) in the training cohort, according to multivariate Cox regression analysis. Higher doses of mononuclear cells (MNCs) demonstrated better OS (HR, 1.517; P = 0.038) and DFS (HR, 1.532; P = 0.027). Older patient age (> 46 years), older donor age (≥ 50 years) and a higher refined disease risk index (rDRI) were also related to OS. A graft CD8+ T-cell risk system based on graft CD8+ T-cell dose, donor age and rDRI was constructed using a nomogram model after LASSO Cox regression analysis. It showed acceptable discrimination, with a C-index of 0.62 and 0.63, respectively. Graft CD8+ T-cell dose was negatively correlated with donor age (P < 0.001) and positively correlated with a higher lymphocyte percentage in the peripheral blood before mobilisation (P < 0.001). Conclusion A higher CD8+ T-cell dose in peripheral blood-derived grafts improves patients' survival with ATG-based myeloablative haplo-HSCT. Younger donors with higher lymphocyte percentages improved patients' survival with an intermediate rDRI risk.
Collapse
Affiliation(s)
- Panpan Zhu
- Bone Marrow Transplantation Center, the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhouChina
- Institute of HematologyZhejiang UniversityHangzhouChina
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity TherapyHangzhouChina
| | - Luxin Yang
- Bone Marrow Transplantation Center, the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhouChina
- Institute of HematologyZhejiang UniversityHangzhouChina
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity TherapyHangzhouChina
| | - Yibo Wu
- Bone Marrow Transplantation Center, the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhouChina
- Institute of HematologyZhejiang UniversityHangzhouChina
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity TherapyHangzhouChina
| | - Jimin Shi
- Bone Marrow Transplantation Center, the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhouChina
- Institute of HematologyZhejiang UniversityHangzhouChina
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity TherapyHangzhouChina
| | - Xiaoyu Lai
- Bone Marrow Transplantation Center, the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhouChina
- Institute of HematologyZhejiang UniversityHangzhouChina
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity TherapyHangzhouChina
| | - Lizhen Liu
- Bone Marrow Transplantation Center, the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhouChina
- Institute of HematologyZhejiang UniversityHangzhouChina
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity TherapyHangzhouChina
| | - Yishan Ye
- Bone Marrow Transplantation Center, the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhouChina
- Institute of HematologyZhejiang UniversityHangzhouChina
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity TherapyHangzhouChina
| | - Jian Yu
- Bone Marrow Transplantation Center, the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhouChina
- Institute of HematologyZhejiang UniversityHangzhouChina
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity TherapyHangzhouChina
| | - Yanmin Zhao
- Bone Marrow Transplantation Center, the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhouChina
- Institute of HematologyZhejiang UniversityHangzhouChina
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity TherapyHangzhouChina
| | - Xiaolin Yuan
- Bone Marrow Transplantation Center, the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhouChina
- Institute of HematologyZhejiang UniversityHangzhouChina
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity TherapyHangzhouChina
| | - Huarui Fu
- Bone Marrow Transplantation Center, the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhouChina
- Institute of HematologyZhejiang UniversityHangzhouChina
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity TherapyHangzhouChina
| | - Zhen Cai
- Bone Marrow Transplantation Center, the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhouChina
- Institute of HematologyZhejiang UniversityHangzhouChina
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity TherapyHangzhouChina
| | - He Huang
- Bone Marrow Transplantation Center, the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhouChina
- Institute of HematologyZhejiang UniversityHangzhouChina
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity TherapyHangzhouChina
| | - Yi Luo
- Bone Marrow Transplantation Center, the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhouChina
- Institute of HematologyZhejiang UniversityHangzhouChina
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity TherapyHangzhouChina
| |
Collapse
|
30
|
Li X, Wang Y, Yang Q, Song L, Kang L, Hu Z, Wang Z. Microarray-Based CD38 Peptide Probe Screening for Multiple Myeloma Imaging. Mol Pharm 2024; 21:245-254. [PMID: 38096423 DOI: 10.1021/acs.molpharmaceut.3c00808] [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] [Indexed: 01/02/2024]
Abstract
Assessing CD38 expression in vivo has become a significant element in multiple myeloma (MM) therapy, as it can be used to detect lesions and forecast the effectiveness of treatment. Accurate diagnosis requires a multifunctional, high-throughput probe screening platform to develop molecular probes for tumor-targeted multimodal imaging and treatment. Here, we investigated a microarray chip-based strategy for high-throughput screening of peptide probes for CD38. We obtained two new target peptides, CA-1 and CA-2, from a 105 peptide library with a dissociation constant (KD) of 10-7 M. The specificity and affinity of the target peptides were confirmed at the molecular and cellular levels. Peptide probes were labeled with indocyanine green (ICG) dye and 68Ga-DOTA, which were injected into a CD38-positive Ramos tumor-bearing mouse via its tail vein, and small animal fluorescence and positron emission tomography (PET) imaging showed that the peptide probes could show specific enrichment in the tumor tissue. Our study shows that a microchip-based screening of peptide probes can be used as a promising imaging tool for MM diagnosis.
