1
|
Zhou Q, Wang Y. A shadow in the treatment of acute leukemia: lineage switch. BLOOD SCIENCE 2025; 7:e00220. [PMID: 40123764 PMCID: PMC11927657 DOI: 10.1097/bs9.0000000000000220] [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: 07/24/2024] [Accepted: 01/16/2025] [Indexed: 03/25/2025] Open
Abstract
Lineage switch is a rare phenomenon in which acute myeloid leukemia (AML) transforms into acute lymphoblastic leukemia (ALL) and vice versa, sharing the same clonal origin. It is more common for AML to relapse as ALL. Cytogenetics, microenvironment, and preceding therapies are associated with lineage switch. Since the etiology of lineage switch is unclear, presumptions include clonal selection, pluripotent stem cells, and differentiated cell trans-differentiation or re-differentiation. The key point for diagnosing lineage switch is that the relapsed tumor originates from the common cell of the primary leukemia, although it is occasionally derived via clonal evolution. It is very important to distinguish lineage switch from other illnesses, such as secondary leukemia or the blast phase of chronic leukemia. Although direct treatment of the present lineage results in an improved prognosis, the outcome of these patients remains poor, with low survival and rapid progression. Hematopoietic stem cell transplantation can extend survival. Lineage switch risk-adapted management stratification may be beneficial for detecting relapse and more promptly provide suitable therapy. Efficient and toxicity-restricted therapy is being developed to improve the very poor prognosis.
Collapse
Affiliation(s)
- Qiaoyi Zhou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Ying Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- Tianjin Institutes of Health Science, Tianjin 301600, China
| |
Collapse
|
2
|
Nesic K, Parker P, Swisher EM, Krais JJ. DNA repair and the contribution to chemotherapy resistance. Genome Med 2025; 17:62. [PMID: 40420317 PMCID: PMC12107761 DOI: 10.1186/s13073-025-01488-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 05/14/2025] [Indexed: 05/28/2025] Open
Abstract
The DNA damage response comprises a set of imperfect pathways that maintain cell survival following exposure to DNA damaging agents. Cancers frequently exhibit DNA repair pathway alterations that contribute to their intrinsic genome instability. This, in part, facilitates a therapeutic window for many chemotherapeutic agents whose mechanisms of action often converge at the generation of a double-strand DNA break. The development of therapy resistance occurs through countless molecular mechanisms that promote tolerance to DNA damage, often by preventing break formation or increasing repair capacity. This review broadly discusses the DNA damaging mechanisms of action for different classes of chemotherapeutics, how avoidance and repair of double-strand breaks can promote resistance, and strategic directions for counteracting therapy resistance.
Collapse
Affiliation(s)
- Ksenija Nesic
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Phoebe Parker
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | | | - John J Krais
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
3
|
Zhang Y, Lu Y, Mai L, Wen Z, Dai M, Xu S, Lin X, Luo Y, Qiu Y, Chen Y, Dong Z, Chen C, Meng W, Luo X, Lin G, Tam PKH, Pan X. Dynamic heterogeneity towards drug resistance in AML cells is primarily driven by epigenomic mechanism unveiled by multi-omics analysis. J Adv Res 2025:S2090-1232(25)00358-3. [PMID: 40409464 DOI: 10.1016/j.jare.2025.05.038] [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: 02/11/2025] [Revised: 05/06/2025] [Accepted: 05/18/2025] [Indexed: 05/25/2025] Open
Abstract
INTRODUCTION Acute myeloid leukemia (AML) is a hematologic malignancy characterized by aggressive proliferation and chemoresistance, leading to poor patient outcomes. Despite advances in chemotherapy, resistance mechanisms remain inadequately understood, particularly at the cellular and molecular level. OBJECTIVES This study aims to elucidate the cellular and molecular mechanisms underlying drug resistance in AML cells. METHODS A multi-omics approach was employed, integrating single-cell RNA sequencing (scRNA-seq), chromatin accessibility profiling (scATAC-seq), DNA methylation analysis, and whole-exome sequencing (WES). AML cell lines (KG-1a, Kasumi-1, and HL-60) were treated with standard chemotherapeutic agents, including cytarabine (Ara-C), daunorubicin (DNR), azacitidine (AZA), and decitabine (DEC). Additionally, we developed a novel multiplexed scRNA-seq strategy, NAMUL-seq, to enhance the efficiency and scalability of single-cell transcriptomic research. RESULTS We observed substantial cellular heterogeneity and dynamic transcriptomic trajectories in AML cells subjected to various treatments, uncovering a tendency for reprogramming towards a more stem-like state. Notably, Ara-C-resistant KG-1a cells predominantly originated from G2/M phase subpopulations, suggesting a resistance mechanism linked to specific cell cycle stages. Our findings further indicate that rapid Ara-C resistance is primarily driven by epigenomic changes, including alterations in DNA methylation, chromatin architecture, and transcription factor activity, whereas exonic mutations played a minimal role. CONCLUSION This study demonstrates that AML drug resistance is predominantly driven by epigenomic mechanisms rather than genetic mutations. This study provides a detailed cellular and molecular characterization of AML drug response and resistance, identifying potential therapeutic targets and laying the groundwork for future efforts to overcome chemoresistance.
Collapse
Affiliation(s)
- Yulong Zhang
- Department of Biochemistry and Molecular Biology, and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, School of Basic Medical Sciences, Guangzhou, Guangdong, China; Precision Regenerative Medicine Research Centre, Medical Science Division, and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078, China
| | - Yanfang Lu
- Department of Biochemistry and Molecular Biology, and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, School of Basic Medical Sciences, Guangzhou, Guangdong, China; Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan International Joint Laboratory of Kidney Disease and Microenvironment, Henan Provincial Clinical Research Center for Kidney Disease, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Henan 450053, China
| | - Liyao Mai
- Department of Biochemistry and Molecular Biology, and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, School of Basic Medical Sciences, Guangzhou, Guangdong, China; Key Laboratory of Conservation and Application in Biodiversity of South China, School of Life Sciences, Guangzhou University, Guangzhou, Guangdong, China
| | - Zebin Wen
- Department of Biochemistry and Molecular Biology, and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, School of Basic Medical Sciences, Guangzhou, Guangdong, China
| | - Min Dai
- Institute of Genetics and Developmental Biology, Innovation Academy of Seed Design, Chinese Academy of Sciences, Beijing, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Siwen Xu
- Department of Biochemistry and Molecular Biology, and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, School of Basic Medical Sciences, Guangzhou, Guangdong, China
| | - Xianwei Lin
- SequMed Institute of Biomedical Sciences, Guangzhou 510530 Guangdong Province, China
| | - Yongjian Luo
- SequMed Institute of Biomedical Sciences, Guangzhou 510530 Guangdong Province, China
| | - Yinbin Qiu
- Department of Biochemistry and Molecular Biology, and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, School of Basic Medical Sciences, Guangzhou, Guangdong, China
| | - Yuting Chen
- Department of Biochemistry and Molecular Biology, and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, School of Basic Medical Sciences, Guangzhou, Guangdong, China; Precision Regenerative Medicine Research Centre, Medical Science Division, and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078, China
| | - Zhanying Dong
- Department of Biochemistry and Molecular Biology, and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, School of Basic Medical Sciences, Guangzhou, Guangdong, China
| | - Caiming Chen
- Department of Biochemistry and Molecular Biology, and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, School of Basic Medical Sciences, Guangzhou, Guangdong, China; Precision Regenerative Medicine Research Centre, Medical Science Division, and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078, China
| | - Wei Meng
- Department of Biochemistry and Molecular Biology, and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, School of Basic Medical Sciences, Guangzhou, Guangdong, China
| | - Xingguang Luo
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Guanchuan Lin
- Department of Biochemistry and Molecular Biology, and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, School of Basic Medical Sciences, Guangzhou, Guangdong, China.
| | - Paul K H Tam
- Precision Regenerative Medicine Research Centre, Medical Science Division, and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078, China.
| | - Xinghua Pan
- Department of Biochemistry and Molecular Biology, and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, School of Basic Medical Sciences, Guangzhou, Guangdong, China; Precision Regenerative Medicine Research Centre, Medical Science Division, and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078, China; Key Laboratory of Infectious Diseases Research in South China (China Ministry Education), Southern Medical University, Guangzhou, Guangdong 510515, China; Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, Guangdong Province 510515, China.
| |
Collapse
|
4
|
Shang K, Huang D, Liu J, Yu Z, Bian W, Chen J, Zhao Y, Liu L, Jiang J, Wang Y, Duan Y, Ge J, Zhang S, Zhou C, Han Y, Hu Y, Zheng W, Sun J, Huang H, Pei S, Qian P, Sun J. CD97-directed CAR-T cells with enhanced persistence eradicate acute myeloid leukemia in diverse xenograft models. Cell Rep Med 2025:102148. [PMID: 40425009 DOI: 10.1016/j.xcrm.2025.102148] [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: 02/15/2024] [Revised: 11/11/2024] [Accepted: 04/30/2025] [Indexed: 05/29/2025]
Abstract
Chimeric antigen receptor (CAR)-T therapy on acute myeloid leukemia (AML) is hindered by the absence of a suitable tumor-specific antigen. Here, we propose CD97 as a potential target for CAR-T therapy against AML based on its broader and higher expression on AML cells compared to normal hematopoietic stem and progenitor cells (HSPCs). To resolve the fratricide problem caused by CD97 expression on T cells, we knock out CD97 in CAR-T cells using CRISPR-Cas9. Our CD97KO CAR-T cells eliminate both AML cell lines and primary AML cells effectively while showing tolerable toxicity to HSPCs. Furthermore, we mutate the CD3ζ domain of the CAR and find that the optimized CD97 CAR-T cells exhibit persistent anti-tumor activity both in vitro and in multiple xenograft models. Mechanistically, transcriptional profiles reveal that the optimized CAR-T cells delay differentiation and resist exhaustion. Collectively, our study supports CD97 as a promising target for CAR-T therapy against AML.
Collapse
Affiliation(s)
- Kai Shang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; Department of Cell Biology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China; Institute of Hematology, Zhejiang University, Hangzhou 310058, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China
| | - Deyu Huang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; Institute of Hematology, Zhejiang University, Hangzhou 310058, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China; Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jun Liu
- Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Zebin Yu
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; Institute of Hematology, Zhejiang University, Hangzhou 310058, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China; Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Wei Bian
- Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China
| | - Jiangqing Chen
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; Department of Cell Biology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China; Institute of Hematology, Zhejiang University, Hangzhou 310058, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China
| | - Yin Zhao
- Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China
| | - Lina Liu
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Jie Jiang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; Department of Cell Biology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China; Institute of Hematology, Zhejiang University, Hangzhou 310058, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China
| | - Yajie Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; Department of Cell Biology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China; Institute of Hematology, Zhejiang University, Hangzhou 310058, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China
| | - Yanting Duan
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; Department of Cell Biology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China; Institute of Hematology, Zhejiang University, Hangzhou 310058, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China
| | - Jingyu Ge
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Shize Zhang
- Center for Genetic Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Chun Zhou
- School of Public Health, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Yingli Han
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; Institute of Hematology, Zhejiang University, Hangzhou 310058, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China; Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yongxian Hu
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; Institute of Hematology, Zhejiang University, Hangzhou 310058, China
| | - Weiyan Zheng
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Jie Sun
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China
| | - He Huang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; Institute of Hematology, Zhejiang University, Hangzhou 310058, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China.
| | - Shanshan Pei
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; Institute of Hematology, Zhejiang University, Hangzhou 310058, China.
| | - Pengxu Qian
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; Institute of Hematology, Zhejiang University, Hangzhou 310058, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China; Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Jie Sun
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; Department of Cell Biology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China; Institute of Hematology, Zhejiang University, Hangzhou 310058, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China.
| |
Collapse
|
5
|
Wu S, Zhao P, Hou Y, He L, Wang Z, Yang D, Chai Y, Liu J, Shen Y, Li A, Cui X, Zhao D, Luo B, Wang J, Liu L, Liao W, Zhang Y. Design, synthesis, and biological evaluation of 4-(2-fluorophenoxy)pyridine derivatives as novel FLT3-ITD inhibitors. Eur J Med Chem 2025; 289:117492. [PMID: 40081102 DOI: 10.1016/j.ejmech.2025.117492] [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: 02/11/2025] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/15/2025]
Abstract
FMS-like tyrosine kinase 3 (FLT3) is an ideal drug target for the treatment of acute myeloid leukemia (AML). Although several FLT3 inhibitors have been approved or evaluated in clinical trials, selectivity over c-Kit kinase and FLT3 WT remains a major challenge. Herein, we report a series of 4-(2-fluorophenoxy)pyridine derivatives with potent inhibitory activities against FLT3 internal tandem duplication (FLT3-ITD). The representative compound 13v inhibited FLT3-ITD kinase and isogenic BaF3-FLT3-ITD cells with nanomolar IC50 values and achieved selectivity over c-Kit (>53-fold) and FLT WT (19-fold) in transformed BaF3 cells. In addition, compound 13v displayed excellent selectivity against FLT3-ITD driven AML cells compared to other leukemia cells, solid tumors, and normal peripheral blood mononuclear cells. Mechanistic studies revealed that 13v disrupted FLT3 signal transduction and induced G0/G1 cell cycle arrest and apoptosis. Moreover, it also showed good developmental profiles in ADME assays. In in vivo studies, 13v demonstrated desirable pharmacokinetic (PK) profiles and sufficient tumor growth inhibition in a MOLM-13 xenograft model. Taken together, 13v may represent a starting point for the development of improved FLT3-ITD inhibitors.
Collapse
Affiliation(s)
- Shengfei Wu
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 550004, China
| | - Pengjuan Zhao
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 550004, China
| | - Youli Hou
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 550004, China
| | - Lihong He
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 550004, China
| | - Zhongyuan Wang
- Department of Pharmacy, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Dan Yang
- Clinical Medical Research Center, The Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Yunyan Zone, Guiyang, 550004, China
| | - Yue'e Chai
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 550004, China
| | - Junji Liu
- Department of Respiratory Medicine, The Central Hospital of Shaoyang, Shaoyang, 422000, China
| | - Yulong Shen
- Department of Radiation Oncology, The Ninth Medical Center of Chinese PLA General Hospital, Beijing, 100101, China
| | - Aihong Li
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 550004, China
| | - Xing Cui
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 550004, China
| | - Dongsheng Zhao
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 550004, China
| | - Bilan Luo
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Jianta Wang
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 550004, China
| | - Lan Liu
- Department of Respiratory Medicine, The Central Hospital of Shaoyang, Shaoyang, 422000, China.
| | - Weike Liao
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 550004, China.
| | - Yu Zhang
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 550004, China.
| |
Collapse
|
6
|
Chen YJ, Zhao Y, Yao MY, Wang YF, Ma M, Yu CC, Jiang HL, Wei W, Shen J, Xu XW, Xie CY. Concurrent inhibition of p300/CBP and FLT3 enhances cytotoxicity and overcomes resistance in acute myeloid leukemia. Acta Pharmacol Sin 2025; 46:1390-1403. [PMID: 39885312 PMCID: PMC12032420 DOI: 10.1038/s41401-025-01479-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 12/22/2024] [Indexed: 02/01/2025]
Abstract
FMS-like tyrosine kinase-3 (FLT3), a class 3 receptor tyrosine kinase, can be activated by mutations of internal tandem duplication (FLT3-ITD) or point mutations in the tyrosine kinase domain (FLT3-TKD), leading to constitutive activation of downstream signaling cascades, including the JAK/STAT5, PI3K/AKT/mTOR and RAS/MAPK pathways, which promote the progression of leukemic cells. Despite the initial promise of FLT3 inhibitors, the discouraging outcomes in the treatment of FLT3-ITD-positive acute myeloid leukemia (AML) promote the pursuit of more potent and enduring therapeutic approaches. The histone acetyltransferase complex comprising the E1A binding protein P300 and its paralog CREB-binding protein (p300/CBP) is a promising therapeutic target, but the development of effective p300/CBP inhibitors faces challenges due to inherent resistance and low efficacy, often exacerbated by the absence of reliable clinical biomarkers for patient stratification. In this study we investigated the role of p300/CBP in FLT3-ITD AML and evaluated the therapeutic potential of targeting p300/CBP alone or in combination with FLT3 inhibitors. We showed that high expression of p300 was significantly associated with poor prognosis in AML patients and positively correlated with FLT3 expression. We unveiled that the p300/CBP inhibitors A485 or CCS1477 dose-dependently downregulated FLT3 transcription via abrogation of histone acetylation in FLT3-ITD AML cells; in contrast, the FLT3 inhibitor quizartinib reduced the level of H3K27Ac. Concurrent inhibition of p300/CBP and FLT3 enhanced the suppression of FLT3 signaling and H3K27 acetylation, concomitantly reducing the phosphorylation of STAT5, AKT, ERK and the expression of c-Myc, thereby leading to synergistic antileukemic effects both in vitro and in vivo. Moreover, we found that p300/CBP-associated transcripts were highly expressed in quizartinib-resistant AML cells with FLT3-TKD mutation. Targeting p300/CBP with A485 or CCS1477 retained the efficacy of quizartinib, suggesting marked synergy when combined with p300/CBP inhibitors in quizartinib-resistant AML models, as well as primary FLT3-ITD+ AML samples. These results demonstrate a potential therapeutic strategy of combining p300/CBP and FLT3 inhibitors to treat FLT3-ITD and FLT3-TKD AML.
Collapse
Affiliation(s)
- Yu-Jun Chen
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Yu Zhao
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | | | - Ya-Fang Wang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Ming Ma
- Lingang Laboratory, Shanghai, 200031, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | | | - Hua-Liang Jiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
- Drug Discovery and Development Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Wu Wei
- Lingang Laboratory, Shanghai, 200031, China
| | - Jie Shen
- Department of Pharmacy, The SATCM Third Grade Laboratory of Traditional Chinese Medicine Preparations, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Xiao-Wei Xu
- Department of Hematology, Shanghai Jiao Tong University School of Medicine Affiliated Shanghai General Hospital, Shanghai, 200080, China.
| | - Cheng-Ying Xie
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China.
