1
|
Bischof K, Cremaschi A, Eroukhmanoff L, Landskron J, Flage‐Larsen L, Gade A, Bjørge L, Urbanucci A, Taskén K. Patient-derived acellular ascites fluid affects drug responses in ovarian cancer cell lines through the activation of key signalling pathways. Mol Oncol 2025; 19:81-98. [PMID: 39245677 PMCID: PMC11705723 DOI: 10.1002/1878-0261.13726] [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: 03/04/2024] [Revised: 07/03/2024] [Accepted: 08/22/2024] [Indexed: 09/10/2024] Open
Abstract
Malignant ascites is commonly produced in advanced epithelial ovarian cancer (EOC) and serves as unique microenvironment for tumour cells. Acellular ascites fluid (AAF) is rich in signalling molecules and has been proposed to play a role in the induction of chemoresistance. Through in vitro testing of drug sensitivity and by assessing intracellular phosphorylation status in response to mono- and combination treatment of five EOC cell lines after incubation with AAFs derived from 20 different patients, we investigated the chemoresistance-inducing potential of ascites. We show that the addition of AAFs to the culture media of EOC cell lines has the potential to induce resistance to standard-of-care drugs (SCDs). We also show that AAFs induce time- and concentration-dependent activation of downstream signalling to signal transducer and activator of transcription 3 (STAT3), and concomitantly altered phosphorylation of mitogen-activated protein kinase kinase (MEK), phosphoinositide 3-kinase (PI3K)-protein kinase B (AKT) and nuclear factor NF-kappa-B (NFκB). Antibodies targeting the interleukin-6 receptor (IL6R) effectively blocked phosphorylation of STAT3 and STAT1. Treatments with SCDs were effective in reducing cell viability in only a third of 30 clinically relevant conditions examined, defined as combinations of drugs, different cell lines and AAFs. Combinations of SCDs and novel therapeutics such as trametinib, fludarabine or rapamycin were superior in another third. Notably, we could nominate effective treatment combinations in almost all conditions except in 4 out of 30 conditions, in which trametinib or fludarabine showed higher efficacy alone. Taken together, our study underscores the importance of the molecular characterisation of individual patients' AAFs and the impact on treatment resistance as providing clinically meaningful information for future precision treatment approaches in EOC.
Collapse
Affiliation(s)
- Katharina Bischof
- Department of Cancer Immunology, Institute for Cancer ResearchUniversity of OsloNorway
- Division of Cancer Medicine, Department of Gynecological OncologyOslo University HospitalNorway
| | - Andrea Cremaschi
- Centre for Molecular Medicine Norway (NCMM)Nordic EMBL Partnership, University of OsloNorway
- Oslo Centre for Biostatistics and EpidemiologyUniversity of OsloNorway
- Singapore Institute for Clinical Sciences, A*STARSingapore
- Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| | - Lena Eroukhmanoff
- Centre for Molecular Medicine Norway (NCMM)Nordic EMBL Partnership, University of OsloNorway
| | - Johannes Landskron
- Centre for Molecular Medicine Norway (NCMM)Nordic EMBL Partnership, University of OsloNorway
| | - Lise‐Lotte Flage‐Larsen
- Centre for Molecular Medicine Norway (NCMM)Nordic EMBL Partnership, University of OsloNorway
| | - Alexandra Gade
- Centre for Molecular Medicine Norway (NCMM)Nordic EMBL Partnership, University of OsloNorway
| | - Line Bjørge
- Department of Obstetrics and GynaecologyHaukeland University HospitalBergenNorway
- Department of Clinical Science, Centre for Cancer Biomarkers CCBIOUniversity of BergenNorway
| | - Alfonso Urbanucci
- Faculty of Medicine and Health TechnologyTAYS Cancer Centre and FICAN Mid, Tampere UniversityFinland
- Department of Tumor Biology, Institute for Cancer ResearchUniversity of OsloNorway
| | - Kjetil Taskén
- Department of Cancer Immunology, Institute for Cancer ResearchUniversity of OsloNorway
- Institute of Clinical MedicineUniversity of OsloNorway
| |
Collapse
|
2
|
Sahib NRBM, Mohamed JS, Rashid MBMA, Jayalakshmi, Lin YC, Chee YL, Fan BE, De Mel S, Ooi MGM, Jen WY, Chow EKH. A Combinatorial Functional Precision Medicine Platform for Rapid Therapeutic Response Prediction in AML. Cancer Med 2024; 13:e70401. [PMID: 39560206 PMCID: PMC11574777 DOI: 10.1002/cam4.70401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Despite advances made in targeted biomarker-based therapy for acute myeloid leukemia (AML) treatment, remission is often short and followed by relapse and acquired resistance. Functional precision medicine (FPM) efforts have been shown to improve therapy selection guidance by incorporating comprehensive biological data to tailor individual treatment. However, effectively managing complex biological data, while also ensuring rapid conversion of actionable insights into clinical utility remains challenging. METHODS We have evaluated the clinical applicability of quadratic phenotypic optimization platform (QPOP), to predict clinical response to combination therapies in AML and reveal patient-centric insights into combination therapy sensitivities. In this prospective study, 51 primary samples from newly diagnosed (ND) or refractory/relapsed (R/R) AML patients were evaluated by QPOP following ex vivo drug testing. RESULTS Individualized drug sensitivity reports were generated in 55/63 (87.3%) patient samples with a median turnaround time of 5 (4-10) days from sample collection to report generation. To evaluate clinical feasibility, QPOP-predicted response was compared to clinical treatment outcomes and indicated concordant results with 83.3% sensitivity and 90.9% specificity and an overall 86.2% accuracy. Serial QPOP analysis in a FLT3-mutant patient sample indicated decreased FLT3 inhibitor (FLT3i) sensitivity, which is concordant with increasing FLT3 allelic burden and drug resistance development. Forkhead box M1 (FOXM1)-AKT signaling was subsequently identified to contribute to resistance to FLT3i. CONCLUSION Overall, this study demonstrates the feasibility of applying QPOP as a functional combinatorial precision medicine platform to predict therapeutic sensitivities in AML and provides the basis for prospective clinical trials evaluating ex vivo-guided combination therapy.
Collapse
Affiliation(s)
- Noor Rashidha Binte Meera Sahib
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jameelah Sheik Mohamed
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore
| | | | - Jayalakshmi
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | | | - Yen Lin Chee
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore
| | - Bingwen Eugene Fan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Haematology, Tan Tock Seng Hospital, Singapore
- Lee Kong Chain School of Medicine, Nanyang Technological University, Singapore
- Department of Laboratory Medicine, Khoo Teck Puat Hospital, Singapore
| | - Sanjay De Mel
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore
| | - Melissa Gaik Ming Ooi
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore
| | - Wei-Ying Jen
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Edward Kai-Hua Chow
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore
| |
Collapse
|
3
|
Wegmann R, Bonilla X, Casanova R, Chevrier S, Coelho R, Esposito C, Ficek-Pascual J, Goetze S, Gut G, Jacob F, Jacobs A, Kuipers J, Lischetti U, Mena J, Milani ES, Prummer M, Del Castillo JS, Singer F, Sivapatham S, Toussaint NC, Vilinovszki O, Wildschut MHE, Thavayogarajah T, Malani D, Aebersold R, Bacac M, Beerenwinkel N, Beisel C, Bodenmiller B, Heinzelmann-Schwarz V, Koelzer VH, Levesque MP, Moch H, Pelkmans L, Rätsch G, Tolnay M, Wicki A, Wollscheid B, Manz MG, Snijder B, Theocharides APA. Single-cell landscape of innate and acquired drug resistance in acute myeloid leukemia. Nat Commun 2024; 15:9402. [PMID: 39477946 PMCID: PMC11525670 DOI: 10.1038/s41467-024-53535-4] [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: 03/08/2024] [Accepted: 10/10/2024] [Indexed: 11/02/2024] Open
Abstract
Deep single-cell multi-omic profiling offers a promising approach to understand and overcome drug resistance in relapsed or refractory (rr) acute myeloid leukemia (AML). Here, we combine single-cell ex vivo drug profiling (pharmacoscopy) with single-cell and bulk DNA, RNA, and protein analyses, alongside clinical data from 21 rrAML patients. Unsupervised data integration reveals reduced ex vivo response to the Bcl-2 inhibitor venetoclax (VEN) in patients treated with both a hypomethylating agent (HMA) and VEN, compared to those pre-exposed to chemotherapy or HMA alone. Integrative analysis identifies both known and unreported mechanisms of innate and treatment-related VEN resistance and suggests alternative treatments, like targeting increased proliferation with the PLK inhibitor volasertib. Additionally, high CD36 expression in VEN-resistant blasts associates with sensitivity to CD36-targeted antibody treatment ex vivo. This study demonstrates how single-cell multi-omic profiling can uncover drug resistance mechanisms and treatment vulnerabilities, providing a valuable resource for future AML research.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/drug effects
- Single-Cell Analysis
- Sulfonamides/pharmacology
- Sulfonamides/therapeutic use
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- CD36 Antigens/metabolism
- CD36 Antigens/genetics
- Female
- Male
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Middle Aged
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Proto-Oncogene Proteins c-bcl-2/genetics
- Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors
- Aged
Collapse
Affiliation(s)
- Rebekka Wegmann
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Ximena Bonilla
- Department of Computer Science, ETH Zurich, Zurich, Switzerland
| | - Ruben Casanova
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Stéphane Chevrier
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Ricardo Coelho
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Cinzia Esposito
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | | | - Sandra Goetze
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- ETH PHRT Swiss Multi-Omics Center (SMOC), Lausanne, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Gabriele Gut
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Francis Jacob
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Andrea Jacobs
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Jack Kuipers
- Department of Biosystems Science and Engineering, ETH Zurich, Zurich, Switzerland
| | - Ulrike Lischetti
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Julien Mena
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Emanuela S Milani
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Michael Prummer
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- NEXUS Personalized Health Technologies, ETH Zurich, Zurich, Switzerland
| | | | - Franziska Singer
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- NEXUS Personalized Health Technologies, ETH Zurich, Zurich, Switzerland
| | - Sujana Sivapatham
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Nora C Toussaint
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- NEXUS Personalized Health Technologies, ETH Zurich, Zurich, Switzerland
- Swiss Data Science Center, ETH Zürich, Zurich, Switzerland
| | - Oliver Vilinovszki
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Mattheus H E Wildschut
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | | | - Disha Malani
- Harvard Medical School and Dana-Farber Cancer Institute, Boston, USA
| | - Rudolf Aebersold
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Marina Bacac
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Zurich, Zurich, Switzerland
| | - Niko Beerenwinkel
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Zurich, Switzerland
| | - Christian Beisel
- Department of Biosystems Science and Engineering, ETH Zurich, Zurich, Switzerland
| | - Bernd Bodenmiller
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | | | - Viktor H Koelzer
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
- University of Zurich, Faculty of Medicine, Zurich, Switzerland
| | | | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
- University of Zurich, Faculty of Medicine, Zurich, Switzerland
| | - Lucas Pelkmans
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Gunnar Rätsch
- Department of Computer Science, ETH Zurich, Zurich, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- AI Center at ETH Zurich, Zurich, Switzerland
| | - Markus Tolnay
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Andreas Wicki
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
- University of Zurich, Faculty of Medicine, Zurich, Switzerland
| | - Bernd Wollscheid
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Markus G Manz
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland.
| | - Berend Snijder
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland.
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| | | |
Collapse
|
4
|
Qin G, Zhang Y, Tyner JW, Kemp CJ, Shmulevich I. Knowledge graphs facilitate prediction of drug response for acute myeloid leukemia. iScience 2024; 27:110755. [PMID: 39280607 PMCID: PMC11401200 DOI: 10.1016/j.isci.2024.110755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 05/04/2024] [Accepted: 08/14/2024] [Indexed: 09/18/2024] Open
Abstract
Acute myeloid leukemia (AML) is a highly aggressive and heterogeneous disease, underscoring the need for improved therapeutic options and methods to optimally predict responses. With the wealth of available data resources, including clinical features, multiomics analysis, and ex vivo drug screening from AML patients, development of drug response prediction models has become feasible. Knowledge graphs (KGs) embed the relationships between different entities or features, allowing for explanation of a wide breadth of drug sensitivity and resistance mechanisms. We designed AML drug response prediction models guided by KGs. Our models included engineered features, relative gene expression between marker genes for each drug and regulators (e.g., transcription factors). We identified relative gene expression of FGD4-MIR4519, NPC2-GATA2, and BCL2-NFKB2 as predictive features for venetoclax ex vivo drug response. The KG-guided models provided high accuracy in independent test sets, overcame potential platform batch effects, and provided candidate drug sensitivity biomarkers for further validation.
Collapse
Affiliation(s)
- Guangrong Qin
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Yue Zhang
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Jeffrey W. Tyner
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | | | | |
Collapse
|
5
|
Futran AS, Lu T, Amberg-Johnson K, Xu J, Yang X, He S, Boyce S, Bell JA, Pelletier R, Suzuki T, Huang X, Qian H, Fang L, Xing L, Xu Z, Kurtz SE, Tyner JW, Tang W, Guo T, Akinsanya K, Madge D, Jensen KK. Ubiquitin-specific protease 7 inhibitors reveal a differentiated mechanism of p53-driven anti-cancer activity. iScience 2024; 27:109693. [PMID: 38689642 PMCID: PMC11059122 DOI: 10.1016/j.isci.2024.109693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/11/2024] [Accepted: 04/05/2024] [Indexed: 05/02/2024] Open
Abstract
The USP7 deubiquitinase regulates proteins involved in the cell cycle, DNA repair, and epigenetics and has been implicated in cancer progression. USP7 inhibition has been pursued for the development of anti-cancer therapies. Here, we describe the discovery of potent and specific USP7 inhibitors exemplified by FX1-5303. FX1-5303 was used as a chemical probe to study the USP7-mediated regulation of p53 signaling in cells. It demonstrates mechanistic differences compared to MDM2 antagonists, a related class of anti-tumor agents that act along the same pathway. FX1-5303 synergizes with the clinically approved BCL2 inhibitor venetoclax in acute myeloid leukemia (AML) cell lines and ex vivo patient samples and leads to strong tumor growth inhibition in in vivo mouse xenograft models of multiple myeloma and AML. This work introduces new USP7 inhibitors, differentiates their mechanism of action from MDM2 inhibition, and identifies specific opportunities for their use in the treatment of AML.
Collapse
Affiliation(s)
- Alan S. Futran
- Schrödinger, 1540 Broadway 24th Floor, New York, NY, USA
| | - Tao Lu
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | | | - Jiayi Xu
- Schrödinger, 1540 Broadway 24th Floor, New York, NY, USA
| | - Xiaoxiao Yang
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Saidi He
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Sarah Boyce
- Schrödinger, 1540 Broadway 24th Floor, New York, NY, USA
| | | | | | - Takao Suzuki
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Xianhai Huang
- Schrödinger, 1540 Broadway 24th Floor, New York, NY, USA
| | - Heng Qian
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Liping Fang
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Li Xing
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Zhaowu Xu
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Stephen E. Kurtz
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Jeffrey W. Tyner
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Wayne Tang
- Schrödinger, 1540 Broadway 24th Floor, New York, NY, USA
| | - Tao Guo
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | | | - David Madge
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | | |
Collapse
|
6
|
Jessop SJ, Fuentos‐Bolanos N, Mayoh C, Dolman MEM, Tax G, Wong‐Erasmus M, Ajuyah P, Tyrell V, Marshall GM, Ziegler DS, Lau LMS. High throughput screening aids clinical decision-making in refractory acute myeloid leukaemia. Cancer Rep (Hoboken) 2024; 7:e2061. [PMID: 38662349 PMCID: PMC11044912 DOI: 10.1002/cnr2.2061] [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: 10/09/2023] [Revised: 02/29/2024] [Accepted: 03/24/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND Despite advances in therapeutics for adverse-risk acute myeloid leukaemia (AML), overall survival remains poor, especially in refractory disease. Comprehensive tumour profiling and pre-clinical drug testing can identify effective personalised therapies. CASE We describe a case of ETV6-MECOM fusion-positive refractory AML, where molecular analysis and in vitro high throughput drug screening identified a tolerable, novel targeted therapy and provided rationale for avoiding what could have been a toxic treatment regimen. Ruxolitinib combined with hydroxyurea led to disease control and enhanced quality-of-life in a patient unsuitable for intensified chemotherapy or allogeneic stem cell transplantation. CONCLUSION This case report demonstrates the feasibility and role of combination pre-clinical high throughput screening to aid decision making in high-risk leukaemia. It also demonstrates the role a JAK1/2 inhibitor can have in the palliative setting in select patients with AML.
