1
|
Dissecting the Genetic and Non-Genetic Heterogeneity of Acute Myeloid Leukemia Using Next-Generation Sequencing and In Vivo Models. Cancers (Basel) 2022; 14:cancers14092182. [PMID: 35565315 PMCID: PMC9103951 DOI: 10.3390/cancers14092182] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Acute myeloid leukemia (AML) is an extremely aggressive form of blood cancer with high rates of treatment failure. AML arises from the stepwise acquisition of genetic aberrations and is a highly heterogeneous disorder. Recent research has shown that individual AML samples often contain several clones that are defined by a distinct combination of genetic lesions, epigenetic patterns and cell surface marker expression profiles. A better understanding of the clonal dynamics of AML is required to develop novel treatment strategies against this disease. In this review, we discuss the recent developments that have further deepened our understanding of clonal evolution and heterogeneity in AML. Abstract Acute myeloid leukemia (AML) is an extremely aggressive and heterogeneous disorder that results from the transformation of hematopoietic stem cells. Although our understanding of the molecular pathology of AML has greatly improved in the last few decades, the overall and relapse free survival rates among AML patients remain quite poor. This is largely due to evolution of the disease and selection of the fittest, treatment-resistant leukemic clones. There is increasing evidence that most AMLs possess a highly complex clonal architecture and individual leukemias are comprised of genetically, phenotypically and epigenetically distinct clones, which are continually evolving. Advances in sequencing technologies as well as studies using murine AML models have provided further insights into the heterogeneity of leukemias. We will review recent advances in the field of genetic and non-genetic heterogeneity in AML.
Collapse
|
2
|
Noble JN, Mishra A. Development and Significance of Mouse Models in Lymphoma Research. Curr Hematol Malig Rep 2020; 14:119-126. [PMID: 30848424 DOI: 10.1007/s11899-019-00504-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
PURPOSE OF REVIEW Animal models have played an indispensable role in interpreting cancer gene functions, pathogenesis of disease, and in the development of innovative therapeutic approaches targeting aberrant biological pathways in human cancers. RECENT FINDINGS These models have guided the therapeutic targeting of cancer-causing mutations and paved the way for assessing anti-cancer drug responses and the preclinical development of immunotherapies. The mammalian models of cancer utilize genetically edited or transplanted mice that develop fairly accurate disease histopathology. The mouse model also allows us to study the effect of tumor microenvironment in the development of lymphoma. The emergence of patient-derived xenografts provides a better opportunity for recapitulating primary lymphoma characteristics and researching personalized drug therapy. In conclusion, the refinement and advancement of available mouse models in lymphoma significantly minimize the therapeutic translational failures in patients.
Collapse
Affiliation(s)
- Jordan N Noble
- College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Anjali Mishra
- College of Medicine, The Ohio State University, Columbus, OH, 43210, USA. .,Comprehensive Cancer Center, James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH, USA. .,Division of Dermatology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA. .,Division of Hematologic Malignancies and Hematopoietic Stem Cell Transplantation, Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, 233 South 10th Street, Philadephia, PA, 19107, USA.
| |
Collapse
|
3
|
Xu ZT, Ding H, Fu TT, Zhu YL, Wang WP. A Nude Mouse Model of Orthotopic Liver Transplantation of Human Hepatocellular Carcinoma HCCLM3 Cell Xenografts and the Use of Imaging to Evaluate Tumor Progression. Med Sci Monit 2019; 25:8694-8703. [PMID: 31736477 PMCID: PMC6880650 DOI: 10.12659/msm.917648] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background This study aimed to develop a nude mouse model of orthotopic liver transplantation of HCCLM3 human hepatocellular carcinoma (HCC) cell xenografts and the use of imaging and histology to evaluate tumor development and progression. Material/Methods HCCLM3 cells were injected subcutaneously into 25 healthy male athymic BALB/c (nu/nu) nude mice. The tumors that developed were transplanted into the liver of a new set of nude mice. After four weeks and six weeks, the mice were imaged using ultrasound (US), software-assisted contrast-enhanced ultrasound (CEUS), fluorodeoxyglucose-positron emission tomography (FDG-PET). Histology was performed on the liver and liver tumors, and included immunohistochemistry for vascular endothelial growth factor (VEGF), CD31, CD34, and α-smooth muscle actin (α-SMA). Results The success rate for orthotopic tumor transplantation in the mouse liver was 90% (18/20). Liver tumors measured 11.8±2.6 mm in diameter and 525.9±250.8 mm3 in volume on the sixth week. CEUS showed rapid wash-in and washout in the liver tumors, and PET showed low tumor cell metabolism. Bone metastases were present in 45% (9/20) of mice in the sixth week. Immunohistochemistry showed positive expression for VEGF, CD31, CD34, and α-SMA. Conclusions The nude mouse orthotopic liver transplantation model of human HCC was shown to be a reliable model that has the potential for future research on the pathogenesis and progression of HCC and studies on drug development.
Collapse
Affiliation(s)
- Zhi-Ting Xu
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (mainland).,Shanghai Institute of Medical Imaging, Shanghai, China (mainland)
| | - Hong Ding
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (mainland)
| | - Tian-Tian Fu
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (mainland).,Shanghai Institute of Medical Imaging, Shanghai, China (mainland)
| | - Yu-Li Zhu
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (mainland)
| | - Wen-Ping Wang
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (mainland)
| |
Collapse
|
4
|
Almosailleakh M, Schwaller J. Murine Models of Acute Myeloid Leukaemia. Int J Mol Sci 2019; 20:E453. [PMID: 30669675 PMCID: PMC6358780 DOI: 10.3390/ijms20020453] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/08/2019] [Accepted: 01/11/2019] [Indexed: 01/08/2023] Open
Abstract
Acute myeloid leukaemia (AML) is a rare but severe form of human cancer that results from a limited number of functionally cooperating genetic abnormalities leading to uncontrolled proliferation and impaired differentiation of hematopoietic stem and progenitor cells. Before the identification of genetic driver lesions, chemically, irradiation or viral infection-induced mouse leukaemia models provided platforms to test novel chemotherapeutics. Later, transgenic mouse models were established to test the in vivo transforming potential of newly cloned fusion genes and genetic aberrations detected in patients' genomes. Hereby researchers constitutively or conditionally expressed the respective gene in the germline of the mouse or reconstituted the hematopoietic system of lethally irradiated mice with bone marrow virally expressing the mutation of interest. More recently, immune deficient mice have been explored to study patient-derived human AML cells in vivo. Unfortunately, although complementary to each other, none of the currently available strategies faithfully model the initiation and progression of the human disease. Nevertheless, fast advances in the fields of next generation sequencing, molecular technology and bioengineering are continuously contributing to the generation of better mouse models. Here we review the most important AML mouse models of each category, briefly describe their advantages and limitations and show how they have contributed to our understanding of the biology and to the development of novel therapies.