- Lingang Laboratory, Shanghai, 200031, China.
| |
Collapse
|
7
|
Shahar Gabay T, Stolero N, Rabhun N, Sabah R, Raz O, Neumeier Y, Marx Z, Tao L, Biezuner T, Amir S, Adar R, Levy R, Chapal-Ilani N, Evtiugina N, Shlush LI, Shapiro E, Yehudai-Resheff S, Zuckerman T. GMP-like and MLP-like Subpopulations of Hematopoietic Stem and Progenitor Cells Harboring Mutated EZH2 and TP53 at Diagnosis Promote Acute Myeloid Leukemia Relapse: Data of Combined Molecular, Functional, and Genomic Single-Stem-Cell Analyses. Int J Mol Sci 2025; 26:4224. [PMID: 40362463 PMCID: PMC12072498 DOI: 10.3390/ijms26094224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/20/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Acute myeloid leukemia (AML) is associated with unfavorable patient outcomes primarily related to disease relapse. Since specific types of leukemic hematopoietic stem and progenitor cells (HSPCs) are suggested to contribute to AML propagation, this study aimed to identify and explore relapse-initiating HSPC subpopulations present at diagnosis, using single-cell analysis (SCA). We developed unique high-resolution techniques capable of tracking single-HSPC-derived subclones during AML evolution. Each subclone was evaluated for chemo-resistance, in vivo leukemogenic potential, mutational profile, and the cell of origin. In BM samples of 15 AML patients, GMP-like and MLP-like HSPC subpopulations were identified as prevalent at relapse, exhibiting chemo-resistance to commonly used chemotherapy agents cytosine arabinoside (Ara-C) and daunorubicin. Reconstruction of phylogenetic lineage trees combined with genetic analysis of single HSPCs and single-HSPC-derived subclones demonstrated two distinct clusters, originating from MLP-like or GMP-like subpopulations, observed both at diagnosis and relapse. These subpopulations induced leukemia development ex vivo and in vivo. Genetic SCA showed that these relapse-related subpopulations harbored mutated EZH2 and TP53, detected already at diagnosis. This study, using combined molecular, functional, and genomic analyses at the level of single cells, identified patient-specific chemo-resistant HSPC subpopulations at the time of diagnosis, promoting AML relapse.
Collapse
MESH Headings
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/drug therapy
- Humans
- Enhancer of Zeste Homolog 2 Protein/genetics
- Single-Cell Analysis/methods
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/pathology
- Tumor Suppressor Protein p53/genetics
- Mutation
- Animals
- Male
- Female
- Middle Aged
- Mice
- Recurrence
- Adult
- Cytarabine/pharmacology
- Drug Resistance, Neoplasm/genetics
- Aged
- Neoplastic Stem Cells/metabolism
Collapse
Affiliation(s)
- Tal Shahar Gabay
- Hematology Research Center, Clinical Research Institute at Rambam, Rambam Health Care Campus, Haifa 3109601, Israel; (T.S.G.); (R.S.); (S.Y.-R.)
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa 3109601, Israel
| | - Nofar Stolero
- Hematology Research Center, Clinical Research Institute at Rambam, Rambam Health Care Campus, Haifa 3109601, Israel; (T.S.G.); (R.S.); (S.Y.-R.)
| | - Niv Rabhun
- Hematology Research Center, Clinical Research Institute at Rambam, Rambam Health Care Campus, Haifa 3109601, Israel; (T.S.G.); (R.S.); (S.Y.-R.)
| | - Rawan Sabah
- Hematology Research Center, Clinical Research Institute at Rambam, Rambam Health Care Campus, Haifa 3109601, Israel; (T.S.G.); (R.S.); (S.Y.-R.)
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa 3109601, Israel
| | - Ofir Raz
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 761001, Israel; (O.R.); (Y.N.); (L.T.); (R.L.); (E.S.)
| | - Yaara Neumeier
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 761001, Israel; (O.R.); (Y.N.); (L.T.); (R.L.); (E.S.)
- Department of Immunology, Weizmann Institute of Science, Rehovot 761001, Israel; (T.B.); (N.C.-I.); (L.I.S.)
| | - Zipora Marx
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 761001, Israel; (O.R.); (Y.N.); (L.T.); (R.L.); (E.S.)
| | - Liming Tao
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 761001, Israel; (O.R.); (Y.N.); (L.T.); (R.L.); (E.S.)
| | - Tamir Biezuner
- Department of Immunology, Weizmann Institute of Science, Rehovot 761001, Israel; (T.B.); (N.C.-I.); (L.I.S.)
| | - Shiran Amir
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 761001, Israel; (O.R.); (Y.N.); (L.T.); (R.L.); (E.S.)
| | - Rivka Adar
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 761001, Israel; (O.R.); (Y.N.); (L.T.); (R.L.); (E.S.)
| | - Ron Levy
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 761001, Israel; (O.R.); (Y.N.); (L.T.); (R.L.); (E.S.)
| | - Noa Chapal-Ilani
- Department of Immunology, Weizmann Institute of Science, Rehovot 761001, Israel; (T.B.); (N.C.-I.); (L.I.S.)
| | - Natalia Evtiugina
- Hematology Research Center, Clinical Research Institute at Rambam, Rambam Health Care Campus, Haifa 3109601, Israel; (T.S.G.); (R.S.); (S.Y.-R.)
| | - Liran I. Shlush
- Department of Immunology, Weizmann Institute of Science, Rehovot 761001, Israel; (T.B.); (N.C.-I.); (L.I.S.)
| | - Ehud Shapiro
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 761001, Israel; (O.R.); (Y.N.); (L.T.); (R.L.); (E.S.)
| | - Shlomit Yehudai-Resheff
- Hematology Research Center, Clinical Research Institute at Rambam, Rambam Health Care Campus, Haifa 3109601, Israel; (T.S.G.); (R.S.); (S.Y.-R.)
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa 3109601, Israel
| | - Tsila Zuckerman
- Hematology Research Center, Clinical Research Institute at Rambam, Rambam Health Care Campus, Haifa 3109601, Israel; (T.S.G.); (R.S.); (S.Y.-R.)
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa 3109601, Israel
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa 3109601, Israel
| |
Collapse
|
8
|
Cao S, Wang Q, Zhu G. From Chemotherapy to Targeted Therapy: Unraveling Resistance in Acute Myeloid Leukemia Through Genetic and Non-Genetic Insights. Int J Mol Sci 2025; 26:4005. [PMID: 40362245 PMCID: PMC12071668 DOI: 10.3390/ijms26094005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/04/2025] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
Acute myeloid leukemia (AML) is a devastating disease characterized by extensive inter-patient and intra-patient heterogeneity. Despite the introduction of intensive chemotherapy in the 1970s as the standard treatment, the development of mechanism-based targeted therapies since 2017 has been broadening the therapeutic landscape. However, both chemotherapy and targeted therapies continue to face the challenges of primary and secondary resistance. This review summarizes the mechanisms underlying resistance to chemotherapy and targeted therapies in AML and discusses the opportunities and challenges brought by the transition from chemotherapy to precision medicine.
Collapse
Affiliation(s)
| | | | - Ganqian Zhu
- School of Biomedical Sciences, Hunan University, Changsha 410082, China; (S.C.); (Q.W.)
| |
Collapse
|
9
|
Chow RD, Velu P, Deihimi S, Belman J, Youn A, Shah N, Luger SM, Carroll MP, Morrissette J, Bowman RL. Persistent postremission clonal hematopoiesis shapes the relapse trajectories of acute myeloid leukemia. Blood Adv 2025; 9:1888-1899. [PMID: 39938015 PMCID: PMC12008691 DOI: 10.1182/bloodadvances.2024015149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/22/2025] [Accepted: 01/28/2025] [Indexed: 02/14/2025] Open
Abstract
ABSTRACT Mutations found in acute myeloid leukemia (AML) such as DNMT3A, TET2, and ASXL1 can be found in the peripheral blood of healthy adults, a phenomenon termed clonal hematopoiesis (CH). These mutations are thought to represent the earliest genetic events in the evolution of AML. Genomic studies on samples acquired at diagnosis, remission, and at relapse have demonstrated significant stability of CH mutations after induction chemotherapy. Meanwhile, later mutations in genes such as NPM1 and FLT3 have been shown to contract at remission, and in the case of FLT3 often are absent at relapse. We sought to understand how early CH mutations influence subsequent evolutionary trajectories throughout remission and relapse in response to induction chemotherapy. We assembled a retrospective cohort of patients diagnosed with de novo AML at our institution that underwent genomic sequencing at diagnosis, remission, and/or relapse (total N = 182 patients). FLT3 and NPM1 mutations were generally eliminated at complete remission but subsequently reemerged upon relapse, whereas DNMT3A, TET2, and ASXL1 mutations often persisted through remission. CH-related mutations exhibited distinct constellations of co-occurring genetic alterations, with NPM1 and FLT3 mutations enriched in DNMT3Amut AML, whereas CBL and SRSF2 mutations were enriched in TET2mut and ASXL1mut AML, respectively. In the case of NPM1 and FLT3 mutations, these differences vanished at the time of complete remission yet readily reemerged upon relapse, indicating the reproducible nature of these genetic interactions. Thus, CH-associated mutations that likely precede malignant transformation subsequently shape the evolutionary trajectories of AML through diagnosis, therapy, and relapse.
Collapse
Affiliation(s)
- Ryan D. Chow
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Priya Velu
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Pathology and Laboratory Medicine, Weill Cornell School of Medicine, Cornell University, New York, NY
| | - Safoora Deihimi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jonathan Belman
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Angela Youn
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Nisargbhai Shah
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Selina M. Luger
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Martin P. Carroll
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jennifer Morrissette
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Robert L. Bowman
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
10
|
Wang X, Jin J, Yan H, Liu J, Huang S, Bai H, Guo M, Cheng X, Deng T, Ba Y, Gu Y, Gao X, Hu D. The mRNA export pathway licenses viral mimicry response and antitumor immunity by actively exporting nuclear retroelement transcripts. Sci Transl Med 2025; 17:eado4370. [PMID: 40203080 DOI: 10.1126/scitranslmed.ado4370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 08/07/2024] [Accepted: 03/06/2025] [Indexed: 04/11/2025]
Abstract
Nuclear retroelement transcripts (RTs), which can be elicited both transcriptionally and posttranscriptionally, form double-stranded RNA (dsRNA) in cytosol to trigger the viral mimicry response (VMR) and antitumor immunity. However, the strength of the induced VMR varies tremendously across tumor types, and the underlying mechanisms remain poorly understood. Here, we demonstrate that the mRNA export pathway modulates the VMR through actively exporting nuclear RTs for cytosolic dsRNA formation after their induction. Tumor cells hijack this process for immune evasion through aberrant coactivator-associated arginine methyltransferase 1 (CARM1) expression. Mechanistically, we show that the cytoplasmic transportation of RTs by the mRNA export pathway is counteracted by the RNA exosome, which cleaves multiple transcripts within this pathway, including those encoding the essential DExD-box helicase 39A (DDX39A) and the adaptor protein ALYREF. CARM1 enhances the RNA exosome activity to attenuate the nuclear export of RTs by the mRNA export pathway through two synergistic mechanisms: (i) transcriptionally activating several RNA exosome components and (ii) posttranslationally methylating arginine 6 of the RNA exosome subunit EXOSC1, which protects it from proteasome-mediated degradation. Collectively, our study highlights the critical active regulatory role of the mRNA export pathway in transporting nuclear RTs into the cytosol for triggering the VMR and tumor immunity. Furthermore, we propose that enhancing the mRNA export pathway activity, either through CARM1 inhibition or RNA exosome modulation, could reinforce the therapeutic agent-induced VMR, thus holding the promise for overcoming tumor immune evasion and immunotherapy resistance.
Collapse
Affiliation(s)
- Xiaoqiang Wang
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Jiaxing Jin
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Han Yan
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Jinhua Liu
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Shan Huang
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Hui Bai
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Mingrui Guo
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Xinyue Cheng
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Ting Deng
- Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Yi Ba
- Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Yong Gu
- Clinical Research Center, Hainan Hospital, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Hainan, 570203, China
| | - Xin Gao
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Deqing Hu
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
- Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| |
Collapse
|
11
|
Elmenawi S, Fawzy M. 15 Years Old ALK Gene from Birth to Adolescence; Where to in NBL. Curr Oncol Rep 2025; 27:431-445. [PMID: 40064818 PMCID: PMC11976753 DOI: 10.1007/s11912-025-01650-w] [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] [Accepted: 02/03/2025] [Indexed: 04/09/2025]
Abstract
PURPOSE OF REVIEW This review provides a comprehensive understanding of the ALK gene, encompassing its prevalence, genetic alterations, and significance in neuroblastoma diagnosis, outcome prediction, and targeted therapy utilization. The insights presented aim to inform future research directions and clinical practices in this field. RECENT FINDINGS High risk neuroblastoma, comprising approximately 50% of all cases, presents a particularly poor prognosis. In 2008, the discovery of ALK aberrations in neuroblastoma marked a significant breakthrough, leading to the recognition of ALK as a target for tumors with activating ALK alterations. This discovery has paved the way for the development of various ALK inhibitors, which have shown promising clinical efficacy. ALK amplification, often observed alongside MYCN amplification, has been associated with unfavorable outcomes in patients. Activating mutations in the kinase domain of ALK, particularly at hotspot positions F1174, R1275, and F1245, have been identified. These mutations can occur at clonal or subclonal levels, posing challenges for early detection and potentially influencing disease progression and therapy resistance. The availability of ALK inhibitors, initially developed for adult cancers, has expedited the translation of this knowledge into targeted therapies for neuroblastoma. However, resistance to ALK inhibitors can emerge as a result of treatment or preexist as subclones within the tumor prior to therapy. Future trials should focus on identifying additional targets complementing ALK inhibition to enhance treatment efficacy and overcome acquired resistance. Furthermore, the utilization of circulating tumor DNA as a non-invasive approach for longitudinal monitoring of ALK-positive neuroblastoma patients, in combination with radiographic evaluation of treatment response, holds promise for understanding dynamic tumor changes over time.
Collapse
Affiliation(s)
- Salma Elmenawi
- Clinical Research Department, Children's Cancer Hospital Egypt, 57357, 1-Sekket Elemam-Sayeda Zeinab, Cairo, Egypt.
| | - Mohamed Fawzy
- Pediatric Oncology Department, Children's Cancer Hospital Egypt, 57357, 1-Sekket Elemam-Sayeda Zeinab, Cairo, Egypt
- Pediatric Oncology Department, National Cancer Institute, Cairo, Egypt
| |
Collapse
|
12
|
Morgenstern Y, Lee J, Na Y, Lieng BY, Ly NS, Gwynne WD, Hurren R, Ma L, Ling D, Gronda M, Arruda A, Frisch A, Zuckerman T, Ofran Y, Minden MD, Zhang L, O'Brien C, Quaile AT, Montenegro-Burke JR, Schimmer AD. Acute myeloid leukemia drug-tolerant persister cells survive chemotherapy by transiently increasing plasma membrane rigidity, that also increases their sensitivity to immune cell killing. Haematologica 2025; 110:893-903. [PMID: 39568440 PMCID: PMC11962361 DOI: 10.3324/haematol.2024.286018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 11/14/2024] [Indexed: 11/22/2024] Open
Abstract
Resistance to chemotherapy remains a major hurdle to the cure of patients with acute myeloid leukemia (AML). Recent studies indicate that a minority of malignant cells, termed drug-tolerant persisters (DTP), stochastically upregulate stress pathways to evade cell death upon acute exposure to chemotherapy without acquiring new genetic mutations. This chemoresistant state is transient and the cells return to the baseline state after removal of chemotherapy. Nevertheless, the mechanisms employed by DTP to resist chemotherapy are not well understood and it is largely unknown whether these mechanisms are also seen in patients receiving chemotherapy. Here, we used leukemia cell lines, primary AML patients' samples and samples from patients with AML receiving systemic chemotherapy to study the DTP state. We demonstrated that a subset of AML cells transiently increases membrane rigidity to resist killing due to acute exposure to daunorubicin and Ara-C. Upon removal of the chemotherapy, membrane rigidity returned to baseline and the cells regained chemosensitivity. Although resistant to chemotherapy, the increased membrane rigidity rendered AML cells more susceptible to T-cell-mediated killing. Thus, we identified a novel mechanism by which DTP leukemic cells evade chemotherapy and a strategy to eradicate these persistent cells.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/metabolism
- Drug Resistance, Neoplasm
- Cell Membrane/metabolism
- Cell Membrane/drug effects
- Cell Line, Tumor
- Daunorubicin/pharmacology
- Cytarabine/pharmacology
- Antineoplastic Agents/pharmacology
Collapse
Affiliation(s)
- Yael Morgenstern
- Princess Margaret Cancer Centre, University Health Network, Toronto
| | - JongBok Lee
- Toronto General Hospital Research Institute, University Health Network, Toronto
| | - Yoosu Na
- Toronto General Hospital Research Institute, University Health Network, Toronto
| | - Brandon Y Lieng
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto
| | - Nicholas S Ly
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto
| | - William D Gwynne
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto
| | - Rose Hurren
- Princess Margaret Cancer Centre, University Health Network, Toronto
| | - Li Ma
- Princess Margaret Cancer Centre, University Health Network, Toronto
| | - Dakai Ling
- Princess Margaret Cancer Centre, University Health Network, Toronto
| | - Marcela Gronda
- Princess Margaret Cancer Centre, University Health Network, Toronto
| | - Andrea Arruda
- Princess Margaret Cancer Centre, University Health Network, Toronto
| | - Avraham Frisch
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
| | - Tsila Zuckerman
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
| | - Yishai Ofran
- Hematology and Stem cell transplantation department and the Eisenberg R-D Authority, Shaare Zedek medical center, Hebrew University Jerusalem, Israel
| | - Mark D Minden
- Princess Margaret Cancer Centre, University Health Network, Toronto
| | - Li Zhang
- Toronto General Hospital Research Institute, University Health Network, Toronto
| | | | - Andrew T Quaile
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto
| | | | - Aaron D Schimmer
- Princess Margaret Cancer Centre, University Health Network, Toronto.
| |
Collapse
|
13
|
Wang X, Wang K, Zhang W, Tang Z, Zhang H, Cheng Y, Zhou D, Zhang C, Zhong WZ, Ma Q, Xu J, Hu Z. Clonal expansion dictates the efficacy of mitochondrial lineage tracing in single cells. Genome Biol 2025; 26:70. [PMID: 40134031 PMCID: PMC11938731 DOI: 10.1186/s13059-025-03540-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 03/11/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Mitochondrial DNA (mtDNA) variants hold promise as endogenous barcodes for tracking human cell lineages, but their efficacy as reliable lineage markers are hindered by the complex dynamics of mtDNA in somatic tissues. RESULTS Here, we use computational modeling and single-cell genomics to thoroughly interrogate the origin and clonal dynamics of mtDNA variants across various biological settings. Our findings reveal that the majority of mtDNA variants which are specifically present in a cell subpopulation, termed subpopulation-specific variants, are pre-existing heteroplasmies in the first cell instead of de novo somatic mutations during divisions. Moreover, subpopulation-specific variants demonstrate limited discriminatory power among different genuine lineages under weak clonal expansion; however, certain subpopulation-specific variants with consistently high frequencies among a subpopulation are capable of faithfully labeling cell lineages in scenarios of stringent clonal expansion, such as strongly expanded T cell populations in diseased conditions and clonal hematopoiesis in aged individuals. Inspired by our simulations, we introduce a lineage informative score, facilitating the identification of reliable mitochondrial lineage tracing markers across different modalities of single-cell genomic data. CONCLUSIONS Combining computational modeling and single-cell sequencing, our study reveals that the performance of mitochondrial lineage tracing is highly dependent on the extent of clonal expansion, which thus should be considered when applying mitochondrial lineage tracing.