Collapse
Affiliation(s)
- S. J. Jessop
- Children's Cancer InstituteLowy Cancer Research Centre, UNSW SydneyKensingtonNew South WalesAustralia
- Department for Haematology/OncologyWomen's and Children's HospitalSouth AustraliaAustralia
- Adelaide Medical SchoolUniversity of AdelaideSouth AustraliaAustralia
| | - N. Fuentos‐Bolanos
- Children's Cancer InstituteLowy Cancer Research Centre, UNSW SydneyKensingtonNew South WalesAustralia
- Kids Cancer CentreSydney Children's HospitalNew South WalesAustralia
| | - C. Mayoh
- Children's Cancer InstituteLowy Cancer Research Centre, UNSW SydneyKensingtonNew South WalesAustralia
- School of Clinical MedicineUNSW Medicine & Health, UNSW SydneyKensingtonNew South WalesAustralia
| | - M. E. M. Dolman
- Children's Cancer InstituteLowy Cancer Research Centre, UNSW SydneyKensingtonNew South WalesAustralia
- School of Clinical MedicineUNSW Medicine & Health, UNSW SydneyKensingtonNew South WalesAustralia
| | - G. Tax
- Children's Cancer InstituteLowy Cancer Research Centre, UNSW SydneyKensingtonNew South WalesAustralia
- School of Clinical MedicineUNSW Medicine & Health, UNSW SydneyKensingtonNew South WalesAustralia
| | - M. Wong‐Erasmus
- Children's Cancer InstituteLowy Cancer Research Centre, UNSW SydneyKensingtonNew South WalesAustralia
| | - P. Ajuyah
- Children's Cancer InstituteLowy Cancer Research Centre, UNSW SydneyKensingtonNew South WalesAustralia
| | - V. Tyrell
- Children's Cancer InstituteLowy Cancer Research Centre, UNSW SydneyKensingtonNew South WalesAustralia
| | - G. M. Marshall
- Children's Cancer InstituteLowy Cancer Research Centre, UNSW SydneyKensingtonNew South WalesAustralia
- Kids Cancer CentreSydney Children's HospitalNew South WalesAustralia
| | - D. S. Ziegler
- Children's Cancer InstituteLowy Cancer Research Centre, UNSW SydneyKensingtonNew South WalesAustralia
- Kids Cancer CentreSydney Children's HospitalNew South WalesAustralia
- School of Clinical MedicineUNSW Medicine & Health, UNSW SydneyKensingtonNew South WalesAustralia
| | - L. M. S. Lau
- Children's Cancer InstituteLowy Cancer Research Centre, UNSW SydneyKensingtonNew South WalesAustralia
- Kids Cancer CentreSydney Children's HospitalNew South WalesAustralia
| |
Collapse
|
7
|
Mirzaie M, Gholizadeh E, Miettinen JJ, Ianevski F, Ruokoranta T, Saarela J, Manninen M, Miettinen S, Heckman CA, Jafari M. Designing patient-oriented combination therapies for acute myeloid leukemia based on efficacy/toxicity integration and bipartite network modeling. Oncogenesis 2024; 13:11. [PMID: 38429288 PMCID: PMC10907624 DOI: 10.1038/s41389-024-00510-9] [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: 06/16/2023] [Revised: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 03/03/2024] Open
Abstract
Acute myeloid leukemia (AML), a heterogeneous and aggressive blood cancer, does not respond well to single-drug therapy. A combination of drugs is required to effectively treat this disease. Computational models are critical for combination therapy discovery due to the tens of thousands of two-drug combinations, even with approved drugs. While predicting synergistic drugs is the focus of current methods, few consider drug efficacy and potential toxicity, which are crucial for treatment success. To find effective new drug candidates, we constructed a bipartite network using patient-derived tumor samples and drugs. The network is based on drug-response screening and summarizes all treatment response heterogeneity as drug response weights. This bipartite network is then projected onto the drug part, resulting in the drug similarity network. Distinct drug clusters were identified using community detection methods, each targeting different biological processes and pathways as revealed by enrichment and pathway analysis of the drugs' protein targets. Four drugs with the highest efficacy and lowest toxicity from each cluster were selected and tested for drug sensitivity using cell viability assays on various samples. Results show that ruxolitinib-ulixertinib and sapanisertib-LY3009120 are the most effective combinations with the least toxicity and the best synergistic effect on blast cells. These findings lay the foundation for personalized and successful AML therapies, ultimately leading to the development of drug combinations that can be used alongside standard first-line AML treatment.
Collapse
Affiliation(s)
- Mehdi Mirzaie
- Department of Biochemistry and Developmental Biology, University of Helsinki, Helsinki, Finland
| | - Elham Gholizadeh
- Department of Biochemistry and Developmental Biology, University of Helsinki, Helsinki, Finland
| | - Juho J Miettinen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Filipp Ianevski
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Tanja Ruokoranta
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Jani Saarela
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | | | - Susanna Miettinen
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tays Research Services, Wellbeing Services County of Pirkanmaa, Tampere University Hospital, Tampere, Finland
| | - Caroline A Heckman
- Institute for Molecular Medicine Finland - FIMM, HiLIFE - Helsinki Institute of Life Science, iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland.
| | - Mohieddin Jafari
- Department of Biochemistry and Developmental Biology, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
8
|
Ali KA, Shah RD, Dhar A, Myers NM, Nguyen C, Paul A, Mancuso JE, Scott Patterson A, Brody JP, Heiser D. Ex vivo discovery of synergistic drug combinations for hematologic malignancies. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024; 29:100129. [PMID: 38101570 DOI: 10.1016/j.slasd.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/13/2023] [Accepted: 12/09/2023] [Indexed: 12/17/2023]
Abstract
Combination therapies have improved outcomes for patients with acute myeloid leukemia (AML). However, these patients still have poor overall survival. Although many combination therapies are identified with high-throughput screening (HTS), these approaches are constrained to disease models that can be grown in large volumes (e.g., immortalized cell lines), which have limited translational utility. To identify more effective and personalized treatments, we need better strategies for screening and exploring potential combination therapies. Our objective was to develop an HTS platform for identifying effective combination therapies with highly translatable ex vivo disease models that use size-limited, primary samples from patients with leukemia (AML and myelodysplastic syndrome). We developed a system, ComboFlow, that comprises three main components: MiniFlow, ComboPooler, and AutoGater. MiniFlow conducts ex vivo drug screening with a miniaturized flow-cytometry assay that uses minimal amounts of patient sample to maximize throughput. ComboPooler incorporates computational methods to design efficient screens of pooled drug combinations. AutoGater is an automated gating classifier for flow cytometry that uses machine learning to rapidly analyze the large datasets generated by the assay. We used ComboFlow to efficiently screen more than 3000 drug combinations across 20 patient samples using only 6 million cells per patient sample. In this screen, ComboFlow identified the known synergistic combination of bortezomib and panobinostat. ComboFlow also identified a novel drug combination, dactinomycin and fludarabine, that synergistically killed leukemic cells in 35 % of AML samples. This combination also had limited effects in normal, hematopoietic progenitors. In conclusion, ComboFlow enables exploration of massive landscapes of drug combinations that were previously inaccessible in ex vivo models. We envision that ComboFlow can be used to discover more effective and personalized combination therapies for cancers amenable to ex vivo models.
Collapse
Affiliation(s)
- Kamran A Ali
- Notable Labs, 320 Hatch Dr, Foster City, CA, 94404, USA; Department of Biomedical Engineering, University of California, Irvine, 3120 Natural Sciences II, Irvine, CA, 92697, USA.
| | - Reecha D Shah
- Notable Labs, 320 Hatch Dr, Foster City, CA, 94404, USA
| | - Anukriti Dhar
- Notable Labs, 320 Hatch Dr, Foster City, CA, 94404, USA
| | - Nina M Myers
- Notable Labs, 320 Hatch Dr, Foster City, CA, 94404, USA
| | | | - Arisa Paul
- Notable Labs, 320 Hatch Dr, Foster City, CA, 94404, USA
| | | | | | - James P Brody
- Department of Biomedical Engineering, University of California, Irvine, 3120 Natural Sciences II, Irvine, CA, 92697, USA
| | - Diane Heiser
- Notable Labs, 320 Hatch Dr, Foster City, CA, 94404, USA
| |
Collapse
|
9
|
Yu S, Zhang Y, Yu G, Wang Y, Shao R, Du X, Xu N, Lin D, Zhao W, Zhang X, Xiao J, Sun Z, Deng L, Liang X, Zhang H, Guo Z, Dai M, Shi P, Huang F, Fan Z, Liu Q, Lin R, Jiang X, Xuan L, Liu Q, Jin H. Sorafenib plus triplet therapy with venetoclax, azacitidine and homoharringtonine for refractory/relapsed acute myeloid leukemia with FLT3-ITD: A multicenter phase 2 study. J Intern Med 2024; 295:216-228. [PMID: 37899297 DOI: 10.1111/joim.13738] [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] [Indexed: 10/31/2023]
Abstract
BACKGROUND Patients with relapsed or refractory acute myeloid leukemia (R/R AML) and FLT3-internal tandem duplication (FLT3-ITD) respond infrequently to salvage chemotherapy. OBJECTIVE To investigate the efficacy of sorafenib plus triplet therapy with venetoclax, azacitidine, and homoharringtonine (VAH) as a salvage therapy in this population. METHODS This multicenter, single-arm, phase 2 study was conducted at 12 hospitals across China. Eligible patients had R/R AML with FLT3-ITD (aged 18-65 years) who were treated with VAH. The primary endpoint was composite complete remission (CRc) after two cycles. Secondary outcomes included the overall response rate (ORR), safety, and survival. RESULTS Between July 9, 2020, and March 19, 2022, 58 patients were assessed for eligibility, 51 of whom were enrolled. The median patient age was 47 years (interquartile range [IQR] 31-57). CRc was 76.5% with ORR of 82.4%. At a median follow-up of 17.7 months (IQR, 8.7-24.7), the median duration of CRc was not reached (NR), overall survival was 18.1 months (95% confidence interval [CI], 11.8-NR) and event-free survival was 11.4 months (95% CI, 5.6-NR). Grade 3 or 4 adverse events occurring in ≥10% of patients included neutropenia in 47 (92.2%), thrombocytopenia in 41 (80.4%), anemia in 35 (68.6%), febrile neutropenia in 29 (56.9%), pneumonia in 13 (25.5%), and sepsis in 6 (11.8%) patients. Treatment-related death occurred in two (3.9%) patients. CONCLUSIONS The sorafenib plus VAH regimen was well tolerated and highly active against R/R AML with FLT3-ITD. This regimen may be a suitable therapeutic option for this population, but larger population trials are needed to be explored. TRIAL REGISTRATION Clinical Trials Registry: NCT04424147.
Collapse
Affiliation(s)
- Sijian Yu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Clinical Medical Research Center of Hematology Diseases of Guangdong Province, Guangzhou, China
| | - Yu Zhang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Clinical Medical Research Center of Hematology Diseases of Guangdong Province, Guangzhou, China
| | - Guopan Yu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Clinical Medical Research Center of Hematology Diseases of Guangdong Province, Guangzhou, China
| | - Yu Wang
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
| | - Ruoyang Shao
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Clinical Medical Research Center of Hematology Diseases of Guangdong Province, Guangzhou, China
| | - Xin Du
- Peking Department of Hematology and Shenzhen Bone Marrow Transplantation Public Service Platform, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Na Xu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Clinical Medical Research Center of Hematology Diseases of Guangdong Province, Guangzhou, China
| | - Dongjun Lin
- Department of Hematology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Weihua Zhao
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiong Zhang
- Department of Hematology, Maoming People's Hospital, Maoming, China
| | - Jie Xiao
- Department of Hematology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhiqiang Sun
- Department of Hematology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Lan Deng
- Department of Hematology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinquan Liang
- Department of Hematology, The First People's Hospital of Chenzhou, Chenzhou, China
| | - Hongyu Zhang
- Department of Hematology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Ziwen Guo
- Department of Hematology, Zhongshan City People's Hospital, Zhongshan, China
| | - Min Dai
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Clinical Medical Research Center of Hematology Diseases of Guangdong Province, Guangzhou, China
| | - Pengcheng Shi
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Clinical Medical Research Center of Hematology Diseases of Guangdong Province, Guangzhou, China
| | - Fen Huang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Clinical Medical Research Center of Hematology Diseases of Guangdong Province, Guangzhou, China
| | - Zhiping Fan
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Clinical Medical Research Center of Hematology Diseases of Guangdong Province, Guangzhou, China
| | - Qiong Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Clinical Medical Research Center of Hematology Diseases of Guangdong Province, Guangzhou, China
| | - Ren Lin
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Clinical Medical Research Center of Hematology Diseases of Guangdong Province, Guangzhou, China
| | - Xuejie Jiang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Clinical Medical Research Center of Hematology Diseases of Guangdong Province, Guangzhou, China
| | - Li Xuan
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Clinical Medical Research Center of Hematology Diseases of Guangdong Province, Guangzhou, China
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Clinical Medical Research Center of Hematology Diseases of Guangdong Province, Guangzhou, China
| | - Hua Jin
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Clinical Medical Research Center of Hematology Diseases of Guangdong Province, Guangzhou, China
| |
Collapse
|
10
|
Mohanty V, Baran N, Huang Y, Ramage CL, Cooper LM, He S, Iqbal R, Daher M, Tyner JW, Mills GB, Konopleva M, Chen K. Transcriptional and phenotypic heterogeneity underpinning venetoclax resistance in AML. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.27.577579. [PMID: 38352538 PMCID: PMC10862759 DOI: 10.1101/2024.01.27.577579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/03/2024]
Abstract
The venetoclax BCL2 inhibitor in combination with hypomethylating agents represents a cornerstone of induction therapy for older AML patients, unfit for intensive chemotherapy. Like other targeted therapies, venetoclax-based therapies suffer from innate and acquired resistance. While several mechanisms of resistance have been identified, the heterogeneity of resistance mechanism across patient populations is poorly understood. Here we utilized integrative analysis of transcriptomic and ex-vivo drug response data in AML patients to identify four transcriptionally distinct VEN resistant clusters (VR_C1-4), with distinct phenotypic, genetic and drug response patterns. VR_C1 was characterized by enrichment for differentiated monocytic- and cDC-like blasts, transcriptional activation of PI3K-AKT-mTOR signaling axis, and energy metabolism pathways. They showed sensitivity to mTOR and CDK inhibition. VR_C2 was enriched for NRAS mutations and associated with distinctive transcriptional suppression of HOX expression. VR_C3 was characterized by enrichment for TP53 mutations and higher infiltration by cytotoxic T cells. This cluster showed transcriptional expression of erythroid markers, suggesting tumor cells mimicking erythroid differentiation, activation of JAK-STAT signaling, and sensitivity to JAK inhibition, which in a subset of cases synergized with venetoclax. VR_C4 shared transcriptional similarities with venetoclax-sensitive patients, with modest over-expression of interferon signaling. They were also characterized by high rates of DNMT3A mutations. Finally, we projected venetoclax-resistance states onto single cells profiled from a patient who relapsed under venetoclax therapy capturing multiple resistance states in the tumor and shifts in their abundance under venetoclax selection, suggesting that single tumors may consist of cells mimicking multiple VR_Cs contributing to intra-tumor heterogeneity. Taken together, our results provide a strategy to evaluate inter- and intra-tumor heterogeneity of venetoclax resistance mechanisms and provide insights into approaches to navigate further management of patients who failed therapy with BCL2 inhibitors.
Collapse
Affiliation(s)
- Vakul Mohanty
- Department of Bioinformatics and Computational biology, The University of Texas MD Anderson Cancer Center
| | - Natalia Baran
- Department of Leukemia, The University of Texas MD Anderson Cancer Center
| | - Yuefan Huang
- Department of Bioinformatics and Computational biology, The University of Texas MD Anderson Cancer Center
| | - Cassandra L Ramage
- Department of Leukemia, The University of Texas MD Anderson Cancer Center
| | - Laurie M Cooper
- Department of Leukemia, The University of Texas MD Anderson Cancer Center
| | - Shan He
- Department of Bioinformatics and Computational biology, The University of Texas MD Anderson Cancer Center
| | - Ramiz Iqbal
- Department of Bioinformatics and Computational biology, The University of Texas MD Anderson Cancer Center
| | - May Daher
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center
| | - Jeffrey W Tyner
- Department of Cell, Developmental & Cancer Biology, Knight Cancer Institute, Oregon Health & Science University
| | - Gordon B Mills
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University
| | - Marina Konopleva
- Department of Medicine (Oncology) and Molecular Pharmacology, Albert Einstein College of Medicine
| | - Ken Chen
- Department of Bioinformatics and Computational biology, The University of Texas MD Anderson Cancer Center
| |
Collapse
|
11
|
Pino JC, Posso C, Joshi SK, Nestor M, Moon J, Hansen JR, Hutchinson-Bunch C, Gritsenko MA, Weitz KK, Watanabe-Smith K, Long N, McDermott JE, Druker BJ, Liu T, Tyner JW, Agarwal A, Traer E, Piehowski PD, Tognon CE, Rodland KD, Gosline SJC. Mapping the proteogenomic landscape enables prediction of drug response in acute myeloid leukemia. Cell Rep Med 2024; 5:101359. [PMID: 38232702 PMCID: PMC10829797 DOI: 10.1016/j.xcrm.2023.101359] [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: 05/25/2023] [Revised: 10/20/2023] [Accepted: 12/10/2023] [Indexed: 01/19/2024]
Abstract
Acute myeloid leukemia is a poor-prognosis cancer commonly stratified by genetic aberrations, but these mutations are often heterogeneous and fail to consistently predict therapeutic response. Here, we combine transcriptomic, proteomic, and phosphoproteomic datasets with ex vivo drug sensitivity data to help understand the underlying pathophysiology of AML beyond mutations. We measure the proteome and phosphoproteome of 210 patients and combine them with genomic and transcriptomic measurements to identify four proteogenomic subtypes that complement existing genetic subtypes. We build a predictor to classify samples into subtypes and map them to a "landscape" that identifies specific drug response patterns. We then build a drug response prediction model to identify drugs that target distinct subtypes and validate our findings on cell lines representing various stages of quizartinib resistance. Our results show how multiomics data together with drug sensitivity data can inform therapy stratification and drug combinations in AML.