Collapse
MESH Headings
- Animals
- Bone Marrow Transplantation
- Carcinogens/administration & dosage
- Cell Transformation, Viral
- Disease Models, Animal
- Gene Editing
- Heterografts
- Humans
- Immunocompromised Host
- Leukemia, Myeloid, Acute/etiology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Mice
- Mice, Transgenic
- Radiation, Ionizing
Collapse
Affiliation(s)
- Marwa Almosailleakh
- Department of Biomedicine, University Children's Hospital beider Basel (UKBB), University of Basel, 4031 Basel, Switzerland.
| | - Juerg Schwaller
- Department of Biomedicine, University Children's Hospital beider Basel (UKBB), University of Basel, 4031 Basel, Switzerland.
| |
Collapse
|
5
|
Xue W, Li W, Shang Y, Zhang Y, Lan X, Wang G, Li Z, Zhang X, Song Y, Wu B, Dong M, Wang X, Zhang M. One method to establish Epstein-Barr virus-associated NK/T cell lymphoma mouse models. J Cell Mol Med 2018; 23:1509-1516. [PMID: 30484952 PMCID: PMC6349153 DOI: 10.1111/jcmm.14057] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 10/22/2018] [Accepted: 11/05/2018] [Indexed: 01/18/2023] Open
Abstract
Novel nude mice model of human NK/T cell lymphoma were established by subcutaneously injecting two NK/T cell lymphoma cell lines into the right axillary region of mice and successful passages were completed by injecting cell suspension which was obtained through a 70‐μm cell strainer. These mice models and corresponding cell clones have been successfully developed for more than 8 generations. The survival rates of both resuscitation and transplantation in NKYS and YT models were 90% and 70% correspondingly. Pathologically, the tumour cells in all passages of the lymphoma‐bearing mice and cell lines obtained from tumours were parallel to initial cell lines. Immunologically, the tumour cells expressed the characteristics of the primary and essential NK/T lymphomas. The novel mice models maintained the essential features of human NK/T cell lymphoma, and they would be ideal tools in vivo for further research of human NK/T cell lymphoma.
Collapse
Affiliation(s)
- Weili Xue
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Jonint International Research Laboratory of Lymphoma, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Weiming Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Jonint International Research Laboratory of Lymphoma, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Yufeng Shang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Jonint International Research Laboratory of Lymphoma, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanjie Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Jonint International Research Laboratory of Lymphoma, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xuan Lan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Jonint International Research Laboratory of Lymphoma, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guannan Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhaoming Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Jonint International Research Laboratory of Lymphoma, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, China
| | - Xudong Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Jonint International Research Laboratory of Lymphoma, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, China
| | - Yue Song
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Jonint International Research Laboratory of Lymphoma, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Baopeng Wu
- The Boiler & Pressure Vessel Safety Inspection Institute of Henan Province, Zhengzhou, China
| | - Meng Dong
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Jonint International Research Laboratory of Lymphoma, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinhua Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Jonint International Research Laboratory of Lymphoma, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Jonint International Research Laboratory of Lymphoma, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, China
| |
Collapse
|
6
|
Lopez-Lastra S, Di Santo JP. Modeling Natural Killer Cell Targeted Immunotherapies. Front Immunol 2017; 8:370. [PMID: 28405194 PMCID: PMC5370275 DOI: 10.3389/fimmu.2017.00370] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 03/14/2017] [Indexed: 01/01/2023] Open
Abstract
Animal models have extensively contributed to our understanding of human immunobiology and to uncover the underlying pathological mechanisms occurring in the development of diseases. However, mouse models do not reproduce the genetic and molecular complexity inherent in human disease conditions. Human immune system (HIS) mouse models that are susceptible to human pathogens and can recapitulate human hematopoiesis and tumor immunobiology provide one means to bridge the interspecies gap. Natural killer cells are the founding member of the innate lymphoid cell family. They exert a rapid and strong immune response against tumor and pathogen-infected cells. Their antitumor features have long been exploited for therapeutic purposes in the context of cancer. In this review, we detail the development of highly immunodeficient mouse strains and the models currently used in cancer research. We summarize the latest improvements in adoptive natural killer (NK) cell therapies and the development of novel NK cell sources. Finally, we discuss the advantages of HIS mice to study the interactions between human NK cells and human cancers and to develop new therapeutic strategies.
Collapse
Affiliation(s)
- Silvia Lopez-Lastra
- Innate Immunity Unit, Institut Pasteur, Paris, France
- Inserm U1223, Paris, France
- Université Paris-Sud (Paris-Saclay), Paris, France
| | - James P. Di Santo
- Innate Immunity Unit, Institut Pasteur, Paris, France
- Inserm U1223, Paris, France
| |
Collapse
|
7
|
ZHANG JINGCHENG, LUO JIA, LIU FANRONG, WU DONGMEI, ZHONG QINGLING, ZENG LIANGTAO, WU ZIQING, ZHAO TONG, WU LIQING, HAO HUA. Diabetes mellitus potentiates diffuse large B-cell lymphoma via high levels of CCL5. Mol Med Rep 2014; 10:1231-6. [DOI: 10.3892/mmr.2014.2341] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Accepted: 05/29/2014] [Indexed: 11/06/2022] Open
|
8
|
Tassone P, Neri P, Burger R, Di Martino MT, Leone E, Amodio N, Caraglia M, Tagliaferri P. Mouse models as a translational platform for the development of new therapeutic agents in multiple myeloma. Curr Cancer Drug Targets 2013; 12:814-22. [PMID: 22671927 PMCID: PMC3587184 DOI: 10.2174/156800912802429292] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 11/25/2011] [Accepted: 12/08/2011] [Indexed: 12/22/2022]
Abstract
Mouse models of multiple myeloma (MM) are basic tools for translational research and play a fundamental role in the development of new therapeutics against plasma cell malignancies. All available models, including transplantable murine tumors in syngenic mice, xenografts of established human cell lines in immunocompromised mice and transgenic models that mirror specific steps of MM pathogenesis, have demonstrated some weaknesses in predicting clinical results, particularly for new drugs targeting the human bone marrow microenvironment (huBMM). The recent interest to models recapitulating the in vivo growth of primary MM cells in a human (SCID-hu) or humanized (SCID-synth-hu) host recipient has provided powerful platforms for the investigation of new compounds targeting MM and/or its huBMM. Here, we review and discuss strengths and weaknesses of the key in vivo models that are currently utilized in the MM preclinical investigation.