Collapse
Affiliation(s)
- Xin Wang
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Kun Wang
- School of Mathematical Sciences, Xiamen University, Xiamen, China
| | - Weixing Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Zhongjie Tang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Hao Zhang
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yuying Cheng
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- School of Life Sciences, Henan University, Kaifeng, China
| | - Da Zhou
- School of Mathematical Sciences, Xiamen University, Xiamen, China
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, China
| | - Chao Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Wen-Zhao Zhong
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Qing Ma
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
- Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| | - Jin Xu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, China.
| | - Zheng Hu
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| |
Collapse
|
14
|
Kempter T, Richter-Pechańska P, Michel K, Rausch T, Erarslan-Uysal B, Eckert C, Zimmermann M, Stanulla M, Schrappe M, Cario G, Köhrer S, Attarbaschi A, Korbel JO, Kunz JB, Kulozik AE. Subclonal TP53 and KRAS variants combined with poor treatment response identify ultrahigh-risk pediatric patients with T-ALL. Blood Adv 2025; 9:1267-1279. [PMID: 39808796 PMCID: PMC11950767 DOI: 10.1182/bloodadvances.2024014209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/28/2024] [Accepted: 11/14/2024] [Indexed: 01/16/2025] Open
Abstract
ABSTRACT Variations in the TP53 and KRAS genes indicate a particularly adverse prognosis in relapsed pediatric T-cell acute lymphoblastic leukemia (T-ALL). We hypothesized that these variations might be subclonally present at disease onset and contribute to relapse risk. To test this, we examined 2 cohorts of children diagnosed with T-ALL: cohort 1 with 81 patients who relapsed and 79 who matched nonrelapsing controls, and cohort 2 with 226 consecutive patients, 30 of whom relapsed. In cohort 1, targeted sequencing revealed TP53 clonal and subclonal variants in 6 of 81 relapsing patients but none in the nonrelapsing group (P = .014). KRAS alterations were found in 9 of 81 relapsing patients compared with 2 of 79 nonrelapsing patients (P = .032). Survival analysis showed that none of the relapsed patients with TP53 and/or KRAS alterations survived, whereas 19 of 67 relapsed patients without such variants did, with a minimum follow-up time of 3 years (P = .023). In cohort 2, none of the relapsing patients but 10 of 196 nonrelapsing patients carried TP53 or KRAS variants, indicating that mutation status alone does not predict poor prognosis. All 10 nonrelapsing patients with mutations had a favorable early treatment response. Among the total cohort of 386 patients, 188 showed poor treatment response, of whom 69 relapsed. Of these poor responders, 9 harbored TP53 or KRAS variants. In conclusion, subclonal TP53 and KRAS alterations identified at the time of initial diagnosis, along with a poor treatment response, characterize a subset of children with T-ALL who face a dismal prognosis and who may benefit from alternative treatment approaches.
Collapse
Affiliation(s)
- Tamara Kempter
- Department of Pediatric Oncology, Hematology, and Immunology and Hopp Children’s Cancer Center Heidelberg, University of Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, University of Heidelberg, Heidelberg, Germany
| | - Paulina Richter-Pechańska
- Department of Pediatric Oncology, Hematology, and Immunology and Hopp Children’s Cancer Center Heidelberg, University of Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, University of Heidelberg, Heidelberg, Germany
| | - Katarzyna Michel
- Department of Pediatric Oncology, Hematology, and Immunology and Hopp Children’s Cancer Center Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Tobias Rausch
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, University of Heidelberg, Heidelberg, Germany
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Büşra Erarslan-Uysal
- Department of Pediatric Oncology, Hematology, and Immunology and Hopp Children’s Cancer Center Heidelberg, University of Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, University of Heidelberg, Heidelberg, Germany
| | - Cornelia Eckert
- Department of Pediatric Oncology/Hematology, Charité University School of Medicine Berlin, Berlin, Germany
| | - Martin Zimmermann
- Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Martin Stanulla
- Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Martin Schrappe
- Department of Pediatrics, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Gunnar Cario
- Department of Pediatrics, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Stefan Köhrer
- Labdia Labordiagnostik, Vienna, Austria
- St. Anna Children’s Cancer Research Institute, Vienna, Austria
| | - Andishe Attarbaschi
- St. Anna Children’s Cancer Research Institute, Vienna, Austria
- Department of Pediatric Hematology and Oncology, St. Anna Children’s Hospital, Medical University of Vienna, Vienna, Austria
| | - Jan O. Korbel
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, University of Heidelberg, Heidelberg, Germany
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Bridging Research Division on Mechanisms of Genomic Variation and Data Science, German Cancer Research Center, Heidelberg, Germany
| | - Joachim B. Kunz
- Department of Pediatric Oncology, Hematology, and Immunology and Hopp Children’s Cancer Center Heidelberg, University of Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, University of Heidelberg, Heidelberg, Germany
| | - Andreas E. Kulozik
- Department of Pediatric Oncology, Hematology, and Immunology and Hopp Children’s Cancer Center Heidelberg, University of Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, University of Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Leukemia, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
15
|
Decker M, Huang BJ, Ware T, Boone C, Tang M, Ybarra J, Ballapuram AC, Taran KA, Chen PY, Amendáriz M, Leung CJ, Harris M, Tjoa K, Hongo H, Abelson S, Rivera J, Ngo N, Herbst DM, Suciu RM, Guijas C, Sedighi K, Andalis T, Roche E, Xie B, Liu Y, Smith CC, Stieglitz E, Niphakis MJ, Cravatt BF, Shannon K. In vivo validation of the palmitoylation cycle as a therapeutic target in NRAS-mutant cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.20.644389. [PMID: 40166265 PMCID: PMC11957127 DOI: 10.1101/2025.03.20.644389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Normal and oncogenic Ras proteins are functionally dependent on one or more lipid modifications1,2. Whereas K-Ras4b farnesylation is sufficient for stable association with the plasma membrane, farnesylated H-Ras, K-Ras4a, and N-Ras traffic to the Golgi where they must undergo palmitoylation before regulated translocation to cell membranes. N-Ras palmitoylation by the DHHC family of palmitoyl acyl transferases (PATs) and depalmitoylation by ABHD17 serine hydrolases is a dynamic process that is essential for the growth of acute myeloid leukemias (AMLs) harboring oncogenic NRAS mutations3-6. Here, we have tested whether co-targeting ABHD17 enzymes and Ras signal output would cooperatively inhibit the proliferation and survival of NRAS-mutant AMLs while sparing normal tissues that retain K-Ras4b function. We show that ABD778, a potent and selective ABHD17 inhibitor with in vivo activity, selectively reduces the growth of NRAS-mutant AML cells in vitro and is synergistic with the allosteric MEK inhibitor PD0325901 (PD901)7,8. Similarly, ABD778 and PD901 significantly extended the survival of recipient mice transplanted with three independent primary mouse AMLs harboring an oncogenic Nras G12D driver mutation. Resistant leukemias that emerged during continuous drug treatment acquired by-pass mutations that confer adaptive drug resistance and increase mitogen activated protein kinase (MAPK) signal output. ABD778 augmented the anti-leukemia activity of the pan-PI3 kinase inhibitor pictilisib9, the K/N-RasG12C inhibitor sotorasib10, and the FLT3 inhibitor gilteritinib11. Co-treatment with ABD778 and gilteritinib restored drug sensitivity in a patient-derived xenograft model of adaptive resistance to FLT3 inhibition. These data validate the palmitoylation cycle as a promising therapeutic target in AML and support exploring it in other NRAS-mutant cancers.
Collapse
Affiliation(s)
- Matthew Decker
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Benjamin J. Huang
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Timothy Ware
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Christopher Boone
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Michelle Tang
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Julia Ybarra
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | | | - Katrine A. Taran
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Pan-Yu Chen
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Marcos Amendáriz
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Camille J. Leung
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Max Harris
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Karensa Tjoa
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Henry Hongo
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Sydney Abelson
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Jose Rivera
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Nhi Ngo
- Lundbeck La Jolla Research Center, Inc., San Diego, CA, USA
| | | | - Radu M. Suciu
- Lundbeck La Jolla Research Center, Inc., San Diego, CA, USA
| | - Carlos Guijas
- Lundbeck La Jolla Research Center, Inc., San Diego, CA, USA
| | - Kimia Sedighi
- Lundbeck La Jolla Research Center, Inc., San Diego, CA, USA
| | - Taylor Andalis
- Lundbeck La Jolla Research Center, Inc., San Diego, CA, USA
| | - Elysia Roche
- Lundbeck La Jolla Research Center, Inc., San Diego, CA, USA
| | - Boer Xie
- Lundbeck La Jolla Research Center, Inc., San Diego, CA, USA
| | - Yunlong Liu
- Center for Center for Medical Genomics, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Catherine C. Smith
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Elliot Stieglitz
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | | | | | - Kevin Shannon
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
16
|
Fu YC, Liang SB, Luo M, Wang XP. Intratumoral heterogeneity and drug resistance in cancer. Cancer Cell Int 2025; 25:103. [PMID: 40102941 PMCID: PMC11917089 DOI: 10.1186/s12935-025-03734-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 03/06/2025] [Indexed: 03/20/2025] Open
Abstract
Intratumoral heterogeneity is the main cause of tumor treatment failure, varying across disease sites (spatial heterogeneity) and polyclonal properties of tumors that evolve over time (temporal heterogeneity). As our understanding of intratumoral heterogeneity, the formation of which is mainly related to the genomic instability, epigenetic modifications, plastic gene expression, and different microenvironments, plays a substantial role in drug-resistant as far as tumor metastasis and recurrence. Understanding the role of intratumoral heterogeneity, it becomes clear that a single therapeutic agent or regimen may only be effective for subsets of cells with certain features, but not for others. This necessitates a shift from our current, unchanging treatment approach to one that is tailored against the killing patterns of cancer cells in different clones. In this review, we discuss recent evidence concerning global perturbations of intratumoral heterogeneity, associations of specific intratumoral heterogeneity in lung cancer, the underlying mechanisms of intratumoral heterogeneity potentially leading to formation, and how it drives drug resistance. Our findings highlight the most up-to-date progress in intratumoral heterogeneity and its role in mediating tumor drug resistance, which could support the development of future treatment strategies.
Collapse
Affiliation(s)
- Yue-Chun Fu
- Department of Clinical Laboratory, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Shao-Bo Liang
- Department of Radiation Oncology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Min Luo
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| | - Xue-Ping Wang
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
17
|
Huang S, Soto AM, Sonnenschein C. The end of the genetic paradigm of cancer. PLoS Biol 2025; 23:e3003052. [PMID: 40100793 DOI: 10.1371/journal.pbio.3003052] [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: 03/20/2025] Open
Abstract
Genome sequencing of cancer and normal tissues, alongside single-cell transcriptomics, continues to produce findings that challenge the idea that cancer is a 'genetic disease', as posited by the somatic mutation theory (SMT). In this prevailing paradigm, tumorigenesis is caused by cancer-driving somatic mutations and clonal expansion. However, results from tumor sequencing, motivated by the genetic paradigm itself, create apparent 'paradoxes' that are not conducive to a pure SMT. But beyond genetic causation, the new results lend credence to old ideas from organismal biology. To resolve inconsistencies between the genetic paradigm of cancer and biological reality, we must complement deep sequencing with deep thinking: embrace formal theory and historicity of biological entities, and (re)consider non-genetic plasticity of cells and tissues. In this Essay, we discuss the concepts of cell state dynamics and tissue fields that emerge from the collective action of genes and of cells in their morphogenetic context, respectively, and how they help explain inconsistencies in the data in the context of SMT.
Collapse
Affiliation(s)
- Sui Huang
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Ana M Soto
- Tufts University School of Medicine, Immunology, Boston, Massachusetts, United States of America
| | - Carlos Sonnenschein
- Tufts University School of Medicine, Immunology, Boston, Massachusetts, United States of America
| |
Collapse
|
18
|
Chen Y, Yin Z, Westover KD, Zhou Z, Shu L. Advances and Challenges in RAS Signaling Targeted Therapy in Leukemia. Mol Cancer Ther 2025; 24:33-46. [PMID: 39404173 DOI: 10.1158/1535-7163.mct-24-0504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/04/2024] [Accepted: 10/08/2024] [Indexed: 01/03/2025]
Abstract
RAS mutations are prevalent in leukemia, including mutations at G12, G13, T58, Q61, K117, and A146. These mutations are often crucial for tumor initiation, maintenance, and recurrence. Although much is known about RAS function in the last 40 years, a substantial knowledge gap remains in understanding the mutation-specific biological activities of RAS in cancer and the approaches needed to target specific RAS mutants effectively. The recent approval of KRASG12C inhibitors, adagrasib and sotorasib, has validated KRAS as a direct therapeutic target and demonstrated the feasibility of selectively targeting specific RAS mutants. Nevertheless, KRASG12C remains the only RAS mutant successfully targeted with FDA-approved inhibitors for cancer treatment in patients, limiting its applicability for other oncogenic RAS mutants, such as G12D, in leukemia. Despite these challenges, new approaches have generated optimism about targeting specific RAS mutations in an allele-dependent manner for cancer therapy, supported by compelling biochemical and structural evidence, which inspires further exploration of RAS allele-specific vulnerabilities. This review will discuss the recent advances and challenges in the development of therapies targeting RAS signaling, highlight emerging therapeutic strategies, and emphasize the importance of allele-specific approaches for leukemia treatment.
Collapse
Affiliation(s)
- Yu Chen
- Department of Immunology, Guizhou Province Key Laboratory for Regenerative Medicine, Clinical Research Center, School of Basic Medicine, Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China
| | - Zhenghao Yin
- Department of Immunology, Guizhou Province Key Laboratory for Regenerative Medicine, Clinical Research Center, School of Basic Medicine, Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China
| | - Kenneth D Westover
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, Texas
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, Texas
| | - Zhiwei Zhou
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, Texas
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, Texas
| | - Liping Shu
- Department of Immunology, Guizhou Province Key Laboratory for Regenerative Medicine, Clinical Research Center, School of Basic Medicine, Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China
- Key Laboratory of Adult Stem Cell Translational Research, Chinese Academy of Medical Sciences, Guiyang, China
| |
Collapse
|
19
|
Leppä AM, Grimes K, Jeong H, Huang FY, Andrades A, Waclawiczek A, Boch T, Jauch A, Renders S, Stelmach P, Müller-Tidow C, Karpova D, Sohn M, Grünschläger F, Hasenfeld P, Benito Garagorri E, Thiel V, Dolnik A, Rodriguez-Martin B, Bullinger L, Mrózek K, Eisfeld AK, Krämer A, Sanders AD, Korbel JO, Trumpp A. Single-cell multiomics analysis reveals dynamic clonal evolution and targetable phenotypes in acute myeloid leukemia with complex karyotype. Nat Genet 2024; 56:2790-2803. [PMID: 39587361 PMCID: PMC11631769 DOI: 10.1038/s41588-024-01999-x] [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: 06/22/2023] [Accepted: 10/15/2024] [Indexed: 11/27/2024]
Abstract
Chromosomal instability is a major driver of intratumoral heterogeneity (ITH), promoting tumor progression. In the present study, we combined structural variant discovery and nucleosome occupancy profiling with transcriptomic and immunophenotypic changes in single cells to study ITH in complex karyotype acute myeloid leukemia (CK-AML). We observed complex structural variant landscapes within individual cells of patients with CK-AML characterized by linear and circular breakage-fusion-bridge cycles and chromothripsis. We identified three clonal evolution patterns in diagnosis or salvage CK-AML (monoclonal, linear and branched polyclonal), with 75% harboring multiple subclones that frequently displayed ongoing karyotype remodeling. Using patient-derived xenografts, we demonstrated varied clonal evolution of leukemic stem cells (LSCs) and further dissected subclone-specific drug-response profiles to identify LSC-targeting therapies, including BCL-xL inhibition. In paired longitudinal patient samples, we further revealed genetic evolution and cell-type plasticity as mechanisms of disease progression. By dissecting dynamic genomic, phenotypic and functional complexity of CK-AML, our findings offer clinically relevant avenues for characterizing and targeting disease-driving LSCs.
Collapse
Affiliation(s)
- Aino-Maija Leppä
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Karen Grimes
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
| | - Hyobin Jeong
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Republic of Korea
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Frank Y Huang
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Alvaro Andrades
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
| | - Alexander Waclawiczek
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
| | - Tobias Boch
- University Hospital Mannheim, Heidelberg University, Mannheim, Germany
| | - Anna Jauch
- Institute of Human Genetics, University of Heidelberg, Heidelberg, Germany
| | - Simon Renders
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Patrick Stelmach
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Darja Karpova
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
| | - Markus Sohn
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
| | - Florian Grünschläger
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Patrick Hasenfeld
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
| | | | - Vera Thiel
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Anna Dolnik
- Charité Medical Department, Division of Hematology, Oncology and Tumor Immunology, Berlin, Germany
| | | | - Lars Bullinger
- Charité Medical Department, Division of Hematology, Oncology and Tumor Immunology, Berlin, Germany
| | - Krzysztof Mrózek
- Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
- Clara D. Bloomfield Center for Leukemia Outcomes Research, Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Ann-Kathrin Eisfeld
- Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
- Clara D. Bloomfield Center for Leukemia Outcomes Research, Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Alwin Krämer
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Ashley D Sanders
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
- Charité-Universitätsmedizin, Berlin, Germany
| | - Jan O Korbel
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany.
- Bridging Research Division on Mechanisms of Genomic Variation and Data Science, German Cancer Research Center, Heidelberg, Germany.
| | - Andreas Trumpp
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany.
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany.
- German Cancer Consortium (DKTK), Heidelberg, Germany.
| |
Collapse
|
20
|
Carranza-Aranda AS, Jave-Suárez LF, Flores-Hernández FY, Huizar-López MDR, Herrera-Rodríguez SE, Santerre A. In silico and in vitro study of FLT3 inhibitors and their application in acute myeloid leukemia. Mol Med Rep 2024; 30:229. [PMID: 39392050 PMCID: PMC11475230 DOI: 10.3892/mmr.2024.13353] [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: 06/05/2024] [Accepted: 09/04/2024] [Indexed: 10/12/2024] Open
Abstract
Acute myeloid leukemia (AML) is the most common hematological cancer in the adult population worldwide. Approximately 35% of patients with AML present internal tandem duplication (ITD) mutations in the FMS‑like tyrosine kinase 3 (FLT3) receptor associated with poor prognosis, and thus, this receptor is a relevant target for potential therapeutics. Tyrosine kinase inhibitors (TKIs) are used to treat AML; however, their molecular interactions and effects on leukemic cells are poorly understood. The present study aimed to gain insights into the molecular interactions and affinity forces of four TKI drugs (sorafenib, midostaurin, gilteritinib and quizartinib) with the wild‑type (WT)‑FLT3 and ITD‑mutated (ITD‑FLT3) structural models of FLT3, in its inactive aspartic acid‑phenylalanine‑glycine motif (DFG‑out) and active aspartic acid‑phenylalanine‑glycine motif (DFG‑in) conformations. Furthermore, the present study evaluated the effects of the second‑generation TKIs gilteritinib and quizartinib on cancer cell viability, apoptosis and proliferation in the MV4‑11 (ITD‑FLT3) and HL60 (WT‑FLT3) AML cell lines. Peripheral blood mononuclear cells (PBMCs) from a healthy volunteer were included as an FLT3‑negative group. Molecular docking analysis indicated higher affinities of second‑generation TKIs for WT‑FLT3/DFG‑out and WT‑FLT3/DFG‑in compared with those of the first‑generation TKIs. However, the ITD mutation changed the affinity of all TKIs. The in vitro data supported the in silico predictions: MV4‑11 cells presented high selective sensibility to gilteritinib and quizartinib compared with the HL60 cells, whereas the drugs had no effect on PBMCs. Thus, the current study presented novel information about molecular interactions between the FLT3 receptors (WT or ITD‑mutated) and some of their inhibitors. It also paves the way for the search for novel inhibitory molecules with potential use against AML.