Collapse
Affiliation(s)
- James C Pino
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Camilo Posso
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Sunil K Joshi
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Michael Nestor
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Jamie Moon
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Joshua R Hansen
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Chelsea Hutchinson-Bunch
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Marina A Gritsenko
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Karl K Weitz
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Kevin Watanabe-Smith
- Division of Oncological Sciences, Oregon Health & Science University, Portland, OR, USA
| | - Nicola Long
- Division of Oncological Sciences, Oregon Health & Science University, Portland, OR, USA
| | - Jason E McDermott
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA; Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Brian J Druker
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA; Division of Oncological Sciences, Oregon Health & Science University, Portland, OR, USA
| | - Tao Liu
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Jeffrey W Tyner
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA; Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Anupriya Agarwal
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA; Division of Oncological Sciences, Oregon Health & Science University, Portland, OR, USA; Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Elie Traer
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Paul D Piehowski
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Cristina E Tognon
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Karin D Rodland
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA; Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, OR, USA.
| | - Sara J C Gosline
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA.
| |
Collapse
|
12
|
Calderon A, Han C, Karma S, Wang E. Non-genetic mechanisms of drug resistance in acute leukemias. Trends Cancer 2024; 10:38-51. [PMID: 37839973 DOI: 10.1016/j.trecan.2023.09.003] [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: 06/20/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 10/17/2023]
Abstract
Acute leukemia is characterized by clonal heterogeneity that contributes to poor drug responses in patients. Despite treatment advances, the occurrence of relapse remains a major barrier to achieving cures as current therapeutic approaches are inadequate to effectively prevent or overcome resistance. Given that only a few genetic mutations are associated with relapse in acute leukemia patients, there is a growing focus on 'non-genetic' mechanisms that affect the hallmarks of cancer to allow leukemic cells to survive post therapy. In this review, we provide an overview of the therapeutic landscape in acute leukemias. Importantly, we discuss non-genetic mechanisms exploited by leukemic cells to promote their survival after treatment. Last, we present current strategies to prevent or overcome drug resistance in this disease.
Collapse
Affiliation(s)
| | - Cuijuan Han
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Sadik Karma
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA; Graduate Program in Genetics and Development, UConn Health, Farmington, CT, USA
| | - Eric Wang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA; Department of Genetics and Genome Sciences, UConn Health, Farmington, CT, USA; Institute for Systems Genomics, University of Connecticut Health Center, Farmington, CT, USA.
| |
Collapse
|
13
|
Ayuda-Durán P, Hermansen JU, Giliberto M, Yin Y, Hanes R, Gordon S, Kuusanmäki H, Brodersen AM, Andersen AN, Taskén K, Wennerberg K, Enserink JM, Skånland SS. Standardized assays to monitor drug sensitivity in hematologic cancers. Cell Death Discov 2023; 9:435. [PMID: 38040674 PMCID: PMC10692209 DOI: 10.1038/s41420-023-01722-5] [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: 07/13/2023] [Revised: 10/21/2023] [Accepted: 11/13/2023] [Indexed: 12/03/2023] Open
Abstract
The principle of drug sensitivity testing is to expose cancer cells to a library of different drugs and measure its effects on cell viability. Recent technological advances, continuous approval of targeted therapies, and improved cell culture protocols have enhanced the precision and clinical relevance of such screens. Indeed, drug sensitivity testing has proven diagnostically valuable for patients with advanced hematologic cancers. However, different cell types behave differently in culture and therefore require optimized drug screening protocols to ensure that their ex vivo drug sensitivity accurately reflects in vivo drug responses. For example, primary chronic lymphocytic leukemia (CLL) and multiple myeloma (MM) cells require unique microenvironmental stimuli to survive in culture, while this is less the case for acute myeloid leukemia (AML) cells. Here, we present our optimized and validated protocols for culturing and drug screening of primary cells from AML, CLL, and MM patients, and a generic protocol for cell line models. We also discuss drug library designs, reproducibility, and quality controls. We envision that these protocols may serve as community guidelines for the use and interpretation of assays to monitor drug sensitivity in hematologic cancers and thus contribute to standardization. The read-outs may provide insight into tumor biology, identify or confirm treatment resistance and sensitivity in real time, and ultimately guide clinical decision-making.
Collapse
Affiliation(s)
- Pilar Ayuda-Durán
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Johanne U Hermansen
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- K. G. Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Mariaserena Giliberto
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- K. G. Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Yanping Yin
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- K. G. Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Haematology, Oslo University Hospital, Oslo, Norway
| | - Robert Hanes
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Section for Biochemistry and Molecular Biology, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Sandra Gordon
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Heikki Kuusanmäki
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Andrea M Brodersen
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Section for Biochemistry and Molecular Biology, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Aram N Andersen
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Section for Biochemistry and Molecular Biology, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Kjetil Taskén
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- K. G. Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Krister Wennerberg
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Jorrit M Enserink
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Section for Biochemistry and Molecular Biology, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Sigrid S Skånland
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
- K. G. Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
14
|
Chang SH, Ice RJ, Chen M, Sidorov M, Woo RWL, Rodriguez-Brotons A, Jian D, Kim HK, Kim A, Stone DE, Nazarian A, Oh A, Tranah GJ, Nosrati M, de Semir D, Dar AA, Desprez PY, Kashani-Sabet M, Soroceanu L, McAllister SD. Pan-Cancer Pharmacogenomic Analysis of Patient-Derived Tumor Cells Using Clinically Relevant Drug Exposures. Mol Cancer Ther 2023; 22:1100-1111. [PMID: 37440705 DOI: 10.1158/1535-7163.mct-22-0486] [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: 07/22/2022] [Revised: 12/11/2022] [Accepted: 07/10/2023] [Indexed: 07/15/2023]
Abstract
As a result of tumor heterogeneity and solid cancers harboring multiple molecular defects, precision medicine platforms in oncology are most effective when both genetic and pharmacologic determinants of a tumor are evaluated. Expandable patient-derived xenograft (PDX) mouse tumor and corresponding PDX culture (PDXC) models recapitulate many of the biological and genetic characteristics of the original patient tumor, allowing for a comprehensive pharmacogenomic analysis. Here, the somatic mutations of 23 matched patient tumor and PDX samples encompassing four cancers were first evaluated using next-generation sequencing (NGS). 19 antitumor agents were evaluated across 78 patient-derived tumor cultures using clinically relevant drug exposures. A binarization threshold sensitivity classification determined in culture (PDXC) was used to identify tumors that best respond to drug in vivo (PDX). Using this sensitivity classification, logic models of DNA mutations were developed for 19 antitumor agents to predict drug response. We determined that the concordance of somatic mutations across patient and corresponding PDX samples increased as variant allele frequency increased. Notable individual PDXC responses to specific drugs, as well as lineage-specific drug responses were identified. Robust responses identified in PDXC were recapitulated in vivo in PDX-bearing mice and logic modeling determined somatic gene mutation(s) defining response to specific antitumor agents. In conclusion, combining NGS of primary patient tumors, high-throughput drug screen using clinically relevant doses, and logic modeling, can provide a platform for understanding response to therapeutic drugs targeting cancer.
Collapse
Affiliation(s)
- Stephen H Chang
- University of California at San Francisco, School of Pharmacy, Department of Clinical Pharmacy, San Francisco, California
| | - Ryan J Ice
- California Pacific Medical Center Research Institute, San Francisco, California
| | - Michelle Chen
- California Pacific Medical Center Research Institute, San Francisco, California
| | - Maxim Sidorov
- California Pacific Medical Center Research Institute, San Francisco, California
| | - Rinette W L Woo
- California Pacific Medical Center Research Institute, San Francisco, California
| | | | - Damon Jian
- California Pacific Medical Center Research Institute, San Francisco, California
| | - Han Kyul Kim
- California Pacific Medical Center Research Institute, San Francisco, California
| | - Angela Kim
- California Pacific Medical Center Research Institute, San Francisco, California
| | - David E Stone
- California Pacific Medical Center Research Institute, San Francisco, California
| | - Ari Nazarian
- California Pacific Medical Center Research Institute, San Francisco, California
| | - Alyssia Oh
- California Pacific Medical Center Research Institute, San Francisco, California
| | - Gregory J Tranah
- California Pacific Medical Center Research Institute, San Francisco, California
| | - Mehdi Nosrati
- California Pacific Medical Center Research Institute, San Francisco, California
| | - David de Semir
- California Pacific Medical Center Research Institute, San Francisco, California
| | - Altaf A Dar
- California Pacific Medical Center Research Institute, San Francisco, California
| | - Pierre-Yves Desprez
- California Pacific Medical Center Research Institute, San Francisco, California
| | | | - Liliana Soroceanu
- California Pacific Medical Center Research Institute, San Francisco, California
| | - Sean D McAllister
- California Pacific Medical Center Research Institute, San Francisco, California
| |
Collapse
|
15
|
Janizek JD, Dincer AB, Celik S, Chen H, Chen W, Naxerova K, Lee SI. Uncovering expression signatures of synergistic drug responses via ensembles of explainable machine-learning models. Nat Biomed Eng 2023; 7:811-829. [PMID: 37127711 PMCID: PMC11149694 DOI: 10.1038/s41551-023-01034-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 04/01/2023] [Indexed: 05/03/2023]
Abstract
Machine learning may aid the choice of optimal combinations of anticancer drugs by explaining the molecular basis of their synergy. By combining accurate models with interpretable insights, explainable machine learning promises to accelerate data-driven cancer pharmacology. However, owing to the highly correlated and high-dimensional nature of transcriptomic data, naively applying current explainable machine-learning strategies to large transcriptomic datasets leads to suboptimal outcomes. Here by using feature attribution methods, we show that the quality of the explanations can be increased by leveraging ensembles of explainable machine-learning models. We applied the approach to a dataset of 133 combinations of 46 anticancer drugs tested in ex vivo tumour samples from 285 patients with acute myeloid leukaemia and uncovered a haematopoietic-differentiation signature underlying drug combinations with therapeutic synergy. Ensembles of machine-learning models trained to predict drug combination synergies on the basis of gene-expression data may improve the feature attribution quality of complex machine-learning models.
Collapse
Affiliation(s)
- Joseph D Janizek
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA, USA
| | - Ayse B Dincer
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA
| | - Safiye Celik
- Recursion Pharmaceuticals, Salt Lake City, UT, USA
| | - Hugh Chen
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA
| | - William Chen
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA
| | - Kamila Naxerova
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Su-In Lee
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA.
| |
Collapse
|
16
|
Hermansen JU, Yin Y, Urban A, Myklebust CV, Karlsen L, Melvold K, Tveita AA, Taskén K, Munthe LA, Tjønnfjord GE, Skånland SS. A tumor microenvironment model of chronic lymphocytic leukemia enables drug sensitivity testing to guide precision medicine. Cell Death Discov 2023; 9:125. [PMID: 37055391 PMCID: PMC10101987 DOI: 10.1038/s41420-023-01426-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 04/03/2023] [Indexed: 04/15/2023] Open
Abstract
The microenvironment of chronic lymphocytic leukemia (CLL) cells in lymph nodes, spleen, and bone marrow provides survival, proliferation, and drug resistance signals. Therapies need to be effective in these compartments, and pre-clinical models of CLL that are used to test drug sensitivity must mimic the tumor microenvironment to reflect clinical responses. Ex vivo models have been developed that capture individual or multiple aspects of the CLL microenvironment, but they are not necessarily compatible with high-throughput drug screens. Here, we report on a model that has reasonable associated costs, can be handled in a regularly equipped cell lab, and is compatible with ex vivo functional assays including drug sensitivity screens. The CLL cells are cultured with fibroblasts that express the ligands APRIL, BAFF and CD40L for 24 h. The transient co-culture was shown to support survival of primary CLL cells for at least 13 days, and mimic in vivo drug resistance signals. Ex vivo sensitivity and resistance to the Bcl-2 antagonist venetoclax correlated with in vivo responses. The assay was used to identify treatment vulnerabilities and guide precision medicine for a patient with relapsed CLL. Taken together, the presented CLL microenvironment model enables clinical implementation of functional precision medicine in CLL.
Collapse
Affiliation(s)
- Johanne U Hermansen
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- K. G. Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Yanping Yin
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- K. G. Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Haematology, Oslo University Hospital, Oslo, Norway
| | - Aleksandra Urban
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- K. G. Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Camilla V Myklebust
- K. G. Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Linda Karlsen
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- K. G. Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Katrine Melvold
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- K. G. Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Anders A Tveita
- K. G. Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Kjetil Taskén
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- K. G. Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ludvig A Munthe
- K. G. Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Geir E Tjønnfjord
- K. G. Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Haematology, Oslo University Hospital, Oslo, Norway
| | - Sigrid S Skånland
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
- K. G. Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
17
|
Strachan DC, Gu CJ, Kita R, Anderson EK, Richardson MA, Yam G, Pimm G, Roselli J, Schweickert A, Terrell M, Rashid R, Gonzalez AK, Oviedo HH, Alozie MC, Ilangovan T, Marcogliese AN, Tada H, Santaguida MT, Stevens AM. Ex Vivo Drug Sensitivity Correlates with Clinical Response and Supports Personalized Therapy in Pediatric AML. Cancers (Basel) 2022; 14:cancers14246240. [PMID: 36551725 PMCID: PMC9777060 DOI: 10.3390/cancers14246240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/01/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease that accounts for ~20% of all childhood leukemias, and more than 40% of children with AML relapse within three years of diagnosis. Although recent efforts have focused on developing a precise medicine-based approach towards treating AML in adults, there remains a critical gap in therapies designed specifically for children. Here, we present ex vivo drug sensitivity profiles for children with de novo AML using an automated flow cytometry platform. Fresh diagnostic blood or bone marrow aspirate samples were screened for sensitivity in response to 78 dose conditions by measuring the reduction in leukemic blasts relative to the control. In pediatric patients treated with conventional chemotherapy, comprising cytarabine, daunorubicin and etoposide (ADE), ex vivo drug sensitivity results correlated with minimal residual disease (r = 0.63) and one year relapse-free survival (r = 0.70; AUROC = 0.94). In the de novo ADE analysis cohort of 13 patients, AML cells showed greater sensitivity to bortezomib/panobinostat compared with ADE, and comparable sensitivity between venetoclax/azacitidine and ADE ex vivo. Two patients showed a differential response between ADE and bortezomib/panobinostat, thus supporting the incorporation of ex vivo drug sensitivity testing in clinical trials to further evaluate the predictive utility of this platform in children with AML.
Collapse
Affiliation(s)
| | | | | | | | | | - George Yam
- Notable Labs, Foster City, CA 94404, USA
| | | | | | | | - Maci Terrell
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Raushan Rashid
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alan K. Gonzalez
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hailey H. Oviedo
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Michelle C. Alozie
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Tamilini Ilangovan
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | - Alexandra M. Stevens
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Correspondence:
| |
Collapse
|
18
|
Wang H, Chan KYY, Cheng CK, Ng MH, Lee PY, Cheng FWT, Lam GKS, Chow TW, Ha SY, Chiang AK, Leung WH, Leung AY, Wang CC, Zhang T, Zhang XB, So CC, Yuen YP, Sun Q, Zhang C, Xu Y, Cheung JTK, Ng WH, Tang PMK, Kang W, To KF, Lee WYW, Wong RS, Poon ENY, Zhao Q, Huang J, Chen C, Yuen PMP, Li CK, Leung AWK, Leung KT. Pharmacogenomic Profiling of Pediatric Acute Myeloid Leukemia to Identify Therapeutic Vulnerabilities and Inform Functional Precision Medicine. Blood Cancer Discov 2022; 3:516-535. [PMID: 35960210 PMCID: PMC9894568 DOI: 10.1158/2643-3230.bcd-22-0011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 05/31/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
Despite the expanding portfolio of targeted therapies for adults with acute myeloid leukemia (AML), direct implementation in children is challenging due to inherent differences in underlying genetics. Here we established the pharmacologic profile of pediatric AML by screening myeloblast sensitivity to approved and investigational agents, revealing candidates of immediate clinical relevance. Drug responses ex vivo correlated with patient characteristics, exhibited age-specific alterations, and concorded with activities in xenograft models. Integration with genomic data uncovered new gene-drug associations, suggesting actionable therapeutic vulnerabilities. Transcriptome profiling further identified gene-expression signatures associated with on- and off-target drug responses. We also demonstrated the feasibility of drug screening-guided treatment for children with high-risk AML, with two evaluable cases achieving remission. Collectively, this study offers a high-dimensional gene-drug clinical data set that could be leveraged to research the unique biology of pediatric AML and sets the stage for realizing functional precision medicine for the clinical management of the disease. SIGNIFICANCE We conducted integrated drug and genomic profiling of patient biopsies to build the functional genomic landscape of pediatric AML. Age-specific differences in drug response and new gene-drug interactions were identified. The feasibility of functional precision medicine-guided management of children with high-risk AML was successfully demonstrated in two evaluable clinical cases. This article is highlighted in the In This Issue feature, p. 476.