Collapse
Affiliation(s)
- P Tassone
- Medical Oncology, Magna Græcia University, Viale Europa, Campus Salvatore Venuta, 88100 Catanzaro, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Ablain J, Nasr R, Zhu J, Bazarbachi A, Lallemand-Breittenbach V, de Thé H. How animal models of leukaemias have already benefited patients. Mol Oncol 2013; 7:224-31. [PMID: 23453906 DOI: 10.1016/j.molonc.2013.01.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 01/22/2013] [Indexed: 10/27/2022] Open
Abstract
The relative genetic simplicity of leukaemias, the development of which likely relies on a limited number of initiating events has made them ideal for disease modelling, particularly in the mouse. Animal models provide incomparable insights into the mechanisms of leukaemia development and allow exploration of the molecular pillars of disease maintenance, an aspect often biased in cell lines or ex vivo systems. Several of these models, which faithfully recapitulate the characteristics of the human disease, have been used for pre-clinical purposes and have been instrumental in predicting therapy response in patients. We plea for a wider use of genetically defined animal models in the design of clinical trials, with a particular focus on reassessment of existing cancer or non-cancer drugs, alone or in combination.
Collapse
Affiliation(s)
- Julien Ablain
- Université Paris Diderot, Sorbonne Paris Cité, Hôpital St. Louis 1, Avenue Claude Vellefaux, 75475 Paris cedex 10, France
| | | | | | | | | | | |
Collapse
|
10
|
Höpken UE, Rehm A. Homeostatic chemokines guide lymphoma cells to tumor growth-promoting niches within secondary lymphoid organs. J Mol Med (Berl) 2012; 90:1237-45. [PMID: 22577036 DOI: 10.1007/s00109-012-0906-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 04/23/2012] [Accepted: 04/26/2012] [Indexed: 02/27/2023]
Abstract
The interaction between lymphoid tumor cells and their tissue microenvironment is thought to promote dissemination and progression of lymphoma. Those type of interactions consists of at least three cornerstones, among them mesenchymal- or bone marrow-derived stromal cells, cells of the innate or adaptive immune response, and the lymphoma cells themselves. The molecular pathways of crosstalk between the lymphoma cells and their nursing stroma are not well understood and their dissection is challenging because of (1) the complexity of stroma cell subpopulations, (2) kinetic and developmental transitions/switches of stroma composition, and (3) inherent technical difficulties to isolate and analyze defined stroma cell subsets. However, recent studies of bone marrow stroma interaction with leukemia or lymphoma cells have revealed therapeutic targets involved in regulating tumor cell mobilization. Release of tumor cells from their supportive niches could be effectuated by inhibition of homing and retention signals. The present review focuses on the effects of homing receptors and cytokines attributed to lymphoid tissue formation in tumor-stroma interactions within secondary lymphoid tissues. We discuss possible cellular and molecular mechanisms of lymphoma-stroma crosstalk and highlight novel therapeutic strategies based on the disruption of tumor-stroma interaction in secondary lymphoid organs.
Collapse
Affiliation(s)
- Uta E Höpken
- Department of Tumor Genetics and Immunogenetics, Max-Delbrück-Center for Molecular Medicine, MDC, Berlin 13125, Germany.
| | | |
Collapse
|
11
|
Noninvasive bioluminescent imaging of primary patient acute lymphoblastic leukemia: a strategy for preclinical modeling. Blood 2011; 118:e112-7. [PMID: 21856863 DOI: 10.1182/blood-2011-04-346528] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The efficient engraftment in immune-deficient mice achieved with both acute lymphoblastic leukemia (ALL) cell lines and primary samples has facilitated identification of the antileukemia activity of a wide variety of agents. Despite widespread usage, however, little is known about the early ALL localization and engraftment kinetics in this model, limiting experimental read-outs primarily to survival and endpoint analysis at high disease burden. In this study, we report that bioluminescent imaging can be reproducibly achieved with primary human ALL samples. This approach provides a noninvasive, longitudinal measure of leukemia burden and localization that enhances the sensitivity of treatment response detection and provides greater insight into the mechanism of action of antileukemia agents. In addition, this study reveals significant cell line- and species-related differences in leukemia migration, especially early in expansion, which may confound observations between various leukemia models. Overall, this study demonstrates that the use of bioluminescent primary ALL allows the detection and quantitation of treatment effects at earlier, previously unquantifiable disease burdens and thus provides the means to standardize and expedite the evaluation of anti-ALL activity in preclinical xenograft studies.
Collapse
|
12
|
Sullivan C, Peng C, Chen Y, Li D, Li S. Targeted therapy of chronic myeloid leukemia. Biochem Pharmacol 2010; 80:584-91. [PMID: 20470758 DOI: 10.1016/j.bcp.2010.05.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Revised: 04/29/2010] [Accepted: 05/04/2010] [Indexed: 11/12/2022]
Abstract
Inhibition of BCR-ABL with kinase inhibitors has become a well-accepted strategy for targeted therapy of Philadelphia-positive (Ph(+)) chronic myeloid leukemia (CML) and has been shown to be highly effective in controlling the disease. However, BCR-ABL kinase inhibitors do not efficiently kill leukemic stem cells (LSCs), indicating that this therapeutic strategy does not lead to a cure of CML. Development of curative therapies of CML require the identification of genes/pathways that play critical roles in survival and self-renewal of LSCs. Targeting of these key BCR-ABL downstream genes provides an opportunity to eradicate LSCs, as shown in our work that identifies the Alox5 gene as a key regulator of the function of CML LSCs. Immediate clinical trials are necessary to test the effectiveness of targeting a key BCR-ABL downstream gene in eradicating LSCs in CML patients. In this review, we will discuss current targeted therapies of CML using BCR-ABL kinase inhibitors, with a focus on the importance of developing a targeted therapy of CML through identification of target genes in CML LSCs.
Collapse
Affiliation(s)
- Con Sullivan
- Maine Institute for Human Genetics and Health, 246 Sylvan Road, Bangor, ME 04401, USA
| | | | | | | | | |
Collapse
|
13
|
Nasr R, de Thé H. Eradication of acute promyelocytic leukemia-initiating cells by PML/RARA-targeting. Int J Hematol 2010; 91:742-7. [PMID: 20455087 DOI: 10.1007/s12185-010-0582-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2010] [Accepted: 04/19/2010] [Indexed: 12/20/2022]
Abstract
Acute promyelocytic leukemia (APL) is characterized by a t(15;17) translocation that yields a PML/RARA fusion protein. Expression of PML/RARA, a potent transcriptional repressor, induces APL in mice. Both retinoic acid (RA) and arsenic trioxide directly target PML/RARA-mediated transcriptional repression and protein stability, inducing rapid differentiation of the promyelocytes and clinical remission in most APL patients. RA also triggers growth arrest and progressive clearance of leukemia initiating cells (LIC), both ex vivo and in vivo. Suboptimal RA concentrations or expression of the PLZF/RARA variant allows complete RA-induced differentiation, but neither LIC clearance nor disease remission. Thus, RA-induced differentiation and LIC clearance may be uncoupled. The RA/arsenic trioxide association, which dramatically synergizes for PML/RARA degradation but not for differentiation, rapidly clears LIC in a proteasome-dependent manner, resulting in APL eradication in murine models and patients. Collectively, these results demonstrate that LIC clearance, which mirrors PML/RARA degradation, is the primary basis for APL cure by the RA/arsenic trioxide association, rather than differentiation. Oncogene degradation could be a generally applicable therapeutic strategy to clear LICs in several types of tumors.