Collapse
Affiliation(s)
- Ahtziri S. Carranza-Aranda
- Biomedicine and Ecology Molecular Markers Laboratory, Department of Cellular and Molecular Biology, Biological and Agricultural Sciences Campus, University of Guadalajara, Zapopan, Jalisco 44600, Mexico
| | - Luis Felipe Jave-Suárez
- Division of Immunology, Western Biomedical Research Center, Mexican Social Security Institute, Guadalajara, Jalisco 44340, Mexico
| | - Flor Y. Flores-Hernández
- Medical and Pharmaceutical Biotechnology Unit, Center for Research and Assistance in Technology and Design of The State of Jalisco, Guadalajara, Jalisco 44270, Mexico
| | - María Del Rosario Huizar-López
- Biomedicine and Ecology Molecular Markers Laboratory, Department of Cellular and Molecular Biology, Biological and Agricultural Sciences Campus, University of Guadalajara, Zapopan, Jalisco 44600, Mexico
| | - Sara E. Herrera-Rodríguez
- Medical and Pharmaceutical Biotechnology Unit, Center for Research and Assistance in Technology and Design of The State of Jalisco, Merida, Yucatan 97302, Mexico
| | - Anne Santerre
- Biomedicine and Ecology Molecular Markers Laboratory, Department of Cellular and Molecular Biology, Biological and Agricultural Sciences Campus, University of Guadalajara, Zapopan, Jalisco 44600, Mexico
| |
Collapse
|
21
|
Bonilla G, Morris A, Kundu S, DuCasse A, Kirkpatrick G, Jeffries NE, Chetal K, Yvanovich EE, Milosevic J, Zhao T, Xia J, Barghout R, Scadden D, Mansour MK, Kingston RE, Sykes DB, Mercier FE, Sadreyev RI. Leukemia aggressiveness is driven by chromatin remodeling and expression changes of core regulators. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.29.582846. [PMID: 38496490 PMCID: PMC10942317 DOI: 10.1101/2024.02.29.582846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Molecular mechanisms driving clonal aggressiveness in leukemia are not fully understood. We tracked and analyzed MLL-rearranged leukemic clones independently evolving towards higher aggressiveness. More aggressive subclones lost their growth differential ex vivo but restored it upon secondary transplantation, suggesting molecular memory of aggressiveness. Development of aggressiveness was associated with clone-specific gradual modulation of chromatin states and expression levels across the genome, with a surprising preferential trend of reversing the earlier changes between normal and leukemic progenitors. To focus on the core aggressiveness program, we identified genes with consistent changes of expression and chromatin marks that were maintained in vivo and ex vivo in both clones. Overexpressing selected core genes (Smad1 as aggressiveness driver, Irx5 and Plag1 as suppressors) affected leukemic progenitor growth in the predicted way and had convergent downstream effects on central transcription factors and repressive epigenetic modifiers, suggesting a broader regulatory network of leukemic aggressiveness.
Collapse
|
22
|
Bowman RL, Dunbar AJ, Mishra T, Xiao W, Waarts MR, Maestre IF, Eisman SE, Cai L, Mowla S, Shah N, Youn A, Bennett L, Fontenard S, Gounder S, Gandhi A, Bowman M, O'Connor K, Zaroogian Z, Sánchez-Vela P, Martinez Benitez AR, Werewski M, Park Y, Csete IS, Krishnan A, Lee D, Boorady N, Potts CR, Jenkins MT, Cai SF, Carroll MP, Meyer SE, Miles LA, Ferrell PB, Trowbridge JJ, Levine RL. In vivo models of subclonal oncogenesis and dependency in hematopoietic malignancy. Cancer Cell 2024; 42:1955-1969.e7. [PMID: 39532065 PMCID: PMC11561369 DOI: 10.1016/j.ccell.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 08/20/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
Cancer evolution is a multifaceted process leading to dysregulation of cellular expansion and differentiation through somatic mutations and epigenetic dysfunction. Clonal expansion and evolution is driven by cell-intrinsic and -extrinsic selective pressures, which can be captured with increasing resolution by single-cell and bulk DNA sequencing. Despite the extensive genomic alterations revealed in profiling studies, there remain limited experimental systems to model and perturb evolutionary processes. Here, we integrate multi-recombinase tools for reversible, sequential mutagenesis from premalignancy to leukemia. We demonstrate that inducible Flt3 mutations differentially cooperate with Dnmt3a, Idh2, and Npm1 mutant alleles, and that changing the order of mutations influences cellular and transcriptional landscapes. We next use a generalizable, reversible approach to demonstrate that mutation reversion results in rapid leukemic regression with distinct differentiation patterns depending upon co-occurring mutations. These studies provide a path to experimentally model sequential mutagenesis, investigate mechanisms of transformation and probe oncogenic dependency in disease evolution.
Collapse
Affiliation(s)
- Robert L Bowman
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Cancer Biology, Perelman Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Andrew J Dunbar
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Leukemia Service, Department of Medicine and Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Tanmay Mishra
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Wenbin Xiao
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Michael R Waarts
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Louis V. Gerstner Jr Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Inés Fernández Maestre
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Louis V. Gerstner Jr Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Shira E Eisman
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Louise Cai
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Shoron Mowla
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Nisargbhai Shah
- Department of Cancer Biology, Perelman Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Angela Youn
- Department of Cancer Biology, Perelman Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Laura Bennett
- Department of Cell and Developmental Biology, Perelman Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Suean Fontenard
- Department of Cell and Developmental Biology, Perelman Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shreeya Gounder
- Department of Cancer Biology, Perelman Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anushka Gandhi
- Department of Cancer Biology, Perelman Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael Bowman
- Department of Cancer Biology, Perelman Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kavi O'Connor
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Zachary Zaroogian
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Pablo Sánchez-Vela
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anthony R Martinez Benitez
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Matthew Werewski
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Young Park
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Isabelle S Csete
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Aishwarya Krishnan
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Darren Lee
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Nayla Boorady
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Chad R Potts
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37212 USA
| | - Matthew T Jenkins
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37212 USA
| | - Sheng F Cai
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Leukemia Service, Department of Medicine and Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Martin P Carroll
- Department of Medicine, Perelman Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sara E Meyer
- Department of Cancer Biology, Thomas Jefferson University, Sidney Kimmel Cancer Center, Philadelphia, PA 19107, USA
| | - Linde A Miles
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - P Brent Ferrell
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37212 USA
| | | | - Ross L Levine
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
23
|
Oksza-Orzechowski K, Quinten E, Shafighi S, Kiełbasa SM, van Kessel HW, de Groen RAL, Vermaat JSP, Sepúlveda Yáñez JH, Navarrete MA, Veelken H, van Bergen CAM, Szczurek E. CaClust: linking genotype to transcriptional heterogeneity of follicular lymphoma using BCR and exomic variants. Genome Biol 2024; 25:286. [PMID: 39501370 PMCID: PMC11536712 DOI: 10.1186/s13059-024-03417-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 10/08/2024] [Indexed: 11/09/2024] Open
Abstract
Tumours exhibit high genotypic and transcriptional heterogeneity. Both affect cancer progression and treatment, but have been predominantly studied separately in follicular lymphoma. To comprehensively investigate the evolution and genotype-to-phenotype maps in follicular lymphoma, we introduce CaClust, a probabilistic graphical model integrating deep whole exome, single-cell RNA and B-cell receptor sequencing data to infer clone genotypes, cell-to-clone mapping, and single-cell genotyping. CaClust outperforms a state-of-the-art model on simulated and patient data. In-depth analyses of single cells from four samples showcase effects of driver mutations, follicular lymphoma evolution, possible therapeutic targets, and single-cell genotyping that agrees with an independent targeted resequencing experiment.
Collapse
Affiliation(s)
| | - Edwin Quinten
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Shadi Shafighi
- Faculty of Mathematics, Informatics and Mechanics, University of Warsaw, Warsaw, Poland
- Cancer Research UK, Cambridge Institute, Cambridge, UK
| | - Szymon M Kiełbasa
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, Netherlands
| | - Hugo W van Kessel
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Ruben A L de Groen
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Joost S P Vermaat
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Julieta H Sepúlveda Yáñez
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
- Facultad de Ciencias de la Salud, Universidad de Magallanes, Punta Arenas, Chile
| | | | - Hendrik Veelken
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Ewa Szczurek
- Faculty of Mathematics, Informatics and Mechanics, University of Warsaw, Warsaw, Poland.
- Institute of AI for Health, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
| |
Collapse
|
24
|
Calvo X. Acute myeloid leukemia at first relapse: approaching the precipice. Haematologica 2024; 109:3463-3465. [PMID: 38961747 PMCID: PMC11532678 DOI: 10.3324/haematol.2024.285676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 06/21/2024] [Indexed: 07/05/2024] Open
Abstract
Not available.
Collapse
Affiliation(s)
- Xavier Calvo
- Laboratori de Citologia Hematològica, Servei de Patologia, Hospital del Mar, Grup de Recerca Translacional en Neoplàsies Hematològiques (GRETNHE), Hospital del Mar Research Institute (IMIM), Barcelona.
| |
Collapse
|
25
|
Zhang H, Zhong L, Wang M, Wan P, Chu X, Chen S, Zhou Z, Shao X, Liu B. p110CUX1 promotes acute myeloid leukemia progression via regulating pyridoxal phosphatase expression and activating PI3K/AKT/mTOR signaling pathway. Mol Carcinog 2024; 63:2063-2077. [PMID: 38994801 DOI: 10.1002/mc.23793] [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: 03/05/2024] [Revised: 06/07/2024] [Accepted: 07/01/2024] [Indexed: 07/13/2024]
Abstract
As an evolutionarily conserved transcription factor, Cut-like homeobox 1 (CUX1) plays crucial roles in embryonic and nervous system development, cell differentiation, and DNA damage repair. One of its major isoforms, p110CUX1, exhibits stable DNA binding capabilities and contributes to the regulation of cell cycle progression, proliferation, migration, and invasion. While p110CUX1 has been implicated in the progression of various malignant tumors, its involvement in acute myeloid leukemia (AML) remains uncertain. This study aims to elucidate the role of p110CUX1 in AML. Our findings reveal heightened expression levels of both p110CUX1 and pyridoxal phosphatase (PDXP) in AML cell lines. Overexpression of p110CUX1 promotes AML cell proliferation while inhibiting apoptosis and differentiation, whereas knockdown of PDXP yields contrasting effects. Mechanistically, p110CUX1 appears to facilitate AML development by upregulating PDXP expression and activating the PI3K/AKT/mTOR signaling pathway. Animal experimental corroborate the pro-AML effect of p110CUX1. These results provide experimental evidence supporting the involvement of the p110CUX1-PDXP-PI3K/AKT/mTOR axis in AML progression. Hence, targeting p110CUX1 may hold promise as a therapeutic strategy for AML.
Collapse
Affiliation(s)
- Hongyan Zhang
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Liang Zhong
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Meng Wang
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Peng Wan
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Xuan Chu
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Shuyu Chen
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Ziwei Zhou
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Xin Shao
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Beizhong Liu
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
26
|
Bataller A, Kantarjian H, Bazinet A, Kadia T, Daver N, DiNardo CD, Borthakur G, Loghavi S, Patel K, Tang G, Sasaki K, Short NJ, Yilmaz M, Issa GC, Alvarado Y, Montalban-Bravo G, Maiti A, Abbas HA, Takahashi K, Pierce S, Jabbour E, Garcia-Manero G, Ravandi F. Outcomes and genetic dynamics of acute myeloid leukemia at first relapse. Haematologica 2024; 109:3543-3556. [PMID: 38695144 PMCID: PMC11532689 DOI: 10.3324/haematol.2024.285057] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/19/2024] [Indexed: 11/05/2024] Open
Abstract
Patients with relapsed acute myeloid leukemia (AML) experience dismal outcomes. We performed a comprehensive analysis of patients with relapsed AML to determine the genetic dynamics and factors predicting survival. We analyzed 875 patients with newly diagnosed AML who received intensive treatment or low-intensity treatment. Of these patients, 197 subsequently relapsed. Data were available for 164 of these patients, with a median time from complete remission/complete remission with incomplete blood count recovery to relapse of 6.5 months. Thirty-five of the 164 patients (21%) experienced relapse after allogeneic hematopoietic stem cell transplantation. At relapse, mutations in genes involved in pathway signaling tended to disappear, whereas clonal hematopoiesis-related mutations or TP53 tended to persist. Patients with normal karyotypes tended to acquire cytogenetic abnormalities at relapse. Patients treated intensively had a higher rate of emergence of TP53 mutations (16%), compared to patients given low-intensity treatment (1%, P=0.009). The overall response rates were 38% and 35% for patients treated with salvage intensive treatment or low-intensity treatment, respectively. Seventeen patients (10%) underwent allogeneic stem cell transplantation after salvage therapy. The median overall survival duration after relapse was 5.3 months, with a 1-year overall survival rate of 17.6%. Complex karyotype (hazard ratio [HR]=2.14, P<0.001), a KMT2A rearrangement (HR=3.52, P=0.011), time in remission <12 months (HR=1.71, P=0.011), and an elevated white blood cell count at relapse (HR=2.38, P=0.005) were independent risk factors for overall survival duration. More effective frontline and maintenance therapies are warranted to prevent relapsed AML.
Collapse
Affiliation(s)
- Alex Bataller
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alexandre Bazinet
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tapan Kadia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Naval Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Courtney D DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gautam Borthakur
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sanam Loghavi
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Keyur Patel
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Guilin Tang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Koji Sasaki
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nicholas J Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Musa Yilmaz
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ghayas C Issa
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yesid Alvarado
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Abhishek Maiti
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hussein A Abbas
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Koichi Takahashi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sherry Pierce
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elias Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
27
|
Ansari AS, Kucharski C, Kc R, Nisakar D, Rahim R, Jiang X, Brandwein J, Uludağ H. Lipopolymer/siRNA complexes engineered for optimal molecular and functional response with chemotherapy in FLT3-mutated acute myeloid leukemia. Acta Biomater 2024; 188:297-314. [PMID: 39236794 DOI: 10.1016/j.actbio.2024.08.053] [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: 06/27/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/07/2024]
Abstract
Approximately 25% of newly diagnosed AML patients display an internal tandem duplication (ITD) in the fms-like tyrosine kinase 3 (FLT3) gene. Although both multi-targeted and FLT3 specific tyrosine kinase inhibitors (TKIs) are being utilized for clinical therapy, drug resistance, short remission periods, and high relapse rates are challenges that still need to be tackled. RNA interference (RNAi), mediated by short interfering RNA (siRNA), presents a mechanistically distinct therapeutic platform with the potential of personalization due to its gene sequence-driven mechanism of action. This study explored the use of a non-viral approach for delivery of FLT3 siRNA (siFLT3) in FLT3-ITD positive AML cell lines and primary cells as well as the feasibility of combining this treatment with drugs currently used in the clinic. Treatment of AML cell lines with FLT3 siRNA nanocomplexes resulted in prominent reduction in cell proliferation rates and induction of apoptosis. Quantitative analysis of relative mRNA transcript levels revealed downregulation of the FLT3 gene, which was accompanied by a similar decline in FLT3 protein levels. Moreover, an impact on leukemic stem cells was observed in a small pool of primary AML samples through significantly reduced colony numbers. An absence of a molecular response post-treatment with lipopolymer/siFLT3 complexes in peripheral blood mononuclear cells, obtained from healthy individuals, denoted a passive selectivity of the complexes towards malignant cells. The effect of combining lipopolymer/siFLT3 complexes with daunorubucin and FLT3 targeting TKI gilteritinib led to a significant augmentation of anti-leukemic activity. These findings demonstrate the promising potential of RNAi implemented with lipopolymer complexes for AML molecular therapy. The study prospectively supports the addition of RNAi therapy to current treatment modalities available to target the heterogeneity prevalent in AML. STATEMENT OF SIGNIFICANCE: We show that a clinically validated target, the FLT3 gene, can be eradicated in leukemia cells using non-viral RNAi. We validated these lipopolymers as effective vehicles to deliver nucleic acids to leukemic cells. The potency of the lipopolymers was superior to that of the 'gold-standard' delivery agent, lipid nanoparticles (LNPs), which are not effective in leukemia cells at clinically relevant doses. Mechanistic studies were undertaken to probe structure-function relationships for effective biomaterial formulations. Cellular and molecular responses to siRNA treatment have been characterized in cell models, including leukemia patient-derived cells. The use of the siRNA therapy with clinically used chemotherapy was demonstrated.
Collapse
MESH Headings
- Humans
- fms-Like Tyrosine Kinase 3/genetics
- fms-Like Tyrosine Kinase 3/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/therapy
- RNA, Small Interfering/pharmacology
- Cell Line, Tumor
- Mutation/genetics
- Apoptosis/drug effects
- Cell Proliferation/drug effects
- Polymers/chemistry
- Polymers/pharmacology
- Aniline Compounds
- Pyrazines
Collapse
Affiliation(s)
- Aysha S Ansari
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Cezary Kucharski
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Remant Kc
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Daniel Nisakar
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Ramea Rahim
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Xiaoyan Jiang
- Terry Fox Laboratory, BC Cancer Research Institute and Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Joseph Brandwein
- Division of Hematology, Department of Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Hasan Uludağ
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Alberta, Canada; Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
28
|
Leder K, Sun R, Wang Z, Zhang X. Parameter estimation from single patient, single time-point sequencing data of recurrent tumors. J Math Biol 2024; 89:51. [PMID: 39382689 DOI: 10.1007/s00285-024-02149-x] [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: 04/10/2024] [Revised: 08/09/2024] [Accepted: 09/22/2024] [Indexed: 10/10/2024]
Abstract
In this study, we develop consistent estimators for key parameters that govern the dynamics of tumor cell populations when subjected to pharmacological treatments. While these treatments often lead to an initial reduction in the abundance of drug-sensitive cells, a population of drug-resistant cells frequently emerges over time, resulting in cancer recurrence. Samples from recurrent tumors present as an invaluable data source that can offer crucial insights into the ability of cancer cells to adapt and withstand treatment interventions. To effectively utilize the data obtained from recurrent tumors, we derive several large number limit theorems, specifically focusing on the metrics that quantify the clonal diversity of cancer cell populations at the time of cancer recurrence. These theorems then serve as the foundation for constructing our estimators. A distinguishing feature of our approach is that our estimators only require a single time-point sequencing data from a single tumor, thereby enhancing the practicality of our approach and enabling the understanding of cancer recurrence at the individual level.