Collapse
Affiliation(s)
- Xuejie Li
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Yuanzhuo Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Qi Yang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Lele Song
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Lei Kang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Zhiyuan Hu
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- School of Nanoscience and Technology, Sino-Danish College, University of Chinese Academy of Sciences, Beijing 100049, China
- School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430205, China
| | - Zihua Wang
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| |
Collapse
|
31
|
Abstract
Motixafortide (APHEXDATM) is a selective C-X-C motif chemokine receptor 4 (CXCR4) inhibitor being developed by BioLineRx under licence from Biokine Therapeutics for the mobilization of haematopoietic stem cells (HSCs) and the treatment of various cancers. On 11 September 2023, motixafortide was approved in the USA for use in combination with filgrastim [granulocyte colony stimulating factor (G-CSF)] to mobilize HSCs to the peripheral blood for collection and subsequent autologous transplantation in patients with multiple myeloma. Motixafortide has been granted Orphan Drug Designation for the treatment of pancreatic cancer in the EU and the USA, and for the treatment of acute myeloid leukaemia in the USA. Clinical development is ongoing for the mobilization of CD34+ HSCs for gene therapy in patients with sickle cell disease. This article summarizes the milestones in the development of motixafortide leading to this first approval.
Collapse
Affiliation(s)
- Sheridan M Hoy
- Springer Nature, Private Bag 65901, Mairangi Bay, Auckland, 0754, New Zealand.
| |
Collapse
|
32
|
Crees ZD, Rettig MP, Bashey A, Devine SM, Jaglowski S, Wan F, Zhou A, Harding M, Vainstein-Haras A, Sorani E, Gliko-Kabir I, Grossman BJ, Westervelt P, DiPersio JF, Uy GL. Hematopoietic stem cell mobilization for allogeneic stem cell transplantation by motixafortide, a novel CXCR4 inhibitor. Blood Adv 2023; 7:5210-5214. [PMID: 37327120 PMCID: PMC10500469 DOI: 10.1182/bloodadvances.2023010407] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 06/18/2023] Open
Affiliation(s)
- Zachary D. Crees
- Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - Michael P. Rettig
- Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - Asad Bashey
- Blood and Marrow Transplant Program, Northside Hospital, Atlanta, GA
| | - Steven M. Devine
- Center for International Blood and Marrow Transplant Research, National Marrow Donor Program, Minneapolis, MN
| | - Samantha Jaglowski
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Fei Wan
- Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - Amy Zhou
- Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - Melinda Harding
- Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | | | | | | | - Brenda J. Grossman
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Peter Westervelt
- Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - John F. DiPersio
- Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - Geoffrey L. Uy
- Division of Oncology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
33
|
Liang KL, Laurenti E, Taghon T. Circulating IRF8-expressing CD123 +CD127 + lymphoid progenitors: key players in human hematopoiesis. Trends Immunol 2023; 44:678-692. [PMID: 37591714 PMCID: PMC7614993 DOI: 10.1016/j.it.2023.07.004] [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: 06/23/2023] [Revised: 07/12/2023] [Accepted: 07/12/2023] [Indexed: 08/19/2023]
Abstract
Lymphopoiesis is the process in which B and T cells, and innate lymphoid cells (ILCs) develop from hematopoietic progenitors that exhibit early lymphoid priming. The branching points where lymphoid-primed human progenitors are further specified to B/T/ILC differentiation trajectories remain unclear. Here, we discuss the emerging role of interferon regulatory factor (IRF)8 as a key factor to bridge human lymphoid and dendritic cell (DC) differentiation, and the current evidence for the existence of circulating and tissue-resident CD123+CD127+ lymphoid progenitors. We propose a model whereby DC/B/T/ILC lineage programs in circulating CD123+CD127+ lymphoid progenitors are expressed in balance. Upon tissue seeding, the tissue microenvironment tilts this molecular balance towards a specific lineage, thereby determining in vivo lineage fates. Finally, we discuss the translational implication of these lymphoid precursors.
Collapse
Affiliation(s)
- Kai Ling Liang
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent, Ghent, Belgium
| | - Elisa Laurenti
- Department of Haematology, University of Cambridge, Cambridge, UK; Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK
| | - Tom Taghon
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent, Ghent, Belgium.
| |
Collapse
|
34
|
Ilic D, Liovic M. Industry updates from the field of stem cell research and regenerative medicine in April 2023. Regen Med 2023; 18:591-599. [PMID: 37340951 DOI: 10.2217/rme-2023-0105] [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] [Indexed: 06/22/2023] Open
Abstract
Latest developments in the field of stem cell research and regenerative medicine compiled from publicly available information and press releases from non-academic institutions in April 2023.
Collapse
Affiliation(s)
- Dusko Ilic
- Stem Cell Laboratories, Guy's Assisted Conception Unit, Department of Women & Children's Health, Faculty of Life Sciences & Medicine, King's College London, London, SE1 9RT, UK
| | - Mirjana Liovic
- Medical Center for Molecular Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, 1000, Slovenia
| |
Collapse
|