Collapse
Affiliation(s)
- Rihab Nasr
- Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| | | |
Collapse
|
14
|
Abstract
Chromosomal aberrations occur with great frequency and some specificity in leukemia and other hematologic malignancies. The most common outcome of these rearrangements is the formation of a fusion gene, comprising portions of 2 genes normally present in the cell. These fusion proteins are presumed to be oncogenic; in many cases, animal models have proven them to be oncogenic. One of the most promiscuous fusion partner genes is the newly identified NUP98 gene, located on chromosome 11p15.5, which to date has been observed fused to 15 different fusion partners. NUP98 encodes a 98 kD protein that is an important component of the nuclear pore complex, which mediates nucleo-cytoplasmic transport of protein and RNA. The fusion partners of NUP98 form 2 distinct groups: homeobox genes and non-homeobox genes. All NUP98 fusions join the N-terminal GLFG repeats of NUP98 to the C-terminal portion of the partner gene, which, in the case of the homeobox gene partners, includes the homeodomain. Clinical findings are reviewed here, along with the findings of several in vivo and in vitro models have been employed to investigate the mechanisms by which NUP98 fusion genes contribute to the pathogenesis of leukemia.
Collapse
MESH Headings
- Acute Disease
- Antineoplastic Agents/pharmacology
- Cell Transformation, Neoplastic/genetics
- Chromosome Breakage
- Chromosomes, Human, Pair 11/genetics
- DNA Topoisomerases, Type II/physiology
- Enzyme Inhibitors/pharmacology
- Genes, Homeobox
- Hematologic Neoplasms/genetics
- Hematologic Neoplasms/metabolism
- Homeodomain Proteins/genetics
- Homeodomain Proteins/physiology
- Humans
- Leukemia/genetics
- Leukemia/metabolism
- Models, Genetic
- Neoplasm Proteins/genetics
- Neoplasm Proteins/physiology
- Nuclear Pore/physiology
- Nuclear Pore Complex Proteins/genetics
- Nuclear Pore Complex Proteins/physiology
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/physiology
- Protein Structure, Tertiary
- Topoisomerase II Inhibitors
- Translocation, Genetic
Collapse
Affiliation(s)
- Christopher Slape
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Navy 8, Room 5101, Bethesda, Maryland, MD 20889-5105, USA
| | | |
Collapse
|
15
|
Kennedy JA, Barabé F. Investigating human leukemogenesis: from cell lines to in vivo models of human leukemia. Leukemia 2008; 22:2029-40. [DOI: 10.1038/leu.2008.206] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
16
|
Abstract
Chromosomal translocations are important genetic perturbations frequently associated with hematologic malignancies; characterization of these events has been a rich source of insights into the mechanisms that lead to malignant transformation. The t(10;11)(p13;q14-21) results in a recently identified rare but recurring chromosomal translocation seen in patients with ALL as well as AML, and results in the production of a CALM-AF10 fusion gene. Although the details by which the CALM-AF10 fusion protein exerts its leukemogenic effect remain unclear, emerging data suggests that the CALM-AF10 fusion impairs differentiation of hematopoietic cells, at least in part via an upregulation of HOXA cluster genes. This review discusses the normal structure and function of CALM and AF10, describes the spectrum of clinical findings seen in patients with CALM-AF10 fusions, summarizes recently published CALM-AF10 mouse models and highlights the role of HOXA cluster gene activation in CALM-AF10 leukemia.
Collapse
Affiliation(s)
- D Caudell
- Genetics Branch, National Cancer Institute, National Institutes for Health, Bethesda, MD 20889-5105, USA
| | | |
Collapse
|
17
|
Caudell D, Zhang Z, Chung YJ, Aplan PD. Expression of a CALM-AF10 fusion gene leads to Hoxa cluster overexpression and acute leukemia in transgenic mice. Cancer Res 2007; 67:8022-31. [PMID: 17804713 PMCID: PMC1986634 DOI: 10.1158/0008-5472.can-06-3749] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
To assess the role of the CALM-AF10 fusion gene in leukemic transformation in vivo, we generated transgenic mice that expressed a CALM-AF10 fusion gene. Depending on the transgenic line, at least 40% to 50% of the F(1) generation mice developed acute leukemia at a median age of 12 months. Leukemic mice typically had enlarged spleens, invasion of parenchymal organs with malignant cells, and tumors with myeloid markers such as myeloperoxidase, Mac1, and Gr1. Although most leukemias were acute myeloid leukemia, many showed lymphoid features, such as CD3 staining, or clonal Tcrb or Igh gene rearrangements. Mice were clinically healthy for the first 9 months of life and had normal peripheral blood hemograms but showed impaired thymocyte differentiation, manifested by decreased CD4(+)/CD8(+) cells and increased immature CD4(-)/CD8(-) cells in the thymus. Hematopoietic tissues from both clinically healthy and leukemic CALM-AF10 mice showed up-regulation of Hoxa cluster genes, suggesting a potential mechanism for the impaired differentiation. The long latency period and incomplete penetrance suggest that additional genetic events are needed to complement the CALM-AF10 transgene and complete the process of leukemic transformation.