Collapse
Affiliation(s)
- Kevin Leder
- Department of Industrial and Systems Engineering, University of Minnesota, Twin Cities, MN, 55455, USA
| | - Ruping Sun
- Department of Laboratory Medicine & Pathology Masonic Cancer Center, University of Minnesota, Twin Cities, MN, 55455, USA
| | - Zicheng Wang
- School of Data Science, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), China
| | - Xuanming Zhang
- Department of Industrial and Systems Engineering, University of Minnesota, Twin Cities, MN, 55455, USA.
| |
Collapse
|
29
|
Kurucova T, Reblova K, Janovska P, Porc JP, Navrkalova V, Pavlova S, Malcikova J, Plevova K, Tichy B, Doubek M, Bryja V, Kotaskova J, Pospisilova S. Unveiling the dynamics and molecular landscape of a rare chronic lymphocytic leukemia subpopulation driving refractoriness: insights from single-cell RNA sequencing. Mol Oncol 2024; 18:2541-2553. [PMID: 38770541 PMCID: PMC11459043 DOI: 10.1002/1878-0261.13663] [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: 12/14/2023] [Revised: 03/28/2024] [Accepted: 05/03/2024] [Indexed: 05/22/2024] Open
Abstract
Early identification of resistant cancer cells is currently a major challenge, as their expansion leads to refractoriness. To capture the dynamics of these cells, we made a comprehensive analysis of disease progression and treatment response in a chronic lymphocytic leukemia (CLL) patient using a combination of single-cell and bulk genomic methods. At diagnosis, the patient presented with unfavorable genetic markers, including notch receptor 1 (NOTCH1) mutation and loss(11q). The initial and subsequent treatment lines did not lead to a durable response and the patient developed refractory disease. Refractory CLL cells featured substantial dysregulation in B-cell phenotypic markers such as human leukocyte antigen (HLA) genes, immunoglobulin (IG) genes, CD19 molecule (CD19), membrane spanning 4-domains A1 (MS4A1; previously known as CD20), CD79a molecule (CD79A) and paired box 5 (PAX5), indicating B-cell de-differentiation and disease transformation. We described the clonal evolution and characterized in detail two cell populations that emerged during the refractory disease phase, differing in the presence of high genomic complexity. In addition, we successfully tracked the cells with high genomic complexity back to the time before treatment, where they formed a rare subpopulation. We have confirmed that single-cell RNA sequencing enables the characterization of refractory cells and the monitoring of their development over time.
Collapse
Affiliation(s)
- Terezia Kurucova
- Central European Institute of Technology, Center of Molecular MedicineMasaryk UniversityBrnoCzech Republic
- Department of Experimental Biology, Faculty of ScienceMasaryk UniversityBrnoCzech Republic
| | - Kamila Reblova
- Central European Institute of Technology, Center of Molecular MedicineMasaryk UniversityBrnoCzech Republic
- Department of Internal Medicine, Hematology and Oncology, Faculty of MedicineMasaryk University and University Hospital BrnoCzech Republic
| | - Pavlina Janovska
- Department of Experimental Biology, Faculty of ScienceMasaryk UniversityBrnoCzech Republic
| | - Jakub Pawel Porc
- Central European Institute of Technology, Center of Molecular MedicineMasaryk UniversityBrnoCzech Republic
- Institute of Medical Genetics and Genomics, Faculty of MedicineMasaryk University and University Hospital BrnoCzech Republic
| | - Veronika Navrkalova
- Central European Institute of Technology, Center of Molecular MedicineMasaryk UniversityBrnoCzech Republic
- Department of Internal Medicine, Hematology and Oncology, Faculty of MedicineMasaryk University and University Hospital BrnoCzech Republic
- Institute of Medical Genetics and Genomics, Faculty of MedicineMasaryk University and University Hospital BrnoCzech Republic
| | - Sarka Pavlova
- Central European Institute of Technology, Center of Molecular MedicineMasaryk UniversityBrnoCzech Republic
- Department of Internal Medicine, Hematology and Oncology, Faculty of MedicineMasaryk University and University Hospital BrnoCzech Republic
- Institute of Medical Genetics and Genomics, Faculty of MedicineMasaryk University and University Hospital BrnoCzech Republic
| | - Jitka Malcikova
- Central European Institute of Technology, Center of Molecular MedicineMasaryk UniversityBrnoCzech Republic
- Department of Internal Medicine, Hematology and Oncology, Faculty of MedicineMasaryk University and University Hospital BrnoCzech Republic
- Institute of Medical Genetics and Genomics, Faculty of MedicineMasaryk University and University Hospital BrnoCzech Republic
| | - Karla Plevova
- Central European Institute of Technology, Center of Molecular MedicineMasaryk UniversityBrnoCzech Republic
- Department of Internal Medicine, Hematology and Oncology, Faculty of MedicineMasaryk University and University Hospital BrnoCzech Republic
- Institute of Medical Genetics and Genomics, Faculty of MedicineMasaryk University and University Hospital BrnoCzech Republic
| | - Boris Tichy
- Central European Institute of Technology, Center of Molecular MedicineMasaryk UniversityBrnoCzech Republic
- Institute of Medical Genetics and Genomics, Faculty of MedicineMasaryk University and University Hospital BrnoCzech Republic
| | - Michael Doubek
- Central European Institute of Technology, Center of Molecular MedicineMasaryk UniversityBrnoCzech Republic
- Department of Internal Medicine, Hematology and Oncology, Faculty of MedicineMasaryk University and University Hospital BrnoCzech Republic
- Institute of Medical Genetics and Genomics, Faculty of MedicineMasaryk University and University Hospital BrnoCzech Republic
| | - Vitezslav Bryja
- Department of Experimental Biology, Faculty of ScienceMasaryk UniversityBrnoCzech Republic
| | - Jana Kotaskova
- Central European Institute of Technology, Center of Molecular MedicineMasaryk UniversityBrnoCzech Republic
- Department of Internal Medicine, Hematology and Oncology, Faculty of MedicineMasaryk University and University Hospital BrnoCzech Republic
- Institute of Medical Genetics and Genomics, Faculty of MedicineMasaryk University and University Hospital BrnoCzech Republic
| | - Sarka Pospisilova
- Central European Institute of Technology, Center of Molecular MedicineMasaryk UniversityBrnoCzech Republic
- Department of Internal Medicine, Hematology and Oncology, Faculty of MedicineMasaryk University and University Hospital BrnoCzech Republic
- Institute of Medical Genetics and Genomics, Faculty of MedicineMasaryk University and University Hospital BrnoCzech Republic
| |
Collapse
|
30
|
Mo CK, Liu J, Chen S, Storrs E, Targino da Costa ALN, Houston A, Wendl MC, Jayasinghe RG, Iglesia MD, Ma C, Herndon JM, Southard-Smith AN, Liu X, Mudd J, Karpova A, Shinkle A, Goedegebuure SP, Abdelzaher ATMA, Bo P, Fulghum L, Livingston S, Balaban M, Hill A, Ippolito JE, Thorsson V, Held JM, Hagemann IS, Kim EH, Bayguinov PO, Kim AH, Mullen MM, Shoghi KI, Ju T, Reimers MA, Weimholt C, Kang LI, Puram SV, Veis DJ, Pachynski R, Fuh KC, Chheda MG, Gillanders WE, Fields RC, Raphael BJ, Chen F, Ding L. Tumour evolution and microenvironment interactions in 2D and 3D space. Nature 2024; 634:1178-1186. [PMID: 39478210 PMCID: PMC11525187 DOI: 10.1038/s41586-024-08087-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 09/19/2024] [Indexed: 11/02/2024]
Abstract
To study the spatial interactions among cancer and non-cancer cells1, we here examined a cohort of 131 tumour sections from 78 cases across 6 cancer types by Visium spatial transcriptomics (ST). This was combined with 48 matched single-nucleus RNA sequencing samples and 22 matched co-detection by indexing (CODEX) samples. To describe tumour structures and habitats, we defined 'tumour microregions' as spatially distinct cancer cell clusters separated by stromal components. They varied in size and density among cancer types, with the largest microregions observed in metastatic samples. We further grouped microregions with shared genetic alterations into 'spatial subclones'. Thirty five tumour sections exhibited subclonal structures. Spatial subclones with distinct copy number variations and mutations displayed differential oncogenic activities. We identified increased metabolic activity at the centre and increased antigen presentation along the leading edges of microregions. We also observed variable T cell infiltrations within microregions and macrophages predominantly residing at tumour boundaries. We reconstructed 3D tumour structures by co-registering 48 serial ST sections from 16 samples, which provided insights into the spatial organization and heterogeneity of tumours. Additionally, using an unsupervised deep-learning algorithm and integrating ST and CODEX data, we identified both immune hot and cold neighbourhoods and enhanced immune exhaustion markers surrounding the 3D subclones. These findings contribute to the understanding of spatial tumour evolution through interactions with the local microenvironment in 2D and 3D space, providing valuable insights into tumour biology.
Collapse
Affiliation(s)
- Chia-Kuei Mo
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Jingxian Liu
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Siqi Chen
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Erik Storrs
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Andre Luiz N Targino da Costa
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Andrew Houston
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Michael C Wendl
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Reyka G Jayasinghe
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Michael D Iglesia
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Cong Ma
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - John M Herndon
- Department of Surgery, Washington University in St Louis, St Louis, MO, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
| | - Austin N Southard-Smith
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Xinhao Liu
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Jacqueline Mudd
- Department of Surgery, Washington University in St Louis, St Louis, MO, USA
| | - Alla Karpova
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Andrew Shinkle
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - S Peter Goedegebuure
- Department of Surgery, Washington University in St Louis, St Louis, MO, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
| | - Abdurrahman Taha Mousa Ali Abdelzaher
- Department of Surgery, Washington University in St Louis, St Louis, MO, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
| | - Peng Bo
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
| | - Lauren Fulghum
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Samantha Livingston
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Metin Balaban
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Angela Hill
- Department of Surgery, Washington University in St Louis, St Louis, MO, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
| | - Joseph E Ippolito
- Department of Radiology, Washington University in St Louis, St Louis, MO, USA
| | | | - Jason M Held
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
- Division of Medical Oncology, Department of Internal Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Ian S Hagemann
- Department of Pathology and Immunology, Washington University in St Louis, St Louis, MO, USA
- Department of Obstetrics and Gynecology, Washington University in St Louis, St Louis, MO, USA
| | - Eric H Kim
- Division of Urological Surgery, Department of Surgery, Washington University, St Louis, MO, USA
| | - Peter O Bayguinov
- Department of Neuroscience, Washington University School of Medicine, St Louis, MO, USA
| | - Albert H Kim
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
- Department of Neurosurgery, Washington University School of Medicine, St Louis, MO, USA
| | - Mary M Mullen
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St Louis, MO, USA
| | - Kooresh I Shoghi
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
- Department of Radiology, Washington University in St Louis, St Louis, MO, USA
| | - Tao Ju
- Department of Computer Science and Engineering, Washington University in St Louis, St Louis, MO, USA
| | - Melissa A Reimers
- Division of Medical Oncology, Department of Internal Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Cody Weimholt
- Department of Pathology and Immunology, Washington University in St Louis, St Louis, MO, USA
| | - Liang-I Kang
- Department of Pathology and Immunology, Washington University in St Louis, St Louis, MO, USA
| | - Sidharth V Puram
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
- Department of Genetics, Washington University in St Louis, St Louis, MO, USA
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine, St Louis, MO, USA
| | - Deborah J Veis
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, Washington University in St Louis, St Louis, MO, USA
| | - Russell Pachynski
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
| | - Katherine C Fuh
- Department of Obstetrics and Gynecology, Washington University in St Louis, St Louis, MO, USA
- Department of Obstetrics and Gynecology, University of California, San Francisco, San Francisco, CA, USA
| | - Milan G Chheda
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
| | - William E Gillanders
- Department of Surgery, Washington University in St Louis, St Louis, MO, USA.
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA.
| | - Ryan C Fields
- Department of Surgery, Washington University in St Louis, St Louis, MO, USA.
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA.
| | - Benjamin J Raphael
- Department of Computer Science, Princeton University, Princeton, NJ, USA.
| | - Feng Chen
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA.
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA.
| | - Li Ding
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA.
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA.
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA.
- Department of Genetics, Washington University in St Louis, St Louis, MO, USA.
| |
Collapse
|
31
|
Straube J, Janardhanan Y, Haldar R, Bywater MJ. Immune control in acute myeloid leukemia. Exp Hematol 2024; 138:104256. [PMID: 38876254 DOI: 10.1016/j.exphem.2024.104256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/09/2024] [Accepted: 06/10/2024] [Indexed: 06/16/2024]
Abstract
Acute myeloid leukemia (AML) is a genetically heterogeneous disease, in that a multitude of oncogenic drivers and chromosomal abnormalities have been identified and associated with the leukemic transformation of myeloid blasts. However, little is known as to how individual mutations influence the interaction between the immune system and AML cells and the efficacy of the immune system in AML disease control. In this review, we will discuss how AML cells potentially activate the immune system and what evidence there is to support the role of the immune system in controlling this disease. We will specifically examine the importance of antigen presentation in fostering an effective anti-AML immune response, explore the disruption of immune responses during AML disease progression, and discuss the emerging role of the oncoprotein MYC in driving immune suppression in AML.
Collapse
Affiliation(s)
- Jasmin Straube
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; The University of Queensland, Brisbane, Queensland, Australia
| | | | - Rohit Haldar
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Megan J Bywater
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; The University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
32
|
Boutzen H, Murison A, Oriecuia A, Bansal S, Arlidge C, Wang JCY, Lupien M, Kaufmann KB, Dick JE. Identification of leukemia stem cell subsets with distinct transcriptional, epigenetic and functional properties. Leukemia 2024; 38:2090-2101. [PMID: 39169113 PMCID: PMC11436360 DOI: 10.1038/s41375-024-02358-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 07/02/2024] [Accepted: 07/17/2024] [Indexed: 08/23/2024]
Abstract
The leukemia stem cell (LSC) compartment is a complex reservoir fueling disease progression in acute myeloid leukemia (AML). The existence of heterogeneity within this compartment is well documented but prior studies have focused on genetic heterogeneity without being able to address functional heterogeneity. Understanding this heterogeneity is critical for the informed design of therapies targeting LSC, but has been hampered by LSC scarcity and the lack of reliable cell surface markers for viable LSC isolation. To overcome these challenges, we turned to the patient-derived OCI-AML22 cell model. This model includes functionally, transcriptionally and epigenetically characterized LSC broadly representative of LSC found in primary AML samples. Focusing on the pool of LSC, we used an integrated approach combining xenograft assays with single-cell analysis to identify two LSC subtypes with distinct transcriptional, epigenetic and functional properties. These LSC subtypes differed in depth of quiescence, differentiation potential, repopulation capacity, sensitivity to chemotherapy and could be isolated based on CD112 expression. A majority of AML patient samples had transcriptional signatures reflective of either LSC subtype, and some even showed coexistence within an individual sample. This work provides a framework for investigating the LSC compartment and designing combinatorial therapeutic strategies in AML.
Collapse
Affiliation(s)
- Héléna Boutzen
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 0A3, Canada.
| | - Alex Murison
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 0A3, Canada
| | - Alexa Oriecuia
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 0A3, Canada
| | - Suraj Bansal
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 0A3, Canada
| | - Christopher Arlidge
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 0A3, Canada
| | - Jean C Y Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 0A3, Canada
- Division of Medical Oncology and Hematology, Department of Medicine, University Health Network, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Mathieu Lupien
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 0A3, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5S 1A4, Canada
| | - Kerstin B Kaufmann
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 0A3, Canada.
| | - John E Dick
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 0A3, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
33
|
Zhang Q, Xu Z, Han R, Wang Y, Ye Z, Zhu J, Cai Y, Zhang F, Zhao J, Yao B, Qin Z, Qiao N, Huang R, Feng J, Wang Y, Rui W, He F, Zhao Y, Ding C. Proteogenomic characterization of skull-base chordoma. Nat Commun 2024; 15:8338. [PMID: 39333076 PMCID: PMC11436687 DOI: 10.1038/s41467-024-52285-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 08/29/2024] [Indexed: 09/29/2024] Open
Abstract
Skull-base chordoma is a rare, aggressive bone cancer with a high recurrence rate. Despite advances in genomic studies, its molecular characteristics and effective therapies remain unknown. Here, we conduct integrative genomics, transcriptomics, proteomics, and phosphoproteomics analyses of 187 skull-base chordoma tumors. In our study, chromosome instability is identified as a prognostic predictor and potential therapeutic target. Multi-omics data reveals downstream effects of chromosome instability, with RPRD1B as a putative target for radiotherapy-resistant patients. Chromosome 1q gain, associated with chromosome instability and upregulated mitochondrial functions, lead to poorer clinical outcomes. Immune subtyping identify an immune cold subtype linked to chromosome 9p/10q loss and immune evasion. Proteomics-based classification reveals subtypes (P-II and P-III) with high chromosome instability and immune cold features, with P-II tumors showing increased invasiveness. These findings, confirmed in 17 paired samples, provide insights into the biology and treatment of skull-base chordoma.
Collapse
Affiliation(s)
- Qilin Zhang
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200433, China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Ziyan Xu
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200433, China
| | - Rui Han
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200433, China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yunzhi Wang
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200433, China
| | - Zhen Ye
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200433, China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiajun Zhu
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200433, China
| | - Yixin Cai
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200433, China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fan Zhang
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200433, China
| | - Jiangyan Zhao
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200433, China
| | - Boyuan Yao
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200433, China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhaoyu Qin
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200433, China
| | - Nidan Qiao
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200433, China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ruofan Huang
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Oncology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Jinwen Feng
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200433, China
| | - Yongfei Wang
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200433, China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenting Rui
- Department of Radiology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fuchu He
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200433, China.
- Research Unit of Proteomics Driven Cancer Precision Medicine. Chinese Academy of Medical Sciences, Beijing, 102206, China.
| | - Yao Zhao
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200433, China.
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, 200040, China.
- Neurosurgical Institute of Fudan University, Shanghai, 200040, China.
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Chen Ding
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200433, China.
