An Experimental and Computational Protocol to Study Cell Proliferation in Human Acute Myeloid Leukemia Xenografts.
Acute myeloid leukemia (AML) is a highly frequent hematological malignancy, characterized by clinical and biological diversity, along with high relapse and mortality rates. The inherent functional and genetic intra-tumor heterogeneity in AML is thought to play an important role in disease recurrence and resistance to chemotherapy. Patient-derived xenograft (PDX) models preserve important features of the original tumor, allowing, at the same time, experimental manipulation and in vivo amplification of the human cells. Here we present a detailed protocol for the generation of fluorescently labeled AML PDX models to monitor cell proliferation kinetics in vivo, at the single-cell level. Although experimental protocols for cell proliferation studies are well established and widespread, they are not easily applicable to in vivo contexts, and the analysis of related time-series data is often complex to achieve. To overcome these limitations, model-driven approaches can be exploited to investigate different aspects of cell population dynamics. Among the existing approaches, the ProCell framework is able to perform detailed and accurate stochastic simulations of cell proliferation, relying on flow cytometry data. In particular, by providing an initial and a target fluorescence histogram, ProCell automatically assesses the validity of any user-defined scenario of intra-tumor heterogeneity, that is, it is able to infer the proportion of various cell subpopulations (including quiescent cells) and the division interval of proliferating cells. Here we explain the protocol in detail, providing a description of our methodology for the conditional expression of H2B-GFP in human AML xenografts, data processing by flow cytometry, and the final elaboration in ProCell.
6755
- 10.1038/nm0797-730
- Jul 1, 1997
- Nature Medicine
212
- 10.1016/j.swevo.2017.09.001
- Sep 7, 2017
- Swarm and Evolutionary Computation
1966
- 10.1016/j.stem.2014.02.006
- Mar 1, 2014
- Cell Stem Cell
351
- 10.1016/j.ccr.2014.01.031
- Mar 1, 2014
- Cancer Cell
10
- 10.1093/bioinformatics/btz063
- Feb 7, 2019
- Bioinformatics
168
- 10.1002/ijc.23772
- Aug 25, 2008
- International Journal of Cancer
1186
- 10.1038/nrclinonc.2012.61
- Apr 17, 2012
- Nature Reviews Clinical Oncology
4
- 10.1109/cibcb.2019.8791468
- Jul 1, 2019
20
- 10.1016/j.yexcr.2010.02.015
- Feb 18, 2010
- Experimental Cell Research
299
- 10.1038/nature07618
- Jan 1, 2009
- Nature
- Abstract
- 10.1182/blood-2021-150305
- Nov 5, 2021
- Blood
WT1 and DNMT3A Play an Essential Function and Represent Therapeutic Vulnerabilities in Certain AML Samples, As Shown By CRISPR/Cas9 Mediated Knockout in PDX Models In Vivo
- Abstract
- 10.1182/blood-2019-129225
- Nov 13, 2019
- Blood
Global Gene Expression and Mutation Signatures Are Preserved in PDX Models of Pediatric AML and Aid Discovery of Targeted Therapy for Cases with CBFA2T3/GLIS2 Rearrangement
- Abstract
- 10.1182/blood-2018-99-111288
- Nov 29, 2018
- Blood
Intra-Tumor Heterogeneity in Acute Myeloid Leukemia (AML): Results from a Real Life Cohort
- Research Article
- 10.1158/1557-3265.pdx16-b06
- Aug 14, 2016
- Clinical Cancer Research
The major therapeutic barrier in acute myeloid leukemia (AML) is chemotherapy resistance. AML cells resistant to conventional chemotherapy targeting DNA synthesis are thought to be enriched in quiescent leukemic stem cells (LSCs). In order to better understand chemotherapy resistance in AML, we analyzed the response to cytarabine (AraC) through patient-derived xenograft (PDX) models with 20 primary AML patient specimens from two clinical sites and in the context of a French “Innovative models initiative” (IMODI) program. After confirming AML engraftment, highly immunodeficient NOD/LtSz-scid IL2Rγc null (NSG) mice were treated with AraC administered IP for 5 days as a single agent at 60 mg/kg daily, which correlates with human dosing. In all mice treated with this regimen, there was a significant but variable cytoreductive effect (4- to 46-fold reduction of tumor cell burden; 2- to 13-fold induction of apoptosis) at 3 days post-treatment. This in vivo AraC response in PDX models has been compared to clinicobiological data of their matched patients (including overall survival, FAB classification, and age at diagnosis). Furthermore, residual leukemic cells (RLCs), surviving after in vivo AraC treatment, have been characterized for their cell surface phenotype, stem cell frequency, cell cycle and metabolic status. Gene expression of RLCs from three different PDX models showed an enrichment of genes involved in inflammatory, immune and stress/ROS responses. When tested in three independent cohorts of AML patients (Verhaak et al. 2009; TGCA. 2011; Metzeler et al. 2011), the down-regulated gene signature is associated with an unfavorable prognosis in patients treated with intensive chemotherapy. Altogether, these results suggest a novel model of AraC chemotherapy resistance uncovering the control of the oxidative and mitochondrial energy metabolism in vivo and the relevance of PDX models for clinical investigations and new preclinical drug assessment. Further studies of the role of immune and stromal microenvironment will be assessed in this model to extend our findings in a more relevant setting. Citation Format: Thomas Farge{Authors}. Studying cytarabine resistance through PDX models in acute myeloid leukemia. [abstract]. In: Proceedings of the AACR Special Conference: Patient-Derived Cancer Models: Present and Future Applications from Basic Science to the Clinic; Feb 11-14, 2016; New Orleans, LA. Philadelphia (PA): AACR; Clin Cancer Res 2016;22(16_Suppl):Abstract nr B06.