Collapse
Affiliation(s)
- David Caudell
- Genetics Branch, National Cancer Institute, National Institutes for Health, Bethesda, MD
- Comparative Molecular Pathology Unit, National Cancer Institute, National Institutes for Health, Bethesda, MD
- Department of Veterinary Medical Sciences, University of Maryland, College Park, MD
| | - Zhenhua Zhang
- Genetics Branch, National Cancer Institute, National Institutes for Health, Bethesda, MD
| | - Yang Jo Chung
- Genetics Branch, National Cancer Institute, National Institutes for Health, Bethesda, MD
| | - Peter D. Aplan
- Genetics Branch, National Cancer Institute, National Institutes for Health, Bethesda, MD
| |
Collapse
|
18
|
Guo C, Jin X. Chemoprotection effect of multidrug resistance 1 (MDR1) gene transfer to hematopoietic progenitor cells and engrafted in mice with cancer allows intensified chemotherapy. Cancer Invest 2007; 24:659-68. [PMID: 17118775 DOI: 10.1080/07357900600981299] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Increasing the proportion of bone marrow cells expression human multidrug resistance (MDR) 1 gene to prevent or circumvent bone morrow toxicity from chemotherapy agent is a high priority of dose intensification protocols. In this study, we have used a BALB/c mouse tumor-bearing model to investigate the chemoprotection effect of MDR1 gene by transfecting retroviral vectors containing and expressing the MDR gene in vivo. Hematopoietic progenitor cells served as a target of MDR1 gene transfer by the mediation of retrovirus vector and engrafted into the BALB/c mice with 60Co-gamma ray exposure in advance. Doxorubicin (5, 10, and 20 mg/kg) suppressed tumor growth of the xenograft significantly in dose-dependence mode if supported by suitable peripheral WBC. WBCs count revealed that the mice that had received gene-transduced cells showed a significant increase in WBCs count compared with their gene-transduced-naive counterparts. The function and expression of MDR1 gene were detected by flow cytometry, RT-PCR and immunohistochemistry (IC) method. MDRl mRNA expression could be detected in BM. Spleens contained measurable amounts of MDRl mRNA. Tail vein blood and tumor tissue detected MDRl DNA but no MDRl mRNA expression. FACS analysis of infected BM cells obtained 6 weeks later showed high levels of P-gp function. Based on these results we conclude that cytostatic drug resistance gene therapy may provide some degree of chemoprotection so can increase the chemotherapy dose to kill tumor cells.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- Animals
- Antibiotics, Antineoplastic/therapeutic use
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/pathology
- Cell Line, Tumor
- DNA, Complementary/metabolism
- Doxorubicin/therapeutic use
- Flow Cytometry
- Genes, MDR/physiology
- Genetic Vectors
- Hematopoietic Stem Cell Transplantation
- Hematopoietic Stem Cells/metabolism
- Humans
- Immunoenzyme Techniques
- Liver Neoplasms, Experimental/drug therapy
- Liver Neoplasms, Experimental/pathology
- Mice
- Mice, Inbred BALB C
- Polymerase Chain Reaction
- RNA, Messenger/metabolism
- Retroviridae/genetics
- Transfection
- Tumor Burden/drug effects
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Chunbao Guo
- The Laboratory of Surgery, Children's Hospital of Chongqing Medical University, Chongqing, PR China.
| | | |
Collapse
|
19
|
Fischer M, Schwieger M, Horn S, Niebuhr B, Ford A, Roscher S, Bergholz U, Greaves M, Löhler J, Stocking C. Defining the oncogenic function of the TEL/AML1 (ETV6/RUNX1) fusion protein in a mouse model. Oncogene 2005; 24:7579-91. [PMID: 16044150 DOI: 10.1038/sj.onc.1208931] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The t(12;21) translocation, generating the TEL/AML1 fusion protein, is the most common genetic lesion in childhood cancer. Using a bone marrow transplantation model, we demonstrate that TEL/AML1 expression impinges on normal hematopoietic differentiation, leading to the in vivo accumulation and persistence of an early progenitor compartment with a Sca1(+)/Kit(hi)/CD11b(+) phenotype and an increased self-renewal capacity, as documented by replating assays in vitro. Differentiation of these cells is not blocked, but the frequency of mature blood cells arising from TEL/AML1-transduced progenitors is low. Impaired differentiation is prominently observed in the pro-B-cell compartment, resulting in an proportional increase in early progenitors in vivo, consistent with the t(12;21) ALL phenotype. Despite the accumulation of both multipotent and B-cell progenitors in vivo, no leukemia induction was observed during an observation period of over 1 year. These results are consistent with findings in twins with concordant ALL, showing that TEL/AML1 generates a preleukemic clone in utero that persists for several years in a clinically covert fashion. Furthermore, our studies showed that the pointed domain of TEL/AML1, which recruits transcriptional repressors and directs oligomerization with either TEL/AML1 or wild-type TEL, was essential for the observed differentiation impairment and could not be replaced with another oligomerization domain.
Collapse
MESH Headings
- Animals
- B-Lymphocytes
- Bone Marrow Transplantation
- Cell Differentiation
- Cell Transformation, Neoplastic/genetics
- Chromosomes, Human, Pair 12
- Chromosomes, Human, Pair 21
- Core Binding Factor Alpha 2 Subunit/biosynthesis
- Core Binding Factor Alpha 2 Subunit/genetics
- Core Binding Factor Alpha 2 Subunit/physiology
- Hematopoietic Stem Cells
- Humans
- Mice
- Mice, Inbred C57BL
- Oncogene Proteins, Fusion/biosynthesis
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/physiology
- Phenotype
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics
- Preleukemia/genetics
- Preleukemia/physiopathology
- Translocation, Genetic
Collapse
Affiliation(s)
- Meike Fischer
- Molecular Pathology Group, Heinrich-Pette-Institut für Experimentelle Immunologie und Virologie, D-20251 Hamburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Guo CB, Li YC, Jin XQ. Chemoprotection effect of retroviral vector encoding multidrug resistance 1 gene to allow intensified chemotherapy in vivo. Cancer Chemother Pharmacol 2005; 58:40-9. [PMID: 16284790 DOI: 10.1007/s00280-005-0144-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2005] [Accepted: 09/29/2005] [Indexed: 10/25/2022]
Abstract
Increasing the expression of human multidrug resistance (MDR) 1 gene in bone marrow cells to prevent or circumvent bone morrow toxicity from chemotherapy agent is a high priority of dose intensification protocols. In this study, we have used a tumor-bearing model to investigate the chemoprotection effect of MDR1 gene by transfecting retroviral vectors containing and expressing the MDR gene in vivo. Hematopoietic progenitor cells were served as target of MDR1 gene transferred by the mediation of retrovirus vector and engrafted into the BALB/c mice with 60Co-gamma ray exposure in advance. Doxorubicin (5, 10, and 20 mg/kg) suppressed tumor growth of the xenograft significantly in a dose-dependence mode if supported by suitable peripheral WBC. WBC count revealed that the mice that had received gene-transduced cells showed a significant increase in WBC count compared with their gene-transduced naive counterparts. The function and expression of MDR1 gene were detected by flow cytometry, RT-PCR, and immunohistochemistry (IC) method. MDRl mRNA expression could be detected in BM. Spleens contained measurable amounts of MDRl mRNA. Tail vein blood and tumor tissue detected MDRl DNA but no MDRl mRNA expression. FACS analysis of infected BM cells obtained 6 weeks later showed high levels of P-gp function. Based on these results we conclude that cytostatic drug resistance gene therapy may provide some degree of chemoprotection and so can increase the chemotherapy dose to kill tumor cells.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Animals
- Antibiotics, Antineoplastic/therapeutic use
- Bone Marrow Cells/metabolism
- Bone Marrow Transplantation
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/pathology
- Cell Line, Tumor
- DNA, Complementary/metabolism
- Doxorubicin/therapeutic use
- Genes, MDR
- Genetic Therapy
- Genetic Vectors
- Humans
- Liver Neoplasms, Experimental/drug therapy
- Liver Neoplasms, Experimental/pathology
- Mice
- Mice, Inbred BALB C
- RNA, Messenger/metabolism
- Retroviridae/genetics
- Tumor Burden/drug effects
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Chun-Bao Guo
- Laboratory of Surgery, Children's Hospital of Chongqing Medical University, 132 Zhongshan Rd., 400014 Chongqing, People's Republic of China.