- Departments of Cancer Research Institute, Affiliated Cancer Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Translational Biomedical Engineering, Urumqi, 830000, China.
| |
Collapse
|
34
|
Bruno S, Borsi E, Patuelli A, Bandini L, Mancini M, Forte D, Nanni J, Barone M, Grassi A, Cristiano G, Venturi C, Robustelli V, Atzeni G, Mosca C, De Santis S, Monaldi C, Poletti A, Terragna C, Curti A, Cavo M, Soverini S, Ottaviani E. Tracking Response and Resistance in Acute Myeloid Leukemia through Single-Cell DNA Sequencing Helps Uncover New Therapeutic Targets. Int J Mol Sci 2024; 25:10002. [PMID: 39337490 PMCID: PMC11432296 DOI: 10.3390/ijms251810002] [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: 07/30/2024] [Revised: 09/04/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive hematologic neoplasia with a complex polyclonal architecture. Among driver lesions, those involving the FLT3 gene represent the most frequent mutations identified at diagnosis. The development of tyrosine kinase inhibitors (TKIs) has improved the clinical outcomes of FLT3-mutated patients (Pt). However, overcoming resistance to these drugs remains a challenge. To unravel the molecular mechanisms underlying therapy resistance and clonal selection, we conducted a longitudinal analysis using a single-cell DNA sequencing approach (MissionBioTapestri® platform, San Francisco, CA, USA) in two patients with FLT3-mutated AML. To this end, samples were collected at the time of diagnosis, during TKI therapy, and at relapse or complete remission. For Pt #1, disease resistance was associated with clonal expansion of minor clones, and 2nd line TKI therapy with gilteritinib provided a proliferative advantage to the clones carrying NRAS and KIT mutations, thereby responsible for relapse. In Pt #2, clonal architecture was less complex, and 1st line TKI therapy with midostaurin was able to eradicate the leukemic clones. Our results corroborate previous findings about clonal selection driven by TKIs, highlighting the importance of a deeper characterization of individual clonal architectures for choosing the best treatment plan for personalized approaches aimed at optimizing outcomes.
Collapse
Affiliation(s)
- Samantha Bruno
- Department of Medical and Surgical Science (DIMEC), University of Bologna, 40138 Bologna, Italy; (A.P.); (L.B.); (D.F.); (J.N.); (A.G.); (G.C.); (C.V.); (G.A.); (C.M.); (S.D.S.); (C.M.); (A.P.); (M.C.); (S.S.)
| | - Enrica Borsi
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero, Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, 940138 Bologna, Italy; (E.B.); (M.M.); (M.B.); (V.R.); (C.T.); (A.C.); (E.O.)
| | - Agnese Patuelli
- Department of Medical and Surgical Science (DIMEC), University of Bologna, 40138 Bologna, Italy; (A.P.); (L.B.); (D.F.); (J.N.); (A.G.); (G.C.); (C.V.); (G.A.); (C.M.); (S.D.S.); (C.M.); (A.P.); (M.C.); (S.S.)
| | - Lorenza Bandini
- Department of Medical and Surgical Science (DIMEC), University of Bologna, 40138 Bologna, Italy; (A.P.); (L.B.); (D.F.); (J.N.); (A.G.); (G.C.); (C.V.); (G.A.); (C.M.); (S.D.S.); (C.M.); (A.P.); (M.C.); (S.S.)
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero, Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, 940138 Bologna, Italy; (E.B.); (M.M.); (M.B.); (V.R.); (C.T.); (A.C.); (E.O.)
| | - Manuela Mancini
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero, Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, 940138 Bologna, Italy; (E.B.); (M.M.); (M.B.); (V.R.); (C.T.); (A.C.); (E.O.)
| | - Dorian Forte
- Department of Medical and Surgical Science (DIMEC), University of Bologna, 40138 Bologna, Italy; (A.P.); (L.B.); (D.F.); (J.N.); (A.G.); (G.C.); (C.V.); (G.A.); (C.M.); (S.D.S.); (C.M.); (A.P.); (M.C.); (S.S.)
| | - Jacopo Nanni
- Department of Medical and Surgical Science (DIMEC), University of Bologna, 40138 Bologna, Italy; (A.P.); (L.B.); (D.F.); (J.N.); (A.G.); (G.C.); (C.V.); (G.A.); (C.M.); (S.D.S.); (C.M.); (A.P.); (M.C.); (S.S.)
| | - Martina Barone
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero, Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, 940138 Bologna, Italy; (E.B.); (M.M.); (M.B.); (V.R.); (C.T.); (A.C.); (E.O.)
| | - Alessandra Grassi
- Department of Medical and Surgical Science (DIMEC), University of Bologna, 40138 Bologna, Italy; (A.P.); (L.B.); (D.F.); (J.N.); (A.G.); (G.C.); (C.V.); (G.A.); (C.M.); (S.D.S.); (C.M.); (A.P.); (M.C.); (S.S.)
| | - Gianluca Cristiano
- Department of Medical and Surgical Science (DIMEC), University of Bologna, 40138 Bologna, Italy; (A.P.); (L.B.); (D.F.); (J.N.); (A.G.); (G.C.); (C.V.); (G.A.); (C.M.); (S.D.S.); (C.M.); (A.P.); (M.C.); (S.S.)
| | - Claudia Venturi
- Department of Medical and Surgical Science (DIMEC), University of Bologna, 40138 Bologna, Italy; (A.P.); (L.B.); (D.F.); (J.N.); (A.G.); (G.C.); (C.V.); (G.A.); (C.M.); (S.D.S.); (C.M.); (A.P.); (M.C.); (S.S.)
| | - Valentina Robustelli
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero, Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, 940138 Bologna, Italy; (E.B.); (M.M.); (M.B.); (V.R.); (C.T.); (A.C.); (E.O.)
| | - Giulia Atzeni
- Department of Medical and Surgical Science (DIMEC), University of Bologna, 40138 Bologna, Italy; (A.P.); (L.B.); (D.F.); (J.N.); (A.G.); (G.C.); (C.V.); (G.A.); (C.M.); (S.D.S.); (C.M.); (A.P.); (M.C.); (S.S.)
| | - Cristina Mosca
- Department of Medical and Surgical Science (DIMEC), University of Bologna, 40138 Bologna, Italy; (A.P.); (L.B.); (D.F.); (J.N.); (A.G.); (G.C.); (C.V.); (G.A.); (C.M.); (S.D.S.); (C.M.); (A.P.); (M.C.); (S.S.)
| | - Sara De Santis
- Department of Medical and Surgical Science (DIMEC), University of Bologna, 40138 Bologna, Italy; (A.P.); (L.B.); (D.F.); (J.N.); (A.G.); (G.C.); (C.V.); (G.A.); (C.M.); (S.D.S.); (C.M.); (A.P.); (M.C.); (S.S.)
| | - Cecilia Monaldi
- Department of Medical and Surgical Science (DIMEC), University of Bologna, 40138 Bologna, Italy; (A.P.); (L.B.); (D.F.); (J.N.); (A.G.); (G.C.); (C.V.); (G.A.); (C.M.); (S.D.S.); (C.M.); (A.P.); (M.C.); (S.S.)
| | - Andrea Poletti
- Department of Medical and Surgical Science (DIMEC), University of Bologna, 40138 Bologna, Italy; (A.P.); (L.B.); (D.F.); (J.N.); (A.G.); (G.C.); (C.V.); (G.A.); (C.M.); (S.D.S.); (C.M.); (A.P.); (M.C.); (S.S.)
| | - Carolina Terragna
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero, Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, 940138 Bologna, Italy; (E.B.); (M.M.); (M.B.); (V.R.); (C.T.); (A.C.); (E.O.)
| | - Antonio Curti
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero, Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, 940138 Bologna, Italy; (E.B.); (M.M.); (M.B.); (V.R.); (C.T.); (A.C.); (E.O.)
| | - Michele Cavo
- Department of Medical and Surgical Science (DIMEC), University of Bologna, 40138 Bologna, Italy; (A.P.); (L.B.); (D.F.); (J.N.); (A.G.); (G.C.); (C.V.); (G.A.); (C.M.); (S.D.S.); (C.M.); (A.P.); (M.C.); (S.S.)
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero, Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, 940138 Bologna, Italy; (E.B.); (M.M.); (M.B.); (V.R.); (C.T.); (A.C.); (E.O.)
| | - Simona Soverini
- Department of Medical and Surgical Science (DIMEC), University of Bologna, 40138 Bologna, Italy; (A.P.); (L.B.); (D.F.); (J.N.); (A.G.); (G.C.); (C.V.); (G.A.); (C.M.); (S.D.S.); (C.M.); (A.P.); (M.C.); (S.S.)
| | - Emanuela Ottaviani
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero, Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, 940138 Bologna, Italy; (E.B.); (M.M.); (M.B.); (V.R.); (C.T.); (A.C.); (E.O.)
| |
Collapse
|
35
|
Fan J, Li Z, Pei L, Hou Y. Post-transcriptional regulation of DEAD-box RNA helicases in hematopoietic malignancies. Genes Dis 2024; 11:101252. [PMID: 38993792 PMCID: PMC11237855 DOI: 10.1016/j.gendis.2024.101252] [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: 05/31/2023] [Revised: 02/01/2024] [Accepted: 02/11/2024] [Indexed: 07/13/2024] Open
Abstract
Hematopoiesis represents a meticulously regulated and dynamic biological process. Genetic aberrations affecting blood cells, induced by various factors, frequently give rise to hematological tumors. These instances are often accompanied by a multitude of abnormal post-transcriptional regulatory events, including RNA alternative splicing, RNA localization, RNA degradation, and storage. Notably, post-transcriptional regulation plays a pivotal role in preserving hematopoietic homeostasis. The DEAD-Box RNA helicase genes emerge as crucial post-transcriptional regulatory factors, intricately involved in sustaining normal hematopoiesis through diverse mechanisms such as RNA alternative splicing, RNA modification, and ribosome assembly. This review consolidates the existing knowledge on the role of DEAD-box RNA helicases in regulating normal hematopoiesis and underscores the pathogenicity of mutant DEAD-Box RNA helicases in malignant hematopoiesis. Emphasis is placed on elucidating both the positive and negative contributions of DEAD-box RNA helicases within the hematopoietic system.
Collapse
Affiliation(s)
- Jiankun Fan
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Zhigang Li
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Li Pei
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yu Hou
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
36
|
Chow RD, Velu P, Deihimi S, Belman J, Youn A, Shah N, Luger SM, Carroll MP, Morrissette J, Bowman RL. Early drivers of clonal hematopoiesis shape the evolutionary trajectories of de novo acute myeloid leukemia. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.31.24312756. [PMID: 39252918 PMCID: PMC11383471 DOI: 10.1101/2024.08.31.24312756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Mutations commonly found in AML such as DNMT3A, TET2 and ASXL1 can be found in the peripheral blood of otherwise healthy adults - a phenomenon referred to as clonal hematopoiesis (CH). These mutations are thought to represent the earliest genetic events in the evolution of AML. Genomic studies on samples acquired at diagnosis, remission, and at relapse have demonstrated significant stability of CH mutations following induction chemotherapy. Meanwhile, later mutations in genes such as NPM1 and FLT3, have been shown to contract at remission and in the case of FLT3 often are absent at relapse. We sought to understand how early CH mutations influence subsequent evolutionary trajectories throughout remission and relapse in response to induction chemotherapy. Here, we assembled a retrospective cohort of patients diagnosed with de novo AML at our institution that underwent genomic sequencing at diagnosis as well as at the time of remission and/or relapse (total n = 182 patients). Corroborating prior studies, FLT3 and NPM1 mutations were generally eliminated at the time of cytologic complete remission but subsequently reemerged upon relapse, whereas DNMT3A, TET2 and ASXL1 mutations often persisted through remission. Early CH-related mutations exhibited distinct constellations of co-occurring genetic alterations, with NPM1 and FLT3 mutations enriched in DNMT3A mut AML, while CBL and SRSF2 mutations were enriched in TET2 mut and ASXL1 mut AML, respectively. In the case of NPM1 and FLT3 mutations, these differences vanished at the time of complete remission yet readily reemerged upon relapse, indicating the reproducible nature of these genetic interactions. Thus, early CH-associated mutations that precede malignant transformation subsequently shape the evolutionary trajectories of AML through diagnosis, therapy, and relapse.
Collapse
Affiliation(s)
- Ryan D. Chow
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Priya Velu
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell School of Medicine, Cornell University, New York, NY, USA
| | - Safoora Deihimi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jonathan Belman
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Angela Youn
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nisargbhai Shah
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Selina M. Luger
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Martin P. Carroll
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jennifer Morrissette
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robert L Bowman
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
37
|
Tani H, Hifumi T, Ito K, Kuramoto T, Miyoshi N, Fujiki M, Nakayama T. A case report of feline mast cell tumour with intertumoral heterogeneity: Identification of secondary mutations c.998G>C and c.2383G>C in KIT after resistance to toceranib. Vet Med Sci 2024; 10:e70003. [PMID: 39177283 PMCID: PMC11342349 DOI: 10.1002/vms3.70003] [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: 04/24/2024] [Revised: 07/31/2024] [Accepted: 08/07/2024] [Indexed: 08/24/2024] Open
Abstract
A 12-year-old male domestic cat with multiple subcutaneous mast cell tumours (MCTs) presented with a 2-week history of pruritus and raw/bleeding skin from self-trauma at Kagoshima University Veterinary Teaching Hospital. Polymerase chain reaction (PCR) and histopathological analyses revealed intertumoral heterogeneity among tumour locations based on the mutation status of KIT. In addition, the expression pattern of KIT was characterized. After failed treatment with vinblastine (2.0-2.2 mg/m2, intravenous administration, two doses in total) or nimustine (25 mg/m2, intravenous administration, two doses in total), toceranib (2.2-2.6 mg/kg, orally administered, every other day) was administered to treat recurrent MCTs harbouring the KIT exon eight internal tandem duplication mutation, achieving a complete response. However, toceranib resistance developed 2 months after treatment initiation. Subsequent PCR analysis was conducted to identify the mutational status of KIT in each MCT and to detect the presence of secondary mutations associated with the acquisition of toceranib resistance. Secondary KIT mutations (c.998G>C and c.2383G>C), which were not initially detected in tumour cells at diagnosis, were identified after the development of resistance to toceranib. This indicates that the tumour cells in feline MCTs in the same case have diverse characteristics. Our findings encourage further investigation into the development of therapeutic strategies for feline MCTs, particularly focusing on the heterogeneous nature of KIT/KIT and overcoming acquired resistance to toceranib.
Collapse
Affiliation(s)
- Hiroyuki Tani
- Laboratory of Veterinary RadiologyDepartment of Veterinary MedicineCollege of Bioresource SciencesNihon UniversityFujisawaKanagawaJapan
| | - Tatsuro Hifumi
- Laboratory of Veterinary HistopathologyJoint Faculty of Veterinary MedicineKagoshima UniversityKagoshimaJapan
| | | | - Tomohide Kuramoto
- Kagoshima University Veterinary Teaching HospitalJoint Faculty of Veterinary MedicineKagoshima UniversityKagoshimaJapan
| | - Noriaki Miyoshi
- Laboratory of Veterinary HistopathologyJoint Faculty of Veterinary MedicineKagoshima UniversityKagoshimaJapan
| | - Makoto Fujiki
- Kagoshima University Veterinary Teaching HospitalJoint Faculty of Veterinary MedicineKagoshima UniversityKagoshimaJapan
- Laboratory of Veterinary SurgeryJoint Faculty of Veterinary MedicineKagoshima UniversityKagoshimaJapan
| | - Tomohiro Nakayama
- Laboratory of Veterinary RadiologyDepartment of Veterinary MedicineCollege of Bioresource SciencesNihon UniversityFujisawaKanagawaJapan
| |
Collapse
|
38
|
Drouillard D, Halyko M, Cinquegrani E, McAllister D, Peterson FC, Marchese A, Dwinell MB. CXCL12 chemokine dimer signaling modulates acute myelogenous leukemia cell migration through altered receptor internalization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.26.609725. [PMID: 39253415 PMCID: PMC11383031 DOI: 10.1101/2024.08.26.609725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Acute myeloid leukemia (AML) is a malignancy of immature myeloid blast cells with stem-like and chemoresistant cells being retained in the bone marrow through CXCL12-CXCR4 signaling. Current CXCR4 inhibitors mobilize AML cells into the bloodstream where they become more chemosensitive have failed to improve patient survival, likely reflecting persistent receptor localization on target cells. Here we characterize the signaling properties of CXCL12-locked dimer (CXCL12-LD), a bioengineered variant of the dimeric CXCL12 structure. CXCL12-LD binding resulted in lower levels of G protein, β-arrestin, and intracellular calcium mobilization, consistent with the locked dimer being a partial agonist of CXCR4. Further, CXCL12-LD failed to induce chemotaxis in AML cells. Despite these partial agonist properties, CXCL12-LD increased CXCR4 internalization compared to wildtype and locked-monomer forms of CXCL12. Analysis of a previously published AML transcriptomic data showed CXCR4 positive AML cells co-express genes involved in chemoresistance and maintenance of a blast-like state. The CXCL12-LD partial agonist effectively mobilized stem cells into the bloodstream in mice suggesting a potential role for their use in targeting CXCR4. Together, our results suggest that enhanced internalization by CXCL12-LD partial agonist signaling can avoid pharmacodynamic tolerance and may identify new avenues to better target GPCRs.
Collapse
Affiliation(s)
- Donovan Drouillard
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee WI, USA
- Center for Immunology, Medical College of Wisconsin, Milwaukee WI, USA
| | - Michael Halyko
- Center for Immunology, Medical College of Wisconsin, Milwaukee WI, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee WI, USA
| | | | - Donna McAllister
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee WI, USA
- Center for Immunology, Medical College of Wisconsin, Milwaukee WI, USA
| | | | - Adriano Marchese
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee WI, USA
| | - Michael B. Dwinell
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee WI, USA
- Center for Immunology, Medical College of Wisconsin, Milwaukee WI, USA
- Department of Surgery, Medical College of Wisconsin, Milwaukee WI, USA
| |
Collapse
|
39
|
Atzeni R, Massidda M, Pieroni E, Rallo V, Pisu M, Angius A. A Novel Affordable and Reliable Framework for Accurate Detection and Comprehensive Analysis of Somatic Mutations in Cancer. Int J Mol Sci 2024; 25:8044. [PMID: 39125613 PMCID: PMC11311285 DOI: 10.3390/ijms25158044] [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: 06/10/2024] [Revised: 07/11/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Accurate detection and analysis of somatic variants in cancer involve multiple third-party tools with complex dependencies and configurations, leading to laborious, error-prone, and time-consuming data conversions. This approach lacks accuracy, reproducibility, and portability, limiting clinical application. Musta was developed to address these issues as an end-to-end pipeline for detecting, classifying, and interpreting cancer mutations. Musta is based on a Python command-line tool designed to manage tumor-normal samples for precise somatic mutation analysis. The core is a Snakemake-based workflow that covers all key cancer genomics steps, including variant calling, mutational signature deconvolution, variant annotation, driver gene detection, pathway analysis, and tumor heterogeneity estimation. Musta is easy to install on any system via Docker, with a Makefile handling installation, configuration, and execution, allowing for full or partial pipeline runs. Musta has been validated at the CRS4-NGS Core facility and tested on large datasets from The Cancer Genome Atlas and the Beijing Institute of Genomics. Musta has proven robust and flexible for somatic variant analysis in cancer. It is user-friendly, requiring no specialized programming skills, and enables data processing with a single command line. Its reproducibility ensures consistent results across users following the same protocol.