Collapse
Affiliation(s)
- Han Wang
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Kathy Yuen Yee Chan
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Chi Keung Cheng
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Margaret H.L. Ng
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Po Yi Lee
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Frankie Wai Tsoi Cheng
- Department of Paediatrics and Adolescent Medicine, Hong Kong Children's Hospital, Kowloon, Hong Kong
| | - Grace Kee See Lam
- Department of Paediatrics and Adolescent Medicine, Hong Kong Children's Hospital, Kowloon, Hong Kong
| | - Tin Wai Chow
- Department of Paediatrics and Adolescent Medicine, Hong Kong Children's Hospital, Kowloon, Hong Kong
| | - Shau Yin Ha
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Alan K.S. Chiang
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Wing Hang Leung
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Anskar Y.H. Leung
- Department of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Chi Chiu Wang
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Tao Zhang
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Xiao-Bing Zhang
- Department of Medicine, Loma Linda University, Loma Linda, California
| | - Chi Chiu So
- Department of Pathology, Hong Kong Children's Hospital, Kowloon, Hong Kong
| | - Yuet Ping Yuen
- Department of Pathology, Hong Kong Children's Hospital, Kowloon, Hong Kong
| | - Qiwei Sun
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Chi Zhang
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Yaqun Xu
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - John Tak Kit Cheung
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Wing Hei Ng
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Patrick Ming-Kuen Tang
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Ka-Fai To
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Wayne Yuk Wai Lee
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Raymond S.M. Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Ellen Ngar Yun Poon
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Qi Zhao
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Junbin Huang
- Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Chun Chen
- Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Patrick Man Pan Yuen
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Chi-kong Li
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong.,Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Shatin, Hong Kong.,Corresponding Authors: Kam Tong Leung, E-mail: ; Chi-kong Li, Hong Kong Children's Hospital, 1 Shing Cheong Road, Kowloon Bay, Kowloon, Hong Kong. Phone: 852-3513-3176; Fax: 852-2636-0020; E-mail: ; and Alex Wing Kwan Leung, E-mail:
| | - Alex Wing Kwan Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong.,Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Shatin, Hong Kong.,Corresponding Authors: Kam Tong Leung, E-mail: ; Chi-kong Li, Hong Kong Children's Hospital, 1 Shing Cheong Road, Kowloon Bay, Kowloon, Hong Kong. Phone: 852-3513-3176; Fax: 852-2636-0020; E-mail: ; and Alex Wing Kwan Leung, E-mail:
| | - Kam Tong Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong.,Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Shatin, Hong Kong.,Corresponding Authors: Kam Tong Leung, E-mail: ; Chi-kong Li, Hong Kong Children's Hospital, 1 Shing Cheong Road, Kowloon Bay, Kowloon, Hong Kong. Phone: 852-3513-3176; Fax: 852-2636-0020; E-mail: ; and Alex Wing Kwan Leung, E-mail:
| |
Collapse
|
19
|
Ding D, Zheng R, Tian Y, Jimenez R, Hou X, Weroha SJ, Wang L, Shi L, Huang H. Retinoblastoma protein as an intrinsic BRD4 inhibitor modulates small molecule BET inhibitor sensitivity in cancer. Nat Commun 2022; 13:6311. [PMID: 36274096 PMCID: PMC9588789 DOI: 10.1038/s41467-022-34024-y] [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: 02/11/2022] [Accepted: 10/07/2022] [Indexed: 12/25/2022] Open
Abstract
Bromodomain and extraterminal (BET) proteins including BRD4 play important roles in oncogenesis and immune inflammation. Here we demonstrate that cancer cells with loss of the retinoblastoma (RB) tumor suppressor became resistant to small molecule bromodomain inhibitors of BET proteins. We find that RB binds to bromodomain-1 (BD1) of BRD4, but binding is impeded by CDK4/6-mediated RB phosphorylation at serine-249/threonine-252 (S249/T252). ChIP-seq analysis shows RB knockdown increases BRD4 occupancy at genomic loci of genes enriched in cancer-related pathways including the GPCR-GNBIL-CREB axis. S249/T252-phosphorylated RB positively correlates with GNBIL protein level in prostate cancer patient samples. BET inhibitor resistance in RB-deficient cells is abolished by co-administration of CREB inhibitor. Our study identifies RB protein as a bona fide intrinsic inhibitor of BRD4 and demonstrates that RB inactivation confers resistance to small molecule BET inhibitors, thereby revealing a regulatory hub that converges RB upstream signaling onto BRD4 functions in diseases such as cancer.
Collapse
Affiliation(s)
- Donglin Ding
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA
- Department of Urology, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA
| | - Rongbin Zheng
- Basic and Translational Research Division, Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Ye Tian
- Department of Urology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Rafael Jimenez
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA
| | - Xiaonan Hou
- Divison of Oncology, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA
| | - Saravut J Weroha
- Divison of Oncology, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA
| | - Liguo Wang
- Divison of Medical Informatics and Statistics, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA
| | - Lei Shi
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA.
- Department of Radiation Oncology, Cancer Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou, 310000, China.
| | - Haojie Huang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA.
- Department of Urology, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA.
- Mayo Clinic Cancer Center, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA.
| |
Collapse
|
20
|
Bottomly D, Long N, Schultz AR, Kurtz SE, Tognon CE, Johnson K, Abel M, Agarwal A, Avaylon S, Benton E, Blucher A, Borate U, Braun TP, Brown J, Bryant J, Burke R, Carlos A, Chang BH, Cho HJ, Christy S, Coblentz C, Cohen AM, d'Almeida A, Cook R, Danilov A, Dao KHT, Degnin M, Dibb J, Eide CA, English I, Hagler S, Harrelson H, Henson R, Ho H, Joshi SK, Junio B, Kaempf A, Kosaka Y, Laderas T, Lawhead M, Lee H, Leonard JT, Lin C, Lind EF, Liu SQ, Lo P, Loriaux MM, Luty S, Maxson JE, Macey T, Martinez J, Minnier J, Monteblanco A, Mori M, Morrow Q, Nelson D, Ramsdill J, Rofelty A, Rogers A, Romine KA, Ryabinin P, Saultz JN, Sampson DA, Savage SL, Schuff R, Searles R, Smith RL, Spurgeon SE, Sweeney T, Swords RT, Thapa A, Thiel-Klare K, Traer E, Wagner J, Wilmot B, Wolf J, Wu G, Yates A, Zhang H, Cogle CR, Collins RH, Deininger MW, Hourigan CS, Jordan CT, Lin TL, Martinez ME, Pallapati RR, Pollyea DA, Pomicter AD, Watts JM, Weir SJ, Druker BJ, McWeeney SK, Tyner JW. Integrative analysis of drug response and clinical outcome in acute myeloid leukemia. Cancer Cell 2022; 40:850-864.e9. [PMID: 35868306 PMCID: PMC9378589 DOI: 10.1016/j.ccell.2022.07.002] [Citation(s) in RCA: 131] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 05/30/2022] [Accepted: 06/30/2022] [Indexed: 12/17/2022]
Abstract
Acute myeloid leukemia (AML) is a cancer of myeloid-lineage cells with limited therapeutic options. We previously combined ex vivo drug sensitivity with genomic, transcriptomic, and clinical annotations for a large cohort of AML patients, which facilitated discovery of functional genomic correlates. Here, we present a dataset that has been harmonized with our initial report to yield a cumulative cohort of 805 patients (942 specimens). We show strong cross-cohort concordance and identify features of drug response. Further, deconvoluting transcriptomic data shows that drug sensitivity is governed broadly by AML cell differentiation state, sometimes conditionally affecting other correlates of response. Finally, modeling of clinical outcome reveals a single gene, PEAR1, to be among the strongest predictors of patient survival, especially for young patients. Collectively, this report expands a large functional genomic resource, offers avenues for mechanistic exploration and drug development, and reveals tools for predicting outcome in AML.
Collapse
Affiliation(s)
- Daniel Bottomly
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR 97239, USA; Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Nicola Long
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Anna Reister Schultz
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Stephen E Kurtz
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Cristina E Tognon
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Kara Johnson
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Melissa Abel
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Anupriya Agarwal
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA; Department of Molecular & Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA; Division of Oncologic Sciences, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Sammantha Avaylon
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Erik Benton
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR 97239, USA; Oregon Clinical and Translational Research Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Aurora Blucher
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR 97239, USA; Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Uma Borate
- Division of Hematology, Department of Internal Medicine, James Cancer Center, Ohio State University, Columbus, OH 43210, USA
| | - Theodore P Braun
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jordana Brown
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jade Bryant
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Russell Burke
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Amy Carlos
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Integrated Genomics Laboratory, Oregon Health & Science University, Portland, OR 97239, USA
| | - Bill H Chang
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology and Oncology, Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239, USA
| | - Hyun Jun Cho
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Stephen Christy
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Cody Coblentz
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Aaron M Cohen
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR 97239, USA; Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Oregon Clinical and Translational Research Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Amanda d'Almeida
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Rachel Cook
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Alexey Danilov
- Department of Hematology and Hematopoietic Stem Cell Transplant, City of Hope National Medical Center, Duarte, CA 91010, USA
| | | | - Michie Degnin
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - James Dibb
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Christopher A Eide
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Isabel English
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Stuart Hagler
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR 97239, USA; Oregon Clinical and Translational Research Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Heath Harrelson
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR 97239, USA; Oregon Clinical and Translational Research Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Rachel Henson
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Integrated Genomics Laboratory, Oregon Health & Science University, Portland, OR 97239, USA
| | - Hibery Ho
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Sunil K Joshi
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Brian Junio
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Andy Kaempf
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Biostatistics Shared Resource, Oregon Health & Science University, Portland, OR 97239, USA
| | - Yoko Kosaka
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR 97239, USA
| | | | - Matt Lawhead
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR 97239, USA; Oregon Clinical and Translational Research Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Hyunjung Lee
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jessica T Leonard
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Chenwei Lin
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Integrated Genomics Laboratory, Oregon Health & Science University, Portland, OR 97239, USA
| | - Evan F Lind
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Selina Qiuying Liu
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Pierrette Lo
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Marc M Loriaux
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Department of Pathology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Samuel Luty
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Julia E Maxson
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Oncologic Sciences, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Tara Macey
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jacqueline Martinez
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jessica Minnier
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Biostatistics Shared Resource, Oregon Health & Science University, Portland, OR 97239, USA; OHSU-PSU School of Public Health, VA Portland Health Care System, Oregon Health & Science University, Portland, OR 97239, USA
| | - Andrea Monteblanco
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Motomi Mori
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Quinlan Morrow
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Dylan Nelson
- High-Throughput Screening Services Laboratory, Oregon State University, Corvallis, OR 97331, USA
| | - Justin Ramsdill
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR 97239, USA; Oregon Clinical and Translational Research Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Angela Rofelty
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Alexandra Rogers
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Kyle A Romine
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Peter Ryabinin
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR 97239, USA; Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jennifer N Saultz
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - David A Sampson
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Samantha L Savage
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | | | - Robert Searles
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Integrated Genomics Laboratory, Oregon Health & Science University, Portland, OR 97239, USA
| | - Rebecca L Smith
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Stephen E Spurgeon
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Tyler Sweeney
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Ronan T Swords
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Aashis Thapa
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Karina Thiel-Klare
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Elie Traer
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jake Wagner
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Beth Wilmot
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR 97239, USA; Oregon Clinical and Translational Research Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Joelle Wolf
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Guanming Wu
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR 97239, USA; Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Oregon Clinical and Translational Research Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Amy Yates
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR 97239, USA; Oregon Clinical and Translational Research Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Haijiao Zhang
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Oncologic Sciences, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Christopher R Cogle
- Department of Medicine, Division of Hematology and Oncology, University of Florida, Gainesville, FL 32610, USA
| | - Robert H Collins
- Department of Internal Medicine/ Hematology Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390-8565, USA
| | - Michael W Deininger
- Division of Hematology & Hematologic Malignancies, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Christopher S Hourigan
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20814-1476, USA
| | - Craig T Jordan
- Division of Hematology, University of Colorado, Denver, CO 80045, USA
| | - Tara L Lin
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas, Kansas City, KS 66205, USA
| | - Micaela E Martinez
- Clinical Research Services, University of Miami Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA
| | - Rachel R Pallapati
- Clinical Research Services, University of Miami Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA
| | - Daniel A Pollyea
- Division of Hematology, University of Colorado, Denver, CO 80045, USA
| | - Anthony D Pomicter
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Justin M Watts
- Division of Hematology, Department of Medicine, University of Miami Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA
| | - Scott J Weir
- Department of Cancer Biology, Division of Medical Oncology, Department of Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Brian J Druker
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA.
| | - Shannon K McWeeney
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR 97239, USA; Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Oregon Clinical and Translational Research Institute, Oregon Health & Science University, Portland, OR 97239, USA.
| | - Jeffrey W Tyner
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA; Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
21
|
Gosline SJC, Tognon C, Nestor M, Joshi S, Modak R, Damnernsawad A, Posso C, Moon J, Hansen JR, Hutchinson-Bunch C, Pino JC, Gritsenko MA, Weitz KK, Traer E, Tyner J, Druker B, Agarwal A, Piehowski P, McDermott JE, Rodland K. Proteomic and phosphoproteomic measurements enhance ability to predict ex vivo drug response in AML. Clin Proteomics 2022; 19:30. [PMID: 35896960 PMCID: PMC9327422 DOI: 10.1186/s12014-022-09367-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 06/22/2022] [Indexed: 11/23/2022] Open
Abstract
Acute Myeloid Leukemia (AML) affects 20,000 patients in the US annually with a five-year survival rate of approximately 25%. One reason for the low survival rate is the high prevalence of clonal evolution that gives rise to heterogeneous sub-populations of leukemic cells with diverse mutation spectra, which eventually leads to disease relapse. This genetic heterogeneity drives the activation of complex signaling pathways that is reflected at the protein level. This diversity makes it difficult to treat AML with targeted therapy, requiring custom patient treatment protocols tailored to each individual's leukemia. Toward this end, the Beat AML research program prospectively collected genomic and transcriptomic data from over 1000 AML patients and carried out ex vivo drug sensitivity assays to identify genomic signatures that could predict patient-specific drug responses. However, there are inherent weaknesses in using only genetic and transcriptomic measurements as surrogates of drug response, particularly the absence of direct information about phosphorylation-mediated signal transduction. As a member of the Clinical Proteomic Tumor Analysis Consortium, we have extended the molecular characterization of this cohort by collecting proteomic and phosphoproteomic measurements from a subset of these patient samples (38 in total) to evaluate the hypothesis that proteomic signatures can improve the ability to predict response to 26 drugs in AML ex vivo samples. In this work we describe our systematic, multi-omic approach to evaluate proteomic signatures of drug response and compare protein levels to other markers of drug response such as mutational patterns. We explore the nuances of this approach using two drugs that target key pathways activated in AML: quizartinib (FLT3) and trametinib (Ras/MEK), and show how patient-derived signatures can be interpreted biologically and validated in cell lines. In conclusion, this pilot study demonstrates strong promise for proteomics-based patient stratification to assess drug sensitivity in AML.
Collapse
Affiliation(s)
| | - Cristina Tognon
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | | | - Sunil Joshi
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Rucha Modak
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Alisa Damnernsawad
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
- Department of Biology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Camilo Posso
- Pacific Northwest National Laboratory, Seattle, WA, USA
| | - Jamie Moon
- Pacific Northwest National Laboratory, Seattle, WA, USA
| | | | | | - James C Pino
- Pacific Northwest National Laboratory, Seattle, WA, USA
| | | | - Karl K Weitz
- Pacific Northwest National Laboratory, Seattle, WA, USA
| | - Elie Traer
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Jeffrey Tyner
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Brian Druker
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Anupriya Agarwal
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
- Division of Oncological Sciences, Oregon Health & Science University, Portland, OR, USA
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | | | - Jason E McDermott
- Pacific Northwest National Laboratory, Seattle, WA, USA
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Karin Rodland
- Pacific Northwest National Laboratory, Seattle, WA, USA.
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
22
|
Rice WG, Howell SB, Zhang H, Rastgoo N, Local A, Kurtz SE, Lo P, Bottomly D, Wilmot B, McWeeney SK, Druker BJ, Tyner JW. Luxeptinib (CG-806) Targets FLT3 and Clusters of Kinases Operative in Acute Myeloid Leukemia. Mol Cancer Ther 2022; 21:1125-1135. [PMID: 35499387 PMCID: PMC9256809 DOI: 10.1158/1535-7163.mct-21-0832] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 01/25/2022] [Accepted: 04/20/2022] [Indexed: 11/16/2022]
Abstract
Luxeptinib (CG-806) simultaneously targets FLT3 and select other kinase pathways operative in myeloid malignancies. We investigated the range of kinases it inhibits, its cytotoxicity landscape ex vivo with acute myeloid leukemia (AML) patient samples, and its efficacy in xenograft models. Luxeptinib inhibits wild-type (WT) and many of the clinically relevant mutant forms of FLT3 at low nanomolar concentrations. It is a more potent inhibitor of the activity of FLT3-internal tandem duplication, FLT3 kinase domain and gatekeeper mutants than against WT FLT3. Broad kinase screens disclosed that it also inhibits other kinases that can drive oncogenic signaling and rescue pathways, but spares kinases known to be associated with clinical toxicity. In vitro profiling of luxeptinib against 186 AML fresh patient samples demonstrated greater potency relative to other FLT3 inhibitors, including cases with mutations in FLT3, isocitrate dehydrogenase-1/2, ASXL1, NPM1, SRSF2, TP53, or RAS, and activity was documented in a xenograft AML model. Luxeptinib administered continuously orally every 12 hours at a dose that yielded a mean Cmin plasma concentration of 1.0 ± 0.3 μmol/L (SEM) demonstrated strong antitumor activity but no myelosuppression or evidence of tissue damage in mice or dogs in acute toxicology studies. On the basis of these studies, luxeptinib was advanced into a phase I trial for patients with AML and myelodysplastic/myeloproliferative neoplasms.