- Research Article
3
- 10.1158/1538-7445.am2022-3617
- Jun 15, 2022
- Cancer Research
Background: As the most prevalent internal decorations in mammalian mRNA, N6-methyladenosine (m6A) has been reported to be involved in many physiological and pathological processes, including acute myeloid leukemia (AML). METTL3 and METTL14, the well-recognized m6A methyltransferase complex, contribute to AML. METTL16 is a recently identified m6A methyltransferase that has been reported to deposit m6A in a few targets. While, unlike METTL3/14, the biological functions of METTL16 are largely unknown. Here, we explored the function and mechanism of METTL16 in AML pathogenesis and evaluated its therapeutic potential for AML treatment. Methods: We performed CRISPR-Cas9 screen to evaluate the dependency of METTL16 in AML cells. We created METTL16 knockout (KO) cells and conditional KO mice to evaluate its role in leukemogenesis and normal hematopoiesis. We employed bone marrow transplantation (BMT), xenograft, and AML patient-derived xenograft (PDX) models to determine its role in AML development and progression. To identify the targets of METTL16, we performed m6A-seq and RNA-seq, followed m6A-qPCR, CLIP-qPCR, in vitro methyltransferase assays and RNA stability assays. To examine the effect of METTL16 on branched chain amino acid (BCAA) metabolism, we performed metabolic profiling with 13C, 15N-leucine. Results: CRISPR-Cas9 screen showed METTL16 is one of the most essential genes for the survival of AMLs. The AML cells display more robust dependency on METTL16 than METTL3/14. We found METTL16 is highly expressed in AML patients compared to healthy controls. METTL16 KO significantly inhibited AML cell proliferation, promoted cell apoptosis and myeloid differentiation in vitro, which could be totally reversed by forced expression of wild-type METTL16, but not catalytic-dead mutant. METTL16 depletion dramatically inhibited AML progression and prolonged survival of recipient mice in the BMT, xenograft and PDX models. In addition, METTL16 is highly expressed in LSCs contrast to leukemic bulk cells and METTL16 KO significantly attenuates LSC self-renewal in vitro and in vivo. By contrast, the role of METLL16 is largely spared in normal hematopoietic cells. Via integrated analysis of m6A-seq data and RNA-seq data, we identified two bona fide targets of METTL16, BCAT1 and BCAT2, which encode two critical BCAA transaminases in BCAA biosynthesis pathway. METTL16 promotes the expression of BCAT1 and BCAT2 via an m6A dependent manner. Metabolomics with 13C, 15N-leucine tracing showed that METTL16 KO results in suppressed pools of TCA cycle intermediates, some non-essential amino acids and nucleotides. Conclusion: We uncover a tumor-promoting role of METTL16 in AML and LSC self-renewal via reprogramming BCAA metabolism, in which METTL16 functions as an m6A methyltransferase to regulate expression of BCAT1 and BCAT2. Our data suggest that METTL16 is an attractive target for AML therapy. Citation Format: Li Han, Lei Dong, Keith Leung, Zhicong Zhao, Yangchan Li, Ying Qing, Jianhuang Xue, Chao Shen, Zhenhua Chen, Lei Gao, Kitty Wang, Keren Zhou, Wei Li, Brandon Tan, Zheng Zhang, Xi Qin, Rui Su, Xiaolan Deng, Jianjun Chen. METTL16 drives leukemogenesis and maintains leukemia stem cell self-renewal via reprogramming BCAA metabolism [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 3617.