Collapse
Affiliation(s)
- Rossano Atzeni
- Center for Advanced Studies, Research and Development in Sardinia (CRS4), 09050 Pula, Italy; (R.A.); (E.P.); (M.P.)
| | - Matteo Massidda
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy;
| | - Enrico Pieroni
- Center for Advanced Studies, Research and Development in Sardinia (CRS4), 09050 Pula, Italy; (R.A.); (E.P.); (M.P.)
| | - Vincenzo Rallo
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche (CNR), Cittadella Universitaria di Cagliari, 09042 Monserrato, Italy;
| | - Massimo Pisu
- Center for Advanced Studies, Research and Development in Sardinia (CRS4), 09050 Pula, Italy; (R.A.); (E.P.); (M.P.)
| | - Andrea Angius
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche (CNR), Cittadella Universitaria di Cagliari, 09042 Monserrato, Italy;
| |
Collapse
|
40
|
Lai J, Yang Y, Liu Y, Scharpf RB, Karchin R. Assessing the merits: an opinion on the effectiveness of simulation techniques in tumor subclonal reconstruction. BIOINFORMATICS ADVANCES 2024; 4:vbae094. [PMID: 38948008 PMCID: PMC11213631 DOI: 10.1093/bioadv/vbae094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/28/2024] [Accepted: 06/15/2024] [Indexed: 07/02/2024]
Abstract
Summary Neoplastic tumors originate from a single cell, and their evolution can be traced through lineages characterized by mutations, copy number alterations, and structural variants. These lineages are reconstructed and mapped onto evolutionary trees with algorithmic approaches. However, without ground truth benchmark sets, the validity of an algorithm remains uncertain, limiting potential clinical applicability. With a growing number of algorithms available, there is urgent need for standardized benchmark sets to evaluate their merits. Benchmark sets rely on in silico simulations of tumor sequence, but there are no accepted standards for simulation tools, presenting a major obstacle to progress in this field. Availability and implementation All analysis done in the paper was based on publicly available data from the publication of each accessed tool.
Collapse
Affiliation(s)
- Jiaying Lai
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, United States
| | - Yi Yang
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, United States
| | - Yunzhou Liu
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, United States
| | - Robert B Scharpf
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States
- Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD 21231, United States
| | - Rachel Karchin
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, United States
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States
- Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD 21231, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, United States
| |
Collapse
|
41
|
Tiong IS, Hiwase DK, Abro E, Bajel A, Palfreyman E, Beligaswatte A, Reynolds J, Anstee N, Nguyen T, Loo S, Chua CC, Ashby M, Wiltshire KM, Fleming S, Fong CY, Teh TC, Blombery P, Dillon R, Ivey A, Wei AH. Targeting Molecular Measurable Residual Disease and Low-Blast Relapse in AML With Venetoclax and Low-Dose Cytarabine: A Prospective Phase II Study (VALDAC). J Clin Oncol 2024; 42:2161-2173. [PMID: 38427924 PMCID: PMC11191043 DOI: 10.1200/jco.23.01599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 11/21/2023] [Accepted: 12/18/2023] [Indexed: 03/03/2024] Open
Abstract
PURPOSE A prospective phase II study examined the safety and efficacy of venetoclax combined with low-dose cytarabine (LDAC) in AML at first measurable residual disease (MRD) or oligoblastic relapse. METHODS Patients with either MRD (≥1 log10 rise) or oligoblastic relapse (blasts 5%-15%) received venetoclax 600 mg once daily D1-28 plus LDAC once daily D1-10 in 28-day cycles. The primary objective was MRD response in the MRD relapse cohort or complete remission (CR/CRh/CRi) in the oligoblastic relapse cohort. RESULTS Forty-eight adults with either MRD (n = 26) or oligoblastic (n = 22) relapse were enrolled. Median age was 67 years (range, 18-80) and 94% had received previous intensive chemotherapy. Patients received a median of four cycles of therapy; 17% completed ≥12 cycles. Patients with oligoblastic relapse had more grade ≥3 anemia (32% v 4%; P = .02) and infections (36% v 8%; P = .03), whereas grade 4 neutropenia (32 v 23%) or thrombocytopenia (27 v 15%) were comparable with the MRD relapse cohort. Markers of molecular MRD relapse included mutant NPM1 (77%), CBFB::MYH11 (4%), RUNX1::RUNX1T1 (4%), or KMT2A::MLLT3 (4%). Three patients with a log10 rise in IDH1/2 (12%) were included. By cycle 2 in the MRD relapse cohort, a log10 reduction in MRD was observed in 69%; 46% achieved MRD negative remission. In the oligoblastic relapse cohort, 73% achieved CR/CRh/CRi. Overall, 21 (44%) underwent hematopoietic cell transplantation. Median overall survival (OS) was not reached in either cohort. Estimated 2-year OS rate was 67% (95% CI, 50 to 89) in the MRD and 53% (95% CI, 34 to 84) in the oligoblastic relapse cohorts. CONCLUSION For AML in first remission and either MRD or oligoblastic relapse, venetoclax plus LDAC is well tolerated and highly effective.
Collapse
MESH Headings
- Humans
- Aged
- Middle Aged
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Neoplasm, Residual
- Cytarabine/administration & dosage
- Sulfonamides/administration & dosage
- Sulfonamides/adverse effects
- Adult
- Female
- Male
- Bridged Bicyclo Compounds, Heterocyclic/administration & dosage
- Bridged Bicyclo Compounds, Heterocyclic/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Aged, 80 and over
- Prospective Studies
- Nucleophosmin
- Young Adult
- Adolescent
Collapse
Affiliation(s)
- Ing Soo Tiong
- The Alfred Hospital and Monash University, Melbourne, Australia
- Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, Australia
| | - Devendra K. Hiwase
- Royal Adelaide Hospital, Adelaide, Australia
- University of Adelaide, Adelaide, Australia
- South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Emad Abro
- Princess Alexandra Hospital, Queensland, Australia
| | - Ashish Bajel
- Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, Australia
- The University of Melbourne, Melbourne, Australia
| | | | - Ashanka Beligaswatte
- University of Adelaide, Adelaide, Australia
- Flinders Medical Centre, Bedford Park, Australia
| | - John Reynolds
- The Alfred Hospital and Monash University, Melbourne, Australia
| | - Natasha Anstee
- The University of Melbourne, Melbourne, Australia
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Tamia Nguyen
- Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, Australia
- The University of Melbourne, Melbourne, Australia
| | - Sun Loo
- Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, Australia
- The University of Melbourne, Melbourne, Australia
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- The Northern Hospital, Melbourne, Australia
| | - Chong Chyn Chua
- The Alfred Hospital and Monash University, Melbourne, Australia
- The University of Melbourne, Melbourne, Australia
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- The Northern Hospital, Melbourne, Australia
| | - Michael Ashby
- The Alfred Hospital and Monash University, Melbourne, Australia
| | | | - Shaun Fleming
- The Alfred Hospital and Monash University, Melbourne, Australia
| | - Chun Y. Fong
- Austin Health and Olivia Newton John Cancer Research Institute, Melbourne, Australia
| | - Tse-Chieh Teh
- The Alfred Hospital and Monash University, Melbourne, Australia
- Box Hill Hospital, Melbourne, Australia
| | - Piers Blombery
- Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Richard Dillon
- Department of Medical and Molecular Genetics, King's College, London, United Kingdom
- Guy's Hospital, London, United Kingdom
| | - Adam Ivey
- The Alfred Hospital and Monash University, Melbourne, Australia
| | - Andrew H. Wei
- Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, Australia
- The University of Melbourne, Melbourne, Australia
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| |
Collapse
|
42
|
Rathgeber AC, Ludwig LS, Penter L. Single-cell genomics-based immune and disease monitoring in blood malignancies. Clin Hematol Int 2024; 6:62-84. [PMID: 38884110 PMCID: PMC11180218 DOI: 10.46989/001c.117961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 12/25/2023] [Indexed: 06/18/2024] Open
Abstract
Achieving long-term disease control using therapeutic immunomodulation is a long-standing concept with a strong tradition in blood malignancies. Besides allogeneic hematopoietic stem cell transplantation that continues to provide potentially curative treatment for otherwise challenging diagnoses, recent years have seen impressive progress in immunotherapies for leukemias and lymphomas with immune checkpoint blockade, bispecific monoclonal antibodies, and CAR T cell therapies. Despite their success, non-response, relapse, and immune toxicities remain frequent, thus prioritizing the elucidation of the underlying mechanisms and identifying predictive biomarkers. The increasing availability of single-cell genomic tools now provides a system's immunology view to resolve the molecular and cellular mechanisms of immunotherapies at unprecedented resolution. Here, we review recent studies that leverage these technological advancements for tracking immune responses, the emergence of immune resistance, and toxicities. As single-cell immune monitoring tools evolve and become more accessible, we expect their wide adoption for routine clinical applications to catalyze more precise therapeutic steering of personal immune responses.
Collapse
Affiliation(s)
- Anja C. Rathgeber
- Berlin Institute for Medical Systems BiologyMax Delbrück Center for Molecular Medicine
- Department of Hematology, Oncology, and TumorimmunologyCharité - Universitätsmedizin Berlin
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin
| | - Leif S. Ludwig
- Berlin Institute for Medical Systems BiologyMax Delbrück Center for Molecular Medicine
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin
| | - Livius Penter
- Department of Hematology, Oncology, and TumorimmunologyCharité - Universitätsmedizin Berlin
- BIH Biomedical Innovation AcademyBerlin Institute of Health at Charité - Universitätsmedizin Berlin
| |
Collapse
|
43
|
Djordjevic S, Itzykson R, Hague F, Lebon D, Legrand J, Ouled‐Haddou H, Jedraszak G, Harbonnier J, Collet L, Paubelle E, Marolleau J, Garçon L, Boyer T. STIM2 is involved in the regulation of apoptosis and the cell cycle in normal and malignant monocytic cells. Mol Oncol 2024; 18:1571-1592. [PMID: 38234211 PMCID: PMC11161727 DOI: 10.1002/1878-0261.13584] [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: 09/27/2023] [Revised: 11/28/2023] [Accepted: 01/02/2024] [Indexed: 01/19/2024] Open
Abstract
Calcium is a ubiquitous messenger that regulates a wide range of cellular functions, but its involvement in the pathophysiology of acute myeloid leukemia (AML) is not widely investigated. Here, we identified, from an analysis of The Cancer Genome Atlas and genotype-tissue expression databases, stromal interaction molecule 2 (STIM2) as being highly expressed in AML with monocytic differentiation and negatively correlated with overall survival. This was confirmed on a validation cohort of 407 AML patients. We then investigated the role of STIM2 in cell proliferation, differentiation, and survival in two leukemic cell lines with monocytic potential and in normal hematopoietic stem cells. STIM2 expression increased at the RNA and protein levels upon monocyte differentiation. Phenotypically, STIM2 knockdown drastically inhibited cell proliferation and induced genomic stress with DNA double-strand breaks, as shown by increased levels of phosphorylate histone H2AXγ (p-H2AXγ), followed by activation of the cellular tumor antigen p53 pathway, decreased expression of cell cycle regulators such as cyclin-dependent kinase 1 (CDK1)-cyclin B1 and M-phase inducer phosphatase 3 (CDC25c), and a decreased apoptosis threshold with a low antiapoptotic/proapoptotic protein ratio. Our study reports STIM2 as a new actor regulating genomic stability and p53 response in terms of cell cycle and apoptosis of human normal and malignant monocytic cells.
Collapse
Affiliation(s)
| | - Raphaël Itzykson
- Département Hématologie et ImmunologieHôpital Saint‐Louis, Assistance Publique‐Hôpitaux de ParisFrance
- Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRSUniversité Paris CitéFrance
| | - Frédéric Hague
- Laboratoire de Physiologie Cellulaire et Moléculaire UR4667Université Picardie Jules VerneAmiensFrance
| | - Delphine Lebon
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Service d'Hématologie Clinique et de Thérapie CellulaireCHU Amiens‐PicardieFrance
| | - Julien Legrand
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
| | | | - Guillaume Jedraszak
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Laboratoire de Génétique ConstitutionnelleCHU Amiens‐PicardieFrance
| | | | - Louison Collet
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
| | - Etienne Paubelle
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Service d'Hématologie Clinique et de Thérapie CellulaireCHU Amiens‐PicardieFrance
| | - Jean‐Pierre Marolleau
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Service d'Hématologie Clinique et de Thérapie CellulaireCHU Amiens‐PicardieFrance
| | - Loïc Garçon
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Service d'Hématologie BiologiqueCHU Amiens‐PicardieFrance
| | - Thomas Boyer
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Service d'Hématologie BiologiqueCHU Amiens‐PicardieFrance
| |
Collapse
|
44
|
Hall T, Gurbuxani S, Crispino JD. Malignant progression of preleukemic disorders. Blood 2024; 143:2245-2255. [PMID: 38498034 PMCID: PMC11181356 DOI: 10.1182/blood.2023020817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 02/23/2024] [Accepted: 02/29/2024] [Indexed: 03/19/2024] Open
Abstract
ABSTRACT The spectrum of myeloid disorders ranges from aplastic bone marrow failure characterized by an empty bone marrow completely lacking in hematopoiesis to acute myeloid leukemia in which the marrow space is replaced by undifferentiated leukemic blasts. Recent advances in the capacity to sequence bulk tumor population as well as at a single-cell level has provided significant insight into the stepwise process of transformation to acute myeloid leukemia. Using models of progression in the context of germ line predisposition (trisomy 21, GATA2 deficiency, and SAMD9/9L syndrome), premalignant states (clonal hematopoiesis and clonal cytopenia of unknown significance), and myelodysplastic syndrome, we review the mechanisms of progression focusing on the hierarchy of clonal mutation and potential roles of transcription factor alterations, splicing factor mutations, and the bone marrow environment in progression to acute myeloid leukemia. Despite major advances in our understanding, preventing the progression of these disorders or treating them at the acute leukemia phase remains a major area of unmet medical need.
Collapse
Affiliation(s)
- Trent Hall
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Sandeep Gurbuxani
- Section of Hematopathology, Department of Pathology, University of Chicago, Chicago, IL
| | - John D. Crispino
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| |
Collapse
|
45
|
Pearce SM, Cross NA, Smith DP, Clench MR, Flint LE, Hamm G, Goodwin R, Langridge JI, Claude E, Cole LM. Multimodal Mass Spectrometry Imaging of an Osteosarcoma Multicellular Tumour Spheroid Model to Investigate Drug-Induced Response. Metabolites 2024; 14:315. [PMID: 38921450 PMCID: PMC11205347 DOI: 10.3390/metabo14060315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/27/2024] Open
Abstract
A multimodal mass spectrometry imaging (MSI) approach was used to investigate the chemotherapy drug-induced response of a Multicellular Tumour Spheroid (MCTS) 3D cell culture model of osteosarcoma (OS). The work addresses the critical demand for enhanced translatable early drug discovery approaches by demonstrating a robust spatially resolved molecular distribution analysis in tumour models following chemotherapeutic intervention. Advanced high-resolution techniques were employed, including desorption electrospray ionisation (DESI) mass spectrometry imaging (MSI), to assess the interplay between metabolic and cellular pathways in response to chemotherapeutic intervention. Endogenous metabolite distributions of the human OS tumour models were complemented with subcellularly resolved protein localisation by the detection of metal-tagged antibodies using Imaging Mass Cytometry (IMC). The first application of matrix-assisted laser desorption ionization-immunohistochemistry (MALDI-IHC) of 3D cell culture models is reported here. Protein localisation and expression following an acute dosage of the chemotherapy drug doxorubicin demonstrated novel indications for mechanisms of region-specific tumour survival and cell-cycle-specific drug-induced responses. Previously unknown doxorubicin-induced metabolite upregulation was revealed by DESI-MSI of MCTSs, which may be used to inform mechanisms of chemotherapeutic resistance. The demonstration of specific tumour survival mechanisms that are characteristic of those reported for in vivo tumours has underscored the increasing value of this approach as a tool to investigate drug resistance.
Collapse
Affiliation(s)
- Sophie M. Pearce
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, Sheffield S1 1WB, UK; (S.M.P.); (N.A.C.); (D.P.S.); (M.R.C.)
| | - Neil A. Cross
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, Sheffield S1 1WB, UK; (S.M.P.); (N.A.C.); (D.P.S.); (M.R.C.)
| | - David P. Smith
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, Sheffield S1 1WB, UK; (S.M.P.); (N.A.C.); (D.P.S.); (M.R.C.)
| | - Malcolm R. Clench
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, Sheffield S1 1WB, UK; (S.M.P.); (N.A.C.); (D.P.S.); (M.R.C.)
| | - Lucy E. Flint
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, The Discovery Centre (DISC), Biomedical Campus, 1 Francis Crick Ave, Trumpington, Cambridge CB2 0AA, UK; (L.E.F.); (G.H.); (R.G.)
| | - Gregory Hamm
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, The Discovery Centre (DISC), Biomedical Campus, 1 Francis Crick Ave, Trumpington, Cambridge CB2 0AA, UK; (L.E.F.); (G.H.); (R.G.)
| | - Richard Goodwin
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, The Discovery Centre (DISC), Biomedical Campus, 1 Francis Crick Ave, Trumpington, Cambridge CB2 0AA, UK; (L.E.F.); (G.H.); (R.G.)
| | - James I. Langridge
- Waters Corporation, Stamford Avenue, Altrincham Road, Wilmslow, Cheshire SK9 4AX, UK; (J.I.L.); (E.C.)
| | - Emmanuelle Claude
- Waters Corporation, Stamford Avenue, Altrincham Road, Wilmslow, Cheshire SK9 4AX, UK; (J.I.L.); (E.C.)
| | - Laura M. Cole
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, Sheffield S1 1WB, UK; (S.M.P.); (N.A.C.); (D.P.S.); (M.R.C.)
| |
Collapse
|
46
|
Kim N, Hahn S, Choi YJ, Cho H, Chung H, Jang JE, Lyu CJ, Lee ST, Choi JR, Cheong JW, Shin S. Comprehensive insights into AML relapse: genetic mutations, clonal evolution, and clinical outcomes. Cancer Cell Int 2024; 24:174. [PMID: 38764048 PMCID: PMC11103850 DOI: 10.1186/s12935-024-03368-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 05/10/2024] [Indexed: 05/21/2024] Open
Abstract
INTRODUCTION Acute myeloid leukemia (AML) is a complex hematologic malignancy characterized by uncontrolled proliferation of myeloid precursor cells within bone marrow. Despite advances in understanding of its molecular underpinnings, AML remains a therapeutic challenge due to its high relapse rate and clonal evolution. METHODS In this retrospective study, we analyzed data from 24 AML patients diagnosed at a single institution between January 2017 and August 2023. Comprehensive genetic analyses, including chromosomal karyotyping, next-generation sequencing, and gene fusion assays, were performed on bone marrow samples obtained at initial diagnosis and relapse. Clinical data, treatment regimens, and patient outcomes were also documented. RESULTS Mutations in core genes of FLT3, NPM1, DNMT3A, and IDH2 were frequently discovered in diagnostic sample and remained in relapse sample. FLT3-ITD, TP53, KIT, RUNX1, and WT1 mutation were acquired at relapse in one patient each. Gene fusion assays revealed stable patterns, while chromosomal karyotype analyses indicated a greater diversity of mutations in relapsed patients. Clonal evolution patterns varied, with some cases showing linear or branching evolution and others exhibiting no substantial change in core mutations between diagnosis and relapse. CONCLUSIONS Our study integrates karyotype, gene rearrangements, and gene mutation results to provide a further understanding of AML heterogeneity and evolution. We demonstrate the clinical relevance of specific mutations and clonal evolution patterns, emphasizing the need for personalized therapies and measurable residual disease monitoring in AML management. By bridging the gap between genetics and clinical outcome, we move closer to tailored AML therapies and improved patient prognoses.