Collapse
Affiliation(s)
| | - Stephen B. Howell
- Department of Medicine and the Moores Cancer Center, University of California, San Diego, California
| | | | | | | | - Stephen E. Kurtz
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon.,Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon
| | - Pierrette Lo
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon.,Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon
| | - Daniel Bottomly
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon.,Division of Bioinformatics and Computational Biology, Oregon Health & Science University, Portland, Oregon
| | - Beth Wilmot
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon.,Division of Bioinformatics and Computational Biology, Oregon Health & Science University, Portland, Oregon
| | - Shannon K. McWeeney
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon.,Division of Bioinformatics and Computational Biology, Oregon Health & Science University, Portland, Oregon
| | - Brian J. Druker
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon.,Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon.,Corresponding Author: Brian J. Druker, Oregon Health & Science University, 3181 SW Sam Jackson Park Road CR 145 & L592, Portland, OR 97239. Phone: 503-494-5596; Fax: 503-494-3688; E-mail:
| | - Jeffrey W. Tyner
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon.,Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon.,Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
23
|
Baran N, Lodi A, Dhungana Y, Herbrich S, Collins M, Sweeney S, Pandey R, Skwarska A, Patel S, Tremblay M, Kuruvilla VM, Cavazos A, Kaplan M, Warmoes MO, Veiga DT, Furudate K, Rojas-Sutterin S, Haman A, Gareau Y, Marinier A, Ma H, Harutyunyan K, Daher M, Garcia LM, Al-Atrash G, Piya S, Ruvolo V, Yang W, Shanmugavelandy SS, Feng N, Gay J, Du D, Yang JJ, Hoff FW, Kaminski M, Tomczak K, Eric Davis R, Herranz D, Ferrando A, Jabbour EJ, Emilia Di Francesco M, Teachey DT, Horton TM, Kornblau S, Rezvani K, Sauvageau G, Gagea M, Andreeff M, Takahashi K, Marszalek JR, Lorenzi PL, Yu J, Tiziani S, Hoang T, Konopleva M. Inhibition of mitochondrial complex I reverses NOTCH1-driven metabolic reprogramming in T-cell acute lymphoblastic leukemia. Nat Commun 2022; 13:2801. [PMID: 35589701 PMCID: PMC9120040 DOI: 10.1038/s41467-022-30396-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 04/25/2022] [Indexed: 01/05/2023] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is commonly driven by activating mutations in NOTCH1 that facilitate glutamine oxidation. Here we identify oxidative phosphorylation (OxPhos) as a critical pathway for leukemia cell survival and demonstrate a direct relationship between NOTCH1, elevated OxPhos gene expression, and acquired chemoresistance in pre-leukemic and leukemic models. Disrupting OxPhos with IACS-010759, an inhibitor of mitochondrial complex I, causes potent growth inhibition through induction of metabolic shut-down and redox imbalance in NOTCH1-mutated and less so in NOTCH1-wt T-ALL cells. Mechanistically, inhibition of OxPhos induces a metabolic reprogramming into glutaminolysis. We show that pharmacological blockade of OxPhos combined with inducible knock-down of glutaminase, the key glutamine enzyme, confers synthetic lethality in mice harboring NOTCH1-mutated T-ALL. We leverage on this synthetic lethal interaction to demonstrate that IACS-010759 in combination with chemotherapy containing L-asparaginase, an enzyme that uncovers the glutamine dependency of leukemic cells, causes reduced glutaminolysis and profound tumor reduction in pre-clinical models of human T-ALL. In summary, this metabolic dependency of T-ALL on OxPhos provides a rational therapeutic target.
Collapse
Affiliation(s)
- Natalia Baran
- grid.240145.60000 0001 2291 4776Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Alessia Lodi
- grid.89336.370000 0004 1936 9924Department of Nutritional Sciences, Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX USA
| | - Yogesh Dhungana
- grid.240871.80000 0001 0224 711XSt. Jude Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Shelley Herbrich
- grid.240145.60000 0001 2291 4776Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Meghan Collins
- grid.89336.370000 0004 1936 9924Department of Nutritional Sciences, Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX USA
| | - Shannon Sweeney
- grid.89336.370000 0004 1936 9924Department of Nutritional Sciences, Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX USA
| | - Renu Pandey
- grid.89336.370000 0004 1936 9924Department of Nutritional Sciences, Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX USA
| | - Anna Skwarska
- grid.240145.60000 0001 2291 4776Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Shraddha Patel
- grid.240145.60000 0001 2291 4776Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Mathieu Tremblay
- grid.14848.310000 0001 2292 3357Institute for Research in Immunology and Cancer, The University of Montreal, Montréal, QC Canada
| | - Vinitha Mary Kuruvilla
- grid.240145.60000 0001 2291 4776Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Antonio Cavazos
- grid.240145.60000 0001 2291 4776Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Mecit Kaplan
- grid.240145.60000 0001 2291 4776Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Marc O. Warmoes
- grid.240145.60000 0001 2291 4776Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Diogo Troggian Veiga
- grid.249880.f0000 0004 0374 0039The Jackson Laboratory for Genomic Medicine, Farmington, CT USA
| | - Ken Furudate
- grid.240145.60000 0001 2291 4776Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX USA ,grid.257016.70000 0001 0673 6172Department of Oral and Maxillofacial Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori Japan
| | - Shanti Rojas-Sutterin
- grid.14848.310000 0001 2292 3357Institute for Research in Immunology and Cancer, The University of Montreal, Montréal, QC Canada
| | - Andre Haman
- grid.14848.310000 0001 2292 3357Institute for Research in Immunology and Cancer, The University of Montreal, Montréal, QC Canada
| | - Yves Gareau
- grid.14848.310000 0001 2292 3357Institute for Research in Immunology and Cancer, The University of Montreal, Montréal, QC Canada
| | - Anne Marinier
- grid.14848.310000 0001 2292 3357Institute for Research in Immunology and Cancer, The University of Montreal, Montréal, QC Canada
| | - Helen Ma
- grid.240145.60000 0001 2291 4776Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Karine Harutyunyan
- grid.240145.60000 0001 2291 4776Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - May Daher
- grid.240145.60000 0001 2291 4776Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Luciana Melo Garcia
- grid.240145.60000 0001 2291 4776Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Gheath Al-Atrash
- grid.240145.60000 0001 2291 4776Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Sujan Piya
- grid.240145.60000 0001 2291 4776Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Vivian Ruvolo
- grid.240145.60000 0001 2291 4776Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Wentao Yang
- grid.240871.80000 0001 0224 711XDepartment of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Sriram Saravanan Shanmugavelandy
- grid.240145.60000 0001 2291 4776Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Ningping Feng
- grid.240145.60000 0001 2291 4776TRACTION Platform, Therapeutics Discovery Division, University of Texas M. D. Anderson Cancer Center, Houston, USA
| | - Jason Gay
- grid.240145.60000 0001 2291 4776TRACTION Platform, Therapeutics Discovery Division, University of Texas M. D. Anderson Cancer Center, Houston, USA
| | - Di Du
- grid.240145.60000 0001 2291 4776Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Jun J. Yang
- grid.240871.80000 0001 0224 711XDepartment of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Fieke W. Hoff
- grid.240145.60000 0001 2291 4776Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Marcin Kaminski
- grid.240871.80000 0001 0224 711XDepartment of Immunology, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Katarzyna Tomczak
- grid.240145.60000 0001 2291 4776Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - R. Eric Davis
- grid.240145.60000 0001 2291 4776Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Daniel Herranz
- grid.430387.b0000 0004 1936 8796Rutgers Robert Wood Johnson Medical School, Cancer Institute of New Jersey, New Brunswick, NJ USA
| | - Adolfo Ferrando
- grid.21729.3f0000000419368729Irving Cancer Research Center, Columbia University Irving Medical Center, New York, NY USA
| | - Elias J. Jabbour
- grid.240145.60000 0001 2291 4776Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - M. Emilia Di Francesco
- grid.240145.60000 0001 2291 4776Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - David T. Teachey
- grid.25879.310000 0004 1936 8972Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA USA
| | - Terzah M. Horton
- grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Baylor College of Medicine, Houston, TX USA
| | - Steven Kornblau
- grid.240145.60000 0001 2291 4776Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Katayoun Rezvani
- grid.240145.60000 0001 2291 4776Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Guy Sauvageau
- grid.14848.310000 0001 2292 3357Institute for Research in Immunology and Cancer, The University of Montreal, Montréal, QC Canada
| | - Mihai Gagea
- grid.240145.60000 0001 2291 4776Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Michael Andreeff
- grid.240145.60000 0001 2291 4776Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Koichi Takahashi
- grid.240145.60000 0001 2291 4776Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Joseph R. Marszalek
- grid.240145.60000 0001 2291 4776TRACTION Platform, Therapeutics Discovery Division, University of Texas M. D. Anderson Cancer Center, Houston, USA
| | - Philip L. Lorenzi
- grid.240145.60000 0001 2291 4776Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Jiyang Yu
- grid.240871.80000 0001 0224 711XDepartment of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Stefano Tiziani
- grid.89336.370000 0004 1936 9924Department of Nutritional Sciences, Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX USA
| | - Trang Hoang
- grid.14848.310000 0001 2292 3357Institute for Research in Immunology and Cancer, The University of Montreal, Montréal, QC Canada ,grid.14848.310000 0001 2292 3357Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC Canada
| | - Marina Konopleva
- grid.240145.60000 0001 2291 4776Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| |
Collapse
|
24
|
Ianevski A, Giri AK, Aittokallio T. SynergyFinder 3.0: an interactive analysis and consensus interpretation of multi-drug synergies across multiple samples. Nucleic Acids Res 2022; 50:W739-W743. [PMID: 35580060 PMCID: PMC9252834 DOI: 10.1093/nar/gkac382] [Citation(s) in RCA: 248] [Impact Index Per Article: 82.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/16/2022] [Accepted: 04/29/2022] [Indexed: 11/26/2022] Open
Abstract
SynergyFinder (https://synergyfinder.fimm.fi) is a free web-application for interactive analysis and visualization of multi-drug combination response data. Since its first release in 2017, SynergyFinder has become a popular tool for multi-dose combination data analytics, partly because the development of its functionality and graphical interface has been driven by a diverse user community, including both chemical biologists and computational scientists. Here, we describe the latest upgrade of this community-effort, SynergyFinder release 3.0, introducing a number of novel features that support interactive multi-sample analysis of combination synergy, a novel consensus synergy score that combines multiple synergy scoring models, and an improved outlier detection functionality that eliminates false positive results, along with many other post-analysis options such as weighting of synergy by drug concentrations and distinguishing between different modes of synergy (potency and efficacy). Based on user requests, several additional improvements were also implemented, including new data visualizations and export options for multi-drug combinations. With these improvements, SynergyFinder 3.0 supports robust identification of consistent combinatorial synergies for multi-drug combinatorial discovery and clinical translation.
Collapse
Affiliation(s)
- Aleksandr Ianevski
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Finland.,Helsinki Institute for Information Technology (HIIT), Aalto University, Finland
| | - Anil K Giri
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Finland.,Foundation for the Finnish Cancer Institute (FCI), University of Helsinki, Finland
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Finland.,Helsinki Institute for Information Technology (HIIT), Aalto University, Finland.,Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital, Norway.,Centre for Biostatistics and Epidemiology (OCBE), Faculty of Medicine, University of Oslo, Norway
| |
Collapse
|
25
|
Xiang W, Lam YH, Periyasamy G, Chuah C. Application of High Throughput Technologies in the Development of Acute Myeloid Leukemia Therapy: Challenges and Progress. Int J Mol Sci 2022; 23:ijms23052863. [PMID: 35270002 PMCID: PMC8910862 DOI: 10.3390/ijms23052863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/26/2022] [Accepted: 02/28/2022] [Indexed: 11/27/2022] Open
Abstract
Acute myeloid leukemia (AML) is a complex hematological malignancy characterized by extensive heterogeneity in genetics, response to therapy and long-term outcomes, making it a prototype example of development for personalized medicine. Given the accessibility to hematologic malignancy patient samples and recent advances in high-throughput technologies, large amounts of biological data that are clinically relevant for diagnosis, risk stratification and targeted drug development have been generated. Recent studies highlight the potential of implementing genomic-based and phenotypic-based screens in clinics to improve survival in patients with refractory AML. In this review, we will discuss successful applications as well as challenges of most up-to-date high-throughput technologies, including artificial intelligence (AI) approaches, in the development of personalized medicine for AML, and recent clinical studies for evaluating the utility of integrating genomics-guided and drug sensitivity testing-guided treatment approaches for AML patients.
Collapse
Affiliation(s)
- Wei Xiang
- Department of Haematology, Singapore General Hospital, Singapore 169608, Singapore; (W.X.); (Y.H.L.)
| | - Yi Hui Lam
- Department of Haematology, Singapore General Hospital, Singapore 169608, Singapore; (W.X.); (Y.H.L.)
| | - Giridharan Periyasamy
- High Throughput Phenomics Platform, Experimental Drug Development Centre, Agency for Science, Technology and Research (A*STAR), Singapore 139632, Singapore;
| | - Charles Chuah
- Department of Haematology, Singapore General Hospital, Singapore 169608, Singapore; (W.X.); (Y.H.L.)
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore 169857, Singapore
- Correspondence:
| |
Collapse
|
26
|
Letai A. Functional Precision Medicine: Putting Drugs on Patient Cancer Cells and Seeing What Happens. Cancer Discov 2022; 12:290-292. [PMID: 35140175 PMCID: PMC8852353 DOI: 10.1158/2159-8290.cd-21-1498] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
For too long, assays exposing patient tumor cells to drugs to identify active therapies have been dismissed as ineffective. In this issue of Cancer Discovery, two groups independently demonstrate clinical utility of such functional precision medicine assays in hematologic malignancies.See related article by Kornauth et al., p. 372.See related article by Malani et al., p. 388.
Collapse
Affiliation(s)
- Anthony Letai
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
27
|
Associating Drug Sensitivity with Differentiation Status Identifies Effective Combinations for Acute Myeloid Leukemia. Blood Adv 2022; 6:3062-3067. [PMID: 35078224 PMCID: PMC9131911 DOI: 10.1182/bloodadvances.2021006307] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 01/12/2022] [Indexed: 11/20/2022] Open
Abstract
Combined targeting of AML patient cells with p38 MAPK and BCL2 inhibitors overcomes monocytic-associated resistance to VEN. Exploiting complementary drug sensitivity profiles with respect to leukemic differentiation state affords enhanced efficacy in AML.
Using ex vivo drug screening of primary patient specimens, we identified the combination of the p38 MAPK inhibitor doramapimod (DORA) with the BCL2 inhibitor venetoclax (VEN) as demonstrating broad, enhanced efficacy compared with each single agent across 335 acute myeloid leukemia (AML) patient samples while sparing primary stromal cells. Single-agent DORA and VEN sensitivity was associated with distinct, nonoverlapping tumor cell differentiation states. In particular, increased monocytes, M4/M5 French-American-British classification, and CD14+ immunophenotype tracked with sensitivity to DORA and resistance to VEN but were mitigated with the combination. Increased expression of MAPK14 and BCL2, the respective primary targets of DORA and VEN, were observed in monocytic and undifferentiated leukemias, respectively. Enrichment for DORA and VEN sensitivities was observed in AML with monocyte-like and progenitor-like transcriptomic signatures, respectively, and these associations diminished with the combination. The mechanism underlying the combination’s enhanced efficacy may result from inhibition of p38 MAPK-mediated phosphorylation of BCL2, which in turn enhances sensitivity to VEN. These findings suggest exploiting complementary drug sensitivity profiles with respect to leukemic differentiation state, such as dual targeting of p38 MAPK and BCL2, offers opportunity for broad, enhanced efficacy across the clinically challenging heterogeneous landscape of AML.
Collapse
|
28
|
Mengxuan S, Fen Z, Runming J. Novel Treatments for Pediatric Relapsed or Refractory Acute B-Cell Lineage Lymphoblastic Leukemia: Precision Medicine Era. Front Pediatr 2022; 10:923419. [PMID: 35813376 PMCID: PMC9259965 DOI: 10.3389/fped.2022.923419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/02/2022] [Indexed: 12/05/2022] Open
Abstract
With the markedly increased cure rate for children with newly diagnosed pediatric B-cell acute lymphoblastic leukemia (B-ALL), relapse and refractory B-ALL (R/R B-ALL) remain the primary cause of death worldwide due to the limitations of multidrug chemotherapy. As we now have a more profound understanding of R/R ALL, including the mechanism of recurrence and drug resistance, prognostic indicators, genotypic changes and so on, we can use newly emerging technologies to identify operational molecular targets and find sensitive drugs for individualized treatment. In addition, more promising and innovative immunotherapies and molecular targeted drugs that are expected to kill leukemic cells more effectively while maintaining low toxicity to achieve minimal residual disease (MRD) negativity and better bridge hematopoietic stem cell transplantation (HSCT) have also been widely developed. To date, the prognosis of pediatric patients with R/R B-ALL has been enhanced markedly thanks to the development of novel drugs. This article reviews the new advancements of several promising strategies for pediatric R/R B-ALL.