| | | | | |
Collapse
|
21
|
Lallemand-Breitenbach V, Zhu J, Kogan S, Chen Z, de Thé H. Opinion: how patients have benefited from mouse models of acute promyelocytic leukaemia. Nat Rev Cancer 2005; 5:821-7. [PMID: 16175176 DOI: 10.1038/nrc1719] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
One of the challenges of studying anticancer therapies is that effects observed in cell lines or mouse models are not always good indicators of clinical trial results. The mouse model of acute promyelocytic leukaemia has bucked this trend, as targeted therapies such as retinoic acid and arsenic induce differentiation and clearance of leukaemia cells in both mice and humans. This mouse model has also provided important mechanistic insights into the combinatorial effects of these agents and has promoted combined therapies that have shown recent success in the clinic.
Collapse
Affiliation(s)
- Valérie Lallemand-Breitenbach
- Université de Paris, CNRS UMR 7151, Université de Paris VII, Equipe Labellisée de la Ligue contre le Cancer, Hôpital St. Louis, 1 Avenue C. Vellefaux, 75475 Paris CEDEX 10, France
| | | | | | | | | |
Collapse
|
22
|
Dunn DA, Pinkert CA, Kooyman DL. Foundation Review: Transgenic animals and their impact on the drug discovery industry. Drug Discov Today 2005; 10:757-67. [PMID: 15922934 DOI: 10.1016/s1359-6446(05)03452-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The ability to direct genetic changes at the molecular level has resulted in a revolution in biology. Nowhere has this been more apparent than in the production of transgenic animals. Transgenic technology lies at the junction of several enabling techniques in such diverse fields as embryology, cell biology and molecular genetics. A host of techniques have been used to effect change in gene expression and develop new pharmaceutical and nutraceutical compounds cost-effectively. Scientific advances gained by transgenic capabilities enable further understanding of basic biological pathways and yield insights into how changes in fundamental processes can perturb programmed development or culminate in disease pathogenesis.
Collapse
Affiliation(s)
- David A Dunn
- Department of Pathology and Laboratory Medicine, Center for Aging and Developmental Biology, University of Rochester Medical Center, Rochester, NY, USA
| | | | | |
Collapse
|
23
|
Daser A, Rabbitts TH. The versatile mixed lineage leukaemia gene MLL and its many associations in leukaemogenesis. Semin Cancer Biol 2005; 15:175-88. [PMID: 15826832 DOI: 10.1016/j.semcancer.2005.01.007] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The marked association of abnormalities of chromosome 11 long arm, band q23, with human leukaemia led to the identification of the 11q23 gene called MLL (or HTRX, HRX, TRX1, ALL-1). MLL can become fused with one of a remarkable panoply of genes from other chromosome locations in individual leukaemias, leading to either acute myeloid or lymphoid tumours (hence the name MLL for mixed lineage leukaemia). The unusual finding that a single protein could be involved in both myeloid and lymphoid malignancies and that the truncated protein could do so as a fusion with very disparate partners has prompted studies to define the molecular role of MLL-fusions in leukaemogenesis and to the development of MLL-controlled mouse models of leukaemogenesis. These studies have defined MLL-fusion proteins as regulators of gene expression, controlling such elements as HOX genes, and have indicated a variety of mechanisms by which MLL-fusion proteins contribute to leukaemogenesis.
Collapse
Affiliation(s)
- A Daser
- MRC Laboratory of Molecular Biology, Hills Road, Cambridge CB22QH, UK
| | | |
Collapse
|
24
|
Racanicchi S, Maccherani C, Liberatore C, Billi M, Gelmetti V, Panigada M, Rizzo G, Nervi C, Grignani F. Targeting fusion protein/corepressor contact restores differentiation response in leukemia cells. EMBO J 2005; 24:1232-42. [PMID: 15729358 PMCID: PMC556397 DOI: 10.1038/sj.emboj.7600593] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2004] [Accepted: 02/01/2005] [Indexed: 01/05/2023] Open
Abstract
The AML1/ETO and PML/RARalpha leukemia fusion proteins induce acute myeloid leukemia by acting as transcriptional repressors. They interact with corepressors, such as N-CoR and SMRT, that recruit a multiprotein complex containing histone deacetylases on crucial myeloid differentiation genes. This leads to gene repression contributing to generate a differentiation block. We expressed in leukemia cells containing PML/RARalpha and AML1/ETO N-CoR protein fragments derived from fusion protein/corepressor interaction surfaces. This blocks N-CoR/SMRT binding by these fusion proteins, and disrupts the repressor protein complex. In consequence, the expression of genes repressed by these fusion proteins increases and differentiation response to vitamin D3 and retinoic acid is restored in previously resistant cells. The alteration of PML/RARalpha-N-CoR/SMRT connections triggers proteasomal degradation of the fusion protein. The N-CoR fragments are biologically effective also when directly transduced by virtue of a protein transduction domain. Our data indicate that fusion protein activity is permanently required to maintain the leukemia phenotype and show the route to developing a novel therapeutic approach for leukemia, based on its molecular pathogenesis.