- Abstract
1
- 10.1182/blood-2018-99-114085
- Nov 29, 2018
- Blood
Dynamics of Genetic Landscapes and Clonal Evolution between Patients and PDX Models in Acute Myeloid Leukemia
- Abstract
- 10.1182/blood-2023-188391
- Nov 28, 2023
- Blood
Potent and Target Specific Cytotoxicity of PRAME TCR Mimic CAR T Cells in KMT2A-Rearranged AML Patient-Derived Xenograft Model
- Abstract
1
- 10.1182/blood-2019-129475
- Nov 13, 2019
- Blood
Identification of Novel Combination Therapies Active in BCL2 Inhibitor-Resistant Patient-Derived AML Models
- Research Article
1
- 10.1158/1538-7445.sabcs16-p4-06-05
- Feb 14, 2017
- Cancer Research
The phosphoinositide 3-kinase (PI3K) pathway is a key driver of hormone receptor (HR)–positive breast cancer growth and survival. It is estimated that 40-45% of HR+ breast cancers harbor oncogenic mutations in the PIK3CA gene, which encodes the p110α isoform of PI3K. Taselisib (GDC-0032) is a mutant-selective PI3K inhibitor that demonstrates enhanced potency in PIK3CA mutant breast cancer cells and is being developed as a treatment for metastatic breast cancer that targets PIK3CA-mutant, HR-positive, HER2-negative patients. Activating mutations in the ESR1 gene were recently described in metastatic breast cancer. These mutations confer hormone independent growth and may be associated with resistance to aromatase inhibitors. Drugs that selectively bind and antagonize the Estrogen Receptor alpha (ERα) protein and target it for degradation, such as fulvestrant, are referred to as selective estrogen receptor degraders (SERDs). Preclinical activity of the orally bioavailable SERD, GDC-0810, has not been well characterized in ESR1 mutant PDX models. Therefore, our aim was to evaluate the efficacy and pharmacodynamic responses to agents that target ERα and PI3K as monotherapies and in combination, in ESR1 and PIK3CA mutant HR+ breast cancer patient-derived xenograft (PDX) models. We hypothesized that mutational status of ESR1 and PIK3CA may predict the responsiveness of HR+ PDX models to SERDs and PI3K inhibitors in vivo. Characterization of seven PDX models included authentication of hormone receptor status by immunohistochemistry (IHC) and determination of ESR1 and PIK3CA genotype and allele frequency by exome sequencing. For a subset of models that utilize estrogen for growth, mice were supplemented with 17β-estradiol, and cells or tumor fragments were implanted into the fat pad of intact female NOD-SCID or NOD-SCID-IL2Rgamma null mice and treated with fulvestrant, GDC-0810, or taselisib. Both fulvestrant and GDC-0810 were efficacious in ESR1 wild type (WT) and mutant PDX models but to variable degrees ranging from tumor stasis to growth delay, with GDC-0810 resulting in superior single agent activity at relevant clinical exposure in the WHIM20 and WHIM43 ESR1 mutant models. PIK3CA mutations (E542K, E545K, M1004V, and H1047R) were confirmed in six PDX models and PI3K pathway activation verified by strong pS6RP IHC staining. Taselisib induced tumor growth inhibition and tumor regressions in models harboring PIK3CA mutations, and models with no detectable expression of WT p110α were the most sensitive. In the WHIM43 (ESR1 D538G, PIK3CA M1004V), HCI-011 (ESR1 WT, PIK3CA E545K) and HCI-013 (ESR1 Y537S, PIK3CA H1047R) PDX models, combining fulvestrant and taselisib treatment further enhanced tumor growth inhibition with respect to either treatment alone. Our studies demonstrate the diverse anti-tumor responses of HR+ PDX models to SERDs and the PI3K inhibitor taselisib in the context of clinically relevant ESR1 and PIK3CA mutations. Pharmacological and genomic characterization of additional PDX models may aid in strengthening associations between genotype, drug sensitivity and predictive biomarkers of response. Citation Format: Young A, Crocker L, Cheng E, Lacap J, Hamilton P, Oeh J, Ingalla E, Arrazate A, Hager J, Nannini M, Friedman L, Daemen A, Giltnane J, Sampath D. Treatment of ESR1 mutant and PIK3CA mutant patient-derived breast cancer xenograft models reveals differential anti-tumor responses to estrogen receptor degraders and PI3K inhibitors in vivo [abstract]. In: Proceedings of the 2016 San Antonio Breast Cancer Symposium; 2016 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2017;77(4 Suppl):Abstract nr P4-06-05.