Collapse
Affiliation(s)
- Shang Mengxuan
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhou Fen
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jin Runming
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
29
|
Nykänen N, Mäkelä R, Arjonen A, Härmä V, Lewandowski L, Snowden E, Blaesius R, Jantunen I, Kuopio T, Kononen J, Rantala JK. Ex Vivo Drug Screening Informed Targeted Therapy for Metastatic Parotid Squamous Cell Carcinoma. Front Oncol 2021; 11:735820. [PMID: 34604070 PMCID: PMC8481915 DOI: 10.3389/fonc.2021.735820] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 08/23/2021] [Indexed: 11/13/2022] Open
Abstract
The purpose of ex vivo drug screening in the context of precision oncology is to serve as a functional diagnostic method for therapy efficacy modeling directly on patient-derived tumor cells. Here, we report a case study using integrated multiomics ex vivo drug screening approach to assess therapy efficacy in a rare metastatic squamous cell carcinoma of the parotid gland. Tumor cells isolated from lymph node metastasis and distal subcutaneous metastasis were used for imaging-based single-cell resolution drug screening and reverse-phase protein array-based drug screening assays to inform the treatment strategy after standard therapeutic options had been exhausted. The drug targets discovered on the basis of the ex vivo measured drug efficacy were validated with histopathology, genomic profiling, and in vitro cell biology methods, and targeted treatments with durable clinical responses were achieved. These results demonstrate the use of serial ex vivo drug screening to inform adjuvant therapy options prior to and during treatment and highlight HER2 as a potential therapy target also in metastatic squamous cell carcinoma of the salivary glands.
Collapse
Affiliation(s)
| | | | | | - Ville Härmä
- Misvik Biology Oy, Turku, Finland.,Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | | | - Eileen Snowden
- Genomic Sciences, BD Technologies, Research Triangle Park, Durham, NC, United States
| | - Rainer Blaesius
- Genomic Sciences, BD Technologies, Research Triangle Park, Durham, NC, United States
| | - Ismo Jantunen
- Central Finland Health Care District, Jyväskylä, Finland
| | - Teijo Kuopio
- Central Finland Health Care District, Jyväskylä, Finland.,Department of Biological and Environmental Science, Jyväskylä, Finland
| | | | - Juha K Rantala
- Misvik Biology Oy, Turku, Finland.,Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
30
|
Romine KA, Nechiporuk T, Bottomly D, Jeng S, McWeeney SK, Kaempf A, Corces MR, Majeti R, Tyner JW. Monocytic differentiation and AHR signaling as Primary Nodes of BET Inhibitor Response in Acute Myeloid Leukemia. Blood Cancer Discov 2021; 2:518-531. [PMID: 34568834 DOI: 10.1158/2643-3230.bcd-21-0012] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
To understand mechanisms of response to BET inhibitors (BETi), we mined the Beat AML functional genomic dataset and performed genome-wide CRISPR screens on BETi- sensitive and BETi- resistant AML cells. Both strategies revealed regulators of monocytic differentiation, SPI1, JUNB, FOS, and aryl-hydrocarbon receptor signaling (AHR/ARNT), as determinants of BETi response. AHR activation synergized with BETi while inhibition antagonized BETi-mediated cytotoxicity. Consistent with BETi sensitivity dependence on monocytic differentiation, ex vivo sensitivity to BETi in primary AML patient samples correlated with higher expression of monocytic markers CSF1R, LILRs, and VCAN. In addition, HL-60 cell line differentiation enhanced its sensitivity to BETi. Further, screens to rescue BETi sensitivity identified BCL2 and CDK6 as druggable vulnerabilities. Finally, monocytic AML patient samples refractory to venetoclax ex vivo were significantly more sensitive to combined BETi + venetoclax. Together, our work highlights mechanisms that could predict BETi response and identifies combination strategies to overcome resistance.
Collapse
Affiliation(s)
- Kyle A Romine
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA.,Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Tamilla Nechiporuk
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA.,Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Daniel Bottomly
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.,Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Portland, OR, USA
| | - Sophia Jeng
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.,Oregon Clinical and Translational Research Institute, Portland, OR, USA
| | - Shannon K McWeeney
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.,Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Portland, OR, USA.,Oregon Clinical and Translational Research Institute, Portland, OR, USA
| | - Andy Kaempf
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.,Biostatistics Shared Resource, Portland, OR, USA
| | - M Ryan Corces
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA.,Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Ravindra Majeti
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jeffrey W Tyner
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA.,Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.,Division of Hematology & Medical Oncology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
31
|
Liu T, Lam V, Thieme E, Sun D, Wang X, Xu F, Wang L, Danilova OV, Xia Z, Tyner JW, Kurtz SE, Danilov AV. Pharmacologic Targeting of Mcl-1 Induces Mitochondrial Dysfunction and Apoptosis in B-Cell Lymphoma Cells in a TP53- and BAX-Dependent Manner. Clin Cancer Res 2021; 27:4910-4922. [PMID: 34233959 DOI: 10.1158/1078-0432.ccr-21-0464] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 05/17/2021] [Accepted: 06/30/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Bcl-2 has been effectively targeted in lymphoid malignancies. However, resistance is inevitable, and novel approaches to target mitochondrial apoptosis are necessary. AZD5991, a selective BH3-mimetic in clinical trials, inhibits Mcl-1 with high potency. EXPERIMENTAL DESIGN We explored the preclinical activity of AZD5991 in diffuse large B-cell lymphoma (DLBCL) and ibrutinib-resistant mantle cell lymphoma (MCL) cell lines, MCL patient samples, and mice bearing DLBCL and MCL xenografts using flow cytometry, immunoblotting, and Seahorse respirometry assay. Cas9 gene editing and ex vivo functional drug screen assays helped identify mechanisms of resistance to Mcl-1 inhibition. RESULTS Mcl-1 was expressed in DLBCL and MCL cell lines and primary tumors. Treatment with AZD5991 restricted growth of DLBCL cells independent of cell of origin and overcame ibrutinib resistance in MCL cells. Mcl-1 inhibition led to mitochondrial dysfunction as manifested by mitochondrial membrane depolarization, decreased mitochondrial mass, and induction of mitophagy. This was accompanied by impairment of oxidative phosphorylation. TP53 and BAX were essential for sensitivity to Mcl-1, and oxidative phosphorylation was implicated in resistance to Mcl-1 inhibition. Induction of prosurvival proteins (e.g., Bcl-xL) in stromal conditions that mimic the tumor microenvironment rendered protection of primary MCL cells from Mcl-1 inhibition, while BH3-mimetics targeting Bcl-2/xL sensitized lymphoid cells to AZD5991. Treatment with AZD5991 reduced tumor growth in murine lymphoma models and prolonged survival of MCL PDX mice. CONCLUSIONS Selective targeting Mcl-1 is a promising therapeutic approach in lymphoid malignancies. TP53 apoptotic network and metabolic reprogramming underlie susceptibility to Mcl-1 inhibition.
Collapse
Affiliation(s)
- Tingting Liu
- City of Hope National Medical Center, Duarte, California
| | - Vi Lam
- City of Hope National Medical Center, Duarte, California
| | - Elana Thieme
- City of Hope National Medical Center, Duarte, California
| | - Duanchen Sun
- Oregon Health and Science University, Portland, Oregon
| | - Xiaoguang Wang
- City of Hope National Medical Center, Duarte, California
| | - Fei Xu
- Oregon Health and Science University, Portland, Oregon
| | - Lili Wang
- City of Hope National Medical Center, Duarte, California
| | | | - Zheng Xia
- Oregon Health and Science University, Portland, Oregon
| | | | | | | |
Collapse
|
32
|
Antileukemic efficacy of a potent artemisinin combined with sorafenib and venetoclax. Blood Adv 2021; 5:711-724. [PMID: 33560385 DOI: 10.1182/bloodadvances.2020003429] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 12/28/2020] [Indexed: 12/21/2022] Open
Abstract
Artemisinins are active against human leukemia cell lines and have low clinical toxicity in worldwide use as antimalarials. Because multiagent combination regimens are necessary to cure fully evolved leukemias, we sought to leverage our previous finding that artemisinin analogs synergize with kinase inhibitors, including sorafenib (SOR), by identifying additional synergistic antileukemic drugs with low toxicity. Screening of a targeted antineoplastic drug library revealed that B-cell lymphoma 2 (BCL2) inhibitors synergize with artemisinins, and validation assays confirmed that the selective BCL2 inhibitor, venetoclax (VEN), synergized with artemisinin analogs to inhibit growth and induce apoptotic cell death of multiple acute leukemia cell lines in vitro. An oral 3-drug "SAV" regimen (SOR plus the potent artemisinin-derived trioxane diphenylphosphate 838 dimeric analog [ART838] plus VEN) killed leukemia cell lines and primary cells in vitro. Leukemia cells cultured in ART838 had decreased induced myeloid leukemia cell differentiation protein (MCL1) levels and increased levels of DNA damage-inducible transcript 3 (DDIT3; GADD153) messenger RNA and its encoded CCATT/enhancer-binding protein homologous protein (CHOP), a key component of the integrated stress response. Thus, synergy of the SAV combination may involve combined targeting of MCL1 and BCL2 via discrete, tolerable mechanisms, and cellular levels of MCL1 and DDIT3/CHOP may serve as biomarkers for action of artemisinins and SAV. Finally, SAV treatment was tolerable and resulted in deep responses with extended survival in 2 acute myeloid leukemia (AML) cell line xenograft models, both harboring a mixed lineage leukemia gene rearrangement and an FMS-like receptor tyrosine kinase-3 internal tandem duplication, and inhibited growth in 2 AML primagraft models.
Collapse
|
33
|
Ex vivo drug screening defines novel drug sensitivity patterns for informing personalized therapy in myeloid neoplasms. Blood Adv 2021; 4:2768-2778. [PMID: 32569379 DOI: 10.1182/bloodadvances.2020001934] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/18/2020] [Indexed: 12/11/2022] Open
Abstract
Precision medicine approaches such as ex vivo drug sensitivity screening (DSS) are appealing to inform rational drug selection in myelodysplastic syndromes (MDSs) and acute myeloid leukemia, given their marked biologic heterogeneity. We evaluated a novel, fully automated ex vivo DSS platform that uses high-throughput flow cytometry in 54 patients with newly diagnosed or treatment-refractory myeloid neoplasms to evaluate sensitivity (blast cytotoxicity and differentiation) to 74 US Food and Drug Administration-approved or investigational drugs and 36 drug combinations. After piloting the platform in 33 patients, we conducted a prospective feasibility study enrolling 21 patients refractory to hypomethylating agents (HMAs) to determine whether this assay could be performed within a clinically actionable time frame and could accurately predict clinical responses in vivo. When assayed for cytotoxicity, ex vivo drug sensitivity patterns were heterogeneous, but they defined distinct patient clusters with differential sensitivity to HMAs, anthracyclines, histone deacetylase inhibitors, and kinase inhibitors (P < .001 among clusters) and demonstrated synergy between HMAs and venetoclax (P < .01 for combinations vs single agents). In our feasibility study, ex vivo DSS results were available at a median of 15 days after bone marrow biopsy, and they informed personalized therapy, which frequently included venetoclax combinations, kinase inhibitors, differentiative agents, and androgens. In 21 patients with available ex vivo and in vivo clinical response data, the DSS platform had a positive predictive value of 0.92, negative predictive value of 0.82, and overall accuracy of 0.85. These data demonstrate the utility of this approach for identifying potentially useful and often novel therapeutic drugs for patients with myeloid neoplasms refractory to standard therapies.
Collapse
|
34
|
Panina SB, Pei J, Kirienko NV. Mitochondrial metabolism as a target for acute myeloid leukemia treatment. Cancer Metab 2021; 9:17. [PMID: 33883040 PMCID: PMC8058979 DOI: 10.1186/s40170-021-00253-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/30/2021] [Indexed: 02/06/2023] Open
Abstract
Acute myeloid leukemias (AML) are a group of aggressive hematologic malignancies resulting from acquired genetic mutations in hematopoietic stem cells that affect patients of all ages. Despite decades of research, standard chemotherapy still remains ineffective for some AML subtypes and is often inappropriate for older patients or those with comorbidities. Recently, a number of studies have identified unique mitochondrial alterations that lead to metabolic vulnerabilities in AML cells that may present viable treatment targets. These include mtDNA, dependency on oxidative phosphorylation, mitochondrial metabolism, and pro-survival signaling, as well as reactive oxygen species generation and mitochondrial dynamics. Moreover, some mitochondria-targeting chemotherapeutics and their combinations with other compounds have been FDA-approved for AML treatment. Here, we review recent studies that illuminate the effects of drugs and synergistic drug combinations that target diverse biomolecules and metabolic pathways related to mitochondria and their promise in experimental studies, clinical trials, and existing chemotherapeutic regimens.
Collapse
Affiliation(s)
| | - Jingqi Pei
- Department of BioSciences, Rice University, Houston, TX, USA
| | | |
Collapse
|
35
|
Pollyea DA, Pei S, Stevens BM, Smith CA, Jordan CT. The Intriguing Clinical Success of BCL-2 Inhibition in Acute Myeloid Leukemia. ANNUAL REVIEW OF CANCER BIOLOGY 2021. [DOI: 10.1146/annurev-cancerbio-060220-124048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Over the past several decades numerous preclinical and clinical studies have pursued new approaches for the treatment of acute myeloid leukemia (AML). While some degree of clinical response has been demonstrated for many therapies, for the most part, fundamental changes in the treatment landscape have been lacking. Recently, the use of the BCL-2 inhibitor venetoclax has emerged as a potent therapy for a majority of newly diagnosed AML patients. Venetoclax regimens have shown broad response rates with deep and durable remissions, with a superior toxicity profile compared with traditional intensive chemotherapy agents. Numerous ongoing studies are now using venetoclax in combination with a wide range of other agents as investigators seek even more effective and well-tolerated regimens. Notably, however, while the empirical results of BCL-2 inhibition are encouraging, the mechanisms that have led to these successful clinical outcomes remain unclear. Intriguingly, the activity of venetoclax in AML patients appears to go beyond simply modulating canonical antiapoptosis mechanisms; in addition, the efficacy of venetoclax is linked to its combined use with conventional low-intensity backbone therapies. This article will evaluate the state of the field, provide a summary of key considerations, and propose directions for future studies.
Collapse
Affiliation(s)
- Daniel A. Pollyea
- Division of Hematology, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Shanshan Pei
- Division of Hematology, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Brett M. Stevens
- Division of Hematology, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Clayton A. Smith
- Division of Hematology, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Craig T. Jordan
- Division of Hematology, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| |
Collapse
|
36
|
Valent P, Orfao A, Kubicek S, Staber P, Haferlach T, Deininger M, Kollmann K, Lion T, Virgolini I, Winter G, Hantschel O, Kenner L, Zuber J, Grebien F, Moriggl R, Hoermann G, Hermine O, Andreeff M, Bock C, Mughal T, Constantinescu SN, Kralovics R, Sexl V, Skoda R, Superti-Furga G, Jäger U. Precision Medicine in Hematology 2021: Definitions, Tools, Perspectives, and Open Questions. Hemasphere 2021; 5:e536. [PMID: 33623882 PMCID: PMC7892291 DOI: 10.1097/hs9.0000000000000536] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 12/16/2020] [Indexed: 12/20/2022] Open
Abstract
During the past few years, our understanding of molecular mechanisms and cellular interactions relevant to malignant blood cell disorders has improved substantially. New insights include a detailed knowledge about disease-initiating exogenous factors, endogenous (genetic, somatic, epigenetic) elicitors or facilitators of disease evolution, and drug actions and interactions that underlie efficacy and adverse event profiles in defined cohorts of patients. As a result, precision medicine and personalized medicine are rapidly growing new disciplines that support the clinician in making the correct diagnosis, in predicting outcomes, and in optimally selecting patients for interventional therapies. In addition, precision medicine tools are greatly facilitating the development of new drugs, therapeutic approaches, and new multiparametric prognostic scoring models. However, although the emerging roles of precision medicine and personalized medicine in hematology and oncology are clearly visible, several questions remain. For example, it remains unknown how precision medicine tools can be implemented in healthcare systems and whether all possible approaches are also affordable. In addition, there is a need to define terminologies and to relate these to specific and context-related tools and strategies in basic and applied science. To discuss these issues, a working conference was organized in September 2019. The outcomes of this conference are summarized herein and include a proposal for definitions, terminologies, and applications of precision and personalized medicine concepts and tools in hematologic neoplasms. We also provide proposals aimed at reducing costs, thereby making these applications affordable in daily practice.