Collapse
Affiliation(s)
- Serena Racanicchi
- Patologia Generale and Medicina Interna e Scienze Oncologiche, Dipartimento di Medicina Clinica e Sperimentale, Perugia University, Policlinico Monteluce, Perugia, Italy
| | - Chiara Maccherani
- Patologia Generale and Medicina Interna e Scienze Oncologiche, Dipartimento di Medicina Clinica e Sperimentale, Perugia University, Policlinico Monteluce, Perugia, Italy
| | - Concetta Liberatore
- Patologia Generale and Medicina Interna e Scienze Oncologiche, Dipartimento di Medicina Clinica e Sperimentale, Perugia University, Policlinico Monteluce, Perugia, Italy
| | - Monia Billi
- Patologia Generale and Medicina Interna e Scienze Oncologiche, Dipartimento di Medicina Clinica e Sperimentale, Perugia University, Policlinico Monteluce, Perugia, Italy
| | - Vania Gelmetti
- Dipartimento di Istologia ed Embriologia Medica, Università di Roma ‘La Sapienza', Roma, Italy
- Parco Bio-Medico Scientifico San Raffaele di Roma, Roma, Italy
| | - Maddalena Panigada
- Patologia Generale and Medicina Interna e Scienze Oncologiche, Dipartimento di Medicina Clinica e Sperimentale, Perugia University, Policlinico Monteluce, Perugia, Italy
| | - Giovanni Rizzo
- Patologia Generale and Medicina Interna e Scienze Oncologiche, Dipartimento di Medicina Clinica e Sperimentale, Perugia University, Policlinico Monteluce, Perugia, Italy
| | - Clara Nervi
- Dipartimento di Istologia ed Embriologia Medica, Università di Roma ‘La Sapienza', Roma, Italy
- Parco Bio-Medico Scientifico San Raffaele di Roma, Roma, Italy
| | - Francesco Grignani
- Patologia Generale and Medicina Interna e Scienze Oncologiche, Dipartimento di Medicina Clinica e Sperimentale, Perugia University, Policlinico Monteluce, Perugia, Italy
- Medicina Interna e Scienze Oncologiche, Dipartimento di Medicina Clinica e Sperimentale, Perugia University, Policlinico Monteluce, 06100 Perugia, Italy. Tel.: +39 075 572 6264; Fax: +39 075 578 3444; E-mail:
| |
Collapse
|
25
|
Zhu J, Zhou J, Peres L, Riaucoux F, Honoré N, Kogan S, de Thé H. A sumoylation site in PML/RARA is essential for leukemic transformation. Cancer Cell 2005; 7:143-53. [PMID: 15710327 DOI: 10.1016/j.ccr.2005.01.005] [Citation(s) in RCA: 114] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2004] [Revised: 06/18/2004] [Accepted: 01/12/2005] [Indexed: 11/15/2022]
Abstract
Pathogenesis of acute promyelocytic leukemia (APL) has been proposed to involve transcriptional repression through enhanced corepressors binding onto RARA moieties of PML/RARA homodimers. Unexpectedly, we show that the K160 sumoylation site in the PML moiety of PML/RARA is required for efficient immortalization/differentiation arrest ex vivo, implying that RARA homodimerization is insufficient to fully immortalize primary hematopoietic progenitor cells. Similarly, PML/RARAK160R transgenic mice develop myeloproliferative syndromes, but never APL. The Daxx repressor no longer binds PML/RARAK160R, but fusion of these two proteins restores the differentiation block ex vivo. Thus, transcriptional repression dependent on a specific sumoylation site in PML is critical for the APL phenotype, while forced RARA dimerization could control expansion of the myeloid compartment.
Collapse
Affiliation(s)
- Jun Zhu
- CNRS UPR 9051, laboratoire associé N11 du comité de Paris de la Ligue contre le Cancer, affilié à l'Université de Paris VII, Hôpital St. Louis, 1 avenue Claude Vellefaux, 75475 Paris, Cedex 10, France
| | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Selection of mouse models of cancer is often based simply on availability of a mouse strain and a known compatible tumor. Frequently this results in use of tumor models long on history but short on homology and quality control. Other factors including genetics, sex, immunological status, method and site of tumor implantation, technical competence, biological activity of the tumor, protocol sequence and timing, and selection of endpoints interact to produce outcomes in tumor models. Common reliance on survival and tumor burden data in a single mouse model often skews expectations towards high remission and cure rates; a finding seldom duplicated in clinical trials. Inherent limitations of tumor models coupled with the advent of new therapeutic targets reinforce need for careful attention to design, conduct, and stringent selection of in vivo and ex vivo endpoints. Preclinical efficacy testing for anti-tumor therapies should progress through a series of models of increasing sophistication that includes incorporation of genetically engineered animals, and orthotopic and combination therapy models. Pharmacology and safety testing in tumor-bearing animals may also help to improve predictive value of these models for clinical efficacy. Trends in bioinformatics, genetic refinements, and specialized imaging techniques are helping to maintain mice as the most scientifically and economically powerful model of malignant neoplasms.
Collapse
Affiliation(s)
- JoAnn C L Schuh
- Applied Veterinary Pathobiology, Bainbridge Island, Washington 98110-3663, USA.
| |
Collapse
|
27
|
Nimer SD, Moore MAS. Effects of the leukemia-associated AML1-ETO protein on hematopoietic stem and progenitor cells. Oncogene 2004; 23:4249-54. [PMID: 15156180 DOI: 10.1038/sj.onc.1207673] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Insights into the pathogenesis of human leukemia have relied heavily on studies of the identified chromosomal translocations found in this group of malignant diseases. Acquired, balanced translocations in acute myelogenous leukemia (AML) generally involve transcriptional regulatory genes, whereas in the myeloproliferative disorders tyrosine kinases are frequently involved. These rearrangements alter the function of at least one and often both of the involved genes. In this review, we focus on the AML1-ETO (a.k.a. RUNX1-ETO) fusion protein, which is found in t(8;21)+ AML. Expression of AML1-ETO in human hematopoietic stem cells (HSCs) preferentially enhances their maintenance, as opposed to their differentiation. The direct effects of AML1-ETO on human and murine HSCs, and the potentially cooperating events that may contribute to its leukemogenic properties, are discussed.
Collapse
Affiliation(s)
- Stephen D Nimer
- Laboratory of Molecular Aspects of Hematopoiesis, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA.
| | | |
Collapse
|
28
|
Tarantul VZ. Transgenic Mice as an In Vivo Model of Lymphomagenesis. INTERNATIONAL REVIEW OF CYTOLOGY 2004; 236:123-80. [PMID: 15261738 DOI: 10.1016/s0074-7696(04)36004-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This review covers multiple data obtained on genetically modified mice that help to elucidate various intricate molecular mechanisms of lymphomagenesis in humans. We are in a "golden age" of mouse genetics. The mouse is by far the most accessible mammalian system physiologically similar to humans. Transgenic mouse models have illuminated how different genes contribute to human lymphomagenesis. Multiple experiments with transgenic mice have not only confirmed the data obtained for human lymphomas but also gave additional evidence for the role of some genes and cooperative participation of their products in the development of human lymphomas. Genes and gene networks detected on transgenic mice can successfully serve as molecular targets for tumor therapy. This review demonstrates the extraordinary possibilities of transgenic technology, which is presently one of the readily available, efficient, and accurate tools to solve the problem of cancer.
Collapse
Affiliation(s)
- V Z Tarantul
- Institute of Molecular Genetics, Russian Academy of Sciences, Moscow 123182, Russia
| |
Collapse
|
29
|
Damm-Welk C, Fuchs U, Wössmann W, Borkhardt A. Targeting oncogenic fusion genes in leukemias and lymphomas by RNA interference. Semin Cancer Biol 2003; 13:283-92. [PMID: 14563123 DOI: 10.1016/s1044-579x(03)00042-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Leukemias and lymphomas are often characterized by non-random chromosomal translocations that, at the molecular level, induce the activation of specific oncogenes or create novel chimeric genes. They have frequently been regarded as optimal targets for gene-silencing approaches because of the large body of evidence that these single abnormalities directly initiate and maintain the malignant process. Herein, we discuss RNA interference (RNAi)-based approaches for targeting the fusion sites of chromosomal translocations as a future treatment option in leukemias and lymphomas.