- Abstract
1
- 10.1182/blood.v128.22.580.580
- Dec 2, 2016
- Blood
SL-401 Mediates Potent Cytotoxicity Against CD123+ AML and MDS with Excess Blasts and Demonstrates Therapeutic Benefit in PDX Model
- Research Article
10
- 10.1093/bioinformatics/btz063
- Feb 7, 2019
- Bioinformatics
MotivationAcute myeloid leukemia (AML) is one of the most common hematological malignancies, characterized by high relapse and mortality rates. The inherent intra-tumor heterogeneity in AML is thought to play an important role in disease recurrence and resistance to chemotherapy. Although experimental protocols for cell proliferation studies are well established and widespread, they are not easily applicable to in vivo contexts, and the analysis of related time-series data is often complex to achieve. To overcome these limitations, model-driven approaches can be exploited to investigate different aspects of cell population dynamics.ResultsIn this work, we present ProCell, a novel modeling and simulation framework to investigate cell proliferation dynamics that, differently from other approaches, takes into account the inherent stochasticity of cell division events. We apply ProCell to compare different models of cell proliferation in AML, notably leveraging experimental data derived from human xenografts in mice. ProCell is coupled with Fuzzy Self-Tuning Particle Swarm Optimization, a swarm-intelligence settings-free algorithm used to automatically infer the models parameterizations. Our results provide new insights on the intricate organization of AML cells with highly heterogeneous proliferative potential, highlighting the important role played by quiescent cells and proliferating cells characterized by different rates of division in the progression and evolution of the disease, thus hinting at the necessity to further characterize tumor cell subpopulations.Availability and implementationThe source code of ProCell and the experimental data used in this work are available under the GPL 2.0 license on GITHUB at the following URL: https://github.com/aresio/ProCell.Supplementary informationSupplementary data are available at Bioinformatics online.
- Abstract
- 10.1182/blood-2020-140878
- Nov 5, 2020
- Blood
Xeno-MCL: Genomic, Transcriptomic and Pathologic Landscape Associated with Disease Progression, Clonal Evolution and Tissue Tropism in Patient-Derived Xenografts of Mantle Cell Lymphoma
- Abstract
- 10.1182/blood-2022-170348
- Nov 15, 2022
- Blood
Development of a Pediatric AML Patient Derived Xenograft Program
- Abstract
1
- 10.1182/blood-2023-182026
- Nov 28, 2023
- Blood
Synergistic Growth Inhibition of NPM1 Mutant AML PDX By Combined Therapy with BCL-2 Inhibitor Venetoclax (ABT-199) and Menin Inhibitor DS-1594b In Vivo
- Abstract
- 10.1182/blood-2021-152023
- Nov 5, 2021
- Blood
SLC5A3 Transports Myo-Inositol to Support the Growth of Acute Myeloid Leukemia
- New
- Research Article
- 10.1007/978-1-0716-4848-3_12
- Nov 4, 2025
- Methods in molecular biology (Clifton, N.J.)
- New
- Research Article
- 10.1007/978-1-0716-4848-3_19
- Nov 4, 2025
- Methods in molecular biology (Clifton, N.J.)
- New
- Research Article
- 10.1007/978-1-0716-4848-3_25
- Nov 4, 2025
- Methods in molecular biology (Clifton, N.J.)
- New
- Research Article
- 10.1007/978-1-0716-4848-3_3
- Nov 4, 2025
- Methods in molecular biology (Clifton, N.J.)
- New
- Research Article
- 10.1007/978-1-0716-4848-3_28
- Nov 4, 2025
- Methods in molecular biology (Clifton, N.J.)
- New
- Research Article
- 10.1007/978-1-0716-4848-3_14
- Nov 4, 2025
- Methods in molecular biology (Clifton, N.J.)
- New
- Research Article
- 10.1007/978-1-0716-4848-3_26
- Nov 4, 2025
- Methods in molecular biology (Clifton, N.J.)
- New
- Research Article
- 10.1007/978-1-0716-4848-3_13
- Nov 4, 2025
- Methods in molecular biology (Clifton, N.J.)
- New
- Research Article
- 10.1007/978-1-0716-4848-3_22
- Nov 4, 2025
- Methods in molecular biology (Clifton, N.J.)
- New
- Research Article
- 10.1007/978-1-0716-4848-3_7
- Nov 4, 2025
- Methods in molecular biology (Clifton, N.J.)
- Ask R Discovery
- Chat PDF
AI summaries and top papers from 250M+ research sources.