Collapse
Affiliation(s)
- Peter Valent
- Department of Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Austria
| | - Alberto Orfao
- Servicio Central de Citometria, Centro de Investigacion del Cancer (IBMCC; CSIC/USAL), IBSAL, CIBERONC and Department of Medicine, University of Salamanca, Spain
| | - Stefan Kubicek
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Philipp Staber
- Department of Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Austria
| | | | - Michael Deininger
- Division of Hematology and Hematologic Malignancies, University of Utah, Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | - Karoline Kollmann
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Austria
| | - Thomas Lion
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Austria
- Children’s Cancer Research Institute, Vienna, Austria
| | - Irene Virgolini
- Department of Nuclear Medicine, Medical University of Innsbruck, Austria
| | - Georg Winter
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Oliver Hantschel
- Institute of Physiological Chemistry, Faculty of Medicine, Philipps-University of Marburg, Germany
| | - Lukas Kenner
- Pathology of Laboratory Animals, University of Veterinary Medicine, Vienna, Austria
| | - Johannes Zuber
- Research Institute of Molecular Pathology (IMP), Vienna, Austria
| | - Florian Grebien
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Austria
| | - Richard Moriggl
- Institute of Animal Breeding and Genetics, Unit for Functional Cancer Genomics, University of Veterinary Medicine Vienna, Austria
| | - Gregor Hoermann
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Austria
- MLL Munich Leukemia Laboratory, Munich, Germany
| | - Olivier Hermine
- Imagine Institute Université Paris Descartes, Sorbonne, Paris Cité, Paris, France
- Department of Hematology, Necker Hospital, Paris, France
| | - Michael Andreeff
- University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Tariq Mughal
- Division of Hematology & Oncology, Tufts University Medical Center, Boston, Massachusetts, USA
| | - Stefan N. Constantinescu
- de Duve Institute and Ludwig Cancer Research Brussels, Université catholique de Louvain, Brussels, Belgium
| | - Robert Kralovics
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Austria
| | - Radek Skoda
- Departement of Biomedicine, University of Basel, Switzerland
| | - Giulio Superti-Furga
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Ulrich Jäger
- Department of Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Austria
| |
Collapse
|
37
|
Akimov Y, Aittokallio T. Re-defining synthetic lethality by phenotypic profiling for precision oncology. Cell Chem Biol 2021; 28:246-256. [PMID: 33631125 DOI: 10.1016/j.chembiol.2021.01.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 12/28/2020] [Accepted: 01/28/2021] [Indexed: 12/31/2022]
Abstract
High-throughput functional and genomic screening techniques provide systematic means for phenotypic discovery. Using synthetic lethality (SL) as a paradigm for anticancer drug and target discovery, we describe how these screening technologies may offer new possibilities to identify therapeutically relevant and selective SL interactions by addressing some of the challenges that have made robust discovery of SL candidates difficult. We further introduce an extended concept of SL interaction, in which a simultaneous perturbation of two or more cellular components reduces cell viability more than expected by their individual effects, which we feel is highly befitting for anticancer applications. We also highlight the potential benefits and challenges related to computational quantification of synergistic interactions and cancer selectivity. Finally, we explore how tumoral heterogeneity can be exploited to find phenotype-specific SL interactions for precision oncology using high-throughput functional screening and the exciting opportunities these methods provide for the identification of subclonal SL interactions.
Collapse
Affiliation(s)
- Yevhen Akimov
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland; Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital, Oslo, Norway; Centre for Biostatistics and Epidemiology (OCBE), Faculty of Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
38
|
The delta isoform of phosphatidylinositol-3-kinase predominates in chronic myelomonocytic leukemia and can be targeted effectively with umbralisib and ruxolitinib. Exp Hematol 2021; 97:57-65.e5. [PMID: 33617893 DOI: 10.1016/j.exphem.2021.02.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 11/20/2022]
Abstract
Chronic myelomonocytic leukemia (CMML) is a myelodysplastic syndrome/myeloproliferative neoplasm overlap syndrome characterized by monocytic proliferation in the presence of dysplastic bone marrow changes, inflammatory symptoms, and propensity for transformation to acute myeloid leukemia (AML), with a poor prognosis and limited treatment options. Unlike the α and β isoforms, the phosphatidylinositol-3-kinase (PI3K)-δ signaling protein is predominantly expressed by hematopoietic cells and therefore has garnered interest as a potential target for the treatment of lymphomas and leukemias. We revealed a pattern of increased PIK3CD:PIK3CA ratio in monocytic M5 AML patients and cell lines, and this ratio correlated with responsiveness to pharmacological PI3K-δ inhibition in vitro. Because CMML is a disease defined by monocytic clonal proliferation, we tested the PI3K-δ inhibitor umbralisib as a single agent and in combination with the JAK1/2 inhibitor ruxolitinib, in CMML. Our ex vivo experiments with primary CMML patient samples revealed synergistic inhibition of viability and clonogenicity with this combination. Phospho-specific flow cytometry revealed that dual inhibition had the unique ability to decrease STAT5, ERK, AKT, and S6 phosphorylation simultaneously, which offers a mechanistic hypothesis for the enhanced efficacy of the combination treatment. These preclinical data indicate promising activity by co-inhibition of PI3K-δ and JAK1/2 and support the use of ruxolitinib + umbralisib combination therapy in CMML under active clinical investigation.
Collapse
|
39
|
Pikman Y, Tasian SK, Sulis ML, Stevenson K, Blonquist TM, Apsel Winger B, Cooper TM, Pauly M, Maloney KW, Burke MJ, Brown PA, Gossai N, McNeer JL, Shukla NN, Cole PD, Kahn JM, Chen J, Barth MJ, Magee JA, Gennarini L, Adhav AA, Clinton CM, Ocasio-Martinez N, Gotti G, Li Y, Lin S, Imamovic A, Tognon CE, Patel T, Faust HL, Contreras CF, Cremer A, Cortopassi WA, Garrido Ruiz D, Jacobson MP, Dharia NV, Su A, Robichaud AL, Saur Conway A, Tarlock K, Stieglitz E, Place AE, Puissant A, Hunger SP, Kim AS, Lindeman NI, Gore L, Janeway KA, Silverman LB, Tyner JW, Harris MH, Loh ML, Stegmaier K. Matched Targeted Therapy for Pediatric Patients with Relapsed, Refractory, or High-Risk Leukemias: A Report from the LEAP Consortium. Cancer Discov 2021; 11:1424-1439. [PMID: 33563661 DOI: 10.1158/2159-8290.cd-20-0564] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 11/25/2020] [Accepted: 01/14/2021] [Indexed: 11/16/2022]
Abstract
Despite a remarkable increase in the genomic profiling of cancer, integration of genomic discoveries into clinical care has lagged behind. We report the feasibility of rapid identification of targetable mutations in 153 pediatric patients with relapsed/refractory or high-risk leukemias enrolled on a prospective clinical trial conducted by the LEAP Consortium. Eighteen percent of patients had a high confidence Tier 1 or 2 recommendation. We describe clinical responses in the 14% of patients with relapsed/refractory leukemia who received the matched targeted therapy. Further, in order to inform future targeted therapy for patients, we validated variants of uncertain significance, performed ex vivo drug-sensitivity testing in patient leukemia samples, and identified new combinations of targeted therapies in cell lines and patient-derived xenograft models. These data and our collaborative approach should inform the design of future precision medicine trials. SIGNIFICANCE: Patients with relapsed/refractory leukemias face limited treatment options. Systematic integration of precision medicine efforts can inform therapy. We report the feasibility of identifying targetable mutations in children with leukemia and describe correlative biology studies validating therapeutic hypotheses and novel mutations.See related commentary by Bornhauser and Bourquin, p. 1322.This article is highlighted in the In This Issue feature, p. 1307.
Collapse
Affiliation(s)
- Yana Pikman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts
| | - Sarah K Tasian
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics and Abramson Cancer Center at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Maria Luisa Sulis
- Division of Pediatric Hematology/Oncology/Stem Cell Transplantation, Columbia University Irving Medical Center, New York, New York
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kristen Stevenson
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Traci M Blonquist
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Beth Apsel Winger
- Department of Pediatrics, Division of Hematology/Oncology, Benioff Children's Hospital and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Todd M Cooper
- Seattle Children's Hospital, Cancer and Blood Disorders Center, Seattle, Washington
| | - Melinda Pauly
- Division of Hematology/Oncology, Emory University, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Kelly W Maloney
- Children's Hospital Colorado, University of Colorado Cancer Center, Aurora, Colorado
| | - Michael J Burke
- Medical College of Wisconsin, Children's Hospital of Wisconsin, Milwaukee, Wisconsin
| | | | - Nathan Gossai
- Center for Cancer and Blood Disorders, Children's Minnesota, Minneapolis, Minnesota
| | | | - Neerav N Shukla
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Peter D Cole
- Children's Hospital at Montefiore, Bronx, New York
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Justine M Kahn
- Division of Pediatric Hematology/Oncology/Stem Cell Transplantation, Columbia University Irving Medical Center, New York, New York
| | - Jing Chen
- Division of Pediatric Hematology/Oncology/Stem Cell Transplantation, Columbia University Irving Medical Center, New York, New York
- Children's Cancer Institute, Joseph M. Sanzari Children's Hospital, Hackensack University Medical Center, Hackensack, New Jersey
| | | | - Jeffrey A Magee
- Division of Pediatric Hematology/Oncology, Washington University/St. Louis Children's Hospital, St. Louis, Missouri
| | | | - Asmani A Adhav
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Catherine M Clinton
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Giacomo Gotti
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Yuting Li
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Shan Lin
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Alma Imamovic
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts
| | - Cristina E Tognon
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Tasleema Patel
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Haley L Faust
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Cristina F Contreras
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Anjali Cremer
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- University Hospital Frankfurt, Department of Hematology/Oncology, Frankfurt/Main, Germany
| | - Wilian A Cortopassi
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California
| | - Diego Garrido Ruiz
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California
| | - Matthew P Jacobson
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California
| | - Neekesh V Dharia
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts
| | - Angela Su
- INSERM UMR 944, IRSL, St Louis Hospital, Paris, France
| | - Amanda L Robichaud
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Amy Saur Conway
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Katherine Tarlock
- Seattle Children's Hospital, Cancer and Blood Disorders Center, Seattle, Washington
| | - Elliot Stieglitz
- Department of Pediatrics, Division of Hematology/Oncology, Benioff Children's Hospital and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Andrew E Place
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts
| | | | - Stephen P Hunger
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics and Abramson Cancer Center at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Annette S Kim
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Neal I Lindeman
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Lia Gore
- Children's Hospital Colorado, University of Colorado Cancer Center, Aurora, Colorado
| | - Katherine A Janeway
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts
| | - Lewis B Silverman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts
| | - Jeffrey W Tyner
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Marian H Harris
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts
| | - Mignon L Loh
- Department of Pediatrics, Division of Hematology/Oncology, Benioff Children's Hospital and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Kimberly Stegmaier
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts
| |
Collapse
|
40
|
Ianevski A, Lahtela J, Javarappa KK, Sergeev P, Ghimire BR, Gautam P, Vähä-Koskela M, Turunen L, Linnavirta N, Kuusanmäki H, Kontro M, Porkka K, Heckman CA, Mattila P, Wennerberg K, Giri AK, Aittokallio T. Patient-tailored design for selective co-inhibition of leukemic cell subpopulations. SCIENCE ADVANCES 2021; 7:eabe4038. [PMID: 33608276 PMCID: PMC7895436 DOI: 10.1126/sciadv.abe4038] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 01/08/2021] [Indexed: 06/12/2023]
Abstract
The extensive drug resistance requires rational approaches to design personalized combinatorial treatments that exploit patient-specific therapeutic vulnerabilities to selectively target disease-driving cell subpopulations. To solve the combinatorial explosion challenge, we implemented an effective machine learning approach that prioritizes patient-customized drug combinations with a desired synergy-efficacy-toxicity balance by combining single-cell RNA sequencing with ex vivo single-agent testing in scarce patient-derived primary cells. When applied to two diagnostic and two refractory acute myeloid leukemia (AML) patient cases, each with a different genetic background, we accurately predicted patient-specific combinations that not only resulted in synergistic cancer cell co-inhibition but also were capable of targeting specific AML cell subpopulations that emerge in differing stages of disease pathogenesis or treatment regimens. Our functional precision oncology approach provides an unbiased means for systematic identification of personalized combinatorial regimens that selectively co-inhibit leukemic cells while avoiding inhibition of nonmalignant cells, thereby increasing their likelihood for clinical translation.
Collapse
Affiliation(s)
- Aleksandr Ianevski
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Helsinki Institute for Information Technology (HIIT), Department of Computer Science, Aalto University, Espoo, Finland
| | - Jenni Lahtela
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Komal K Javarappa
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Philipp Sergeev
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Bishwa R Ghimire
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Prson Gautam
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Markus Vähä-Koskela
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Laura Turunen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Nora Linnavirta
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Heikki Kuusanmäki
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Biotech Research and Innovation Centre (BRIC) and Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Mika Kontro
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Kimmo Porkka
- Helsinki University Hospital Comprehensive Cancer Center, Hematology Research Unit Helsinki, iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Caroline A Heckman
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Pirkko Mattila
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Krister Wennerberg
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland.
- Biotech Research and Innovation Centre (BRIC) and Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
| | - Anil K Giri
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland.
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland.
- Helsinki Institute for Information Technology (HIIT), Department of Computer Science, Aalto University, Espoo, Finland
- Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital, Oslo, Norway
- Centre for Biostatistics and Epidemiology (OCBE), Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
41
|
Affiliation(s)
- John S Welch
- Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
42
|
Bohannan Z, Pudupakam RS, Koo J, Horwitz H, Tsang J, Polley A, Han EJ, Fernandez E, Park S, Swartzfager D, Qi NSX, Tu C, Rankin WV, Thamm DH, Lee HR, Lim S. Predicting likelihood of in vivo chemotherapy response in canine lymphoma using ex vivo drug sensitivity and immunophenotyping data in a machine learning model. Vet Comp Oncol 2020; 19:160-171. [PMID: 33025640 PMCID: PMC7894155 DOI: 10.1111/vco.12656] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/24/2020] [Accepted: 10/02/2020] [Indexed: 02/06/2023]
Abstract
We report a precision medicine platform that evaluates the probability of chemotherapy drug efficacy for canine lymphoma by combining ex vivo chemosensitivity and immunophenotyping assays with computational modelling. We isolated live cancer cells from fresh fine needle aspirates of affected lymph nodes and collected post‐treatment clinical responses in 261 canine lymphoma patients scheduled to receive at least 1 of 5 common chemotherapy agents (doxorubicin, vincristine, cyclophosphamide, lomustine and rabacfosadine). We used flow cytometry analysis for immunophenotyping and ex vivo chemosensitivity testing. For each drug, 70% of treated patients were randomly selected to train a random forest model to predict the probability of positive Veterinary Cooperative Oncology Group (VCOG) clinical response based on input variables including antigen expression profiles and treatment sensitivity readouts for each patient's cancer cells. The remaining 30% of patients were used to test model performance. Most models showed a test set ROC‐AUC > 0.65, and all models had overall ROC‐AUC > 0.95. Predicted response scores significantly distinguished (P < .001) positive responses from negative responses in B‐cell and T‐cell disease and newly diagnosed and relapsed patients. Patient groups with predicted response scores >50% showed a statistically significant reduction (log‐rank P < .05) in time to complete response when compared to the groups with scores <50%. The computational models developed in this study enabled the conversion of ex vivo cell‐based chemosensitivity assay results into a predicted probability of in vivo therapeutic efficacy, which may help improve treatment outcomes of individual canine lymphoma patients by providing predictive estimates of positive treatment response.
Collapse
Affiliation(s)
| | | | - Jamin Koo
- ImpriMed, Inc., Palo Alto, California, USA.,ImpriMed Korea, Inc., Seoul, Republic of Korea.,Department of Chemical Engineering, Hongik University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | - Chantal Tu
- SAGE Veterinary Centers, Dublin, California, USA
| | | | - Douglas H Thamm
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, USA
| | - Hye-Ryeon Lee
- ImpriMed, Inc., Palo Alto, California, USA.,ImpriMed Korea, Inc., Seoul, Republic of Korea
| | - Sungwon Lim
- ImpriMed, Inc., Palo Alto, California, USA.,ImpriMed Korea, Inc., Seoul, Republic of Korea
| |
Collapse
|
43
|
Mason CC, Fiol CR, Baker MJ, Nadal-Melsio E, Yebra-Fernandez E, Bicalho L, Chowdhury A, Albert M, Reid AG, Claudiani S, Apperley JF, Khorashad JS. Identification of genetic targets in acute myeloid leukaemia for designing targeted therapy. Br J Haematol 2020; 192:137-145. [PMID: 33022753 DOI: 10.1111/bjh.17129] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/03/2020] [Accepted: 09/04/2020] [Indexed: 01/12/2023]
Abstract
Few effective therapies exist for acute myeloid leukaemia (AML), in part due to the molecular heterogeneity of this disease. We sought to identify genes crucial to deregulated AML signal transduction pathways which, if inhibited, could effectively eradicate leukaemia stem cells. Due to difficulties in screening primary cells, most previous studies have performed next-generation sequencing (NGS) library knockdown screens in cell lines. Using carefully considered methods including evaluation at multiple timepoints to ensure equitable gene knockdown, we employed a large NGS short hairpin RNA (shRNA) knockdown screen of nearly 5 000 genes in primary AML cells from six patients to identify genes that are crucial for leukaemic survival. Across various levels of stringency, genome-wide bioinformatic analysis identified a gene in the NOX family, NOX1, to have the most consistent knockdown effectiveness in primary cells (P = 5∙39 × 10-5 , Bonferroni-adjusted), impacting leukaemia cell survival as the top-ranked gene for two of the six AML patients and also showing high effectiveness in three of the other four patients. Further investigation of this pathway highlighted NOX2 as the member of the NOX family with clear knockdown efficacy. We conclude that genes in the NOX family are enticing candidates for therapeutic development in AML.