Collapse
Affiliation(s)
- Christine Damm-Welk
- Pediatric Hematology & Oncology, Children's University Hospital Giessen, Feulgenstr. 12, 35392 Giessen, Germany
| | | | | | | |
Collapse
|
30
|
Abstract
In this issue of Cancer Cell, Forster et al. (2003) have generated mice that recapitulate both the mechanism (sporadic somatic translocation) and the consequences (expression of two translocation fusion genes) leading to an accurate leukemia model.
Collapse
Affiliation(s)
- Haydn Prosser
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | | |
Collapse
|
31
|
Prosser H, Rastan S. Manipulation of the mouse genome: a multiple impact resource for drug discovery and development. Trends Biotechnol 2003; 21:224-32. [PMID: 12727384 DOI: 10.1016/s0167-7799(03)00087-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Few would deny that the pharmaceutical industry's investment in genomics throughout the 1990s has yet to deliver in terms of drugs on the market. The reasons are complex and beyond the scope of this review. The unique ability to manipulate the mouse genome, however, has already had a positive impact on all stages of the drug discovery process and, increasingly, on the drug development process too. We give an overview of some recent applications of so-called 'transgenic' mouse technology in pharmaceutical research and development. We show how genetic manipulation in the mouse can be employed at multiple points in the drug discovery and development process, providing new solutions to old problems.
Collapse
Affiliation(s)
- Haydn Prosser
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, CB10 1SA UK
| | | |
Collapse
|
32
|
Baum C, Düllmann J, Li Z, Fehse B, Meyer J, Williams DA, von Kalle C. Side effects of retroviral gene transfer into hematopoietic stem cells. Blood 2003; 101:2099-114. [PMID: 12511419 DOI: 10.1182/blood-2002-07-2314] [Citation(s) in RCA: 289] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Recent conceptual and technical improvements have resulted in clinically meaningful levels of gene transfer into repopulating hematopoietic stem cells. At the same time, evidence is accumulating that gene therapy may induce several kinds of unexpected side effects, based on preclinical and clinical data. To assess the therapeutic potential of genetic interventions in hematopoietic cells, it will be important to derive a classification of side effects, to obtain insights into their underlying mechanisms, and to use rigorous statistical approaches in comparing data. We here review side effects related to target cell manipulation; vector production; transgene insertion and expression; selection procedures for transgenic cells; and immune surveillance. We also address some inherent differences between hematopoiesis in the most commonly used animal model, the laboratory mouse, and in humans. It is our intention to emphasize the need for a critical and hypothesis-driven analysis of "transgene toxicology," in order to improve safety, efficiency, and prognosis for the yet small but expanding group of patients that could benefit from gene therapy.
Collapse
Affiliation(s)
- Christopher Baum
- Department of Hematology and Oncology, Hannover Medical School, Hannover, Germany.
| | | | | | | | | | | | | |
Collapse
|
33
|
Feller SM, Tuchscherer G, Voss J. High affinity molecules disrupting GRB2 protein complexes as a therapeutic strategy for chronic myelogenous leukaemia. Leuk Lymphoma 2003; 44:411-27. [PMID: 12688310 DOI: 10.1080/1042819021000037930] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Chronic myelogenous leukaemia (CML) is one of the most intensively studied human malignancies. It has been the focus of major efforts to develop potent drugs for several decades, but until recently cure rates remained low. A breakthrough in CML therapy was very likely accomplished with the clinical introduction of STI-571 [imatinib mesylate; Gleevec (USA); Glivec (other countries)] in 2000/2001. Despite the hope that STI-571 has generated for many CML patients, development of resistance to this drug is already apparent in some cases, especially if the CML is diagnosed in its later stages. Therefore, novel drugs which can be used alone or in combination with STI-571 are highly desirable. This review briefly summarises the current understanding and therapy of CML and then discusses in more detail basic laboratory research that attempts to target Grb2, an adaptor protein known to directly interact with the Bcr portion of the Bcr-Abl fusion protein. Blocking the binding of Grb2 to the GDP-releasing protein SoS is well known to abrogate the activation of the GTPase Ras, a major driving force of the central mitogenic (MAP kinase) pathway. Additional Grb2 effector proteins may also contribute to the proliferation-inhibiting effects observed upon uncoupling Grb2 from its downstream signalling system. Since Grb2 is a known signal transducer for several major human oncogenes, this approach may have applications for a wider range of human cancers.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing
- Animals
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Benzamides
- Drug Design
- Enzyme Inhibitors/administration & dosage
- Enzyme Inhibitors/therapeutic use
- Fatty Acids, Unsaturated/pharmacology
- Forecasting
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/metabolism
- GRB2 Adaptor Protein
- Humans
- Imatinib Mesylate
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Macromolecular Substances
- Mice
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/metabolism
- Peptide Fragments/metabolism
- Phosphatidylinositol 3-Kinases/physiology
- Piperazines/administration & dosage
- Piperazines/therapeutic use
- Protein Binding/drug effects
- Proteins/antagonists & inhibitors
- Proteins/chemistry
- Proteins/metabolism
- Pyrimidines/administration & dosage
- Pyrimidines/therapeutic use
- Signal Transduction/drug effects
- Son of Sevenless Proteins/physiology
- Structure-Activity Relationship
- Transcription Factors/physiology
- ras Proteins/antagonists & inhibitors
- src Homology Domains
Collapse
Affiliation(s)
- Stephan M Feller
- Cell Signalling Group, Molecular Oncology Laboratory, Cancer Research UK, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK. stephan.feller@.cancer.org.uk
| | | | | |
Collapse
|
34
|
Abstract
The past decade has seen great strides in our understanding of the genetic basis of human disease. Arguably, the most profound impact has been in the area of cancer genetics, where the explosion of genomic sequence and molecular profiling data has illustrated the complexity of human malignancies. In a tumor cell, dozens of different genes may be aberrant in structure or copy number, and hundreds or thousands of genes may be differentially expressed. A number of familial cancer genes with high-penetrance mutations have been identified, but the contribution of low-penetrance genetic variants or polymorphisms to the risk of sporadic cancer development remains unclear. Studies of the complex somatic genetic events that take place in the emerging cancer cell may aid the search for the more elusive germline variants that confer increased susceptibility. Insights into the molecular pathogenesis of cancer have provided new strategies for treatment, but a deeper understanding of this disease will require new statistical and computational approaches for analysis of the genetic and signaling networks that orchestrate individual cancer susceptibility and tumor behavior.
Collapse
Affiliation(s)
- Allan Balmain
- UCSF Comprehensive Cancer Center and Department of Biochemistry and Biophysics, San Francisco, California 94143, USA.
| | | | | |
Collapse
|