Collapse
Affiliation(s)
- Namsoo Kim
- Department of Laboratory Medicine, Yonsei University College of Medicine, Severance Hospital, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Seungmin Hahn
- Department of Pediatric Hematology-Oncology, Severance Hospital, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Yu Jeong Choi
- Department of Laboratory Medicine, Yonsei University College of Medicine, Severance Hospital, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Hyunsoo Cho
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Severance Hospital, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Haerim Chung
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Severance Hospital, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Ji Eun Jang
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Severance Hospital, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Chuhl Joo Lyu
- Department of Pediatric Hematology-Oncology, Severance Hospital, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Seung-Tae Lee
- Department of Laboratory Medicine, Yonsei University College of Medicine, Severance Hospital, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
- Dxome Co. Ltd, Seongnam-si, Gyeonggi-do, Korea
| | - Jong Rak Choi
- Department of Laboratory Medicine, Yonsei University College of Medicine, Severance Hospital, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
- Dxome Co. Ltd, Seongnam-si, Gyeonggi-do, Korea
| | - June-Won Cheong
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Severance Hospital, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea.
| | - Saeam Shin
- Department of Laboratory Medicine, Yonsei University College of Medicine, Severance Hospital, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea.
| |
Collapse
|
47
|
Lao Z, Ding LW, Sun QY, Jia L, Yan B, Ng AYJ, Capinpin SM, Wang R, Ying L, Chng WJ, Phillip Koeffler H, Koh WP, Yuan JM, Yang H, Goh YT, Grigoropoulos N. A Pre-Leukemic DNA Methylation Signature in Healthy Individuals at Higher Risk for Developing Myeloid Malignancy. Clin Cancer Res 2024; 30:2170-2180. [PMID: 38437679 PMCID: PMC11096012 DOI: 10.1158/1078-0432.ccr-22-3804] [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: 12/28/2022] [Revised: 10/23/2023] [Accepted: 02/29/2024] [Indexed: 03/06/2024]
Abstract
PURPOSE DNA methylation alterations are widespread in acute myelogenous leukemia (AML) and myelodysplastic syndrome (MDS), some of which appear to have evolved independently of somatic mutations in epigenetic regulators. Although the presence of somatic mutations in peripheral blood can predict the risk of development of AML and MDS, its accuracy remains unsatisfactory. EXPERIMENTAL DESIGN We performed global DNA methylation profiling in a case control study nested within the Singapore Chinese Health Study to evaluate whether DNA methylation alterations were associated with AML/MDS development. Targeted deep sequencing and methylated DNA immunoprecipitation sequencing (MeDIP-seq) were performed on peripheral blood collected a median of 9.9 years before diagnosis of AML or MDS, together with age-matched still-healthy individuals as controls. RESULTS Sixty-six individuals who developed AML or MDS displayed significant DNA methylation changes in the peripheral blood compared with 167 age- and gender-matched controls who did not develop AML/MDS during the follow-up period. Alterations in methylation in the differentially methylation regions were associated with increased odds of developing AML/MDS. CONCLUSIONS The epigenetic changes may be acquired independently and before somatic mutations that are relevant for AML/MDS development. The association between methylation changes and the risk of pre-AML/MDS in these individuals was considerably stronger than somatic mutations, suggesting that methylation changes could be used as biomarkers for pre-AML/MDS screening.
Collapse
Affiliation(s)
- Zhentang Lao
- Department of Haematology, Singapore General Hospital, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Ling-Wen Ding
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- Department of Pathology, National University of Singapore; Nanomedicine Translational Research Programme, Yong Yoo Lin School of Medicine, National University of Singapore. Singapore
| | - Qiao-Yang Sun
- Department of Haematology, Singapore General Hospital, Singapore
- Department of Neurology, Singapore General Hospital, National Neuroscience Institute, Singapore
| | - Li Jia
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Benedict Yan
- Department of Laboratory Medicine, National University Hospital, Singapore
| | | | - Sharah Mae Capinpin
- Healthy Longitudinal Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, National University of Singapore, Singapore
| | - Renwei Wang
- Division of Cancer Control and Population Sciences, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Li Ying
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Wee Joo Chng
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- NUS Center for Cancer Research and Dept of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- National University Cancer Institute, National University Health System, Singapore
| | - H Phillip Koeffler
- National University Cancer Institute, National University Health System, Singapore
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Woon-Puay Koh
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jian-Min Yuan
- Division of Cancer Control and Population Sciences, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania. USA
| | - Henry Yang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- Department of Biomedical Informatics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yeow Tee Goh
- Department of Haematology, Singapore General Hospital, Singapore
| | | |
Collapse
|
48
|
Stelljes M, Middeke JM, Bug G, Wagner-Drouet EM, Müller LP, Schmid C, Krause SW, Bethge W, Jost E, Platzbecker U, Klein SA, Schubert J, Niederland J, Kaufmann M, Schäfer-Eckart K, Schaich M, Baldauf H, Stölzel F, Petzold C, Röllig C, Alakel N, Steffen B, Hauptrock B, Schliemann C, Sockel K, Lang F, Kriege O, Schaffrath J, Reicherts C, Berdel WE, Serve H, Ehninger G, Schmidt AH, Bornhäuser M, Mikesch JH, Schetelig J. Remission induction versus immediate allogeneic haematopoietic stem cell transplantation for patients with relapsed or poor responsive acute myeloid leukaemia (ASAP): a randomised, open-label, phase 3, non-inferiority trial. Lancet Haematol 2024; 11:e324-e335. [PMID: 38583455 DOI: 10.1016/s2352-3026(24)00065-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/16/2024] [Accepted: 02/16/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND Whether high-dose cytarabine-based salvage chemotherapy, administered to induce complete remission in patients with poor responsive or relapsed acute myeloid leukaemia scheduled for allogeneic haematopoietic stem-cell transplantation (HSCT) after intensive conditioning confers a survival advantage, is unclear. METHODS To test salvage chemotherapy before allogeneic HSCT, patients aged between 18 and 75 years with non-favourable-risk acute myeloid leukaemia not in complete remission after first induction or untreated first relapse were randomly assigned 1:1 to remission induction with high-dose cytarabine (3 g/m2 intravenously, 1 g/m2 intravenously for patients >60 years or with a substantial comorbidity) twice daily on days 1-3 plus mitoxantrone (10 mg/m2 intravenously) on days 3-5 or immediate allogeneic HSCT for the disease control group. Block randomisation with variable block lengths was used and patients were stratified by age, acute myeloid leukaemia risk, and disease status. The study was open label. The primary endpoint was treatment success, defined as complete remission on day 56 after allogeneic HSCT, with the aim to show non-inferiority for disease control compared with remission induction with a non-inferiority-margin of 5% and one-sided type 1 error of 2·5%. The primary endpoint was analysed in both the intention-to-treat (ITT) population and in the per-protocol population. The trial is completed and was registered at ClinicalTrials.gov, NCT02461537. FINDINGS 281 patients were enrolled between Sept 17, 2015, and Jan 12, 2022. Of 140 patients randomly assigned to disease control, 135 (96%) proceeded to allogeneic HSCT, 97 (69%) after watchful waiting only. Of 141 patients randomly assigned to remission induction, 134 (95%) received salvage chemotherapy and 128 (91%) patients subsequently proceeded to allogeneic HSCT. In the ITT population, treatment success was observed in 116 (83%) of 140 patients in the disease control group versus 112 (79%) of 141 patients with remission induction (test for non-inferiority, p=0·036). Among per-protocol treated patients, treatment success was observed in 116 (84%) of 138 patients with disease control versus 109 (81%) of 134 patients in the remission induction group (test for non-inferiority, p=0·047). The difference in treatment success between disease control and remission induction was estimated as 3·4% (95% CI -5·8 to 12·6) for the ITT population and 2·7% (-6·3 to 11·8) for the per-protocol population. Fewer patients with disease control compared with remission induction had non-haematological adverse events grade 3 or worse (30 [21%] of 140 patients vs 86 [61%] of 141 patients, χ2 test p<0·0001). Between randomisation and the start of conditioning, with disease control two patients died from progressive acute myeloid leukaemia and zero from treatment-related complications, and with remission induction two patients died from progressive acute myeloid leukaemia and two from treatment-related complications. Between randomisation and allogeneic HSCT, patients with disease control spent a median of 27 days less in hospital than those with remission induction, ie, the median time in hospital was 15 days (range 7-64) versus 42 days (27-121, U test p<0·0001), respectively. INTERPRETATION Non-inferiority of disease control could not be shown at the 2·5% significance level. The rate of treatment success was also not statistically better for patients with remission induction. Watchful waiting and immediate transplantation could be an alternative for fit patients with poor response or relapsed acute myeloid leukaemia who have a stem cell donor available. More randomised controlled intention-to-transplant trials are needed to define the optimal treatment before transplantation for patients with active acute myeloid leukaemia. FUNDING DKMS and the Gert and Susanna Mayer Stiftung Foundation.
Collapse
Affiliation(s)
| | | | - Gesine Bug
- Goethe University Frankfurt, Frankfurt am Main, Germany
| | | | - Lutz P Müller
- University Hospital, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Christoph Schmid
- Faculty of Medicine, Augsburg University Hospital, Augsburg, Germany
| | | | | | - Edgar Jost
- University Hospital Aachen & Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf, Aachen, Germany
| | | | | | | | - Judith Niederland
- Helios Klinikum Berlin-Buch, Klinik für Hämatologie und Zelltherapie, Berlin, Germany
| | | | | | | | | | - Friedrich Stölzel
- University Hospital TU Dresden, Dresden, Germany; University Hospital Schleswig-Holstein, Kiel, Germany
| | | | | | - Nael Alakel
- University Hospital TU Dresden, Dresden, Germany
| | - Björn Steffen
- Goethe University Frankfurt, Frankfurt am Main, Germany
| | | | | | - Katja Sockel
- University Hospital TU Dresden, Dresden, Germany
| | - Fabian Lang
- Goethe University Frankfurt, Frankfurt am Main, Germany
| | | | - Judith Schaffrath
- University Hospital, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | | | | | - Hubert Serve
- Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Gerhard Ehninger
- University Hospital TU Dresden, Dresden, Germany; Cellex Cell Professionals, Cologne, Germany
| | | | - Martin Bornhäuser
- University Hospital TU Dresden, Dresden, Germany; National Center for Tumor Diseases, Dresden, Germany
| | | | - Johannes Schetelig
- University Hospital TU Dresden, Dresden, Germany; DKMS gemeinnützige GmbH, Tübingen und Dresden, Germany.
| |
Collapse
|
49
|
Olesinski EA, Bhatia KS, Wang C, Pioso MS, Lin XX, Mamdouh AM, Ng SX, Sandhu V, Jasdanwala SS, Yilma B, Bohl S, Ryan JA, Malani D, Luskin MR, Kallioniemi O, Porkka K, Adamia S, Chng WJ, Osato M, Weinstock DM, Garcia JS, Letai A, Bhatt S. Acquired Multidrug Resistance in AML Is Caused by Low Apoptotic Priming in Relapsed Myeloblasts. Blood Cancer Discov 2024; 5:180-201. [PMID: 38442309 PMCID: PMC11061585 DOI: 10.1158/2643-3230.bcd-24-0001] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 10/05/2023] [Accepted: 12/19/2023] [Indexed: 03/07/2024] Open
Abstract
In many cancers, mortality is associated with the emergence of relapse with multidrug resistance (MDR). Thus far, the investigation of cancer relapse mechanisms has largely focused on acquired genetic mutations. Using acute myeloid leukemia (AML) patient-derived xenografts (PDX), we systematically elucidated a basis of MDR and identified drug sensitivity in relapsed AML. We derived pharmacologic sensitivity for 22 AML PDX models using dynamic BH3 profiling (DBP), together with genomics and transcriptomics. Using in vivo acquired resistant PDXs, we found that resistance to unrelated, narrowly targeted agents in distinct PDXs was accompanied by broad resistance to drugs with disparate mechanisms. Moreover, baseline mitochondrial apoptotic priming was consistently reduced regardless of the class of drug-inducing selection. By applying DBP, we identified drugs showing effective in vivo activity in resistant models. This study implies evasion of apoptosis drives drug resistance and demonstrates the feasibility of the DBP approach to identify active drugs for patients with relapsed AML. SIGNIFICANCE Acquired resistance to targeted therapy remains challenging in AML. We found that reduction in mitochondrial priming and common transcriptomic signatures was a conserved mechanism of acquired resistance across different drug classes in vivo. Drugs active in vivo can be identified even in the multidrug resistant state by DBP.
Collapse
MESH Headings
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/genetics
- Humans
- Apoptosis/drug effects
- Animals
- Mice
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Multiple/genetics
- Drug Resistance, Multiple/drug effects
- Xenograft Model Antitumor Assays
- Granulocyte Precursor Cells/drug effects
- Granulocyte Precursor Cells/pathology
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
Collapse
Affiliation(s)
- Elyse A. Olesinski
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | | | - Chuqi Wang
- Department of Pharmacy, National University of Singapore, Singapore
| | - Marissa S. Pioso
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Xiao Xian Lin
- Department of Pharmacy, National University of Singapore, Singapore
| | - Ahmed M. Mamdouh
- Department of Pharmacy, National University of Singapore, Singapore
| | - Shu Xuan Ng
- Department of Pharmacy, National University of Singapore, Singapore
| | - Vedant Sandhu
- Department of Pharmacy, National University of Singapore, Singapore
| | | | - Binyam Yilma
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Stephan Bohl
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Jeremy A. Ryan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Disha Malani
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Marlise R. Luskin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Olli Kallioniemi
- Institute for Molecular Medicine Finland FIMM, Hi-Life, University of Helsinki, Helsinki, Finland
- Science for Life Laboratory, Department of Oncology and Pathology, Karolinska Institute, Solna, Sweden
| | - Kimmo Porkka
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Department of Hematology, HUS, Helsinki, Finland
| | - Sophia Adamia
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Wee Joo Chng
- Cancer Science Institute of Singapore, National University of Singapore, Centre for Translational Medicine, Singapore
| | - Motomi Osato
- Cancer Science Institute of Singapore, National University of Singapore, Centre for Translational Medicine, Singapore
| | - David M. Weinstock
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Jacqueline S. Garcia
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Anthony Letai
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Shruti Bhatt
- Department of Pharmacy, National University of Singapore, Singapore
| |
Collapse
|
50
|
Nuno K, Azizi A, Koehnke T, Lareau C, Ediriwickrema A, Corces MR, Satpathy AT, Majeti R. Convergent epigenetic evolution drives relapse in acute myeloid leukemia. eLife 2024; 13:e93019. [PMID: 38647535 PMCID: PMC11034943 DOI: 10.7554/elife.93019] [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: 09/26/2023] [Accepted: 03/26/2024] [Indexed: 04/25/2024] Open
Abstract
Relapse of acute myeloid leukemia (AML) is highly aggressive and often treatment refractory. We analyzed previously published AML relapse cohorts and found that 40% of relapses occur without changes in driver mutations, suggesting that non-genetic mechanisms drive relapse in a large proportion of cases. We therefore characterized epigenetic patterns of AML relapse using 26 matched diagnosis-relapse samples with ATAC-seq. This analysis identified a relapse-specific chromatin accessibility signature for mutationally stable AML, suggesting that AML undergoes epigenetic evolution at relapse independent of mutational changes. Analysis of leukemia stem cell (LSC) chromatin changes at relapse indicated that this leukemic compartment underwent significantly less epigenetic evolution than non-LSCs, while epigenetic changes in non-LSCs reflected overall evolution of the bulk leukemia. Finally, we used single-cell ATAC-seq paired with mitochondrial sequencing (mtscATAC) to map clones from diagnosis into relapse along with their epigenetic features. We found that distinct mitochondrially-defined clones exhibit more similar chromatin accessibility at relapse relative to diagnosis, demonstrating convergent epigenetic evolution in relapsed AML. These results demonstrate that epigenetic evolution is a feature of relapsed AML and that convergent epigenetic evolution can occur following treatment with induction chemotherapy.
Collapse
Affiliation(s)
- Kevin Nuno
- Cancer Biology Graduate Program, Stanford University School of MedicineStanfordUnited States
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
- Cancer Institute, Stanford University School of MedicineStanfordUnited States
- Department of Medicine, Division of Hematology, Stanford University School of MedicineStanfordUnited States
| | - Armon Azizi
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
- Cancer Institute, Stanford University School of MedicineStanfordUnited States
- Department of Medicine, Division of Hematology, Stanford University School of MedicineStanfordUnited States
- University of California Irvine School of MedicineIrvineUnited States
| | - Thomas Koehnke
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
- Cancer Institute, Stanford University School of MedicineStanfordUnited States
- Department of Medicine, Division of Hematology, Stanford University School of MedicineStanfordUnited States
| | - Caleb Lareau
- Department of Pathology, Stanford UniversityStanfordUnited States
- Program in Immunology, Stanford UniversityStanfordUnited States
| | - Asiri Ediriwickrema
- Cancer Biology Graduate Program, Stanford University School of MedicineStanfordUnited States
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
- Cancer Institute, Stanford University School of MedicineStanfordUnited States
- Department of Medicine, Division of Hematology, Stanford University School of MedicineStanfordUnited States
| | - M Ryan Corces
- Cancer Biology Graduate Program, Stanford University School of MedicineStanfordUnited States
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
- Cancer Institute, Stanford University School of MedicineStanfordUnited States
- Department of Medicine, Division of Hematology, Stanford University School of MedicineStanfordUnited States
- Gladstone Institute of Neurological DiseaseSan FranciscoUnited States
- Gladstone Institute of Data Science and BiotechnologySan FranciscoUnited States
- Department of Neurology, University of California, San FranciscoSan FranciscoUnited States
| | - Ansuman T Satpathy
- Department of Pathology, Stanford UniversityStanfordUnited States
- Program in Immunology, Stanford UniversityStanfordUnited States
- Parker Institute for Cancer Immunotherapy, Stanford UniversityStanfordUnited States
- Gladstone-UCSF Institute of Genomic ImmunologySan FranciscoUnited States
| | - Ravindra Majeti
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
- Cancer Institute, Stanford University School of MedicineStanfordUnited States
- Department of Medicine, Division of Hematology, Stanford University School of MedicineStanfordUnited States
| |
Collapse
|