Collapse
Affiliation(s)
- Clinton C Mason
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, University of Utah, Salt Lake City, UT, USA
| | | | - Monika J Baker
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, University of Utah, Salt Lake City, UT, USA
| | - Elisabet Nadal-Melsio
- SIHMDS North West London Pathology, Imperial College Healthcare NHS Trust, London, UK
| | - Eva Yebra-Fernandez
- SIHMDS North West London Pathology, Imperial College Healthcare NHS Trust, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Cerella C, Dicato M, Diederich M. BH3 Mimetics in AML Therapy: Death and Beyond? Trends Pharmacol Sci 2020; 41:793-814. [PMID: 33032835 DOI: 10.1016/j.tips.2020.09.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 09/01/2020] [Accepted: 09/10/2020] [Indexed: 12/16/2022]
Abstract
B cell lymphoma 2 (BCL2) homology domain 3 (BH3) mimetics are targeted therapeutic agents that allow response prediction and patient stratification. BH3 mimetics are prototypical activators of the mitochondrial death program in cancer. They emerged as important modulators of cellular mechanisms contributing to poor therapeutic responses, including cancer cell stemness, cancer-specific metabolic routes, paracrine signaling to the tumor microenvironment, and immune modulation. We present an overview of the antagonism between BH3 mimetics and antiapoptotic BCL2 proteins. We focus on acute myeloid leukemia (AML), a cancer with reduced therapeutic options that have recently been improved by BH3 mimetics.
Collapse
Affiliation(s)
- Claudia Cerella
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, L-2540 Luxembourg, Luxembourg
| | - Mario Dicato
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, L-2540 Luxembourg, Luxembourg
| | - Marc Diederich
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 151-742, South Korea.
| |
Collapse
|
45
|
Megías-Vericat JE, Martínez-Cuadrón D, Solana-Altabella A, Montesinos P. Precision medicine in acute myeloid leukemia: where are we now and what does the future hold? Expert Rev Hematol 2020; 13:1057-1065. [PMID: 32869672 DOI: 10.1080/17474086.2020.1818559] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Precision medicine has revolutionized the diagnostic and therapeutic management of acute myeloid leukemia (AML), from standardized schemes based on chemotherapy to tailored approaches according to molecular and genetic profile and targeted therapy. AREAS COVERED The main topics of precision medicine in AML were reviewed in MEDLINE, EMBASE, and Cochrane Central Register databases, and future directions in this therapeutic area were addressed. This review included targeted therapies, drug-sensitivity tests and predictive biomarkers, and genetic studies employing pharmacogenetic and deep sequencing strategies. EXPERT OPINION Precision medicine has opened the door to personalized therapy for specific AML patient populations with promising results. Several targeted therapies have been approved or are being tested for specific mutations (i.e. FLT3, IDH, BCL-2, TP53), obtaining improvements in clinical outcomes and less toxicity as compared with intensive treatment, allowing potential combination therapy. Ongoing trials and real data will establish the role of these molecules in monotherapy or combined in different AML settings (front-line, relapsed/refractory, or post-transplant). Experience in drug-sensitivity predictors and pharmacogenetic biomarkers is encouraging and could be useful tools in the next years, but we need a better understanding of AML biology and pathogenesis as well as confirmatory studies to demonstrate the utility in clinical practice.
Collapse
Affiliation(s)
| | - David Martínez-Cuadrón
- Servicio de Hematología y Hemoterapia, Hospital Universitari i Politècnic La Fe , Valencia, Spain.,CIBERONC, Instituto de Salud Carlos III , Madrid, Spain
| | - Antonio Solana-Altabella
- Servicio de Farmacia, Área del Medicamento, Hospital Universitari i Politècnic La Fe , Valencia, Spain
| | - Pau Montesinos
- Servicio de Hematología y Hemoterapia, Hospital Universitari i Politècnic La Fe , Valencia, Spain.,CIBERONC, Instituto de Salud Carlos III , Madrid, Spain
| |
Collapse
|
46
|
Hellström-Lindberg E, Tobiasson M, Greenberg P. Myelodysplastic syndromes: moving towards personalized management. Haematologica 2020; 105:1765-1779. [PMID: 32439724 PMCID: PMC7327628 DOI: 10.3324/haematol.2020.248955] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 04/24/2020] [Indexed: 02/06/2023] Open
Abstract
The myelodysplastic syndromes (MDS) share their origin in the hematopoietic stem cell but have otherwise very heterogeneous biological and genetic characteristics. Clinical features are dominated by cytopenia and a substantial risk for progression to acute myeloid leukemia. According to the World Health Organization, MDS is defined by cytopenia, bone marrow dysplasia and certain karyotypic abnormalities. The understanding of disease pathogenesis has undergone major development with the implementation of next-generation sequencing and a closer integration of morphology, cytogenetics and molecular genetics is currently paving the way for improved classification and prognostication. True precision medicine is still in the future for MDS and the development of novel therapeutic compounds with a propensity to markedly change patients' outcome lags behind that for many other blood cancers. Treatment of higher-risk MDS is dominated by monotherapy with hypomethylating agents but novel combinations are currently being evaluated in clinical trials. Agents that stimulate erythropoiesis continue to be first-line treatment for the anemia of lower-risk MDS but luspatercept has shown promise as second-line therapy for sideroblastic MDS and lenalidomide is an established second-line treatment for del(5q) lower-risk MDS. The only potentially curative option for MDS is hematopoietic stem cell transplantation, until recently associated with a relatively high risk of transplant-related mortality and relapse. However, recent studies show increased cure rates due to better tools to target the malignant clone with less toxicity. This review provides a comprehensive overview of the current status of the clinical evaluation, biology and therapeutic interventions for this spectrum of disorders.
Collapse
Affiliation(s)
- Eva Hellström-Lindberg
- Karolinska Institutet, Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska University Hospital, Stockholm, Sweden
| | - Magnus Tobiasson
- Karolinska Institutet, Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska University Hospital, Stockholm, Sweden
| | - Peter Greenberg
- Stanford Cancer Institute, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
47
|
Yong S, Kim J, Chung JY, Ra S, kim SS, Kim Y. Heme Oxygenase 1-Targeted Hybrid Nanoparticle for Chemo- and Immuno-Combination Therapy in Acute Myelogenous Leukemia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2000487. [PMID: 32670766 PMCID: PMC7341080 DOI: 10.1002/advs.202000487] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/29/2020] [Indexed: 05/02/2023]
Abstract
Acute myelogenous leukemia (AML) is a fatal blood cancer with high patient mortality. Daunorubicin and cytarabine are first-line chemotherapy for AML, with bone marrow transplantation in most cases. Recently, cancer immunotherapy has been challenged in AML and leukemia-niche myeloid cells are promising targets for the AML immunotherapy. Heme oxygenase 1 (HO1) is an antioxidative and cytoprotective enzyme inducing chemo-resistant AML and has been focused as an immune checkpoint molecule in tumor microenvironments. Herein, lipid-polymer hybrid nanoparticle (hNP) is loaded with tin mesoporphyrin (SnMP), a HO1-inhibitor, and non-covalently modified with an engineered antibody for leukemic cell-targeted delivery. HO1-inhibiting T-hNP (T-hNP/SnMP) enhances chemo-sensitivity in human leukemia cells. In a human AML-bearing orthotopic mouse model, intravenously injected T-hNP not only actively targets to human leukemia cells but passively targets to CD11b+ myeloid cells in a bone marrow niche. The T-hNP/SnMP enhances the chemo-therapeutic effect of daunorubicin and boosts immune response by reprogramming bone marrow myeloid cells resulting from the recruitment of the monocyte-lineage and induction of inflammatory genes. The ex vivo study demonstrates an enhanced immune response of HO1-inhibited bone marrow CD11b+ myeloid cells against apoptotic leukemia cells. Collectively, HO1-inhibiting dual cell-targeted T-hNP/SnMP has a strong potential as a novel therapeutic in AML.
Collapse
Affiliation(s)
- Seok‐Beom Yong
- Department of BioengineeringInstitute for Bioengineering and Biopharmaceutical ResearchBK 21 Plus Future Biopharmaceutical Human Resources Training and Research TeamHanyang UniversitySeoul133‐791Republic of Korea
| | - Jaehyun Kim
- Department of BioengineeringInstitute for Bioengineering and Biopharmaceutical ResearchBK 21 Plus Future Biopharmaceutical Human Resources Training and Research TeamHanyang UniversitySeoul133‐791Republic of Korea
| | - Jee Young Chung
- Department of BioengineeringInstitute for Bioengineering and Biopharmaceutical ResearchBK 21 Plus Future Biopharmaceutical Human Resources Training and Research TeamHanyang UniversitySeoul133‐791Republic of Korea
| | - Sehee Ra
- Department of BioengineeringInstitute for Bioengineering and Biopharmaceutical ResearchBK 21 Plus Future Biopharmaceutical Human Resources Training and Research TeamHanyang UniversitySeoul133‐791Republic of Korea
| | - Seong Su kim
- Department of BioengineeringInstitute for Bioengineering and Biopharmaceutical ResearchBK 21 Plus Future Biopharmaceutical Human Resources Training and Research TeamHanyang UniversitySeoul133‐791Republic of Korea
| | - Yong‐Hee Kim
- Department of BioengineeringInstitute for Bioengineering and Biopharmaceutical ResearchBK 21 Plus Future Biopharmaceutical Human Resources Training and Research TeamHanyang UniversitySeoul133‐791Republic of Korea
| |
Collapse
|
48
|
Collignon A, Hospital MA, Montersino C, Courtier F, Charbonnier A, Saillard C, D'Incan E, Mohty B, Guille A, Adelaïde J, Carbuccia N, Garnier S, Mozziconacci MJ, Zemmour C, Pakradouni J, Restouin A, Castellano R, Chaffanet M, Birnbaum D, Collette Y, Vey N. A chemogenomic approach to identify personalized therapy for patients with relapse or refractory acute myeloid leukemia: results of a prospective feasibility study. Blood Cancer J 2020; 10:64. [PMID: 32488055 PMCID: PMC7266815 DOI: 10.1038/s41408-020-0330-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 04/06/2020] [Accepted: 04/23/2020] [Indexed: 02/05/2023] Open
Abstract
Targeted next-generation sequencing (tNGS) and ex vivo drug sensitivity/resistance profiling (DSRP) have laid foundations defining the functional genomic landscape of acute myeloid leukemia (AML) and premises of personalized medicine to guide treatment options for patients with aggressive and/or chemorefractory hematological malignancies. Here, we have assessed the feasibility of a tailored treatment strategy (TTS) guided by systematic parallel ex vivo DSRP and tNGS for patients with relapsed/refractory AML (number NCT02619071). A TTS issued by an institutional personalized committee could be achieved for 47/55 included patients (85%), 5 based on tNGS only, 6 on DSRP only, while 36 could be proposed on the basis of both, yielding more options and a better rationale. The TSS was available in <21 days for 28 patients (58.3%). On average, 3 to 4 potentially active drugs were selected per patient with only five patient samples being resistant to the entire drug panel. Seventeen patients received a TTS-guided treatment, resulting in four complete remissions, one partial remission, and five decreased peripheral blast counts. Our results show that chemogenomic combining tNGS with DSRP to determine a TTS is a promising approach to propose patient-specific treatment options within 21 days.
Collapse
Affiliation(s)
- A Collignon
- Haematology Department, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille, France
| | - M A Hospital
- Haematology Department, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille, France
| | - C Montersino
- Inserm, CNRS, Institut Paoli-Calmettes, CRCM, TrGET Preclinical Platform, Aix-Marseille Université, Marseille, France
| | - F Courtier
- Inserm, CNRS, Institut Paoli-Calmettes, CRCM, Predictive Oncology, Aix-Marseille Université, Marseille, France
| | - A Charbonnier
- Haematology Department, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille, France
| | - C Saillard
- Haematology Department, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille, France
| | - E D'Incan
- Haematology Department, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille, France
| | - B Mohty
- Haematology Department, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille, France
| | - A Guille
- Inserm, CNRS, Institut Paoli-Calmettes, CRCM, Predictive Oncology, Aix-Marseille Université, Marseille, France
| | - J Adelaïde
- Inserm, CNRS, Institut Paoli-Calmettes, CRCM, Predictive Oncology, Aix-Marseille Université, Marseille, France
| | - N Carbuccia
- Inserm, CNRS, Institut Paoli-Calmettes, CRCM, Predictive Oncology, Aix-Marseille Université, Marseille, France
| | - S Garnier
- Inserm, CNRS, Institut Paoli-Calmettes, CRCM, Predictive Oncology, Aix-Marseille Université, Marseille, France
| | - M J Mozziconacci
- Department of Biopathology, Institut Paoli-Calmettes, Marseille, France
| | - C Zemmour
- Department of Clinical Research & Innovation, Institut Paoli-Calmettes, Biostatistics & Methodology Unit, Aix Marseille Université, INSERM, IRD, SESSTIM, Marseille, France
| | - J Pakradouni
- Department of Clinical Research & Innovation, Sponsor Unit, Institut Paoli-Calmettes, Marseille, France
| | - A Restouin
- Inserm, CNRS, Institut Paoli-Calmettes, CRCM, TrGET Preclinical Platform, Aix-Marseille Université, Marseille, France
| | - R Castellano
- Inserm, CNRS, Institut Paoli-Calmettes, CRCM, TrGET Preclinical Platform, Aix-Marseille Université, Marseille, France
| | - M Chaffanet
- Inserm, CNRS, Institut Paoli-Calmettes, CRCM, Predictive Oncology, Aix-Marseille Université, Marseille, France
| | - D Birnbaum
- Inserm, CNRS, Institut Paoli-Calmettes, CRCM, Predictive Oncology, Aix-Marseille Université, Marseille, France.
| | - Y Collette
- Inserm, CNRS, Institut Paoli-Calmettes, CRCM, TrGET Preclinical Platform, Aix-Marseille Université, Marseille, France.
| | - N Vey
- Haematology Department, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille, France.
| |
Collapse
|
49
|
Dzobo K. Epigenomics-Guided Drug Development: Recent Advances in Solving the Cancer Treatment "jigsaw puzzle". OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2020; 23:70-85. [PMID: 30767728 DOI: 10.1089/omi.2018.0206] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The human epigenome plays a key role in determining cellular identity and eventually function. Drug discovery undertakings have focused mainly on the role of genomics in carcinogenesis, with the focus turning to the epigenome recently. Drugs targeting DNA and histone modifications are under development with some such as 5-azacytidine, decitabine, vorinostat, and panobinostat already approved by the Food and Drug Administration (FDA) and the European Medicines Agency (EMA). This expert review offers a critical analysis of the epigenomics-guided drug discovery and development and the opportunities and challenges for the next decade. Importantly, the coupling of epigenetic editing techniques, such as clustered regularly interspersed short palindromic repeat (CRISPR)-CRISPR-associated protein-9 (Cas9) and APOBEC-coupled epigenetic sequencing (ACE-seq) with epigenetic drug screens, will allow the identification of small-molecule inhibitors or drugs able to reverse epigenetic changes responsible for many diseases. In addition, concrete and sustainable innovation in cancer treatment ought to integrate epigenome targeting drugs with classic therapies such as chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Kevin Dzobo
- 1 International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa.,2 Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
50
|
Panina SB, Pei J, Baran N, Konopleva M, Kirienko NV. Utilizing Synergistic Potential of Mitochondria-Targeting Drugs for Leukemia Therapy. Front Oncol 2020; 10:435. [PMID: 32318340 PMCID: PMC7146088 DOI: 10.3389/fonc.2020.00435] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/11/2020] [Indexed: 12/12/2022] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive group of cancers with high mortality rates and significant relapse risks. Current treatments are insufficient, and new therapies are needed. Recent discoveries suggest that AML may be particularly sensitive to chemotherapeutics that target mitochondria. To further investigate this sensitivity, six compounds that target mitochondria [IACS-010759, rotenone, cytarabine, etoposide, ABT-199 (venetoclax), and carbonyl cyanide m-chlorophenylhydrazone] were each paired with six compounds with other activities, including tyrosine kinase inhibitors (midostaurin and dasatinib), glycolytic inhibitors (2-deoxy-D-glucose, 3-bromopyruvate, and lonidamine), and the microtubule destabilizer vinorelbine. The 36 resulting drug combinations were tested for synergistic cytotoxicity against MOLM-13 and OCI-AML2 AML cell lines. Four combinations (IACS-010759 with vinorelbine, rotenone with 2-deoxy-D-glucose, carbonyl cyanide m-chlorophenylhydrazone with dasatinib, and venetoclax with lonidamine) showed synergistic cytotoxicity in both AML cell lines and were selective for tumor cells, as survival of healthy PBMCs was dramatically higher. Among these drug pairs, IACS-010759/vinorelbine decreased ATP level and impaired mitochondrial respiration and coupling efficiency most profoundly. Some of these four treatments were also effective in K-562, KU812 (chronic myelogenous leukemia) and CCRF-CEM, MOLT-4 (acute lymphoblastic leukemia) cells, suggesting that these treatments may have value in treating other forms of leukemia. Finally, two of the four combinations retained high synergy and strong selectivity in primary AML cells from patient samples, supporting the potential of these treatments for patients.
Collapse
Affiliation(s)
- Svetlana B Panina
- Department of BioSciences, Rice University, Houston, TX, United States
| | - Jingqi Pei
- Department of BioSciences, Rice University, Houston, TX, United States
| | - Natalia Baran
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | |
Collapse
|